Publications
Key Publications
B cells orchestrate tolerance to the neuromyelitis optica autoantigen AQP4
A. M. Afzali, L. Nirschl, C. Sie, M. Pfaller, O. Ulianov, T. Hassler, C. Federle, E. Petrozziello, S. R. Kalluri, H. H. Chen, S. Tyystjarvi, A. Muschaweckh, K. Lammens, C. Delbridge, A. Buttner, K. Steiger, G. Seyhan, O. P. Ottersen, R. Ollinger, R. Rad, S. Jarosch, A. Straub, A. Muhlbauer, S. Grassmann, B. Hemmer, J. P. Bottcher, I. Wagner, M. Kreutzfeldt, D. Merkler, I. B. Pardas, M. Schmidt Supprian, V. R. Buchholz, S. Heink, D. H. Busch, L. Klein, and T. Korn
Nature, 2024
B cells orchestrate tolerance to the neuromyelitis optica autoantigen AQP4
A. M. Afzali, L. Nirschl, C. Sie, M. Pfaller, O. Ulianov, T. Hassler, C. Federle, E. Petrozziello, S. R. Kalluri, H. H. Chen, S. Tyystjarvi, A. Muschaweckh, K. Lammens, C. Delbridge, A. Buttner, K. Steiger, G. Seyhan, O. P. Ottersen, R. Ollinger, R. Rad, S. Jarosch, A. Straub, A. Muhlbauer, S. Grassmann, B. Hemmer, J. P. Bottcher, I. Wagner, M. Kreutzfeldt, D. Merkler, I. B. Pardas, M. Schmidt Supprian, V. R. Buchholz, S. Heink, D. H. Busch, L. Klein, and T. Korn
Neuromyelitis optica is a paradigmatic autoimmune disease of the central nervous system, in which the water-channel protein AQP4 is the target antigen(1). The immunopathology in neuromyelitis optica is largely driven by autoantibodies to AQP4(2). However, the T cell response that is required for the generation of these anti-AQP4 antibodies is not well understood. Here we show that B cells endogenously express AQP4 in response to activation with anti-CD40 and IL-21 and are able to present their endogenous AQP4 to T cells with an AQP4-specific T cell receptor (TCR). A population of thymic B cells emulates a CD40-stimulated B cell transcriptome, including AQP4 (in mice and humans), and efficiently purges the thymic TCR repertoire of AQP4-reactive clones. Genetic ablation of Aqp4 in B cells rescues AQP4-specific TCRs despite sufficient expression of AQP4 in medullary thymic epithelial cells, and B-cell-conditional AQP4-deficient mice are fully competent to raise AQP4-specific antibodies in productive germinal-centre responses. Thus, the negative selection of AQP4-specific thymocytes is dependent on the expression and presentation of AQP4 by thymic B cells. As AQP4 is expressed in B cells in a CD40-dependent (but not AIRE-dependent) manner, we propose that thymic B cells might tolerize against a group of germinal-centre-associated antigens, including disease-relevant autoantigens such as AQP4.
The glioblastoma multiforme tumor site promotes the commitment of tumor-infiltrating lymphocytes to the TH17 lineage in humans
M. Mitsdoerffer, L. Aly, M. Barz, T. Engleitner, C. Sie, C. Delbridge, G. Lepennetier, R. Ollinger, M. Pfaller, B. Wiestler, R. Rad, B. Meyer, B. Knier, F. Schmidt-Graf, J. Gempt, and T. Korn
Proc Natl Acad Sci U S A, 119(34): e2206208119, 2022
The glioblastoma multiforme tumor site promotes the commitment of tumor-infiltrating lymphocytes to the TH17 lineage in humans
M. Mitsdoerffer, L. Aly, M. Barz, T. Engleitner, C. Sie, C. Delbridge, G. Lepennetier, R. Ollinger, M. Pfaller, B. Wiestler, R. Rad, B. Meyer, B. Knier, F. Schmidt-Graf, J. Gempt, and T. Korn
Although glioblastoma multiforme (GBM) is not an invariably cold tumor, checkpoint inhibition has largely failed in GBM. In order to investigate T cell-intrinsic properties that contribute to the resistance of GBM to endogenous or therapeutically enhanced adaptive immune responses, we sorted CD4(+) and CD8(+) T cells from the peripheral blood, normal-appearing brain tissue, and tumor bed of nine treatment-naive patients with GBM. Bulk RNA sequencing of highly pure T cell populations from these different compartments was used to obtain deep transcriptomes of tumor-infiltrating T cells (TILs). While the transcriptome of CD8(+) TILs suggested that they were partly locked in a dysfunctional state, CD4(+) TILs showed a robust commitment to the type 17 T helper cell (TH17) lineage, which was corroborated by flow cytometry in four additional GBM cases. Therefore, our study illustrates that the brain tumor environment in GBM might instruct TH17 commitment of infiltrating T helper cells. Whether these properties of CD4(+) TILs facilitate a tumor-promoting milieu and thus could be a target for adjuvant anti-TH17 cell interventions needs to be further investigated.
IL-24 intrinsically regulates Th17 cell pathogenicity in mice
C. Sie, R. Kant, C. Peter, A. Muschaweckh, M. Pfaller, L. Nirschl, H. D. Moreno, T. Chadimová, G. Lepennetier, T. Kuhlmann, R. Öllinger, T. Engleitner, R. Rad, and T. Korn
J Exp Med, 219(8), 2022
IL-24 intrinsically regulates Th17 cell pathogenicity in mice
C. Sie, R. Kant, C. Peter, A. Muschaweckh, M. Pfaller, L. Nirschl, H. D. Moreno, T. Chadimová, G. Lepennetier, T. Kuhlmann, R. Öllinger, T. Engleitner, R. Rad, and T. Korn
In certain instances, Th17 responses are associated with severe immunopathology. T cell–intrinsic mechanisms that restrict pathogenic effector functions have been described for type 1 and 2 responses but are less well studied for Th17 cells. Here, we report a cell-intrinsic feedback mechanism that controls the pathogenicity of Th17 cells. Th17 cells produce IL-24, which prompts them to secrete IL-10. The IL-10–inducing function of IL-24 is independent of the cell surface receptor of IL-24 on Th17 cells. Rather, IL-24 is recruited to the inner mitochondrial membrane, where it interacts with the NADH dehydrogenase (ubiquinone) 1 α subcomplex subunit 13 (also known as Grim19), a constituent of complex I of the respiratory chain. Together, Grim19 and IL-24 promote the accumulation of STAT3 in the mitochondrial compartment. We propose that IL-24–guided mitochondrial STAT3 constitutes a rheostat to blunt extensive STAT3 deflections in the nucleus, which might then contribute to a robust IL-10 response in Th17 cells and a restriction of immunopathology in experimental autoimmune encephalomyelitis.
Skin and gut imprinted helper T cell subsets exhibit distinct functional phenotypes in central nervous system autoimmunity
M. Hiltensperger, E. Beltran, R. Kant, S. Tyystjarvi, G. Lepennetier, H. Dominguez Moreno, I. J. Bauer, S. Grassmann, S. Jarosch, K. Schober, V. R. Buchholz, S. Kenet, C. Gasperi, R. Ollinger, R. Rad, A. Muschaweckh, C. Sie, L. Aly, B. Knier, G. Garg, A. M. Afzali, L. A. Gerdes, T. Kumpfel, S. Franzenburg, N. Kawakami, B. Hemmer, D. H. Busch, T. Misgeld, K. Dornmair, and T. Korn
Nat Immunol, 22(7): 880-892, 2021
Skin and gut imprinted helper T cell subsets exhibit distinct functional phenotypes in central nervous system autoimmunity
M. Hiltensperger, E. Beltran, R. Kant, S. Tyystjarvi, G. Lepennetier, H. Dominguez Moreno, I. J. Bauer, S. Grassmann, S. Jarosch, K. Schober, V. R. Buchholz, S. Kenet, C. Gasperi, R. Ollinger, R. Rad, A. Muschaweckh, C. Sie, L. Aly, B. Knier, G. Garg, A. M. Afzali, L. A. Gerdes, T. Kumpfel, S. Franzenburg, N. Kawakami, B. Hemmer, D. H. Busch, T. Misgeld, K. Dornmair, and T. Korn
Multidimensional single-cell analyses of T cells have fueled the debate about whether there is extensive plasticity or 'mixed' priming of helper T cell subsets in vivo. Here, we developed an experimental framework to probe the idea that the site of priming in the systemic immune compartment is a determinant of helper T cell-induced immunopathology in remote organs. By site-specific in vivo labeling of antigen-specific T cells in inguinal (i) or gut draining mesenteric (m) lymph nodes, we show that i-T cells and m-T cells isolated from the inflamed central nervous system (CNS) in a model of multiple sclerosis (MS) are distinct. i-T cells were Cxcr6(+), and m-T cells expressed P2rx7. Notably, m-T cells infiltrated white matter, while i-T cells were also recruited to gray matter. Therefore, we propose that the definition of helper T cell subsets by their site of priming may guide an advanced understanding of helper T cell biology in health and disease.
Formation and immunomodulatory function of meningeal B cell aggregates in progressive CNS autoimmunity
M. Mitsdoerffer, G. Di Liberto, S. Dotsch, C. Sie, I. Wagner, M. Pfaller, M. Kreutzfeldt, S. Frassle, L. Aly, B. Knier, D. H. Busch, D. Merkler, and T. Korn
Brain, 144(6): 1697-1710, 2021
Formation and immunomodulatory function of meningeal B cell aggregates in progressive CNS autoimmunity
M. Mitsdoerffer, G. Di Liberto, S. Dotsch, C. Sie, I. Wagner, M. Pfaller, M. Kreutzfeldt, S. Frassle, L. Aly, B. Knier, D. H. Busch, D. Merkler, and T. Korn
Meningeal B lymphocyte aggregates have been described in autopsy material of patients with chronic multiple sclerosis. The presence of meningeal B cell aggregates has been correlated with worse disease. However, the functional role of these meningeal B cell aggregates is not understood. Here, we use a mouse model of multiple sclerosis, the spontaneous opticospinal encephalomyelitis model, which is built on the double transgenic expression of myelin oligodendrocyte glycoprotein-specific T-cell and B-cell receptors, to show that the formation of meningeal B cell aggregates is dependent on the expression of alpha4 integrins by antigen-specific T cells. T cell-conditional genetic ablation of alpha4 integrins in opticospinal encephalomyelitis mice impaired the formation of meningeal B cell aggregates, and surprisingly, led to a higher disease incidence as compared to opticospinal encephalomyelitis mice with alpha4 integrin-sufficient T cells. B cell-conditional ablation of alpha4 integrins in opticospinal encephalomyelitis mice resulted in the entire abrogation of the formation of meningeal B cell aggregates, and opticospinal encephalomyelitis mice with alpha4 integrin-deficient B cells suffered from a higher disease burden than regular opticospinal encephalomyelitis mice. While anti-CD20 antibody-mediated systemic depletion of B cells in opticospinal encephalomyelitis mice after onset of disease failed to efficiently decrease meningeal B cell aggregates without significantly modulating disease progression, treatment with anti-CD19 chimeric antigen receptor-T cells eliminated meningeal B cell aggregates and exacerbated clinical disease in opticospinal encephalomyelitis mice. Since about 20% of B cells in organized meningeal B cell aggregates produced either IL-10 or IL-35, we propose that meningeal B cell aggregates might also have an immunoregulatory function as to the immunopathology in adjacent spinal cord white matter. The immunoregulatory function of meningeal B cell aggregates needs to be considered when designing highly efficient therapies directed against meningeal B cell aggregates for clinical application in multiple sclerosis.
Blimp1 Prevents Methylation of Foxp3 and Loss of Regulatory T Cell Identity at Sites of Inflammation
G. Garg, A. Muschaweckh, H. Moreno, A. Vasanthakumar, S. Floess, G. Lepennetier, R. Oellinger, Y. Zhan, T. Regen, M. Hiltensperger, C. Peter, L. Aly, B. Knier, L. R. Palam, R. Kapur, M. H. Kaplan, A. Waisman, R. Rad, G. Schotta, J. Huehn, A. Kallies, and T. Korn
Cell Rep, 26(7): 1854-1868 e5, 2019
Blimp1 Prevents Methylation of Foxp3 and Loss of Regulatory T Cell Identity at Sites of Inflammation
G. Garg, A. Muschaweckh, H. Moreno, A. Vasanthakumar, S. Floess, G. Lepennetier, R. Oellinger, Y. Zhan, T. Regen, M. Hiltensperger, C. Peter, L. Aly, B. Knier, L. R. Palam, R. Kapur, M. H. Kaplan, A. Waisman, R. Rad, G. Schotta, J. Huehn, A. Kallies, and T. Korn
Foxp3(+) regulatory T (Treg) cells restrict immune pathology in inflamed tissues; however, an inflammatory environment presents a threat to Treg cell identity and function. Here, we establish a transcriptional signature of central nervous system (CNS) Treg cells that accumulate during experimental autoimmune encephalitis (EAE) and identify a pathway that maintains Treg cell function and identity during severe inflammation. This pathway is dependent on the transcriptional regulator Blimp1, which prevents downregulation of Foxp3 expression and "toxic" gain-of-function of Treg cells in the inflamed CNS. Blimp1 negatively regulates IL-6- and STAT3-dependent Dnmt3a expression and function restraining methylation of Treg cell-specific conserved non-coding sequence 2 (CNS2) in the Foxp3 locus. Consequently, CNS2 is heavily methylated when Blimp1 is ablated, leading to a loss of Foxp3 expression and severe disease. These findings identify a Blimp1-dependent pathway that preserves Treg cell stability in inflamed non-lymphoid tissues.
Myeloid-derived suppressor cells control B cell accumulation in the central nervous system during autoimmunity
B. Knier, M. Hiltensperger, C. Sie, L. Aly, G. Lepennetier, T. Engleitner, G. Garg, A. Muschaweckh, M. Mitsdorffer, U. Koedel, B. Hochst, P. Knolle, M. Gunzer, B. Hemmer, R. Rad, D. Merkler, and T. Korn
Nat Immunol, 19(12): 1341-1351, 2018
Myeloid-derived suppressor cells control B cell accumulation in the central nervous system during autoimmunity
B. Knier, M. Hiltensperger, C. Sie, L. Aly, G. Lepennetier, T. Engleitner, G. Garg, A. Muschaweckh, M. Mitsdorffer, U. Koedel, B. Hochst, P. Knolle, M. Gunzer, B. Hemmer, R. Rad, D. Merkler, and T. Korn
Polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) have been characterized in the context of malignancies. Here we show that PMN-MDSCs can restrain B cell accumulation during central nervous system (CNS) autoimmunity. Ly6G(+) cells were recruited to the CNS during experimental autoimmune encephalomyelitis (EAE), interacted with B cells that produced the cytokines GM-CSF and interleukin-6 (IL-6), and acquired properties of PMN-MDSCs in the CNS in a manner dependent on the signal transducer STAT3. Depletion of Ly6G(+) cells or dysfunction of Ly6G(+) cells through conditional ablation of STAT3 led to the selective accumulation of GM-CSF-producing B cells in the CNS compartment, which in turn promoted an activated microglial phenotype and lack of recovery from EAE. The frequency of CD138(+) B cells in the cerebrospinal fluid (CSF) of human subjects with multiple sclerosis was negatively correlated with the frequency of PMN-MDSCs in the CSF. Thus PMN-MDSCs might selectively control the accumulation and cytokine secretion of B cells in the inflamed CNS.
Trans-presentation of IL-6 by dendritic cells is required for the priming of pathogenic TH17 cells
S. Heink, N. Yogev, C. Garbers, M. Herwerth, L. Aly, C. Gasperi, V. Husterer, A. L. Croxford, K. Moller-Hackbarth, H. S. Bartsch, K. Sotlar, S. Krebs, T. Regen, H. Blum, B. Hemmer, T. Misgeld, T. F. Wunderlich, J. Hidalgo, M. Oukka, S. Rose-John, M. Schmidt-Supprian, A. Waisman, and T. Korn
Nat Immunol, 18(1): 74-85, 2017
Trans-presentation of IL-6 by dendritic cells is required for the priming of pathogenic TH17 cells
S. Heink, N. Yogev, C. Garbers, M. Herwerth, L. Aly, C. Gasperi, V. Husterer, A. L. Croxford, K. Moller-Hackbarth, H. S. Bartsch, K. Sotlar, S. Krebs, T. Regen, H. Blum, B. Hemmer, T. Misgeld, T. F. Wunderlich, J. Hidalgo, M. Oukka, S. Rose-John, M. Schmidt-Supprian, A. Waisman, and T. Korn
The cellular sources of interleukin 6 (IL-6) that are relevant for differentiation of the TH17 subset of helper T cells remain unclear. Here we used a novel strategy for the conditional deletion of distinct IL-6-producing cell types to show that dendritic cells (DCs) positive for the signaling regulator Sirpα were essential for the generation of pathogenic TH17 cells. Using their IL-6 receptor α-chain (IL-6Rα), Sirpα+ DCs trans-presented IL-6 to T cells during the process of cognate interaction. While ambient IL-6 was sufficient to suppress the induction of expression of the transcription factor Foxp3 in T cells, trans-presentation of IL-6 by DC-bound IL-6Rα (called 'IL-6 cluster signaling' here) was needed to prevent premature induction of interferon-γ (IFN-γ) expression in T cells and to generate pathogenic TH17 cells in vivo. Our findings should guide therapeutic approaches for the treatment of TH17-cell-mediated autoimmune diseases.
Original Articles
2024
Rapamycin rescues loss-of-function in blood-brain barrier-interacting regulatory T cells
P. Baeten, I. Hamad, C. Hoeks, M. Hiltensperger, B. Van Wijmeersch, V. Popescu, L. Aly, V. Somers, T. Korn, M. Kleinewietfeld, N. Hellings, and B. Broux
JCI Insight, 2024
Rapamycin rescues loss-of-function in blood-brain barrier-interacting regulatory T cells
P. Baeten, I. Hamad, C. Hoeks, M. Hiltensperger, B. Van Wijmeersch, V. Popescu, L. Aly, V. Somers, T. Korn, M. Kleinewietfeld, N. Hellings, and B. Broux
In autoimmunity, FOXP3+ regulatory T cells (Tregs) skew towards a pro-inflammatory, non-suppressive phenotype and are therefore unable to control the exaggerated autoimmune response. This largely impacts the success of autologous Treg therapy which is currently under investigation for autoimmune diseases, including multiple sclerosis (MS). There is a need to ensure in vivo Treg stability before successful application of Treg therapy. Using genetic fate-mapping mice, we demonstrate that inflammatory, cytokine-expressing exFOXP3 T cells accumulate in the central nervous system during experimental autoimmune encephalomyelitis. In a human in vitro model, we discovered that interaction with inflamed blood-brain barrier endothelial cells (BBB-ECs) induces loss-of-function by Tregs. Transcriptome and cytokine analysis revealed that in vitro migrated Tregs have disrupted regenerative potential, a pro-inflammatory Th1/17 signature and upregulate the mTORC1 signaling pathway. In vitro treatment of migrated human Tregs with the clinically-approved mTORC1 inhibitor rapamycin restored suppression. Finally, flow cytometric analysis indicated an enrichment of inflammatory, less suppressive CD49d+ Tregs in the cerebrospinal fluid of people with MS. In sum, interaction with BBB-ECs is sufficient to affect Treg function, and transmigration triggers an additive pro-inflammatory phenotype switch. These insights help improve the efficacy of autologous Treg therapy of MS.
JCI Insight, 2024
TGF-β specifies T(FH) versus T(H)17 cell fates in murine CD4(+) T cells through c-Maf
Y. Chang, L. Bach, M. Hasiuk, L. Wen, T. Elmzzahi, C. Tsui, N. Gutiérrez-Melo, T. Steffen, D. T. Utzschneider, T. Raj, P. J. Jost, S. Heink, J. Cheng, O. T. Burton, J. Zeiträg, D. Alterauge, F. Dahlström, J. C. Becker, M. Kastl, K. Symeonidis, M. van Uelft, M. Becker, S. Reschke, S. Krebs, H. Blum, Z. Abdullah, K. Paeschke, C. Ohnmacht, C. Neumann, A. Liston, F. Meissner, T. Korn, J. Hasenauer, V. Heissmeyer, M. Beyer, A. Kallies, L. T. Jeker, and D. Baumjohann
Sci Immunol, 9(93): eadd4818, 2024
TGF-β specifies T(FH) versus T(H)17 cell fates in murine CD4(+) T cells through c-Maf
Y. Chang, L. Bach, M. Hasiuk, L. Wen, T. Elmzzahi, C. Tsui, N. Gutiérrez-Melo, T. Steffen, D. T. Utzschneider, T. Raj, P. J. Jost, S. Heink, J. Cheng, O. T. Burton, J. Zeiträg, D. Alterauge, F. Dahlström, J. C. Becker, M. Kastl, K. Symeonidis, M. van Uelft, M. Becker, S. Reschke, S. Krebs, H. Blum, Z. Abdullah, K. Paeschke, C. Ohnmacht, C. Neumann, A. Liston, F. Meissner, T. Korn, J. Hasenauer, V. Heissmeyer, M. Beyer, A. Kallies, L. T. Jeker, and D. Baumjohann
T follicular helper (T(FH)) cells are essential for effective antibody responses, but deciphering the intrinsic wiring of mouse T(FH) cells has long been hampered by the lack of a reliable protocol for their generation in vitro. We report that transforming growth factor-β (TGF-β) induces robust expression of T(FH) hallmark molecules CXCR5 and Bcl6 in activated mouse CD4(+) T cells in vitro. TGF-β-induced mouse CXCR5(+) T(FH) cells are phenotypically, transcriptionally, and functionally similar to in vivo-generated T(FH) cells and provide critical help to B cells. The study further reveals that TGF-β-induced CXCR5 expression is independent of Bcl6 but requires the transcription factor c-Maf. Classical TGF-β-containing T helper 17 (T(H)17)-inducing conditions also yield separate CXCR5(+) and IL-17A-producing cells, highlighting shared and distinct cell fate trajectories of T(FH) and T(H)17 cells. We demonstrate that excess IL-2 in high-density T cell cultures interferes with the TGF-β-induced T(FH) cell program, that T(FH) and T(H)17 cells share a common developmental stage, and that c-Maf acts as a switch factor for T(FH) versus T(H)17 cell fates in TGF-β-rich environments in vitro and in vivo.
Sci Immunol, 9(93): eadd4818, 2024
B cells orchestrate tolerance to the neuromyelitis optica autoantigen AQP4
A. M. Afzali, L. Nirschl, C. Sie, M. Pfaller, O. Ulianov, T. Hassler, C. Federle, E. Petrozziello, S. R. Kalluri, H. H. Chen, S. Tyystjarvi, A. Muschaweckh, K. Lammens, C. Delbridge, A. Buttner, K. Steiger, G. Seyhan, O. P. Ottersen, R. Ollinger, R. Rad, S. Jarosch, A. Straub, A. Muhlbauer, S. Grassmann, B. Hemmer, J. P. Bottcher, I. Wagner, M. Kreutzfeldt, D. Merkler, I. B. Pardas, M. Schmidt Supprian, V. R. Buchholz, S. Heink, D. H. Busch, L. Klein, and T. Korn
Nature, 2024
B cells orchestrate tolerance to the neuromyelitis optica autoantigen AQP4
A. M. Afzali, L. Nirschl, C. Sie, M. Pfaller, O. Ulianov, T. Hassler, C. Federle, E. Petrozziello, S. R. Kalluri, H. H. Chen, S. Tyystjarvi, A. Muschaweckh, K. Lammens, C. Delbridge, A. Buttner, K. Steiger, G. Seyhan, O. P. Ottersen, R. Ollinger, R. Rad, S. Jarosch, A. Straub, A. Muhlbauer, S. Grassmann, B. Hemmer, J. P. Bottcher, I. Wagner, M. Kreutzfeldt, D. Merkler, I. B. Pardas, M. Schmidt Supprian, V. R. Buchholz, S. Heink, D. H. Busch, L. Klein, and T. Korn
Neuromyelitis optica is a paradigmatic autoimmune disease of the central nervous system, in which the water-channel protein AQP4 is the target antigen(1). The immunopathology in neuromyelitis optica is largely driven by autoantibodies to AQP4(2). However, the T cell response that is required for the generation of these anti-AQP4 antibodies is not well understood. Here we show that B cells endogenously express AQP4 in response to activation with anti-CD40 and IL-21 and are able to present their endogenous AQP4 to T cells with an AQP4-specific T cell receptor (TCR). A population of thymic B cells emulates a CD40-stimulated B cell transcriptome, including AQP4 (in mice and humans), and efficiently purges the thymic TCR repertoire of AQP4-reactive clones. Genetic ablation of Aqp4 in B cells rescues AQP4-specific TCRs despite sufficient expression of AQP4 in medullary thymic epithelial cells, and B-cell-conditional AQP4-deficient mice are fully competent to raise AQP4-specific antibodies in productive germinal-centre responses. Thus, the negative selection of AQP4-specific thymocytes is dependent on the expression and presentation of AQP4 by thymic B cells. As AQP4 is expressed in B cells in a CD40-dependent (but not AIRE-dependent) manner, we propose that thymic B cells might tolerize against a group of germinal-centre-associated antigens, including disease-relevant autoantigens such as AQP4.
IL-23 stabilizes an effector T(reg) cell program in the tumor microenvironment
T. Wertheimer, P. Zwicky, L. Rindlisbacher, C. Sparano, M. Vermeer, B. M. S. de Melo, C. Haftmann, T. Ruckert, A. Sethi, S. Scharli, A. Huber, F. Ingelfinger, C. Xu, D. Kim, P. Hane, A. Fonseca da Silva, A. Muschaweckh, N. Nunez, S. Krishnarajah, N. Kohler, R. Zeiser, M. Oukka, T. Korn, S. Tugues, and B. Becher
Nat Immunol, 2024
IL-23 stabilizes an effector T(reg) cell program in the tumor microenvironment
T. Wertheimer, P. Zwicky, L. Rindlisbacher, C. Sparano, M. Vermeer, B. M. S. de Melo, C. Haftmann, T. Ruckert, A. Sethi, S. Scharli, A. Huber, F. Ingelfinger, C. Xu, D. Kim, P. Hane, A. Fonseca da Silva, A. Muschaweckh, N. Nunez, S. Krishnarajah, N. Kohler, R. Zeiser, M. Oukka, T. Korn, S. Tugues, and B. Becher
Interleukin-23 (IL-23) is a proinflammatory cytokine mainly produced by myeloid cells that promotes tumor growth in various preclinical cancer models and correlates with adverse outcomes. However, as to how IL-23 fuels tumor growth is unclear. Here, we found tumor-associated macrophages to be the main source of IL-23 in mouse and human tumor microenvironments. Among IL-23-sensing cells, we identified a subset of tumor-infiltrating regulatory T (T(reg)) cells that display a highly suppressive phenotype across mouse and human tumors. The use of three preclinical models of solid cancer in combination with genetic ablation of Il23r in T(reg) cells revealed that they are responsible for the tumor-promoting effect of IL-23. Mechanistically, we found that IL-23 sensing represents a crucial signal driving the maintenance and stabilization of effector T(reg) cells involving the transcription factor Foxp3. Our data support that targeting the IL-23/IL-23R axis in cancer may represent a means of eliciting antitumor immunity.
2023
Association of retinal vessel pathology and brain atrophy in relapsing-remitting multiple sclerosis
E. F. Romahn, T. Wiltgen, M. Bussas, L. Aly, R. Wicklein, C. Noll, A. Berthele, V. Dehmelt, C. Mardin, C. Zimmer, T. Korn, B. Hemmer, J. S. Kirschke, M. Muhlau, and B. Knier
Front Immunol, 14: 1284986, 2023
Association of retinal vessel pathology and brain atrophy in relapsing-remitting multiple sclerosis
E. F. Romahn, T. Wiltgen, M. Bussas, L. Aly, R. Wicklein, C. Noll, A. Berthele, V. Dehmelt, C. Mardin, C. Zimmer, T. Korn, B. Hemmer, J. S. Kirschke, M. Muhlau, and B. Knier
BACKGROUND: Optical coherence tomography angiography (OCTA) allows non-invasive assessment of retinal vessel structures. Thinning and loss of retinal vessels is evident in eyes of patients with multiple sclerosis (MS) and might be associated with a proinflammatory disease phenotype and worse prognosis. We investigated whether changes of the retinal vasculature are linked to brain atrophy and disability in MS. MATERIAL AND METHODS: This study includes one longitudinal observational cohort (n=79) of patients with relapsing-remitting MS. Patients underwent annual assessment of the expanded disability status scale (EDSS), timed 25-foot walk, symbol digit modalities test (SDMT), retinal optical coherence tomography (OCT), OCTA, and brain MRI during a follow-up duration of at least 20 months. We investigated intra-individual associations between changes in the retinal architecture, vasculature, brain atrophy and disability. Eyes with a history of optic neuritis (ON) were excluded. RESULTS: We included 79 patients with a median disease duration of 12 (interquartile range 2 - 49) months and a median EDSS of 1.0 (0 - 2.0). Longitudinal retinal axonal and ganglion cell loss were linked to grey matter atrophy, cortical atrophy, and volume loss of the putamen. We observed an association between vessel loss of the superficial vascular complex (SVC) and both grey and white matter atrophy. Both observations were independent of retinal ganglion cell loss. Moreover, patients with worsening of the EDSS and SDMT revealed a pronounced longitudinal rarefication of the SVC and the deep vascular complex. DISCUSSION: ON-independent narrowing of the retinal vasculature might be linked to brain atrophy and disability in MS. Our findings suggest that retinal OCTA might be a new tool for monitoring neurodegeneration during MS.
Unrestrained cleavage of Roquin-1 by MALT1 induces spontaneous T cell activation and the development of autoimmunity
H. Schmidt, T. Raj, T. J. O'Neill, A. Muschaweckh, F. Giesert, A. Negraschus, K. P. Hoefig, G. Behrens, L. Esser, C. Baumann, R. Feederle, C. Plaza-Sirvent, A. Geerlof, A. Gewies, S. E. Isay, J. Ruland, I. Schmitz, W. Wurst, T. Korn, D. Krappmann, and V. Heissmeyer
Proc Natl Acad Sci U S A, 120(48): e2309205120, 2023
Unrestrained cleavage of Roquin-1 by MALT1 induces spontaneous T cell activation and the development of autoimmunity
H. Schmidt, T. Raj, T. J. O'Neill, A. Muschaweckh, F. Giesert, A. Negraschus, K. P. Hoefig, G. Behrens, L. Esser, C. Baumann, R. Feederle, C. Plaza-Sirvent, A. Geerlof, A. Gewies, S. E. Isay, J. Ruland, I. Schmitz, W. Wurst, T. Korn, D. Krappmann, and V. Heissmeyer
Constitutive activation of the MALT1 paracaspase in conventional T cells of Malt1(TBM/TBM) (TRAF6 Binding Mutant = TBM) mice causes fatal inflammation and autoimmunity, but the involved targets and underlying molecular mechanisms are unknown. We genetically rendered a single MALT1 substrate, the RNA-binding protein (RBP) Roquin-1, insensitive to MALT1 cleavage. These Rc3h1(Mins/Mins) mice showed normal immune homeostasis. Combining Rc3h1(Mins/Mins) alleles with those encoding for constitutively active MALT1 (TBM) prevented spontaneous T cell activation and restored viability of Malt1(TBM/TBM) mice. Mechanistically, we show how antigen/MHC recognition is translated by MALT1 into Roquin cleavage and derepression of Roquin targets. Increasing T cell receptor (TCR) signals inactivated Roquin more effectively, and only high TCR strength enabled derepression of high-affinity targets to promote Th17 differentiation. Induction of experimental autoimmune encephalomyelitis (EAE) revealed increased cleavage of Roquin-1 in disease-associated Th17 compared to Th1 cells in the CNS. T cells from Rc3h1(Mins/Mins) mice did not efficiently induce the high-affinity Roquin-1 target IkappaB(NS) in response to TCR stimulation, showed reduced Th17 differentiation, and Rc3h1(Mins/Mins) mice were protected from EAE. These data demonstrate how TCR signaling and MALT1 activation utilize graded cleavage of Roquin to differentially regulate target mRNAs that control T cell activation and differentiation as well as the development of autoimmunity.
The role of intrathecal free light chains kappa for the detection of autoimmune encephalitis in subacute onset neuropsychiatric syndromes
D. Bertram, T. Tsaktanis, A. Berthele, and T. Korn
Sci Rep, 13(1): 17224, 2023
The role of intrathecal free light chains kappa for the detection of autoimmune encephalitis in subacute onset neuropsychiatric syndromes
D. Bertram, T. Tsaktanis, A. Berthele, and T. Korn
{Intrathecal synthesis of free light chains kappa (FLCK) is increasingly recognized as a marker of inflammatory CNS pathologies. Here, we tested the performance of FLCK in differentiating autoimmune encephalitis (AIE) from non-inflammatory etiologies in subacute onset neuropsychiatric syndromes. Patients undergoing diagnostic work-up for suspected autoimmune encephalitis at our department between 2015 and 2020 were retrospectively assessed for definitive diagnosis, available CSF and blood samples, as well as complete clinical records. Intrathecal FLCK was measured along with established CSF markers of CNS inflammation. The study cohort consisted of 19 patients with antibody-mediated AIE (AIE(+)), 18 patients with suspected AIE but without detectable autoantibodies (AIE(-)), 10 patients with infectious (viral) encephalitis (INE), and 15 patients with degenerative encephalopathies (DGE). 25 age- and sex-matched patients with non-inflammatory neurological diseases (NIND) were used as a control group. All AIE(+) patients exhibited intrathecal synthesis of FLCK compared to only 39% of AIE(-) patients and 81% of patients in the INE group. No intrathecal synthesis of FLCK was found in DGE and NIND patients. While intrathecal FLCK was equally specific for an inflammatory etiology as oligoclonal bands (OCB) in the cerebrospinal fluid (CSF), the sensitivity of intrathecal FLCK for any inflammatory intrathecal process was higher than that of OCB (83% vs. 38%). Intrathecal FLCK synthesis was found to discriminate AIE(+) from non-inflammatory encephalopathies and AIE(-) when the CSF cell count was normal [receiver operating characteristic (ROC) analysis area under the curve (AUC): 0.867
Visualizing the activation of encephalitogenic T cells in the ileal lamina propria by in vivo two-photon imaging
I. J. Bauer, P. Fang, K. F. Lammle, S. Tyystjarvi, D. Alterauge, D. Baumjohann, H. Yoon, T. Korn, H. Wekerle, and N. Kawakami
Proc Natl Acad Sci U S A, 120(30): e2302697120, 2023
Visualizing the activation of encephalitogenic T cells in the ileal lamina propria by in vivo two-photon imaging
I. J. Bauer, P. Fang, K. F. Lammle, S. Tyystjarvi, D. Alterauge, D. Baumjohann, H. Yoon, T. Korn, H. Wekerle, and N. Kawakami
Autoreactive encephalitogenic T cells exist in the healthy immune repertoire but need a trigger to induce CNS inflammation. The underlying mechanisms remain elusive, whereby microbiota were shown to be involved in the manifestation of CNS autoimmunity. Here, we used intravital imaging to explore how microbiota affect the T cells as trigger of CNS inflammation. Encephalitogenic CD4(+) T cells transduced with the calcium-sensing protein Twitch-2B showed calcium signaling with higher frequency than polyclonal T cells in the small intestinal lamina propria (LP) but not in Peyer's patches. Interestingly, nonencephalitogenic T cells specific for OVA and LCMV also showed calcium signaling in the LP, indicating a general stimulating effect of microbiota. The observed calcium signaling was microbiota and MHC class II dependent as it was significantly reduced in germfree animals and after administration of anti-MHC class II antibody, respectively. As a consequence of T cell stimulation in the small intestine, the encephalitogenic T cells start expressing Th17-axis genes. Finally, we show the migration of CD4(+) T cells from the small intestine into the CNS. In summary, our direct in vivo visualization revealed that microbiota induced T cell activation in the LP, which directed T cells to adopt a Th17-like phenotype as a trigger of CNS inflammation.
Reactive oxygen species produced by myeloid cells in psoriasis as a potential biofactor contributing to the development of vascular inflammation
T. Schaller, J. Ringen, B. Fischer, T. Bieler, K. Perius, T. Knopp, K. S. Kommoss, T. Korn, M. Heikenwalder, M. Oelze, A. Daiber, T. Munzel, D. Kramer, P. Wenzel, J. Wild, S. Karbach, and A. Waisman
Biofactors, 49(4): 861-874, 2023
Reactive oxygen species produced by myeloid cells in psoriasis as a potential biofactor contributing to the development of vascular inflammation
T. Schaller, J. Ringen, B. Fischer, T. Bieler, K. Perius, T. Knopp, K. S. Kommoss, T. Korn, M. Heikenwalder, M. Oelze, A. Daiber, T. Munzel, D. Kramer, P. Wenzel, J. Wild, S. Karbach, and A. Waisman
Psoriasis is an immune-mediated inflammatory skin disease driven by interleukin-17A (IL-17A) and associated with cardiovascular dysfunction. We used a severe psoriasis mouse model of keratinocyte IL-17A overexpression (K14-IL-17A(ind/+) , IL-17A(ind/+) control mice) to investigate the activity of neutrophils and a potential cellular interconnection between skin and vasculature. Levels of dermal reactive oxygen species (ROS) and their release by neutrophils were measured by lucigenin-/luminol-based assays, respectively. Quantitative RT-PCR determined neutrophilic activity and inflammation-related markers in skin and aorta. To track skin-derived immune cells, we used PhAM-K14-IL-17A(ind/+) mice allowing us to mark all cells in the skin by photoconversion of a fluorescent protein to analyze their migration into spleen, aorta, and lymph nodes by flow cytometry. Compared to controls, K14-IL-17A(ind/+) mice exhibited elevated ROS levels in the skin and a higher neutrophilic oxidative burst accompanied by the upregulation of several activation markers. In line with these results psoriatic mice displayed elevated expression of genes involved in neutrophil migration (e.g., Cxcl2 and S100a9) in skin and aorta. However, no direct immune cell migration from the psoriatic skin into the aortic vessel wall was observed. Neutrophils of psoriatic mice showed an activated phenotype, but no direct cellular migration from the skin to the vasculature was observed. This suggests that highly active vasculature-invading neutrophils must originate directly from the bone marrow. Hence, the skin-vasculature crosstalk in psoriasis is most likely based on the systemic effects of the autoimmune skin disease, emphasizing the importance of a systemic therapeutic approach for psoriasis patients.
Pyrimidine de novo synthesis inhibition selectively blocks effector but not memory T cell development
S. Scherer, S. G. Oberle, K. Kanev, A. K. Gerullis, M. Wu, G. P. de Almeida, D. J. Puleston, F. Baixauli, L. Aly, A. Greco, T. Nizharadze, N. B. Becker, M. V. Hoesslin, L. V. Donhauser, J. Berner, T. Chu, H. A. McNamara, Z. Esencan, P. Roelli, C. Wurmser, I. Kleiter, Mjgt Vehreschild, C. A. Mayer, P. Knolle, M. Klingenspor, V. Fumagalli, M. Iannacone, M. Prlic, T. Korn, E. L. Pearce, T. Hofer, A. M. Schulz, and D. Zehn
Nat Immunol, 24(3): 501-515, 2023
Pyrimidine de novo synthesis inhibition selectively blocks effector but not memory T cell development
S. Scherer, S. G. Oberle, K. Kanev, A. K. Gerullis, M. Wu, G. P. de Almeida, D. J. Puleston, F. Baixauli, L. Aly, A. Greco, T. Nizharadze, N. B. Becker, M. V. Hoesslin, L. V. Donhauser, J. Berner, T. Chu, H. A. McNamara, Z. Esencan, P. Roelli, C. Wurmser, I. Kleiter, Mjgt Vehreschild, C. A. Mayer, P. Knolle, M. Klingenspor, V. Fumagalli, M. Iannacone, M. Prlic, T. Korn, E. L. Pearce, T. Hofer, A. M. Schulz, and D. Zehn
Blocking pyrimidine de novo synthesis by inhibiting dihydroorotate dehydrogenase is used to treat autoimmunity and prevent expansion of rapidly dividing cell populations including activated T cells. Here we show memory T cell precursors are resistant to pyrimidine starvation. Although the treatment effectively blocked effector T cells, the number, function and transcriptional profile of memory T cells and their precursors were unaffected. This effect occurred in a narrow time window in the early T cell expansion phase when developing effector, but not memory precursor, T cells are vulnerable to pyrimidine starvation. This vulnerability stems from a higher proliferative rate of early effector T cells as well as lower pyrimidine synthesis capacity when compared with memory precursors. This differential sensitivity is a drug-targetable checkpoint that efficiently diminishes effector T cells without affecting the memory compartment. This cell fate checkpoint might therefore lead to new methods to safely manipulate effector T cell responses.
Retinal ganglion cell loss is associated with future disability worsening in early relapsing-remitting multiple sclerosis
J. Wauschkuhn, G. Solorza Buenrostro, L. Aly, S. Asseyer, R. Wicklein, J. M. Hartberger, K. Ruprecht, M. Muhlau, T. Schmitz-Hubsch, C. Chien, A. Berthele, A. U. Brandt, T. Korn, F. Paul, B. Hemmer, H. G. Zimmermann, and B. Knier
Eur J Neurol, 30(4): 982-990, 2023
Retinal ganglion cell loss is associated with future disability worsening in early relapsing-remitting multiple sclerosis
J. Wauschkuhn, G. Solorza Buenrostro, L. Aly, S. Asseyer, R. Wicklein, J. M. Hartberger, K. Ruprecht, M. Muhlau, T. Schmitz-Hubsch, C. Chien, A. Berthele, A. U. Brandt, T. Korn, F. Paul, B. Hemmer, H. G. Zimmermann, and B. Knier
{BACKGROUND AND PURPOSE: Thinning of the retinal combined ganglion cell and inner plexiform layer (GCIP) as measured by optical coherence tomography (OCT) is a common finding in patients with multiple sclerosis. This study aimed to investigate whether a single retinal OCT analysis allows prediction of future disease activity after a first demyelinating event. METHODS: This observational cohort study included 201 patients with recently diagnosed clinically iso lated syndrome or relapsing-remitting multiple sclerosis from two German tertiary referral centers. Individuals underwent neurological examination, magnetic resonance imaging, and OCT at baseline and at yearly follow-up visits. RESULTS: Patients were included at a median disease duration of 2.0 months. During a median follow-up of 59 (interquartile range = 43-71) months, 82% of patients had ongoing disease activity as demonstrated by failing the no evidence of disease activity 3 (NEDA-3) criteria, and 19% presented with confirmed disability worsening. A GCIP threshold of <=77 mum at baseline identified patients with a high risk for NEDA-3 failure (hazard ratio [HR] = 1.7, 95% confidence interval [CI] = 1.1-2.8
2022
Cognate microglia-T cell interactions shape the functional regulatory T cell pool in experimental autoimmune encephalomyelitis pathology
Z. Haimon, G. R. Frumer, J. S. Kim, S. Trzebanski, R. Haffner-Krausz, S. Ben-Dor, Z. Porat, A. Muschaweckh, L. Chappell-Maor, S. Boura-Halfon, T. Korn, and S. Jung
Nat Immunol, 23(12): 1749-1762, 2022
Cognate microglia-T cell interactions shape the functional regulatory T cell pool in experimental autoimmune encephalomyelitis pathology
Z. Haimon, G. R. Frumer, J. S. Kim, S. Trzebanski, R. Haffner-Krausz, S. Ben-Dor, Z. Porat, A. Muschaweckh, L. Chappell-Maor, S. Boura-Halfon, T. Korn, and S. Jung
Microglia, the parenchymal brain macrophages of the central nervous system, have emerged as critical players in brain development and homeostasis. The immune functions of these cells, however, remain less well defined. We investigated contributions of microglia in a relapsing-remitting multiple sclerosis paradigm, experimental autoimmune encephalitis in C57BL/6 x SJL F(1) mice. Fate mapping-assisted translatome profiling during the relapsing-remitting disease course revealed the potential of microglia to interact with T cells through antigen presentation, costimulation and coinhibition. Abundant microglia-T cell aggregates, as observed by histology and flow cytometry, supported the idea of functional interactions of microglia and T cells during remission, with a bias towards regulatory T cells. Finally, microglia-restricted interferon-gamma receptor and major histocompatibility complex mutagenesis significantly affected the functionality of the regulatory T cell compartment in the diseased central nervous system and remission. Collectively, our data establish critical non-redundant cognate and cytokine-mediated interactions of microglia with CD4(+) T cells during autoimmune neuroinflammation.
Association of the retinal vasculature, intrathecal immunity, and disability in multiple sclerosis
C. Noll, M. Hiltensperger, L. Aly, R. Wicklein, A. M. Afzali, C. Mardin, C. Gasperi, A. Berthele, B. Hemmer, T. Korn, and B. Knier
Front Immunol, 13: 997043, 2022
Association of the retinal vasculature, intrathecal immunity, and disability in multiple sclerosis
C. Noll, M. Hiltensperger, L. Aly, R. Wicklein, A. M. Afzali, C. Mardin, C. Gasperi, A. Berthele, B. Hemmer, T. Korn, and B. Knier
{BACKGROUND: Optical coherence tomography angiography (OCT-A) is a novel technique allowing non-invasive assessment of the retinal vasculature. During relapsing remitting multiple sclerosis (RRMS), retinal vessel loss occurs in eyes suffering from acute optic neuritis and recent data suggest that retinal vessel loss might also be evident in non-affected eyes. We investigated whether alterations of the retinal vasculature are linked to the intrathecal immunity and whether they allow prognostication of the future disease course. MATERIAL AND METHODS: This study includes two different patient cohorts recruited at a tertiary German academic multiple sclerosis center between 2018 and 2020 and a cohort of 40 healthy controls. A total of 90 patients with RRMS undergoing lumbar puncture and OCT-A analysis were enrolled into a cross-sectional cohort study to search for associations between the retinal vasculature and the intrathecal immune compartment. We recruited another 86 RRMS patients into a prospective observational cohort study who underwent clinical examination, OCT-A and cerebral magnetic resonance imaging at baseline and during annual follow-up visits to clarify whether alterations of the retinal vessels are linked to RRMS disease activity. Eyes with a history of optic neuritis were excluded from the analysis. RESULTS: Rarefication of the superficial vascular complex occured during RRMS and was linked to higher frequencies of activated B cells and higher levels of the pro-inflammatory cytokines interferon-gamma, tumor necrosis factor alpha and interleukin-17 in the cerebrospinal fluid. During a median follow-up of 23 (interquartile range 14 - 25) months, vessel loss within the superficial (hazard ratio [HR] 1.6 for a 1%-point decrease in vessel density
Normative Data and Conversion Equation for Spectral-Domain Optical Coherence Tomography in an International Healthy Control Cohort
R. Kenney, M. Liu, L. Hasanaj, B. Joseph, A. A. Al-Hassan, L. Balk, R. Behbehani, A. U. Brandt, P. A. Calabresi, E. M. Frohman, T. Frohman, J. Havla, B. Hemmer, H. Jiang, B. Knier, T. Korn, L. Leocani, E. H. Martinez-Lapiscina, A. Papadopoulou, F. Paul, A. Petzold, M. Pisa, P. Villoslada, H. Zimmermann, H. Ishikawa, J. S. Schuman, G. Wollstein, Y. Chen, S. Saidha, L. E. Thorpe, S. L. Galetta, L. J. Balcer, and Imsvisual Consortium
J Neuroophthalmol, 42(4): 442-453, 2022
Normative Data and Conversion Equation for Spectral-Domain Optical Coherence Tomography in an International Healthy Control Cohort
R. Kenney, M. Liu, L. Hasanaj, B. Joseph, A. A. Al-Hassan, L. Balk, R. Behbehani, A. U. Brandt, P. A. Calabresi, E. M. Frohman, T. Frohman, J. Havla, B. Hemmer, H. Jiang, B. Knier, T. Korn, L. Leocani, E. H. Martinez-Lapiscina, A. Papadopoulou, F. Paul, A. Petzold, M. Pisa, P. Villoslada, H. Zimmermann, H. Ishikawa, J. S. Schuman, G. Wollstein, Y. Chen, S. Saidha, L. E. Thorpe, S. L. Galetta, L. J. Balcer, and Imsvisual Consortium
{BACKGROUND: Spectral-domain (SD-) optical coherence tomography (OCT) can reliably measure axonal (peripapillary retinal nerve fiber layer [pRNFL]) and neuronal (macular ganglion cell + inner plexiform layer [GCIPL]) thinning in the retina. Measurements from 2 commonly used SD-OCT devices are often pooled together in multiple sclerosis (MS) studies and clinical trials despite software and segmentation algorithm differences; however, individual pRNFL and GCIPL thickness measurements are not interchangeable between devices. In some circumstances, such as in the absence of a consistent OCT segmentation algorithm across platforms, a conversion equation to transform measurements between devices may be useful to facilitate pooling of data. The availability of normative data for SD-OCT measurements is limited by the lack of a large representative world-wide sample across various ages and ethnicities. Larger international studies that evaluate the effects of age, sex, and race/ethnicity on SD-OCT measurements in healthy control participants are needed to provide normative values that reflect these demographic subgroups to provide comparisons to MS retinal degeneration. METHODS: Participants were part of an 11-site collaboration within the International Multiple Sclerosis Visual System (IMSVISUAL) consortium. SD-OCT was performed by a trained technician for healthy control subjects using Spectralis or Cirrus SD-OCT devices. Peripapillary pRNFL and GCIPL thicknesses were measured on one or both devices. Automated segmentation protocols, in conjunction with manual inspection and correction of lines delineating retinal layers, were used. A conversion equation was developed using structural equation modeling, accounting for clustering, with healthy control data from one site where participants were scanned on both devices on the same day. Normative values were evaluated, with the entire cohort, for pRNFL and GCIPL thicknesses for each decade of age, by sex, and across racial groups using generalized estimating equation (GEE) models, accounting for clustering and adjusting for within-patient, intereye correlations. Change-point analyses were performed to determine at what age pRNFL and GCIPL thicknesses exhibit accelerated rates of decline. RESULTS: The healthy control cohort (n = 546) was 54% male and had a wide distribution of ages, ranging from 18 to 87 years, with a mean (SD) age of 39.3 (14.6) years. Based on 346 control participants at a single site, the conversion equation for pRNFL was Cirrus = -5.0 + (1.0 x Spectralis global value). Based on 228 controls, the equation for GCIPL was Cirrus = -4.5 + (0.9 x Spectralis global value). Standard error was 0.02 for both equations. After the age of 40 years, there was a decline of -2.4 mum per decade in pRNFL thickness ( P < 0.001, GEE models adjusting for sex, race, and country) and -1.4 mum per decade in GCIPL thickness ( P < 0.001). There was a small difference in pRNFL thickness based on sex, with female participants having slightly higher thickness (2.6 mum
The glioblastoma multiforme tumor site promotes the commitment of tumor-infiltrating lymphocytes to the TH17 lineage in humans
M. Mitsdoerffer, L. Aly, M. Barz, T. Engleitner, C. Sie, C. Delbridge, G. Lepennetier, R. Ollinger, M. Pfaller, B. Wiestler, R. Rad, B. Meyer, B. Knier, F. Schmidt-Graf, J. Gempt, and T. Korn
Proc Natl Acad Sci U S A, 119(34): e2206208119, 2022
The glioblastoma multiforme tumor site promotes the commitment of tumor-infiltrating lymphocytes to the TH17 lineage in humans
M. Mitsdoerffer, L. Aly, M. Barz, T. Engleitner, C. Sie, C. Delbridge, G. Lepennetier, R. Ollinger, M. Pfaller, B. Wiestler, R. Rad, B. Meyer, B. Knier, F. Schmidt-Graf, J. Gempt, and T. Korn
Although glioblastoma multiforme (GBM) is not an invariably cold tumor, checkpoint inhibition has largely failed in GBM. In order to investigate T cell-intrinsic properties that contribute to the resistance of GBM to endogenous or therapeutically enhanced adaptive immune responses, we sorted CD4(+) and CD8(+) T cells from the peripheral blood, normal-appearing brain tissue, and tumor bed of nine treatment-naive patients with GBM. Bulk RNA sequencing of highly pure T cell populations from these different compartments was used to obtain deep transcriptomes of tumor-infiltrating T cells (TILs). While the transcriptome of CD8(+) TILs suggested that they were partly locked in a dysfunctional state, CD4(+) TILs showed a robust commitment to the type 17 T helper cell (TH17) lineage, which was corroborated by flow cytometry in four additional GBM cases. Therefore, our study illustrates that the brain tumor environment in GBM might instruct TH17 commitment of infiltrating T helper cells. Whether these properties of CD4(+) TILs facilitate a tumor-promoting milieu and thus could be a target for adjuvant anti-TH17 cell interventions needs to be further investigated.
IL-24 intrinsically regulates Th17 cell pathogenicity in mice
C. Sie, R. Kant, C. Peter, A. Muschaweckh, M. Pfaller, L. Nirschl, H. D. Moreno, T. Chadimová, G. Lepennetier, T. Kuhlmann, R. Öllinger, T. Engleitner, R. Rad, and T. Korn
J Exp Med, 219(8), 2022
IL-24 intrinsically regulates Th17 cell pathogenicity in mice
C. Sie, R. Kant, C. Peter, A. Muschaweckh, M. Pfaller, L. Nirschl, H. D. Moreno, T. Chadimová, G. Lepennetier, T. Kuhlmann, R. Öllinger, T. Engleitner, R. Rad, and T. Korn
In certain instances, Th17 responses are associated with severe immunopathology. T cell–intrinsic mechanisms that restrict pathogenic effector functions have been described for type 1 and 2 responses but are less well studied for Th17 cells. Here, we report a cell-intrinsic feedback mechanism that controls the pathogenicity of Th17 cells. Th17 cells produce IL-24, which prompts them to secrete IL-10. The IL-10–inducing function of IL-24 is independent of the cell surface receptor of IL-24 on Th17 cells. Rather, IL-24 is recruited to the inner mitochondrial membrane, where it interacts with the NADH dehydrogenase (ubiquinone) 1 α subcomplex subunit 13 (also known as Grim19), a constituent of complex I of the respiratory chain. Together, Grim19 and IL-24 promote the accumulation of STAT3 in the mitochondrial compartment. We propose that IL-24–guided mitochondrial STAT3 constitutes a rheostat to blunt extensive STAT3 deflections in the nucleus, which might then contribute to a robust IL-10 response in Th17 cells and a restriction of immunopathology in experimental autoimmune encephalomyelitis.
Ly6D(+)Siglec-H(+) precursors contribute to conventional dendritic cells via a Zbtb46(+)Ly6D(+) intermediary stage
K. Lutz, A. Musumeci, C. Sie, E. Dursun, E. Winheim, J. Bagnoli, C. Ziegenhain, L. Rausch, V. Bergen, M. D. Luecken, R. A. J. Oostendorp, B. U. Schraml, F. J. Theis, W. Enard, T. Korn, and A. B. Krug
Nat Commun, 13(1): 3456, 2022
Ly6D(+)Siglec-H(+) precursors contribute to conventional dendritic cells via a Zbtb46(+)Ly6D(+) intermediary stage
K. Lutz, A. Musumeci, C. Sie, E. Dursun, E. Winheim, J. Bagnoli, C. Ziegenhain, L. Rausch, V. Bergen, M. D. Luecken, R. A. J. Oostendorp, B. U. Schraml, F. J. Theis, W. Enard, T. Korn, and A. B. Krug
Plasmacytoid and conventional dendritic cells (pDC and cDC) are generated from progenitor cells in the bone marrow and commitment to pDCs or cDC subtypes may occur in earlier and later progenitor stages. Cells within the CD11c(+)MHCII(-/lo)Siglec-H(+)CCR9(lo) DC precursor fraction of the mouse bone marrow generate both pDCs and cDCs. Here we investigate the heterogeneity and commitment of subsets in this compartment by single-cell transcriptomics and high-dimensional flow cytometry combined with cell fate analysis: Within the CD11c(+)MHCII(-/lo)Siglec-H(+)CCR9(lo) DC precursor pool cells expressing high levels of Ly6D and lacking expression of transcription factor Zbtb46 contain CCR9(lo)B220(hi) immediate pDC precursors and CCR9(lo)B220(lo) (lo-lo) cells which still generate pDCs and cDCs in vitro and in vivo under steady state conditions. cDC-primed cells within the Ly6D(hi)Zbtb46(-) lo-lo precursors rapidly upregulate Zbtb46 and pass through a Zbtb46(+)Ly6D(+) intermediate stage before acquiring cDC phenotype after cell division. Type I IFN stimulation limits cDC and promotes pDC output from this precursor fraction by arresting cDC-primed cells in the Zbtb46(+)Ly6D(+) stage preventing their expansion and differentiation into cDCs. Modulation of pDC versus cDC output from precursors by external factors may allow for adaptation of DC subset composition at later differentiation stages.
Aquaporin-4 prevents exaggerated astrocytosis and structural damage in retinal inflammation
A. Maisam Afzali, L. Stuve, M. Pfaller, L. Aly, K. Steiger, B. Knier, and T. Korn
J Mol Med (Berl), 100(6): 933-946, 2022
Aquaporin-4 prevents exaggerated astrocytosis and structural damage in retinal inflammation
A. Maisam Afzali, L. Stuve, M. Pfaller, L. Aly, K. Steiger, B. Knier, and T. Korn
Aquaporin-4 (AQP4) is the molecular target of the immune response in neuromyelitis optica (NMO) that leads to severe structural damage in the central nervous system (CNS) and in the retina. Conversely, AQP4 might be upregulated in astrocytes as a compensatory event in multiple sclerosis. Thus, the functional relevance of AQP4 in neuroinflammation needs to be defined. Here, we tested the role of AQP4 in the retina in MOG(35-55)-induced experimental autoimmune encephalomyelitis (EAE) using optical coherence tomography (OCT), OCT angiography, immunohistology, flow cytometry, and gene expression analysis in wild-type and Aqp4(-/-) mice. No direct infiltrates of inflammatory cells were detected in the retina. Yet, early retinal expression of TNF and Iba1 suggested that the retina participated in the inflammatory response during EAE in a similar way in wild-type and Aqp4(-/-) mice. While wild-type mice rapidly cleared retinal swelling, Aqp4(-/-) animals exhibited a sustainedly increased retinal thickness associated with retinal hyperperfusion, albumin extravasation, and upregulation of GFAP as a hallmark of retinal scarring at later stages of EAE. Eventually, the loss of retinal ganglion cells was higher in Aqp4(-/-) mice than in wild-type mice. Therefore, AQP4 expression might be critical for retinal Muller cells to clear the interstitial space from excess vasogenic edema and prevent maladaptive scarring in the retina during remote inflammatory processes of the CNS. KEY MESSAGES : Genetic ablation of AQP4 leads to a functional derangement of the retinal gliovascular unit with retinal hyperperfusion during autoimmune CNS inflammation. Genetic ablation of AQP4 results in a structural impairment of the blood retina barrier with extravasation of albumin during autoimmune CNS inflammation. Eventually, the lack of AQP4 in the retina during an inflammatory event prompts the exaggerated upregulation of GFAP as a hallmark of scarring as well as loss of retinal ganglion cells.
CD4(+)c-Met(+)Itgalpha4(+) T cell subset promotes murine neuroinflammation
M. Benkhoucha, N. L. Tran, G. Breville, I. Senoner, P. F. Bradfield, T. Papayannopoulou, D. Merkler, T. Korn, and P. H. Lalive
J Neuroinflammation, 19(1): 103, 2022
CD4(+)c-Met(+)Itgalpha4(+) T cell subset promotes murine neuroinflammation
M. Benkhoucha, N. L. Tran, G. Breville, I. Senoner, P. F. Bradfield, T. Papayannopoulou, D. Merkler, T. Korn, and P. H. Lalive
OBJECTIVE: c-Met, a tyrosine kinase receptor, is the unique receptor for hepatocyte growth factor (HGF). The HGF/c-Met axis is reported to modulate cell migration, maturation, cytokine production, and antigen presentation. Here, we report that CD4(+)c-Met(+) T cells are detected at increased levels in experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis (MS). METHODS: c-Met expression by CD4(+) T cells was analyzed mostly by flow cytometry and by immunohistochemistry from mice and human PBMCs. The in vivo role of CD4(+)c-Met(+) T cells was assessed in EAE. RESULTS: CD4(+)c-Met(+) T cells found in the CNS during EAE peak disease are characterized by a pro-inflammatory phenotype skewed towards a Th1 and Th17 polarization, with enhanced adhesion and transmigration capacities correlating with increased expression of integrin alpha4 (Itgalpha4). The adoptive transfer of Itgalpha4-expressing CD4(+)Valpha3.2(+)c-Met(+) T cells induces increased disease severity compared to CD4(+)Valpha3.2(+)c-Met(-) T cells. Finally, CD4(+)c-Met(+) T cells are detected in the brain of MS patients, as well as in the blood with a higher level of Itgalpha4. These results highlight c-Met as an immune marker of highly pathogenic pro-inflammatory and pro-migratory CD4(+) T lymphocytes associated with neuroinflammation.
Dynamics of Retinal Vessel Loss After Acute Optic Neuritis in Patients With Relapsing Multiple Sclerosis
L. Aly, C. Noll, R. Wicklein, E. Wolf, E. F. Romahn, J. Wauschkuhn, S. Hosari, C. Mardin, A. Berthele, B. Hemmer, T. Korn, and B. Knier
Neurol Neuroimmunol Neuroinflamm, 9(3), 2022
Dynamics of Retinal Vessel Loss After Acute Optic Neuritis in Patients With Relapsing Multiple Sclerosis
L. Aly, C. Noll, R. Wicklein, E. Wolf, E. F. Romahn, J. Wauschkuhn, S. Hosari, C. Mardin, A. Berthele, B. Hemmer, T. Korn, and B. Knier
BACKGROUND AND OBJECTIVES: Rarefication of the retinal vasculature as measured by optical coherence tomography angiography (OCT-A) is a novel finding in patients with multiple sclerosis (MS). This study aimed to analyze longitudinal dynamics of the retinal vasculature following an acute inflammatory relapse including acute optic neuritis (ON) and to search for associations with alterations of the retinal architecture and visual function. METHODS: This prospective longitudinal cohort study included patients with relapsing-remitting MS or clinically isolated syndrome having an acute ON (n = 20) or a non-ON relapse (n = 33). Patients underwent examinations at baseline and after 7, 14, 28, 90, and 180 days with OCT, OCT-A, and assessment of the high- (HCVA) and low-contrast visual acuity (LCVA). RESULTS: Retinal vessel loss of the superficial vascular complex (SVC) evolves early after ON and reaches a plateau between 90 and 180 days (relative vessel loss 15% +/- 8% [mean +/- SD]). In addition, an 18% +/- 18% intraindividual increase of the foveal avascular zone (FAZ) is evident within 180 days after acute ON. Both SVC thinning and FAZ enlargement were associated with worse HCVA and LCVA. Rarefication of the SVC evolved simultaneously to thinning of the common ganglion cell and inner plexiform layer (GCIP) after ON. No alterations of the deep vascular complex were seen in eyes with ON, and no alterations of the retinal vasculature were recognized in patients having acute non-ON relapses. DISCUSSION: Rarefication of the SVC and growing of the FAZ evolve rapidly after ON and are linked to persistent visual disability. ON-related SVC thinning might be closely linked to GCIP atrophy and might occur due to an altered local metabolic activity within inner retinal layers.
Optical coherence tomography angiography indicates subclinical retinal disease in neuromyelitis optica spectrum disorders
L. Aly, E. M. Strauss, N. Feucht, I. Weiss, A. Berthele, M. Mitsdoerffer, C. Haass, B. Hemmer, M. Maier, T. Korn, and B. Knier
Mult Scler, 28(4): 522-531, 2022
Optical coherence tomography angiography indicates subclinical retinal disease in neuromyelitis optica spectrum disorders
L. Aly, E. M. Strauss, N. Feucht, I. Weiss, A. Berthele, M. Mitsdoerffer, C. Haass, B. Hemmer, M. Maier, T. Korn, and B. Knier
BACKGROUND: Neuromyelitis optica spectrum disorders (NMOSD) are neuroinflammatory diseases of the central nervous system. Patients suffer from recurring relapses and it is unclear whether relapse-independent disease activity occurs and whether this is of clinical relevance. OBJECTIVE: To detect disease-specific alterations of the retinal vasculature that reflect disease activity during NMOSD. METHODS: Cross-sectional analysis of 16 patients with NMOSD, 21 patients with relapsing-remitting multiple sclerosis, and 21 healthy controls using retinal optical coherence tomography (OCT), optical coherence tomography angiography (OCT-A), measurement of glial fibrillary acidic protein (GFAP) serum levels, and assessment of visual acuity. RESULTS: Patients with NMOSD but not multiple sclerosis revealed lower foveal thickness (FT) (p = 0.02) measures and an increase of the foveal avascular zone (FAZ) (p = 0.02) compared to healthy controls independent to optic neuritis. Reduced FT (p = 0.01), enlarged FAZ areas (p = 0.0001), and vessel loss of the superficial vascular complex (p = 0.01) were linked to higher serum GFAP levels and superficial vessel loss was associated with worse visual performance in patients with NMOSD irrespective of optic neuritis. CONCLUSION: Subclinical parafoveal retinal vessel loss might occur during NMOSD and might be linked to astrocyte damage and poor visual performance. OCT-A may be a tool to study subclinical disease activity during NMOSD.
2021
Interleukin-23 receptor expressing gammadelta T cells locally promote early atherosclerotic lesion formation and plaque necrosis in mice
J. Gil-Pulido, N. Amezaga, I. Jorgacevic, H. D. Manthey, M. Rosch, T. Brand, P. Cidlinsky, S. Schafer, A. Beilhack, A. E. Saliba, K. Lorenz, L. Boon, I. Prinz, A. Waisman, T. Korn, C. Cochain, and A. Zernecke
Cardiovasc Res, 2021
Interleukin-23 receptor expressing gammadelta T cells locally promote early atherosclerotic lesion formation and plaque necrosis in mice
J. Gil-Pulido, N. Amezaga, I. Jorgacevic, H. D. Manthey, M. Rosch, T. Brand, P. Cidlinsky, S. Schafer, A. Beilhack, A. E. Saliba, K. Lorenz, L. Boon, I. Prinz, A. Waisman, T. Korn, C. Cochain, and A. Zernecke
AIMS: Atherosclerosis is a chronic inflammatory disease of the vessel wall controlled by local and systemic immune responses. The role of interleukin-23 receptor (IL-23R), expressed in adaptive immune cells (mainly T helper 17 cells) and gammadelta T cells, in atherosclerosis is only incompletely understood. Here we investigated the vascular cell types expressing IL-23R and addressed the function of IL-23R and gammadelta T cells in atherosclerosis. METHOD AND RESULTS: IL-23R+ cells were frequently found in the aortic root in contrast to the aorta in low density lipoprotein receptor deficient IL-23R reporter mice (Ldlr-/-Il23rgfp/+), and mostly identified as gammadelta T cells that express IL-17 and GM-CSF. scRNA-seq confirmed gammadelta T cells as the main cell type expressing Il23r and Il17a in the aorta. Ldlr-/-Il23rgfp/gfp mice deficient in IL-23R showed a loss of IL-23R+ cells in the vasculature, and had reduced atherosclerotic lesion formation in the aortic root compared to Ldlr-/- controls after 6 weeks of high fat diet feeding. In contrast, Ldlr-/-Tcrdelta-/- mice lacking all gammadelta T cells displayed unaltered early atherosclerotic lesion formation compared to Ldlr-/- mice. In both HFD-fed Ldlr-/-Il23rgfp/gfp and Ldlr-/-Tcrdelta-/- mice a reduction in the plaque necrotic core area was noted as well as an expansion of splenic regulatory T cells. In vitro, exposure of bone marrow-derived macrophages to both IL-17A and GM-CSF induced cell necrosis, and necroptotic RIP3K and MLKL expression, as well as inflammatory mediators. CONCLUSIONS: IL-23R+ gammadelta T cells are predominantly found in the aortic root rather than the aorta and promote early atherosclerotic lesion formation, plaque necrosis and inflammation at this site. Targeting IL-23R may thus be explored as a therapeutic approach to mitigate atherosclerotic lesion development. TRANSLATIONAL PERSPECTIVE: The mechanisms and cell types contributing to early inflammation and lesion formation are incompletely understood. Here we demonstrate that the aortic root harbors a population of IL23R-dependent gammadelta T cells that can release IL-17 and GM-CSF, and both cytokines together induce macrophage inflammation and necroptosis. IL-23R+ gammadelta T cells locally promote early lesion formation in the aortic root and contribute to the expansion of the necrotic core, a hallmark of vulnerable atherosclerotic lesions. Targeting IL-23R or IL-23 itself could thus be further explored as a therapeutic option in early atherosclerosis.
Keratinocyte-intrinsic BCL10/MALT1 activity initiates and amplifies psoriasiform skin inflammation
Z. Kurgyis, L. Vornholz, K. Pechloff, L. V. Kemeny, T. Wartewig, A. Muschaweckh, A. Joshi, K. Kranen, L. Hartjes, S. Mockel, K. Steiger, E. Hameister, T. Volz, M. Mellett, L. E. French, T. Biedermann, T. Korn, and J. Ruland
Sci Immunol, 6(65): eabi4425, 2021
Keratinocyte-intrinsic BCL10/MALT1 activity initiates and amplifies psoriasiform skin inflammation
Z. Kurgyis, L. Vornholz, K. Pechloff, L. V. Kemeny, T. Wartewig, A. Muschaweckh, A. Joshi, K. Kranen, L. Hartjes, S. Mockel, K. Steiger, E. Hameister, T. Volz, M. Mellett, L. E. French, T. Biedermann, T. Korn, and J. Ruland
[Figure: see text].
IL-6 signaling in macrophages is required for immunotherapy-driven regression of tumors
E. Beyranvand Nejad, C. Labrie, M. J. van Elsas, J. W. Kleinovink, H. W. Mittrucker, Klmc Franken, S. Heink, T. Korn, R. Arens, T. van Hall, and S. H. van der Burg
J Immunother Cancer, 9(4), 2021
IL-6 signaling in macrophages is required for immunotherapy-driven regression of tumors
E. Beyranvand Nejad, C. Labrie, M. J. van Elsas, J. W. Kleinovink, H. W. Mittrucker, Klmc Franken, S. Heink, T. Korn, R. Arens, T. van Hall, and S. H. van der Burg
BACKGROUND: High serum interleukin (IL-6) levels may cause resistance to immunotherapy by modulation of myeloid cells in the tumor microenvironment. IL-6 signaling blockade is tested in cancer, but as this inflammatory cytokine has pleiotropic effects, this treatment is not always effective. METHODS: IL-6 and IL-6R blockade was applied in an IL-6-mediated immunotherapy-resistant TC-1 tumor model (TC-1.IL-6) and immunotherapy-sensitive TC-1. CONTROL: Effects on therapeutic vaccination-induced tumor regression, recurrence and survival as well on T cells and myeloid cells in the tumor microenvironment were studied. The effects of IL-6 signaling in macrophages under therapy conditions were studied in Il6ra (fl/fl)xLysM(cre+) mice. RESULTS: Our therapeutic vaccination protocol elicits a strong tumor-specific CD8(+) T-cell response, leading to enhanced intratumoral T-cell infiltration and recruitment of tumoricidal macrophages. Blockade of IL-6 signaling exacerbated tumor outgrowth, reflected by fewer complete regressions and more recurrences after therapeutic vaccination, especially in TC-1.IL-6 tumor-bearing mice. Early IL-6 signaling blockade partly inhibited the development of the vaccine-induced CD8(+) T-cell response. However, the main mechanism was the malfunction of macrophages during therapy-induced tumor regression. Therapy efficacy was impaired in Il6ra (fl/fl)xLysM(cre+) but not cre-negative control mice, while no differences in the vaccine-induced CD8(+) T-cell response were found between these mice. IL-6 signaling blockade resulted in decreased expression of suppressor of cytokine signaling 3, essential for effective M1-type function in macrophages, and increased expression of the phagocytic checkpoint molecule signal-regulatory protein alpha by macrophages. CONCLUSION: IL-6 signaling is critical for macrophage function under circumstances of immunotherapy-induced tumor tissue destruction, in line with the acute inflammatory functions of IL-6 signaling described in infections.
Combined Treatment With Pembrolizumab and Allogenic BK Virus-Specific T Cells in Progressive Multifocal Leukoencephalopathy: A Case Report
R. Wicklein, S. Heidegger, M. Verbeek, B. Eiz-Vesper, B. Maecker-Kolhoff, J. S. Kirschke, A. Page, T. Korn, B. Hemmer, and M. Deschauer
Neurol Neuroimmunol Neuroinflamm, 8(5), 2021
Combined Treatment With Pembrolizumab and Allogenic BK Virus-Specific T Cells in Progressive Multifocal Leukoencephalopathy: A Case Report
R. Wicklein, S. Heidegger, M. Verbeek, B. Eiz-Vesper, B. Maecker-Kolhoff, J. S. Kirschke, A. Page, T. Korn, B. Hemmer, and M. Deschauer
OBJECTIVE: We report a combination of BK virus-specific T cells and pembrolizumab as a treatment option in progressive multifocal leukoencephalopathy (PML). RESULTS: A 57-year-old male patient diagnosed with PML presented a fast-progressing right hemiparesis, aphasia, and cognitive deficits. Brain MRI showed a severe leukoencephalopathy with diffusion restriction. The patient was treated with 10 doses of pembrolizumab (2 mg/kg body weight) in differing intervals and 2 partially human leukocyte antigen-matched allogenic BK virus-specific T cell transfusions after the fifth pembrolizumab treatment. Although pembrolizumab alone decreased the viral load but failed to control the virus, BK-specific T cell transfer further enhanced the decline of JC virus copies in the CSF. Moreover, the regression of leukoencephalopathy and disappearance of diffusion restriction in subsequent brain MRI were observed. The combined treatment resulted in a clinical stabilization with improvements of the cognitive and speech deficits. DISCUSSION: This case supports the hypothesis that pembrolizumab is more efficient in the presence of an appropriate number of functional antigen-specific T cells. Thus, the combined treatment of pembrolizumab and virus-specific T cells should be further evaluated as a treatment option for PML in future clinical trials.
Skin and gut imprinted helper T cell subsets exhibit distinct functional phenotypes in central nervous system autoimmunity
M. Hiltensperger, E. Beltran, R. Kant, S. Tyystjarvi, G. Lepennetier, H. Dominguez Moreno, I. J. Bauer, S. Grassmann, S. Jarosch, K. Schober, V. R. Buchholz, S. Kenet, C. Gasperi, R. Ollinger, R. Rad, A. Muschaweckh, C. Sie, L. Aly, B. Knier, G. Garg, A. M. Afzali, L. A. Gerdes, T. Kumpfel, S. Franzenburg, N. Kawakami, B. Hemmer, D. H. Busch, T. Misgeld, K. Dornmair, and T. Korn
Nat Immunol, 22(7): 880-892, 2021
Skin and gut imprinted helper T cell subsets exhibit distinct functional phenotypes in central nervous system autoimmunity
M. Hiltensperger, E. Beltran, R. Kant, S. Tyystjarvi, G. Lepennetier, H. Dominguez Moreno, I. J. Bauer, S. Grassmann, S. Jarosch, K. Schober, V. R. Buchholz, S. Kenet, C. Gasperi, R. Ollinger, R. Rad, A. Muschaweckh, C. Sie, L. Aly, B. Knier, G. Garg, A. M. Afzali, L. A. Gerdes, T. Kumpfel, S. Franzenburg, N. Kawakami, B. Hemmer, D. H. Busch, T. Misgeld, K. Dornmair, and T. Korn
Multidimensional single-cell analyses of T cells have fueled the debate about whether there is extensive plasticity or 'mixed' priming of helper T cell subsets in vivo. Here, we developed an experimental framework to probe the idea that the site of priming in the systemic immune compartment is a determinant of helper T cell-induced immunopathology in remote organs. By site-specific in vivo labeling of antigen-specific T cells in inguinal (i) or gut draining mesenteric (m) lymph nodes, we show that i-T cells and m-T cells isolated from the inflamed central nervous system (CNS) in a model of multiple sclerosis (MS) are distinct. i-T cells were Cxcr6(+), and m-T cells expressed P2rx7. Notably, m-T cells infiltrated white matter, while i-T cells were also recruited to gray matter. Therefore, we propose that the definition of helper T cell subsets by their site of priming may guide an advanced understanding of helper T cell biology in health and disease.
Formation and immunomodulatory function of meningeal B cell aggregates in progressive CNS autoimmunity
M. Mitsdoerffer, G. Di Liberto, S. Dotsch, C. Sie, I. Wagner, M. Pfaller, M. Kreutzfeldt, S. Frassle, L. Aly, B. Knier, D. H. Busch, D. Merkler, and T. Korn
Brain, 144(6): 1697-1710, 2021
Formation and immunomodulatory function of meningeal B cell aggregates in progressive CNS autoimmunity
M. Mitsdoerffer, G. Di Liberto, S. Dotsch, C. Sie, I. Wagner, M. Pfaller, M. Kreutzfeldt, S. Frassle, L. Aly, B. Knier, D. H. Busch, D. Merkler, and T. Korn
Meningeal B lymphocyte aggregates have been described in autopsy material of patients with chronic multiple sclerosis. The presence of meningeal B cell aggregates has been correlated with worse disease. However, the functional role of these meningeal B cell aggregates is not understood. Here, we use a mouse model of multiple sclerosis, the spontaneous opticospinal encephalomyelitis model, which is built on the double transgenic expression of myelin oligodendrocyte glycoprotein-specific T-cell and B-cell receptors, to show that the formation of meningeal B cell aggregates is dependent on the expression of alpha4 integrins by antigen-specific T cells. T cell-conditional genetic ablation of alpha4 integrins in opticospinal encephalomyelitis mice impaired the formation of meningeal B cell aggregates, and surprisingly, led to a higher disease incidence as compared to opticospinal encephalomyelitis mice with alpha4 integrin-sufficient T cells. B cell-conditional ablation of alpha4 integrins in opticospinal encephalomyelitis mice resulted in the entire abrogation of the formation of meningeal B cell aggregates, and opticospinal encephalomyelitis mice with alpha4 integrin-deficient B cells suffered from a higher disease burden than regular opticospinal encephalomyelitis mice. While anti-CD20 antibody-mediated systemic depletion of B cells in opticospinal encephalomyelitis mice after onset of disease failed to efficiently decrease meningeal B cell aggregates without significantly modulating disease progression, treatment with anti-CD19 chimeric antigen receptor-T cells eliminated meningeal B cell aggregates and exacerbated clinical disease in opticospinal encephalomyelitis mice. Since about 20% of B cells in organized meningeal B cell aggregates produced either IL-10 or IL-35, we propose that meningeal B cell aggregates might also have an immunoregulatory function as to the immunopathology in adjacent spinal cord white matter. The immunoregulatory function of meningeal B cell aggregates needs to be considered when designing highly efficient therapies directed against meningeal B cell aggregates for clinical application in multiple sclerosis.
Aryl Hydrocarbon Receptor Plasma Agonist Activity Correlates With Disease Activity in Progressive MS
T. Tsaktanis, T. Beyer, L. Nirschl, M. Linnerbauer, V. Grummel, M. Bussas, E. Tjon, M. Muhlau, T. Korn, B. Hemmer, F. J. Quintana, and V. Rothhammer
Neurol Neuroimmunol Neuroinflamm, 8(2), 2021
Aryl Hydrocarbon Receptor Plasma Agonist Activity Correlates With Disease Activity in Progressive MS
T. Tsaktanis, T. Beyer, L. Nirschl, M. Linnerbauer, V. Grummel, M. Bussas, E. Tjon, M. Muhlau, T. Korn, B. Hemmer, F. J. Quintana, and V. Rothhammer
OBJECTIVE: The relationship between serum aryl hydrocarbon receptor (AHR) agonistic activity levels with disease severity, its modulation over the course of relapsing-remitting MS (RRMS), and its regulation in progressive MS (PMS) are unknown. Here, we report the analysis of AHR agonistic activity levels in cross-sectional and longitudinal serum samples of patients with RRMS and PMS. METHODS: In a cross-sectional investigation, a total of 36 control patients diagnosed with noninflammatory diseases, 84 patients with RRMS, 35 patients with secondary progressive MS (SPMS), and 41 patients with primary progressive MS (PPMS) were included in this study. AHR activity was measured in a cell-based luciferase assay and correlated with age, sex, the presence of disease-modifying therapies, Expanded Disability Status Scale scores, and disease duration. In a second longitudinal investigation, we analyzed AHR activity in 13 patients diagnosed with RRMS over a period from 4 to 10 years and correlated AHR agonistic activity with white matter atrophy and lesion load volume changes. RESULTS: In RRMS, AHR ligand levels were globally decreased and associated with disease duration and neurologic disability. In SPMS and PPMS, serum AHR agonistic activity was decreased and correlated with disease severity. Finally, in longitudinal serum samples of patients with RRMS, decreased AHR agonistic activity was linked to progressive CNS atrophy and increased lesion load. CONCLUSIONS: These findings suggest that serum AHR agonist levels negatively correlate with disability in RRMS and PMS and decrease longitudinally in correlation with MRI markers of disease progression. Thus, serum AHR agonistic activity may serve as novel biomarker for disability progression in MS.
2020
Regulatory T Cells Restrain Interleukin-2- and Blimp-1-Dependent Acquisition of Cytotoxic Function by CD4(+) T Cells
A. Sledzinska, M. Vila de Mucha, K. Bergerhoff, A. Hotblack, D. F. Demane, E. Ghorani, A. U. Akarca, M. A. V. Marzolini, I. Solomon, F. A. Vargas, M. Pule, M. Ono, B. Seddon, G. Kassiotis, C. E. Ariyan, T. Korn, T. Marafioti, G. M. Lord, H. Stauss, R. G. Jenner, K. S. Peggs, and S. A. Quezada
Immunity, 52(1): 151-166 e6, 2020
Regulatory T Cells Restrain Interleukin-2- and Blimp-1-Dependent Acquisition of Cytotoxic Function by CD4(+) T Cells
A. Sledzinska, M. Vila de Mucha, K. Bergerhoff, A. Hotblack, D. F. Demane, E. Ghorani, A. U. Akarca, M. A. V. Marzolini, I. Solomon, F. A. Vargas, M. Pule, M. Ono, B. Seddon, G. Kassiotis, C. E. Ariyan, T. Korn, T. Marafioti, G. M. Lord, H. Stauss, R. G. Jenner, K. S. Peggs, and S. A. Quezada
In addition to helper and regulatory potential, CD4(+) T cells also acquire cytotoxic activity marked by granzyme B (GzmB) expression and the ability to promote rejection of established tumors. Here, we examined the molecular and cellular mechanisms underpinning the differentiation of cytotoxic CD4(+) T cells following immunotherapy. CD4(+) transfer into lymphodepleted animals or regulatory T (Treg) cell depletion promoted GzmB expression by tumor-infiltrating CD4(+), and this was prevented by interleukin-2 (IL-2) neutralization. Transcriptional analysis revealed a polyfunctional helper and cytotoxic phenotype characterized by the expression of the transcription factors T-bet and Blimp-1. While T-bet ablation restricted interferon-gamma (IFN-gamma) production, loss of Blimp-1 prevented GzmB expression in response to IL-2, suggesting two independent programs required for polyfunctionality of tumor-reactive CD4(+) T cells. Our findings underscore the role of Treg cells, IL-2, and Blimp-1 in controlling the differentiation of cytotoxic CD4(+) T cells and offer a pathway to enhancement of anti-tumor activity through their manipulation.
Cutting Edge: IL-6-Driven Immune Dysregulation Is Strictly Dependent on IL-6R alpha-Chain Expression
I. A. Mufazalov, D. Andruszewski, C. Schelmbauer, S. Heink, M. Blanfeld, J. Masri, Y. Tang, R. Schuler, C. Eich, F. T. Wunderlich, S. H. Karbach, J. A. Bluestone, T. Korn, and A. Waisman
J Immunol, 204(4): 747-751, 2020
Cutting Edge: IL-6-Driven Immune Dysregulation Is Strictly Dependent on IL-6R alpha-Chain Expression
I. A. Mufazalov, D. Andruszewski, C. Schelmbauer, S. Heink, M. Blanfeld, J. Masri, Y. Tang, R. Schuler, C. Eich, F. T. Wunderlich, S. H. Karbach, J. A. Bluestone, T. Korn, and A. Waisman
IL-6 binds to the IL-6R alpha-chain (IL-6Ralpha) and signals via the signal transducer gp130. Recently, IL-6 was found to also bind to the cell surface glycoprotein CD5, which would then engage gp130 in the absence of IL-6Ralpha. However, the biological relevance of this alternative pathway is under debate. In this study, we developed a mouse model, in which murine IL-6 is overexpressed in a CD11c-Cre-dependent manner. Transgenic mice developed a lethal immune dysregulation syndrome with increased numbers of Ly-6G(+) neutrophils and Ly-6C(hi) monocytes/macrophages. IL-6 overexpression promoted activation of CD4(+) T cells while suppressing CD5(+) B-1a cell development. However, additional ablation of IL-6Ralpha protected IL-6-overexpressing mice from IL-6-triggered inflammation and fully phenocopied IL-6Ralpha-deficient mice without IL-6 overexpression. Mechanistically, IL-6Ralpha deficiency completely prevented downstream activation of STAT3 in response to IL-6. Altogether, our data clarify that IL-6Ralpha is the only biologically relevant receptor for IL-6 in mice.
Salt generates antiinflammatory Th17 cells but amplifies pathogenicity in proinflammatory cytokine microenvironments
J. Matthias, S. Heink, F. Picard, J. Zeitrag, A. Kolz, Y. Y. Chao, D. Soll, G. P. de Almeida, E. Glasmacher, I. D. Jacobsen, T. Riedel, A. Peters, S. Floess, J. Huehn, D. Baumjohann, M. Huber, T. Korn, and C. E. Zielinski
J Clin Invest, 130(9): 4587-4600, 2020
Salt generates antiinflammatory Th17 cells but amplifies pathogenicity in proinflammatory cytokine microenvironments
J. Matthias, S. Heink, F. Picard, J. Zeitrag, A. Kolz, Y. Y. Chao, D. Soll, G. P. de Almeida, E. Glasmacher, I. D. Jacobsen, T. Riedel, A. Peters, S. Floess, J. Huehn, D. Baumjohann, M. Huber, T. Korn, and C. E. Zielinski
Th cells integrate signals from their microenvironment to acquire distinct specialization programs for efficient clearance of diverse pathogens or for immunotolerance. Ionic signals have recently been demonstrated to affect T cell polarization and function. Sodium chloride (NaCl) was proposed to accumulate in peripheral tissues upon dietary intake and to promote autoimmunity via the Th17 cell axis. Here, we demonstrate that high-NaCl conditions induced a stable, pathogen-specific, antiinflammatory Th17 cell fate in human T cells in vitro. The p38/MAPK pathway, involving NFAT5 and SGK1, regulated FoxP3 and IL-17A expression in high-NaCl conditions. The NaCl-induced acquisition of an antiinflammatory Th17 cell fate was confirmed in vivo in an experimental autoimmune encephalomyelitis (EAE) mouse model, which demonstrated strongly reduced disease symptoms upon transfer of T cells polarized in high-NaCl conditions. However, NaCl was coopted to promote murine and human Th17 cell pathogenicity, if T cell stimulation occurred in a proinflammatory and TGF-beta-low cytokine microenvironment. Taken together, our findings reveal a context-dependent, dichotomous role for NaCl in shaping Th17 cell pathogenicity. NaCl might therefore prove beneficial for the treatment of chronic inflammatory diseases in combination with cytokine-blocking drugs.
Protective effects of 4-aminopyridine in experimental optic neuritis and multiple sclerosis
M. Dietrich, V. Koska, C. Hecker, P. Gottle, A. M. Hilla, A. Heskamp, K. Lepka, A. Issberner, A. Hallenberger, C. Baksmeier, J. Steckel, L. Balk, B. Knier, T. Korn, J. Havla, E. H. Martinez-Lapiscina, N. Sola-Valls, P. Manogaran, E. D. Olbert, S. Schippling, A. Cruz-Herranz, H. Yiu, J. Button, N. G. Caldito, C. von Gall, A. K. Mausberg, M. Stettner, H. G. Zimmermann, F. Paul, A. U. Brandt, P. Kury, N. Goebels, O. Aktas, C. Berndt, S. Saidha, A. J. Green, P. A. Calabresi, D. Fischer, H. P. Hartung, and P. Albrecht
Brain, 143(4): 1127-1142, 2020
Protective effects of 4-aminopyridine in experimental optic neuritis and multiple sclerosis
M. Dietrich, V. Koska, C. Hecker, P. Gottle, A. M. Hilla, A. Heskamp, K. Lepka, A. Issberner, A. Hallenberger, C. Baksmeier, J. Steckel, L. Balk, B. Knier, T. Korn, J. Havla, E. H. Martinez-Lapiscina, N. Sola-Valls, P. Manogaran, E. D. Olbert, S. Schippling, A. Cruz-Herranz, H. Yiu, J. Button, N. G. Caldito, C. von Gall, A. K. Mausberg, M. Stettner, H. G. Zimmermann, F. Paul, A. U. Brandt, P. Kury, N. Goebels, O. Aktas, C. Berndt, S. Saidha, A. J. Green, P. A. Calabresi, D. Fischer, H. P. Hartung, and P. Albrecht
Chronic disability in multiple sclerosis is linked to neuroaxonal degeneration. 4-aminopyridine (4-AP) is used and licensed as a symptomatic treatment to ameliorate ambulatory disability in multiple sclerosis. The presumed mode of action is via blockade of axonal voltage gated potassium channels, thereby enhancing conduction in demyelinated axons. In this study, we provide evidence that in addition to those symptomatic effects, 4-AP can prevent neuroaxonal loss in the CNS. Using in vivo optical coherence tomography imaging, visual function testing and histologic assessment, we observed a reduction in retinal neurodegeneration with 4-AP in models of experimental optic neuritis and optic nerve crush. These effects were not related to an anti-inflammatory mode of action or a direct impact on retinal ganglion cells. Rather, histology and in vitro experiments indicated 4-AP stabilization of myelin and oligodendrocyte precursor cells associated with increased nuclear translocation of the nuclear factor of activated T cells. In experimental optic neuritis, 4-AP potentiated the effects of immunomodulatory treatment with fingolimod. As extended release 4-AP is already licensed for symptomatic multiple sclerosis treatment, we performed a retrospective, multicentre optical coherence tomography study to longitudinally compare retinal neurodegeneration between 52 patients on continuous 4-AP therapy and 51 matched controls. In line with the experimental data, during concurrent 4-AP therapy, degeneration of the macular retinal nerve fibre layer was reduced over 2 years. These results indicate disease-modifying effects of 4-AP beyond symptomatic therapy and provide support for the design of a prospective clinical study using visual function and retinal structure as outcome parameters.
Regulatory myeloid cells paralyze T cells through cell-cell transfer of the metabolite methylglyoxal
T. Baumann, A. Dunkel, C. Schmid, S. Schmitt, M. Hiltensperger, K. Lohr, V. Laketa, S. Donakonda, U. Ahting, B. Lorenz-Depiereux, J. E. Heil, J. Schredelseker, L. Simeoni, C. Fecher, N. Korber, T. Bauer, N. Huser, D. Hartmann, M. Laschinger, K. Eyerich, S. Eyerich, M. Anton, M. Streeter, T. Wang, B. Schraven, D. Spiegel, F. Assaad, T. Misgeld, H. Zischka, P. J. Murray, A. Heine, M. Heikenwalder, T. Korn, C. Dawid, T. Hofmann, P. A. Knolle, and B. Hochst
Nat Immunol, 21(5): 555-566, 2020
Regulatory myeloid cells paralyze T cells through cell-cell transfer of the metabolite methylglyoxal
T. Baumann, A. Dunkel, C. Schmid, S. Schmitt, M. Hiltensperger, K. Lohr, V. Laketa, S. Donakonda, U. Ahting, B. Lorenz-Depiereux, J. E. Heil, J. Schredelseker, L. Simeoni, C. Fecher, N. Korber, T. Bauer, N. Huser, D. Hartmann, M. Laschinger, K. Eyerich, S. Eyerich, M. Anton, M. Streeter, T. Wang, B. Schraven, D. Spiegel, F. Assaad, T. Misgeld, H. Zischka, P. J. Murray, A. Heine, M. Heikenwalder, T. Korn, C. Dawid, T. Hofmann, P. A. Knolle, and B. Hochst
Regulatory myeloid immune cells, such as myeloid-derived suppressor cells (MDSCs), populate inflamed or cancerous tissue and block immune cell effector functions. The lack of mechanistic insight into MDSC suppressive activity and a marker for their identification has hampered attempts to overcome T cell inhibition and unleash anti-cancer immunity. Here, we report that human MDSCs were characterized by strongly reduced metabolism and conferred this compromised metabolic state to CD8(+) T cells, thereby paralyzing their effector functions. We identified accumulation of the dicarbonyl radical methylglyoxal, generated by semicarbazide-sensitive amine oxidase, to cause the metabolic phenotype of MDSCs and MDSC-mediated paralysis of CD8(+) T cells. In a murine cancer model, neutralization of dicarbonyl activity overcame MDSC-mediated T cell suppression and, together with checkpoint inhibition, improved the efficacy of cancer immune therapy. Our results identify the dicarbonyl methylglyoxal as a marker metabolite for MDSCs that mediates T cell paralysis and can serve as a target to improve cancer immune therapy.
IgG Fc sialylation is regulated during the germinal center reaction following immunization with different adjuvants
Y. C. Bartsch, S. Eschweiler, A. Leliavski, H. B. Lunding, S. Wagt, J. Petry, G. M. Lilienthal, J. Rahmoller, N. de Haan, A. Holscher, R. Erapaneedi, A. D. Giannou, L. Aly, R. Sato, L. A. de Neef, A. Winkler, D. Braumann, J. Hobusch, K. Kuhnigk, V. Kremer, M. Steinhaus, V. Blanchard, T. Gemoll, J. K. Habermann, M. Collin, G. Salinas, R. A. Manz, H. Fukuyama, T. Korn, A. Waisman, N. Yogev, S. Huber, B. Rabe, S. Rose-John, H. Busch, F. Berberich-Siebelt, C. Holscher, M. Wuhrer, and M. Ehlers
J Allergy Clin Immunol, 146(3): 652-666 e11, 2020
IgG Fc sialylation is regulated during the germinal center reaction following immunization with different adjuvants
Y. C. Bartsch, S. Eschweiler, A. Leliavski, H. B. Lunding, S. Wagt, J. Petry, G. M. Lilienthal, J. Rahmoller, N. de Haan, A. Holscher, R. Erapaneedi, A. D. Giannou, L. Aly, R. Sato, L. A. de Neef, A. Winkler, D. Braumann, J. Hobusch, K. Kuhnigk, V. Kremer, M. Steinhaus, V. Blanchard, T. Gemoll, J. K. Habermann, M. Collin, G. Salinas, R. A. Manz, H. Fukuyama, T. Korn, A. Waisman, N. Yogev, S. Huber, B. Rabe, S. Rose-John, H. Busch, F. Berberich-Siebelt, C. Holscher, M. Wuhrer, and M. Ehlers
BACKGROUND: Effector functions of IgG Abs are regulated by their Fc N-glycosylation pattern. IgG Fc glycans that lack galactose and terminal sialic acid residues correlate with the severity of inflammatory (auto)immune disorders and have also been linked to protection against viral infection and discussed in the context of vaccine-induced protection. In contrast, sialylated IgG Abs have shown immunosuppressive effects. OBJECTIVE: We sought to investigate IgG glycosylation programming during the germinal center (GC) reaction following immunization of mice with a foreign protein antigen and different adjuvants. METHODS: Mice were analyzed for GC T-cell, B-cell, and plasma cell responses, as well as for antigen-specific serum IgG subclass titers and Fc glycosylation patterns. RESULTS: Different adjuvants induce distinct IgG(+) GC B-cell responses with specific transcriptomes and expression levels of the alpha2,6-sialyltransferase responsible for IgG sialylation that correspond to distinct serum IgG Fc glycosylation patterns. Low IgG Fc sialylation programming in GC B cells was overall highly dependent on the Foxp3(-) follicular helper T (TFH) cell-inducing cytokine IL-6, here in particular induced by water-in-oil adjuvants and Mycobacterium tuberculosis. Furthermore, low IgG Fc sialylation programming was dependent on adjuvants that induced IL-27 receptor-dependent IFN-gamma(+) TFH1 cells, IL-6/IL-23-dependent IL-17A(+) TFH17 cells, and high ratios of TFH cells to Foxp3(+) follicular regulatory T cells. Here, the 2 latter were dependent on M tuberculosis and its cord factor. CONCLUSION: This study's findings regarding adjuvant-dependent GC responses and IgG glycosylation programming may aid in the development of novel vaccination strategies to induce IgG Abs with both high affinity and defined Fc glycosylation patterns in the GC.
Inner retinal layer thinning in radiologically isolated syndrome predicts conversion to multiple sclerosis
L. Aly, J. Havla, G. Lepennetier, T. F. M. Andlauer, C. Sie, E. M. Strauss, M. M. Hoshi, T. Kumpfel, M. Hiltensperger, M. Mitsdoerffer, M. Muhlau, C. Zimmer, B. Hemmer, T. Korn, and B. Knier
Eur J Neurol, 27(11): 2217-2224, 2020
Inner retinal layer thinning in radiologically isolated syndrome predicts conversion to multiple sclerosis
L. Aly, J. Havla, G. Lepennetier, T. F. M. Andlauer, C. Sie, E. M. Strauss, M. M. Hoshi, T. Kumpfel, M. Hiltensperger, M. Mitsdoerffer, M. Muhlau, C. Zimmer, B. Hemmer, T. Korn, and B. Knier
BACKGROUND AND PURPOSE: Individuals with radiologically isolated syndrome (RIS) are at increased risk of converting to multiple sclerosis (MS). Early identification of later converters is crucial for optimal treatment decisions. The purpose of this study was to assess the predictive potential of optical coherence tomography (OCT) measures in individuals with RIS regarding conversion to MS. METHODS: This prospective observational cohort study included 36 individuals with RIS and 36 healthy controls recruited from two German MS centers. All individuals received baseline OCT and clinical examination and were longitudinally followed over up to 6 years. The primary outcome measure was the conversion to MS. RESULTS: During clinical follow-up of 46 (26-58) months (median, 25%-75% interquartile range), eight individuals with RIS converted to MS. Individuals converting to MS showed a thinning of the peripapillary retinal nerve fiber layer (pRNFL) and the common ganglion cell and inner plexiform layer (GCIP) at baseline and during follow-up. Individuals with a pRNFL of 99 microm or lower or a GCIP of 1.99 mm(3) or lower were at a 7.5- and 8.0-fold risk for MS conversion, respectively, compared to individuals with higher measures. After correction for other known risk factors, Cox proportional hazards regression revealed a hazard ratio of 1.08 for conversion to MS for each 1 microm decline in pRNFL. CONCLUSIONS: Reduction of the pRNFL might be a novel and independent risk factor for conversion to MS in individuals with RIS. OCT might be useful for risk stratification and therapeutic decision-making in individuals with RIS.
2019
Retinal inner nuclear layer volume reflects inflammatory disease activity in multiple sclerosis; a longitudinal OCT study
L. J. Balk, D. Coric, B. Knier, H. G. Zimmermann, R. Behbehani, R. Alroughani, E. H. Martinez-Lapiscina, A. U. Brandt, B. Sanchez-Dalmau, A. Vidal-Jordana, P. Albrecht, V. Koska, J. Havla, M. Pisa, R. C. Nolan, L. Leocani, F. Paul, O. Aktas, X. Montalban, L. J. Balcer, P. Villoslada, O. Outteryck, T. Korn, A. Petzold, and Imsvisual consortium
Mult Scler J Exp Transl Clin, 5(3): 2055217319871582, 2019
Retinal inner nuclear layer volume reflects inflammatory disease activity in multiple sclerosis; a longitudinal OCT study
L. J. Balk, D. Coric, B. Knier, H. G. Zimmermann, R. Behbehani, R. Alroughani, E. H. Martinez-Lapiscina, A. U. Brandt, B. Sanchez-Dalmau, A. Vidal-Jordana, P. Albrecht, V. Koska, J. Havla, M. Pisa, R. C. Nolan, L. Leocani, F. Paul, O. Aktas, X. Montalban, L. J. Balcer, P. Villoslada, O. Outteryck, T. Korn, A. Petzold, and Imsvisual consortium
{Background: The association of peripapillary retinal nerve fibre layer (pRNFL) and ganglion cell-inner plexiform layer (GCIPL) thickness with neurodegeneration in multiple sclerosis (MS) is well established. The relationship of the adjoining inner nuclear layer (INL) with inflammatory disease activity is less well understood. Objective: The objective of this paper is to investigate the relationship of INL volume changes with inflammatory disease activity in MS.Methods In this longitudinal, multi-centre study, optical coherence tomography (OCT) and clinical data (disability status, relapses and MS optic neuritis (MSON)) were collected in 785 patients with MS (68.3% female) and 92 healthy controls (63.4% female) from 11 MS centres between 2010 and 2017 and pooled retrospectively. Data on pRNFL, GCIPL and INL were obtained at each centre. Results: There was a significant increase in INL volume in eyes with new MSON during the study (N = 61/1562
Cell-type-specific profiling of brain mitochondria reveals functional and molecular diversity
C. Fecher, L. Trovo, S. A. Muller, N. Snaidero, J. Wettmarshausen, S. Heink, O. Ortiz, I. Wagner, R. Kuhn, J. Hartmann, R. M. Karl, A. Konnerth, T. Korn, W. Wurst, D. Merkler, S. F. Lichtenthaler, F. Perocchi, and T. Misgeld
Nat Neurosci, 2019
Cell-type-specific profiling of brain mitochondria reveals functional and molecular diversity
C. Fecher, L. Trovo, S. A. Muller, N. Snaidero, J. Wettmarshausen, S. Heink, O. Ortiz, I. Wagner, R. Kuhn, J. Hartmann, R. M. Karl, A. Konnerth, T. Korn, W. Wurst, D. Merkler, S. F. Lichtenthaler, F. Perocchi, and T. Misgeld
Mitochondria vary in morphology and function in different tissues; however, little is known about their molecular diversity among cell types. Here we engineered MitoTag mice, which express a Cre recombinase-dependent green fluorescent protein targeted to the outer mitochondrial membrane, and developed an isolation approach to profile tagged mitochondria from defined cell types. We determined the mitochondrial proteome of the three major cerebellar cell types (Purkinje cells, granule cells and astrocytes) and identified hundreds of mitochondrial proteins that are differentially regulated. Thus, we provide markers of cell-type-specific mitochondria for the healthy and diseased mouse and human central nervous systems, including in amyotrophic lateral sclerosis and Alzheimer's disease. Based on proteomic predictions, we demonstrate that astrocytic mitochondria metabolize long-chain fatty acids more efficiently than neuronal mitochondria. We also characterize cell-type differences in mitochondrial calcium buffering via the mitochondrial calcium uniporter (Mcu) and identify regulator of microtubule dynamics protein 3 (Rmdn3) as a determinant of endoplasmic reticulum-mitochondria proximity in Purkinje cells. Our approach enables exploring mitochondrial diversity in many in vivo contexts.
Dendritic Cell Accumulation in the Gut and Central Nervous System Is Differentially Dependent on alpha4 Integrins
C. Sie, L. G. Perez, M. Kreutzfeldt, M. Potthast, C. Ohnmacht, D. Merkler, S. Huber, A. Krug, and T. Korn
J Immunol, 203(6): 1417-1427, 2019
Dendritic Cell Accumulation in the Gut and Central Nervous System Is Differentially Dependent on alpha4 Integrins
C. Sie, L. G. Perez, M. Kreutzfeldt, M. Potthast, C. Ohnmacht, D. Merkler, S. Huber, A. Krug, and T. Korn
Homing of pathogenic CD4(+) T cells to the CNS is dependent on alpha4 integrins. However, it is uncertain whether alpha4 integrins are also required for the migration of dendritic cell (DC) subsets, which sample Ags from nonlymphoid tissues to present it to T cells. In this study, after genetic ablation of Itga4 in DCs and monocytes in mice via the promoters of Cd11c and Lyz2 (also known as LysM), respectively, the recruitment of alpha4 integrin-deficient conventional and plasmacytoid DCs to the CNS was unaffected, whereas alpha4 integrin-deficient, monocyte-derived DCs accumulated less efficiently in the CNS during experimental autoimmune encephalomyelitis in a competitive setting than their wild-type counterparts. In a noncompetitive setting, alpha4 integrin deficiency on monocyte-derived DCs was fully compensated. In contrast, in small intestine and colon, the fraction of alpha4 integrin-deficient CD11b(+)CD103(+) DCs was selectively reduced in steady-state. Yet, T cell-mediated inflammation and host defense against Citrobacter rodentium were not impaired in the absence of alpha4 integrins on DCs. Thus, inflammatory conditions can promote an environment that is indifferent to alpha4 integrin expression by DCs.
Brain-resident memory T cells generated early in life predispose to autoimmune disease in mice
K. Steinbach, I. Vincenti, K. Egervari, M. Kreutzfeldt, F. van der Meer, N. Page, B. Klimek, I. Rossitto-Borlat, G. Di Liberto, A. Muschaweckh, I. Wagner, K. Hammad, C. Stadelmann, T. Korn, O. Hartley, D. D. Pinschewer, and D. Merkler
Sci Transl Med, 11(498), 2019
Brain-resident memory T cells generated early in life predispose to autoimmune disease in mice
K. Steinbach, I. Vincenti, K. Egervari, M. Kreutzfeldt, F. van der Meer, N. Page, B. Klimek, I. Rossitto-Borlat, G. Di Liberto, A. Muschaweckh, I. Wagner, K. Hammad, C. Stadelmann, T. Korn, O. Hartley, D. D. Pinschewer, and D. Merkler
Epidemiological studies associate viral infections during childhood with the risk of developing autoimmune disease during adulthood. However, the mechanistic link between these events remains elusive. We report that transient viral infection of the brain in early life, but not at a later age, precipitates brain autoimmune disease elicited by adoptive transfer of myelin-specific CD4(+) T cells at sites of previous infection in adult mice. Early-life infection of mouse brains imprinted a chronic inflammatory signature that consisted of brain-resident memory T cells expressing the chemokine (C-C motif) ligand 5 (CCL5). Blockade of CCL5 signaling via C-C chemokine receptor type 5 prevented the formation of brain lesions in a mouse model of autoimmune disease. In mouse and human brain, CCL5(+) TRM were located predominantly to sites of microglial activation. This study uncovers how transient brain viral infections in a critical window in life might leave persisting chemotactic cues and create a long-lived permissive environment for autoimmunity.
CSF Protein Concentration Shows No Correlation With Brain Volume Measures
A. Wuschek, S. Grahl, V. Pongratz, T. Korn, J. Kirschke, C. Zimmer, B. Hemmer, and M. Muhlau
Front Neurol, 10: 463, 2019
CSF Protein Concentration Shows No Correlation With Brain Volume Measures
A. Wuschek, S. Grahl, V. Pongratz, T. Korn, J. Kirschke, C. Zimmer, B. Hemmer, and M. Muhlau
Background: CSF protein concentrations vary greatly among individuals. Accounting for brain volume may lower the variance and increase the diagnostic value of CSF protein concentrations. Objective: To determine the relation between CSF protein concentrations and brain volume. Methods: Brain volumes (total intracranial, gray matter, white matter volumes) derived from brain MRI and CSF protein concentrations (total protein, albumin, albumin CSF/serum ratio) of 29 control patients and 497 patients with clinically isolated syndrome or multiple sclerosis were studied. Finding: We found significant positive correlations of CSF protein concentrations with intracranial, gray matter, and white matter volumes. None of the correlations remained significant after correction for age and sex. Conclusion: Accounting for brain volume derived from brain MRI is unlikely to improve the diagnostic value of protein concentrations in CSF.
Optimal intereye difference thresholds by optical coherence tomography in multiple sclerosis: An international study
R. C. Nolan-Kenney, M. Liu, O. Akhand, P. A. Calabresi, F. Paul, A. Petzold, L. Balk, A. U. Brandt, E. H. Martinez-Lapiscina, S. Saidha, P. Villoslada, A. A. Al-Hassan, R. Behbehani, E. M. Frohman, T. Frohman, J. Havla, B. Hemmer, H. Jiang, B. Knier, T. Korn, L. Leocani, A. Papadopoulou, M. Pisa, H. Zimmermann, S. L. Galetta, L. J. Balcer, and Consortium International Multiple Sclerosis Visual System
Ann Neurol, 85(5): 618-629, 2019
Optimal intereye difference thresholds by optical coherence tomography in multiple sclerosis: An international study
R. C. Nolan-Kenney, M. Liu, O. Akhand, P. A. Calabresi, F. Paul, A. Petzold, L. Balk, A. U. Brandt, E. H. Martinez-Lapiscina, S. Saidha, P. Villoslada, A. A. Al-Hassan, R. Behbehani, E. M. Frohman, T. Frohman, J. Havla, B. Hemmer, H. Jiang, B. Knier, T. Korn, L. Leocani, A. Papadopoulou, M. Pisa, H. Zimmermann, S. L. Galetta, L. J. Balcer, and Consortium International Multiple Sclerosis Visual System
OBJECTIVE: To determine the optimal thresholds for intereye differences in retinal nerve fiber and ganglion cell + inner plexiform layer thicknesses for identifying unilateral optic nerve lesions in multiple sclerosis. Current international diagnostic criteria for multiple sclerosis do not include the optic nerve as a lesion site despite frequent involvement. Optical coherence tomography detects retinal thinning associated with optic nerve lesions. METHODS: In this multicenter international study at 11 sites, optical coherence tomography was measured for patients and healthy controls as part of the International Multiple Sclerosis Visual System Consortium. High- and low-contrast acuity were also collected in a subset of participants. Presence of an optic nerve lesion for this study was defined as history of acute unilateral optic neuritis. RESULTS: Among patients (n = 1,530), receiver operating characteristic curve analysis demonstrated an optimal peripapillary retinal nerve fiber layer intereye difference threshold of 5mum and ganglion cell + inner plexiform layer threshold of 4mum for identifying unilateral optic neuritis (n = 477). Greater intereye differences in acuities were associated with greater intereye retinal layer thickness differences (p <= 0.001). INTERPRETATION: Intereye differences of 5mum for retinal nerve fiber layer and 4mum for macular ganglion cell + inner plexiform layer are robust thresholds for identifying unilateral optic nerve lesions. These thresholds may be useful in establishing the presence of asymptomatic and symptomatic optic nerve lesions in multiple sclerosis and could be useful in a new version of the diagnostic criteria. Our findings lend further validation for utilizing the visual system in a multiple sclerosis clinical trial setting. Ann Neurol 2019;85:618-629.
Blimp1 Prevents Methylation of Foxp3 and Loss of Regulatory T Cell Identity at Sites of Inflammation
G. Garg, A. Muschaweckh, H. Moreno, A. Vasanthakumar, S. Floess, G. Lepennetier, R. Oellinger, Y. Zhan, T. Regen, M. Hiltensperger, C. Peter, L. Aly, B. Knier, L. R. Palam, R. Kapur, M. H. Kaplan, A. Waisman, R. Rad, G. Schotta, J. Huehn, A. Kallies, and T. Korn
Cell Rep, 26(7): 1854-1868 e5, 2019
Blimp1 Prevents Methylation of Foxp3 and Loss of Regulatory T Cell Identity at Sites of Inflammation
G. Garg, A. Muschaweckh, H. Moreno, A. Vasanthakumar, S. Floess, G. Lepennetier, R. Oellinger, Y. Zhan, T. Regen, M. Hiltensperger, C. Peter, L. Aly, B. Knier, L. R. Palam, R. Kapur, M. H. Kaplan, A. Waisman, R. Rad, G. Schotta, J. Huehn, A. Kallies, and T. Korn
Foxp3(+) regulatory T (Treg) cells restrict immune pathology in inflamed tissues; however, an inflammatory environment presents a threat to Treg cell identity and function. Here, we establish a transcriptional signature of central nervous system (CNS) Treg cells that accumulate during experimental autoimmune encephalitis (EAE) and identify a pathway that maintains Treg cell function and identity during severe inflammation. This pathway is dependent on the transcriptional regulator Blimp1, which prevents downregulation of Foxp3 expression and "toxic" gain-of-function of Treg cells in the inflamed CNS. Blimp1 negatively regulates IL-6- and STAT3-dependent Dnmt3a expression and function restraining methylation of Treg cell-specific conserved non-coding sequence 2 (CNS2) in the Foxp3 locus. Consequently, CNS2 is heavily methylated when Blimp1 is ablated, leading to a loss of Foxp3 expression and severe disease. These findings identify a Blimp1-dependent pathway that preserves Treg cell stability in inflamed non-lymphoid tissues.
RelB Deficiency in Dendritic Cells Protects from Autoimmune Inflammation Due to Spontaneous Accumulation of Tissue T Regulatory Cells
N. Andreas, M. Potthast, A. L. Geiselhoringer, G. Garg, R. de Jong, J. Riewaldt, D. Russkamp, M. Riemann, J. P. Girard, S. Blank, K. Kretschmer, C. Schmidt-Weber, T. Korn, F. Weih, and C. Ohnmacht
J Immunol, 203(10): 2602-2613, 2019
RelB Deficiency in Dendritic Cells Protects from Autoimmune Inflammation Due to Spontaneous Accumulation of Tissue T Regulatory Cells
N. Andreas, M. Potthast, A. L. Geiselhoringer, G. Garg, R. de Jong, J. Riewaldt, D. Russkamp, M. Riemann, J. P. Girard, S. Blank, K. Kretschmer, C. Schmidt-Weber, T. Korn, F. Weih, and C. Ohnmacht
Foxp3(+) regulatory T cells are well-known immune suppressor cells in various settings. In this study, we provide evidence that knockout of the relB gene in dendritic cells (DCs) of C57BL/6 mice results in a spontaneous and systemic accumulation of Foxp3(+) T regulatory T cells (Tregs) partially at the expense of microbiota-reactive Tregs. Deletion of nfkb2 does not fully recapitulate this phenotype, indicating that alternative NF-kappaB activation via the RelB/p52 complex is not solely responsible for Treg accumulation. Deletion of RelB in DCs further results in an impaired oral tolerance induction and a marked type 2 immune bias among accumulated Foxp3(+) Tregs reminiscent of a tissue Treg signature. Tissue Tregs were fully functional, expanded independently of IL-33, and led to an almost complete Treg-dependent protection from experimental autoimmune encephalomyelitis. Thus, we provide clear evidence that RelB-dependent pathways regulate the capacity of DCs to quantitatively and qualitatively impact on Treg biology and constitute an attractive target for treatment of autoimmune diseases but may come at risk for reduced immune tolerance in the intestinal tract.
Optical coherence tomography angiography indicates associations of the retinal vascular network and disease activity in multiple sclerosis
N. Feucht, M. Maier, G. Lepennetier, M. Pettenkofer, C. Wetzlmair, T. Daltrozzo, P. Scherm, C. Zimmer, M. M. Hoshi, B. Hemmer, T. Korn, and B. Knier
Mult Scler, 25(2): 224-234, 2019
Optical coherence tomography angiography indicates associations of the retinal vascular network and disease activity in multiple sclerosis
N. Feucht, M. Maier, G. Lepennetier, M. Pettenkofer, C. Wetzlmair, T. Daltrozzo, P. Scherm, C. Zimmer, M. M. Hoshi, B. Hemmer, T. Korn, and B. Knier
BACKGROUND:: Patients with multiple sclerosis (MS) and clinically isolated syndrome (CIS) may show alterations of retinal layer architecture as measured by optical coherence tomography. Little is known about changes in the retinal vascular network during MS. OBJECTIVE:: To characterize retinal vessel structures in patients with MS and CIS and to test for associations with MS disease activity. METHOD:: In all, 42 patients with MS or CIS and 50 healthy controls underwent retinal optical coherence tomography angiography (OCT-A) with analysis of the superficial and deep vascular plexuses and the choriocapillaries. We tested OCT-A parameters for associations with retinal layer volumes, history of optic neuritis (ON), and the retrospective disease activity. RESULTS:: Inner retinal layer volumes correlated positively with the density of both the superficial and deep vascular plexuses. Eyes of MS/CIS patients with a history of ON revealed reduced vessel densities of the superficial and deep vascular plexuses as compared to healthy controls. Higher choriocapillary vessel densities were associated with ongoing inflammatory disease activity during 24 months prior to OCT-A examination in MS and CIS patients. CONCLUSION:: Optic neuritis is associated with rarefaction of the superficial and deep retinal vessels. Alterations of the choriocapillaries might be linked to disease activity in MS.
2018
Myeloid-derived suppressor cells control B cell accumulation in the central nervous system during autoimmunity
B. Knier, M. Hiltensperger, C. Sie, L. Aly, G. Lepennetier, T. Engleitner, G. Garg, A. Muschaweckh, M. Mitsdorffer, U. Koedel, B. Hochst, P. Knolle, M. Gunzer, B. Hemmer, R. Rad, D. Merkler, and T. Korn
Nat Immunol, 19(12): 1341-1351, 2018
Myeloid-derived suppressor cells control B cell accumulation in the central nervous system during autoimmunity
B. Knier, M. Hiltensperger, C. Sie, L. Aly, G. Lepennetier, T. Engleitner, G. Garg, A. Muschaweckh, M. Mitsdorffer, U. Koedel, B. Hochst, P. Knolle, M. Gunzer, B. Hemmer, R. Rad, D. Merkler, and T. Korn
Polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) have been characterized in the context of malignancies. Here we show that PMN-MDSCs can restrain B cell accumulation during central nervous system (CNS) autoimmunity. Ly6G(+) cells were recruited to the CNS during experimental autoimmune encephalomyelitis (EAE), interacted with B cells that produced the cytokines GM-CSF and interleukin-6 (IL-6), and acquired properties of PMN-MDSCs in the CNS in a manner dependent on the signal transducer STAT3. Depletion of Ly6G(+) cells or dysfunction of Ly6G(+) cells through conditional ablation of STAT3 led to the selective accumulation of GM-CSF-producing B cells in the CNS compartment, which in turn promoted an activated microglial phenotype and lack of recovery from EAE. The frequency of CD138(+) B cells in the cerebrospinal fluid (CSF) of human subjects with multiple sclerosis was negatively correlated with the frequency of PMN-MDSCs in the CSF. Thus PMN-MDSCs might selectively control the accumulation and cytokine secretion of B cells in the inflamed CNS.
Mitophagy in Intestinal Epithelial Cells Triggers Adaptive Immunity during Tumorigenesis
P. K. Ziegler, J. Bollrath, C. K. Pallangyo, T. Matsutani, O. Canli, T. De Oliveira, M. A. Diamanti, N. Muller, J. Gamrekelashvili, T. Putoczki, D. Horst, A. K. Mankan, M. G. Oner, S. Muller, J. Muller-Hocker, T. Kirchner, J. Slotta-Huspenina, M. M. Taketo, T. Reinheckel, S. Drose, A. C. Larner, W. S. Wels, M. Ernst, T. F. Greten, M. C. Arkan, T. Korn, D. Wirth, and F. R. Greten
Cell, 174(1): 88-101 e16, 2018
Mitophagy in Intestinal Epithelial Cells Triggers Adaptive Immunity during Tumorigenesis
P. K. Ziegler, J. Bollrath, C. K. Pallangyo, T. Matsutani, O. Canli, T. De Oliveira, M. A. Diamanti, N. Muller, J. Gamrekelashvili, T. Putoczki, D. Horst, A. K. Mankan, M. G. Oner, S. Muller, J. Muller-Hocker, T. Kirchner, J. Slotta-Huspenina, M. M. Taketo, T. Reinheckel, S. Drose, A. C. Larner, W. S. Wels, M. Ernst, T. F. Greten, M. C. Arkan, T. Korn, D. Wirth, and F. R. Greten
In colorectal cancer patients, a high density of cytotoxic CD8(+) T cells in tumors is associated with better prognosis. Using a Stat3 loss-of-function approach in two wnt/beta-catenin-dependent autochthonous models of sporadic intestinal tumorigenesis, we unravel a complex intracellular process in intestinal epithelial cells (IECs) that controls the induction of a CD8(+) T cell based adaptive immune response. Elevated mitophagy in IECs causes iron(II)-accumulation in epithelial lysosomes, in turn, triggering lysosomal membrane permeabilization. Subsequent release of proteases into the cytoplasm augments MHC class I presentation and activation of CD8(+) T cells via cross-dressing of dendritic cells. Thus, our findings highlight a so-far-unrecognized link between mitochondrial function, lysosomal integrity, and MHC class I presentation in IECs and suggest that therapies triggering mitophagy or inducing LMP in IECs may prove successful in shifting the balance toward anti-tumor immunity in colorectal cancer.
Blockade of MCAM/CD146 impedes CNS infiltration of T cells over the choroid plexus
J. Breuer, E. Korpos, M. J. Hannocks, T. Schneider-Hohendorf, J. Song, L. Zondler, S. Herich, K. Flanagan, T. Korn, A. Zarbock, T. Kuhlmann, L. Sorokin, H. Wiendl, and N. Schwab
J Neuroinflammation, 15(1): 236, 2018
Blockade of MCAM/CD146 impedes CNS infiltration of T cells over the choroid plexus
J. Breuer, E. Korpos, M. J. Hannocks, T. Schneider-Hohendorf, J. Song, L. Zondler, S. Herich, K. Flanagan, T. Korn, A. Zarbock, T. Kuhlmann, L. Sorokin, H. Wiendl, and N. Schwab
BACKGROUND: Very late antigen 4 (VLA-4; integrin alpha4beta1) is critical for transmigration of T helper (TH) 1 cells into the central nervous system (CNS) under inflammatory conditions such as multiple sclerosis (MS). We have previously shown that VLA-4 and melanoma cell adhesion molecule (MCAM) are important for trans-endothelial migration of human TH17 cells in vitro and here investigate their contribution to pathogenic CNS inflammation. METHODS: Antibody blockade of VLA-4 and MCAM is assessed in murine models of CNS inflammation in conjunction with conditional ablation of alpha4-integrin expression in T cells. Effects of VLA-4 and MCAM blockade on lymphocyte migration are further investigated in the human system via in vitro T cell transmigration assays. RESULTS: Compared to the broad effects of VLA-4 blockade on encephalitogenic T cell migration over endothelial barriers, MCAM blockade impeded encephalitogenic T cell migration in murine models of MS that especially depend on CNS migration across the choroid plexus (CP). In transgenic mice lacking T cell alpha4-integrin expression (CD4::Itga4(-/-)), MCAM blockade delayed disease onset. Migration of MCAM-expressing T cells through the CP into the CNS was restricted, where laminin 411 (composed of alpha4, beta1, gamma1 chains), the proposed major ligand of MCAM, is detected in the endothelial basement membranes of murine CP tissue. This finding was translated to the human system; blockade of MCAM with a therapeutic antibody reduced in vitro transmigration of MCAM-expressing T cells across a human fibroblast-derived extracellular matrix layer and a brain-derived endothelial monolayer, both expressing laminin alpha4. Laminin alpha4 was further detected in situ in CP endothelial-basement membranes in MS patients' brain tissue. CONCLUSIONS: Our findings suggest that MCAM-laminin 411 interactions facilitate trans-endothelial migration of MCAM-expressing T cells into the CNS, which seems to be highly relevant to migration via the CP and to potential future clinical applications in neuroinflammatory disorders.
IL-23 (Interleukin-23)-Producing Conventional Dendritic Cells Control the Detrimental IL-17 (Interleukin-17) Response in Stroke
M. Gelderblom, M. Gallizioli, P. Ludewig, V. Thom, P. Arunachalam, B. Rissiek, C. Bernreuther, M. Glatzel, T. Korn, T. V. Arumugam, J. Sedlacik, C. Gerloff, E. Tolosa, A. M. Planas, and T. Magnus
Stroke, 49(1): 155-164, 2018
IL-23 (Interleukin-23)-Producing Conventional Dendritic Cells Control the Detrimental IL-17 (Interleukin-17) Response in Stroke
M. Gelderblom, M. Gallizioli, P. Ludewig, V. Thom, P. Arunachalam, B. Rissiek, C. Bernreuther, M. Glatzel, T. Korn, T. V. Arumugam, J. Sedlacik, C. Gerloff, E. Tolosa, A. M. Planas, and T. Magnus
BACKGROUND AND PURPOSE: Inflammatory mechanisms can exacerbate ischemic tissue damage and worsen clinical outcome in patients with stroke. Both alphabeta and gammadelta T cells are established mediators of tissue damage in stroke, and the role of dendritic cells (DCs) in inducing the early events of T cell activation and differentiation in stroke is not well understood. METHODS: In a murine model of experimental stroke, we defined the immune phenotype of infiltrating DC subsets based on flow cytometry of surface markers, the expression of ontogenetic markers, and cytokine levels. We used conditional DC depletion, bone marrow chimeric mice, and IL-23 (interleukin-23) receptor-deficient mice to further explore the functional role of DCs. RESULTS: We show that the ischemic brain was rapidly infiltrated by IRF4(+)/CD172a(+) conventional type 2 DCs and that conventional type 2 DCs were the most abundant subset in comparison with all other DC subsets. Twenty-four hours after ischemia onset, conventional type 2 DCs became the major source of IL-23, promoting neutrophil infiltration by induction of IL-17 (interleukin-17) in gammadelta T cells. Functionally, the depletion of CD11c(+) cells or the genetic disruption of the IL-23 signaling abrogated both IL-17 production in gammadelta T cells and neutrophil infiltration. Interruption of the IL-23/IL-17 cascade decreased infarct size and improved neurological outcome after stroke. CONCLUSIONS: Our results suggest a central role for interferon regulatory factor 4-positive IL-23-producing conventional DCs in the IL-17-dependent secondary tissue damage in stroke.
Association of Retinal Ganglion Cell Layer Thickness With Future Disease Activity in Patients With Clinically Isolated Syndrome
H. G. Zimmermann, B. Knier, T. Oberwahrenbrock, J. Behrens, C. Pfuhl, L. Aly, M. Kaminski, M. M. Hoshi, S. Specovius, R. M. Giess, M. Scheel, M. Muhlau, J. Bellmann-Strobl, K. Ruprecht, B. Hemmer, T. Korn, F. Paul, and A. U. Brandt
JAMA Neurol, 75(9): 1071-1079, 2018
Association of Retinal Ganglion Cell Layer Thickness With Future Disease Activity in Patients With Clinically Isolated Syndrome
H. G. Zimmermann, B. Knier, T. Oberwahrenbrock, J. Behrens, C. Pfuhl, L. Aly, M. Kaminski, M. M. Hoshi, S. Specovius, R. M. Giess, M. Scheel, M. Muhlau, J. Bellmann-Strobl, K. Ruprecht, B. Hemmer, T. Korn, F. Paul, and A. U. Brandt
Importance: Clinically isolated syndrome (CIS) describes a first clinical incident suggestive of multiple sclerosis (MS). Identifying patients with CIS who have a high risk of future disease activity and subsequent MS diagnosis is crucial for patient monitoring and the initiation of disease-modifying therapy. Objective: To investigate the association of retinal optical coherence tomography (OCT) results with future disease activity in patients with CIS. Design, Setting, and Participants: This prospective, longitudinal cohort study took place between January 2011 and May 2017 at 2 German tertiary referral centers. A total of 179 patients with CIS were screened (80 in Berlin and 99 in Munich). Patients underwent neurological examination, magnetic resonance imaging (MRI), and OCT. Only eyes with no previous optic neuritis were considered for OCT analysis. Main Outcomes and Measures: The primary outcome was not meeting the no evidence of disease activity (NEDA-3) criteria; secondary outcomes were MS diagnosis (by the 2010 McDonald criteria) and worsening of disability. The primary measure was OCT-derived ganglion cell and inner plexiform layer thickness; the secondary measures included peripapillary retinal nerve fiber layer thickness, inner nuclear layer thickness, and MRI-derived T2-weighted lesions. Results: A total of 97 of the 179 screened patients (54.2%) were enrolled in the study at a median of 93 (interquartile range [IQR], 62-161) days after a first demyelinating event. The median follow-up duration (Kaplan-Meier survival time) was 729 (IQR, 664-903) days. Of 97 patients with CIS (mean age 33.6 [7.9] years; 61 [62.9%] female), 58 (59%) did not meet NEDA-3 criteria during the follow-up period. A Kaplan-Meier analysis showed a significant probability difference in not meeting NEDA-3 criteria by ganglion cell and inner plexiform later thickness (thinnest vs thickest tertile: hazard ratio [HR], 3.33 [95% CI, 1.70-6.55; P < .001; log-rank P = .001). A follow-up diagnosis of MS was more likely for patients with low ganglion cell and inner plexiform layer thickness (thinnest vs thickest tertile: HR, 4.05 [95% CI, 1.93-8.50]; P < .001). Low peripapillary retinal nerve fiber layer thickness likewise indicated risk of not meeting NEDA-3 criteria (thinnest vs thickest tertile: HR, 2.46 [95% CI, 1.29-4.66]; P = .01; log-rank P = .02). Inner nuclear layer thickness and T2-weighted lesion count were not associated with not meeting NEDA-3 criteria. Conclusions and Relevance: Retinal ganglion cell and inner plexiform layer thickness might prove a valuable imaging marker for anticipating future disease activity and diagnosis of MS in patients with CIS, which can potentially support patient monitoring and initiation of disease-modifying therapy.
2017
Fatigue in multiple sclerosis: Associations with clinical, MRI and CSF parameters
V. Biberacher, P. Schmidt, R. C. Selter, V. Pernpeinter, M. C. Kowarik, B. Knier, D. Buck, M. M. Hoshi, T. Korn, A. Berthele, J. S. Kirschke, C. Zimmer, B. Hemmer, and M. Muhlau
Mult Scler, 2017
Fatigue in multiple sclerosis: Associations with clinical, MRI and CSF parameters
V. Biberacher, P. Schmidt, R. C. Selter, V. Pernpeinter, M. C. Kowarik, B. Knier, D. Buck, M. M. Hoshi, T. Korn, A. Berthele, J. S. Kirschke, C. Zimmer, B. Hemmer, and M. Muhlau
BACKGROUND: Damage of different brain structures has been related to fatigue. Alternatively, functional alterations of central nervous system (CNS) cells by the inflammatory milieu within the CNS may be responsible for the development of fatigue. AIM: To investigate the effect of structural brain damage and inflammatory cerebrospinal fluid (CSF) changes on fatigue in multiple sclerosis (MS). METHODS: We determined the association of different clinical, CSF and magnetic resonance imaging (MRI) parameters with prevalence and severity of fatigue, as measured by the Fatigue Scale for Motor and Cognitive Functions in 68 early MS patients (discovery cohort). We validated our findings in two MS cohorts: the MRI validation cohort ( N = 233) for the clinical and MRI parameters, and the CSF validation cohort ( N = 81) for the clinical and CSF parameters. RESULTS: Fatigue was associated with clinical disability. Fatigue did not correlate with any CSF parameter but correlated negatively with total and cortical grey matter volume. However, when controlling for Expanded Disability Status Scale (EDSS) in a multivariate model, these associations lost significance. CONCLUSION: Disability and disease duration best explain fatigue severity but none of the tested MRI or CSF parameter was reliably associated with fatigue.
Deletional tolerance prevents AQP4-directed autoimmunity in mice
A. L. Vogel, B. Knier, K. Lammens, S. R. Kalluri, T. Kuhlmann, J. L. Bennett, and T. Korn
Eur J Immunol, 47(3): 458-469, 2017
Deletional tolerance prevents AQP4-directed autoimmunity in mice
A. L. Vogel, B. Knier, K. Lammens, S. R. Kalluri, T. Kuhlmann, J. L. Bennett, and T. Korn
Neuromyelitis optica (NMO) is an autoimmune disorder of the central nervous system (CNS) mediated by antibodies to the water channel protein AQP4 expressed in astrocytes. The contribution of AQP4-specific T cells to the class switch recombination of pathogenic AQP4-specific antibodies and the inflammation of the blood-brain barrier is incompletely understood, as immunogenic naturally processed T-cell epitopes of AQP4 are unknown. By immunizing Aqp4(-/-) mice with full-length murine AQP4 protein followed by recall with overlapping peptides, we here identify AQP4(201-220) as the major immunogenic IA(b) -restricted epitope of AQP4. We show that WT mice do not harbor AQP4(201-220)-specific T-cell clones in their natural repertoire due to deletional tolerance. However, immunization with AQP4(201-220) of Rag1(-/-) mice reconstituted with the mature T-cell repertoire of Aqp4(-/-) mice elicits an encephalomyelitic syndrome. Similarly to the T-cell repertoire, the B-cell repertoire of WT mice is "purged" of AQP4-specific B cells, and robust serum responses to AQP4 are only mounted in Aqp4(-/-) mice. While AQP4(201-220)-specific T cells alone induce encephalomyelitis, NMO-specific lesional patterns in the CNS and the retina only occur in the additional presence of anti-AQP4 antibodies. Thus, failure of deletional T-cell and B-cell tolerance against AQP4 is a prerequisite for clinically manifest NMO.
Trans-presentation of IL-6 by dendritic cells is required for the priming of pathogenic TH17 cells
S. Heink, N. Yogev, C. Garbers, M. Herwerth, L. Aly, C. Gasperi, V. Husterer, A. L. Croxford, K. Moller-Hackbarth, H. S. Bartsch, K. Sotlar, S. Krebs, T. Regen, H. Blum, B. Hemmer, T. Misgeld, T. F. Wunderlich, J. Hidalgo, M. Oukka, S. Rose-John, M. Schmidt-Supprian, A. Waisman, and T. Korn
Nat Immunol, 18(1): 74-85, 2017
Trans-presentation of IL-6 by dendritic cells is required for the priming of pathogenic TH17 cells
S. Heink, N. Yogev, C. Garbers, M. Herwerth, L. Aly, C. Gasperi, V. Husterer, A. L. Croxford, K. Moller-Hackbarth, H. S. Bartsch, K. Sotlar, S. Krebs, T. Regen, H. Blum, B. Hemmer, T. Misgeld, T. F. Wunderlich, J. Hidalgo, M. Oukka, S. Rose-John, M. Schmidt-Supprian, A. Waisman, and T. Korn
The cellular sources of interleukin 6 (IL-6) that are relevant for differentiation of the TH17 subset of helper T cells remain unclear. Here we used a novel strategy for the conditional deletion of distinct IL-6-producing cell types to show that dendritic cells (DCs) positive for the signaling regulator Sirpα were essential for the generation of pathogenic TH17 cells. Using their IL-6 receptor α-chain (IL-6Rα), Sirpα+ DCs trans-presented IL-6 to T cells during the process of cognate interaction. While ambient IL-6 was sufficient to suppress the induction of expression of the transcription factor Foxp3 in T cells, trans-presentation of IL-6 by DC-bound IL-6Rα (called 'IL-6 cluster signaling' here) was needed to prevent premature induction of interferon-γ (IFN-γ) expression in T cells and to generate pathogenic TH17 cells in vivo. Our findings should guide therapeutic approaches for the treatment of TH17-cell-mediated autoimmune diseases.
IL-1beta and IL-23 Promote Extrathymic Commitment of CD27(+)CD122(-) gammadelta T Cells to gammadeltaT17 Cells
A. Muschaweckh, F. Petermann, and T. Korn
J Immunol, 199(8): 2668-2679, 2017
IL-1beta and IL-23 Promote Extrathymic Commitment of CD27(+)CD122(-) gammadelta T Cells to gammadeltaT17 Cells
A. Muschaweckh, F. Petermann, and T. Korn
gammadeltaT17 cells are a subset of gammadelta T cells committed to IL-17 production and are characterized by the expression of IL-23R and CCR6 and lack of CD27 expression. gammadeltaT17 cells are believed to arise within a narrow time window during prenatal thymic development. In agreement with this concept, we show in this study that adult Rag1(-/-) recipient mice of Il23r(gfp/+) (IL-23R reporter) bone marrow selectively lack IL-23R(+) gammadeltaT17 cells. Despite their absence in secondary lymphoid tissues during homeostasis, gammadeltaT17 cells emerge in bone marrow chimeric mice upon induction of skin inflammation by topical treatment with imiquimod cream (Aldara). We demonstrate that IL-1beta and IL-23 together are able to promote the development of bona fide gammadeltaT17 cells from peripheral CD122(-)IL-23R(-) gammadelta T cells, whereas CD122(+) gammadelta T cells fail to convert into gammadeltaT17 cells and remain stable IFN-gamma producers (gammadeltaT1 cells). IL-23 is instrumental in expanding extrathymically generated gammadeltaT17 cells. In particular, TCR-Vgamma4(+) chain-expressing CD122(-)IL-23R(-) gammadelta T cells are induced to express IL-23R and IL-17 outside the thymus during skin inflammation. In contrast, TCR-Vgamma1(+) gammadelta T cells largely resist this process because prior TCR engagement in the thymus has initiated their commitment to the gammadeltaT1 lineage. In summary, our data reveal that the peripheral pool of gammadelta T cells retains a considerable degree of plasticity because it harbors "naive" precursors, which can be induced to produce IL-17 and replenish peripheral niches that are usually occupied by thymus-derived gammadeltaT17 cells.
Effectors of Th1 and Th17 cells act on astrocytes and augment their neuroinflammatory properties
C. K. Prajeeth, J. Kronisch, R. Khorooshi, B. Knier, H. Toft-Hansen, V. Gudi, S. Floess, J. Huehn, T. Owens, T. Korn, and M. Stangel
J Neuroinflammation, 14(1): 204, 2017
Effectors of Th1 and Th17 cells act on astrocytes and augment their neuroinflammatory properties
C. K. Prajeeth, J. Kronisch, R. Khorooshi, B. Knier, H. Toft-Hansen, V. Gudi, S. Floess, J. Huehn, T. Owens, T. Korn, and M. Stangel
BACKGROUND: Autoreactive Th1 and Th17 cells are believed to mediate the pathology of multiple sclerosis in the central nervous system (CNS). Their interaction with microglia and astrocytes in the CNS is crucial for the regulation of the neuroinflammation. Previously, we have shown that only Th1 but not Th17 effectors activate microglia. However, it is not clear which cells are targets of Th17 effectors in the CNS. METHODS: To understand the effects driven by Th17 cells in the CNS, we induced experimental autoimmune encephalomyelitis in wild-type mice and CD4(+) T cell-specific integrin alpha4-deficient mice where trafficking of Th1 cells into the CNS was affected. We compared microglial and astrocyte response in the brain and spinal cord of these mice. We further treated astrocytes with supernatants from highly pure Th1 and Th17 cultures and assessed the messenger RNA expression of neurotrophic factors, cytokines and chemokines, using real-time PCR. Data obtained was analyzed using the Kruskal-Wallis test. RESULTS: We observed in alpha4-deficient mice weak microglial activation but comparable astrogliosis to that of wild-type mice in the regions of the brain populated with Th17 infiltrates, suggesting that Th17 cells target astrocytes and not microglia. In vitro, in response to supernatants from Th1 and Th17 cultures, astrocytes showed altered expression of neurotrophic factors, pro-inflammatory cytokines and chemokines. Furthermore, increased expression of chemokines in Th1- and Th17-treated astrocytes enhanced recruitment of microglia and transendothelial migration of Th17 cells in vitro. CONCLUSION: Our results demonstrate the delicate interaction between T cell subsets and glial cells and how they communicate to mediate their effects. Effectors of Th1 act on both microglia and astrocytes whereas Th17 effectors preferentially target astrocytes to promote neuroinflammation.
Microstructural visual system changes in AQP4-antibody-seropositive NMOSD
F. C. Oertel, J. Kuchling, H. Zimmermann, C. Chien, F. Schmidt, B. Knier, J. Bellmann-Strobl, T. Korn, M. Scheel, A. Klistorner, K. Ruprecht, F. Paul, and A. U. Brandt
Neurol Neuroimmunol Neuroinflamm, 4(3): e334, 2017
Microstructural visual system changes in AQP4-antibody-seropositive NMOSD
F. C. Oertel, J. Kuchling, H. Zimmermann, C. Chien, F. Schmidt, B. Knier, J. Bellmann-Strobl, T. Korn, M. Scheel, A. Klistorner, K. Ruprecht, F. Paul, and A. U. Brandt
OBJECTIVE: To trace microstructural changes in patients with aquaporin-4 antibody (AQP4-ab)-seropositive neuromyelitis optica spectrum disorders (NMOSDs) by investigating the afferent visual system in patients without clinically overt visual symptoms or visual pathway lesions. METHODS: Of 51 screened patients with NMOSD from a longitudinal observational cohort study, we compared 6 AQP4-ab-seropositive NMOSD patients with longitudinally extensive transverse myelitis (LETM) but no history of optic neuritis (ON) or other bout (NMOSD-LETM) to 19 AQP4-ab-seropositive NMOSD patients with previous ON (NMOSD-ON) and 26 healthy controls (HCs). Foveal thickness (FT), peripapillary retinal nerve fiber layer (pRNFL) thickness, and ganglion cell and inner plexiform layer (GCIPL) thickness were measured with optical coherence tomography (OCT). Microstructural changes in the optic radiation (OR) were investigated using diffusion tensor imaging (DTI). Visual function was determined by high-contrast visual acuity (VA). OCT results were confirmed in a second independent cohort. RESULTS: FT was reduced in both patients with NMOSD-LETM (p = 3.52e(-14)) and NMOSD-ON (p = 1.24e(-16)) in comparison with HC. Probabilistic tractography showed fractional anisotropy reduction in the OR in patients with NMOSD-LETM (p = 0.046) and NMOSD-ON (p = 1.50e(-5)) compared with HC. Only patients with NMOSD-ON but not NMOSD-LETM showed neuroaxonal damage in the form of pRNFL and GCIPL thinning. VA was normal in patients with NMOSD-LETM and was not associated with OCT or DTI parameters. CONCLUSIONS: Patients with AQP4-ab-seropositive NMOSD without a history of ON have microstructural changes in the afferent visual system. The localization of retinal changes around the Muller-cell rich fovea supports a retinal astrocytopathy.
Association of Retinal Architecture, Intrathecal Immunity, and Clinical Course in Multiple Sclerosis
B. Knier, G. Leppenetier, C. Wetzlmair, L. Aly, M. M. Hoshi, V. Pernpeintner, V. Biberacher, A. Berthele, M. Muhlau, C. Zimmer, B. Hemmer, and T. Korn
JAMA Neurol, 74(7): 847-856, 2017
Association of Retinal Architecture, Intrathecal Immunity, and Clinical Course in Multiple Sclerosis
B. Knier, G. Leppenetier, C. Wetzlmair, L. Aly, M. M. Hoshi, V. Pernpeintner, V. Biberacher, A. Berthele, M. Muhlau, C. Zimmer, B. Hemmer, and T. Korn
Importance: Biomarkers to estimate long-term outcomes in patients with multiple sclerosis (MS) and to assign patients to individual treatment regimens are urgently needed. Objective: To assess whether retinal layer volumes are correlated with immune cell subsets and immunoglobulin indices in the cerebrospinal fluid and whether retinal layer volumes alone or in combination with intrathecal variables are associated with worsening of disease in patients with relapsing-remitting MS. Design, Setting, and Participants: This observational cohort study included 312 patients with relapsing-remitting MS in 2 independent cohorts (72 patients with short disease duration [cohort 1] and 240 patients with longer disease duration [cohort 2]) treated at a single German university hospital from April 15, 2013, through November 11, 2015. Main Outcomes and Measures: The common ganglion cell and inner plexiform layer (GCIPL) and inner nuclear layer (INL) volumes were tested for association with the immunoglobulin indices and the frequencies of immune cells in the cerebrospinal fluid (including B cells, T cells, and natural killer cells) (cohort 1). Volumes of GCIPL alone (cohorts 1 and 2) or GCIPL corrected for intrathecal B-cell frequencies (cohort 1) were tested for their association with worsening disability. Results: A total of 312 patients (212 women [67.9%] and 100 men [32.1%]; median age, 34.0 years [interquartile range (IQR), 28.0-42.0 years]) were available for analysis. In cohort 1 (50 women [69.4%] and 22 men [30.6%]; median age, 31.0 years [IQR, 26.3-38.3 years]), with short disease durations (median, 1.0 months [IQR, 1.0-2.0 months]), low GCIPL volumes were associated with increased intrathecal B-cell frequencies (median, 1.96% [IQR, 1.45%-4.20%]) and intrathecal IgG synthesis (median cerebrospinal fluid/serum IgG index, 0.78 [IQR, 0.53-1.07]). The INL volumes correlated with the frequencies of intrathecal CD56bright natural killer cells (r = 0.28; P = .007). Individuals with low GCIPL volumes (<1.99 mm3) had a 6.4-fold risk for worsening disability during follow-up compared with patients with higher GCIPL values (95% CI, 1.7-24.2; P = .007). This finding was reproduced in cohort 2 (162 women [67.5%] and 78 men [32.5%]; median age, 34.0 years [IQR, 29.0-42.0 years]) consisting of patients with longer disease durations (median, 36.0 months [IQR, 21.0-60.0 months]) (hazard ratio, 2.4; 95% CI, 1.2-4.8; P = .02). In both cohorts, INL volumes correlated with the prospective increase in T2 lesion load and the number of gadolinium-enhancing lesions. Conclusions and Relevance: Retinal layers reflect different aspects of disease activity during MS. Loss of GCIPL is associated with intrathecal B-cell immunity and constitutes an independent risk factor for worsening disability, whereas high INL volumes are associated with activity on magnetic resonance imaging in the brain parenchyma. Thus, retinal optical coherence tomography might be a means to support stratification of patients with MS for different therapeutic regimens.
2016
Optical coherence tomography indicates disease activity prior to clinical onset of central nervous system demyelination
B. Knier, A. Berthele, D. Buck, P. Schmidt, C. Zimmer, M. Muhlau, B. Hemmer, and T. Korn
Mult Scler, 22(7): 893-900, 2016
Optical coherence tomography indicates disease activity prior to clinical onset of central nervous system demyelination
B. Knier, A. Berthele, D. Buck, P. Schmidt, C. Zimmer, M. Muhlau, B. Hemmer, and T. Korn
BACKGROUND: Establishing biomarkers for predicting disease activity in demyelinating disease of the central nervous system is crucial for designing appropriate disease modifiying treatment strategies. OBJECTIVE: To investigate retinal findings and disease activity in patients with radiologically isolated and clinically isolated syndromes. METHODS: We performed retinal optical coherence tomography and cerebral magnetic resonance imaging in healthy control individuals (n=19), in individuals with non-specific white matter lesions (n=18), and in patients with clinically isolated syndromes (n=18) and radiologically isolated syndromes (n=20). RESULTS: Reduced volume of retinal nerve fibre layer and increased volume of inner nuclear layer at baseline correlated with subsequent disease activity as measured by an increase in cerebral T2 lesion load in patients with radiologically isolated syndromes. Reduced volume of retinal nerve fibre layer and increased volumes of inner and outer nuclear layer were associated with progression into multiple sclerosis in patients with clinically isolated syndromes. CONCLUSION: Patients with radiologically and clinically isolated syndromes behave similarly concerning paraclinical disease activity in cerebral magnetic resonance imaging. In both conditions, reduction of retinal nerve fibre layer and increased inner nuclear layer and outer nuclear layer volumes predict disease activity and are associated with progression into multiple sclerosis.
Retinal inner nuclear layer volume reflects response to immunotherapy in multiple sclerosis
B. Knier, P. Schmidt, L. Aly, D. Buck, A. Berthele, M. Muhlau, C. Zimmer, B. Hemmer, and T. Korn
Brain, 2016
Retinal inner nuclear layer volume reflects response to immunotherapy in multiple sclerosis
B. Knier, P. Schmidt, L. Aly, D. Buck, A. Berthele, M. Muhlau, C. Zimmer, B. Hemmer, and T. Korn
Assessment of inflammatory disease activity during multiple sclerosis is crucial for selecting appropriate disease-modifying therapies. Previous studies suggested that the retinal inner nuclear layer reflects inflammatory disease severity within the central nervous system. In our study, correlations of longitudinal retinal layer changes as measured by retinal optical coherence tomography with ongoing disease activity were evaluated in 108 multiple sclerosis patients without therapy, on first-line therapy, or on second-line therapy. Healthy subjects served as controls. Inner nuclear layer volume at baseline correlated positively with paraclinical disease activity during the subsequent 12 months. Longitudinal thinning of the inner nuclear layer and thickening of total macular volume were associated with reduced inflammatory disease activity. Reduction in inner nuclear layer volume after 1 year indicated efficient control of inflammatory disease activity including 'no evidence of disease activity'. In conclusion, the retinal inner nuclear layer could serve as biomarker to monitor sustained control of autoimmune central nervous system inflammation by therapeutic interventions.
Antigen-dependent competition shapes the local repertoire of tissue-resident memory CD8+ T cells
A. Muschaweckh, V. R. Buchholz, A. Fellenzer, C. Hessel, P. A. Konig, S. Tao, R. Tao, M. Heikenwalder, D. H. Busch, T. Korn, W. Kastenmuller, I. Drexler, and G. Gasteiger
J Exp Med, 213(13): 3075-3086, 2016
Antigen-dependent competition shapes the local repertoire of tissue-resident memory CD8+ T cells
A. Muschaweckh, V. R. Buchholz, A. Fellenzer, C. Hessel, P. A. Konig, S. Tao, R. Tao, M. Heikenwalder, D. H. Busch, T. Korn, W. Kastenmuller, I. Drexler, and G. Gasteiger
Tissue-resident memory CD8+ T cells (TRM) constitute a major component of the immune-surveillance system in nonlymphoid organs. Local, noncognate factors are both necessary and sufficient to support the programming of TRM cell fate in tissue-infiltrating T cells. Recent evidence suggests that TCR signals received in infected nonlymphoid tissues additionally contribute to TRM cell formation. Here, we asked how antigen-dependent pathways influence the generation of skin-resident memory T cells that arise from a polyclonal repertoire of cells induced by infection with an antigenically complex virus and recombinant vaccine vector. We found that CD8+ T cells of different specificities underwent antigen-dependent competition in the infected tissue, which shaped the composition of the local pool of TRM cells. This local cross-competition was active for T cells recognizing antigens that are coexpressed by infected cells. In contrast, TRM cell development remained largely undisturbed by the presence of potential competitors when antigens expressed in the same tissue were segregated through infection with antigenically distinct viral quasispecies. Functionally, local cross-competition might serve as a gatekeeping mechanism to regulate access to the resident memory niche and to fine-tune the local repertoire of antiviral TRM cells.
Fulminant Central Nervous System Nocardiosis in a Patient Treated With Alemtuzumab for Relapsing-Remitting Multiple Sclerosis
H. Penkert, C. Delbridge, N. Wantia, B. Wiestler, and T. Korn
JAMA Neurol, 73(6): 757-9, 2016
Brain-resident memory T cells represent an autonomous cytotoxic barrier to viral infection
K. Steinbach, I. Vincenti, M. Kreutzfeldt, N. Page, A. Muschaweckh, I. Wagner, I. Drexler, D. Pinschewer, T. Korn, and D. Merkler
J Exp Med, 213(8): 1571-87, 2016
Brain-resident memory T cells represent an autonomous cytotoxic barrier to viral infection
K. Steinbach, I. Vincenti, M. Kreutzfeldt, N. Page, A. Muschaweckh, I. Wagner, I. Drexler, D. Pinschewer, T. Korn, and D. Merkler
Tissue-resident memory T cells (TRM) persist at sites of prior infection and have been shown to enhance pathogen clearance by recruiting circulating immune cells and providing bystander activation. Here, we characterize the functioning of brain-resident memory T cells (bTRM) in an animal model of viral infection. bTRM were subject to spontaneous homeostatic proliferation and were largely refractory to systemic immune cell depletion. After viral reinfection in mice, bTRM rapidly acquired cytotoxic effector function and prevented fatal brain infection, even in the absence of circulating CD8(+) memory T cells. Presentation of cognate antigen on MHC-I was essential for bTRM-mediated protective immunity, which involved perforin- and IFN-γ-dependent effector mechanisms. These findings identify bTRM as an organ-autonomous defense system serving as a paradigm for TRM functioning as a self-sufficient first line of adaptive immunity.
Interleukin-23-Dependent γ/δ T Cells Produce Interleukin-17 and Accumulate in the Enthesis, Aortic Valve, and Ciliary Body in Mice
A. Reinhardt, T. Yevsa, T. Worbs, S. Lienenklaus, I. Sandrock, L. Oberdorfer, T. Korn, S. Weiss, R. Forster, and I. Prinz
Arthritis Rheumatol, 68(10): 2476-86, 2016
Interleukin-23-Dependent γ/δ T Cells Produce Interleukin-17 and Accumulate in the Enthesis, Aortic Valve, and Ciliary Body in Mice
A. Reinhardt, T. Yevsa, T. Worbs, S. Lienenklaus, I. Sandrock, L. Oberdorfer, T. Korn, S. Weiss, R. Forster, and I. Prinz
OBJECTIVE: The spondyloarthritides (SpA) are a group of rheumatic diseases characterized by ossification and inflammation of entheseal tissue, the region where tendon attaches to bone. Interleukin-23 (IL-23) is involved in the pathogenesis of SpA by acting on IL-23 receptor (IL-23R) expressed on enthesis-resident lymphocytes. Upon IL-23 binding, CD3+CD4-CD8- tissue-resident lymphocytes secrete IL-17A and IL-22, leading to inflammation, bone loss, and ossification. Knowledge about enthesis-resident lymphocytes remains fragmentary, and the contribution of entheseal γ/δ T cells in particular is not clear. This study was undertaken to investigate the presence of γ/δ T cells in the enthesis. METHODS: We used 2-photon microscopy and flow cytometry to analyze entheseal lymphocytes from C57BL/6, Tcrd-H2BeGFP, Rorc-GFP, and IL-23R-eGFP mice. To analyze entheseal γ/δ T cells in IL-23-induced inflammation, Tcrd-H2BeGFP mice were crossed with mice of the susceptible B10.RIII background. Hydrodynamic injection of IL-23 minicircle DNA was performed for overexpression of IL-23 and induction of inflammation. Light-sheet fluorescence microscopy was used to visualize arthritic inflammation. RESULTS: Activated Vγ6+CD27- γ/δ T cells were abundant in uninflamed entheseal tissue and constituted the large majority of retinoic acid receptor-related orphan nuclear receptor γt (RORγt)+IL-23R+ enthesis-resident lymphocytes. Fetal thymus-dependent γ/δ T cells were the main source of IL-17A at the enthesis. Under inflammatory conditions, γ/δ T cells increased in number at the Achilles tendon enthesis, aortic root, and adjacent to the ciliary body. CONCLUSION: Entheseal γ/δ T cells are derived from fetal thymus and are maintained as self-renewing tissue-resident cells. As main IL-17A producers within tissues exposed to mechanical stress including enthesis, γ/δ T cells are key players in the pathogenesis of IL-23-induced local inflammation.
[Monitoring of blood parameters under course-modified MS therapy : Substance-specific relevance and current recommendations for action]
L. Klotz, A. Berthele, W. Bruck, A. Chan, P. Flachenecker, R. Gold, A. Haghikia, K. Hellwig, B. Hemmer, R. Hohlfeld, T. Korn, T. Kumpfel, M. Lang, V. Limmroth, R. A. Linker, U. Meier, S. G. Meuth, F. Paul, A. Salmen, M. Stangel, B. Tackenberg, H. Tumani, C. Warnke, M. S. Weber, T. Ziemssen, F. Zipp, and H. Wiendl
Nervenarzt, 87(6): 645-59, 2016
[Monitoring of blood parameters under course-modified MS therapy : Substance-specific relevance and current recommendations for action]
L. Klotz, A. Berthele, W. Bruck, A. Chan, P. Flachenecker, R. Gold, A. Haghikia, K. Hellwig, B. Hemmer, R. Hohlfeld, T. Korn, T. Kumpfel, M. Lang, V. Limmroth, R. A. Linker, U. Meier, S. G. Meuth, F. Paul, A. Salmen, M. Stangel, B. Tackenberg, H. Tumani, C. Warnke, M. S. Weber, T. Ziemssen, F. Zipp, and H. Wiendl
With the approval of various substances for the immunotherapy of multiple sclerosis (MS), treatment possibilities have improved significantly over the last few years. Indeed, the choice of individually tailored preparations and treatment monitoring for the treating doctor is becoming increasingly more complex. This is particularly applicable for monitoring for a treatment-induced compromise of the immune system. The following article by members of the German Multiple Sclerosis Skills Network (KKNMS) and the task force "Provision Structures and Therapeutics" summarizes the practical recommendations for approved immunotherapy for mild to moderate and for (highly) active courses of MS. The focus is on elucidating the substance-specific relevance of particular laboratory parameters with regard to the mechanism of action and the side effects profile. To enable appropriate action to be taken in clinical practice, any blood work changes that can be expected, in addition to any undesirable laboratory findings and their causes and relevance, should be elucidated.
Blood coagulation factor XII drives adaptive immunity during neuroinflammation via CD87-mediated modulation of dendritic cells
K. Gobel, S. Pankratz, C. M. Asaridou, A. M. Herrmann, S. Bittner, M. Merker, T. Ruck, S. Glumm, F. Langhauser, P. Kraft, T. F. Krug, J. Breuer, M. Herold, C. C. Gross, D. Beckmann, A. Korb-Pap, M. K. Schuhmann, S. Kuerten, I. Mitroulis, C. Ruppert, M. W. Nolte, C. Panousis, L. Klotz, B. Kehrel, T. Korn, H. F. Langer, T. Pap, B. Nieswandt, H. Wiendl, T. Chavakis, C. Kleinschnitz, and S. G. Meuth
Nat Commun, 7: 11626, 2016
Blood coagulation factor XII drives adaptive immunity during neuroinflammation via CD87-mediated modulation of dendritic cells
K. Gobel, S. Pankratz, C. M. Asaridou, A. M. Herrmann, S. Bittner, M. Merker, T. Ruck, S. Glumm, F. Langhauser, P. Kraft, T. F. Krug, J. Breuer, M. Herold, C. C. Gross, D. Beckmann, A. Korb-Pap, M. K. Schuhmann, S. Kuerten, I. Mitroulis, C. Ruppert, M. W. Nolte, C. Panousis, L. Klotz, B. Kehrel, T. Korn, H. F. Langer, T. Pap, B. Nieswandt, H. Wiendl, T. Chavakis, C. Kleinschnitz, and S. G. Meuth
Aberrant immune responses represent the underlying cause of central nervous system (CNS) autoimmunity, including multiple sclerosis (MS). Recent evidence implicated the crosstalk between coagulation and immunity in CNS autoimmunity. Here we identify coagulation factor XII (FXII), the initiator of the intrinsic coagulation cascade and the kallikrein-kinin system, as a specific immune cell modulator. High levels of FXII activity are present in the plasma of MS patients during relapse. Deficiency or pharmacologic blockade of FXII renders mice less susceptible to experimental autoimmune encephalomyelitis (a model of MS) and is accompanied by reduced numbers of interleukin-17A-producing T cells. Immune activation by FXII is mediated by dendritic cells in a CD87-dependent manner and involves alterations in intracellular cyclic AMP formation. Our study demonstrates that a member of the plasmatic coagulation cascade is a key mediator of autoimmunity. FXII inhibition may provide a strategy to combat MS and other immune-related disorders.
2015
Nanoparticle-based autoantigen delivery to Treg-inducing liver sinusoidal endothelial cells enables control of autoimmunity in mice
A. Carambia, B. Freund, D. Schwinge, O. T. Bruns, S. C. Salmen, H. Ittrich, R. Reimer, M. Heine, S. Huber, C. Waurisch, A. Eychmuller, D. C. Wraith, T. Korn, P. Nielsen, H. Weller, C. Schramm, S. Luth, A. W. Lohse, J. Heeren, and J. Herkel
J Hepatol, 62(6): 1349-56, 2015
Nanoparticle-based autoantigen delivery to Treg-inducing liver sinusoidal endothelial cells enables control of autoimmunity in mice
A. Carambia, B. Freund, D. Schwinge, O. T. Bruns, S. C. Salmen, H. Ittrich, R. Reimer, M. Heine, S. Huber, C. Waurisch, A. Eychmuller, D. C. Wraith, T. Korn, P. Nielsen, H. Weller, C. Schramm, S. Luth, A. W. Lohse, J. Heeren, and J. Herkel
BACKGROUND & AIMS: It is well-known that the liver can induce immune tolerance, yet this knowledge could, thus far, not be translated into effective treatments for autoimmune diseases. We have previously shown that liver sinusoidal endothelial cells (LSECs) could substantially contribute to hepatic tolerance through their ability to induce CD4+ Foxp3+ regulatory T cells (Tregs). Here, we explored whether the Treg-inducing potential of LSECs could be harnessed for the treatment of autoimmune disease. METHODS: We engineered a polymeric nanoparticle (NP) carrier for the selective delivery of autoantigen peptides to LSECs in vivo. In the well-characterized autoimmune disease model of experimental autoimmune encephalomyelitis (EAE), we investigated whether administration of LSEC-targeting autoantigen peptide-loaded NPs could protect mice from autoimmune disease. RESULTS: We demonstrate that NP-based autoantigen delivery to LSECs could completely and permanently prevent the onset of clinical EAE. More importantly, in a therapeutic approach, mice with already established EAE improved rapidly and substantially following administration of a single dose of autoantigen peptide-loaded NPs, whereas the control group deteriorated. Treatment efficacy seemed to depend on Tregs. The Treg frequencies in the spleens of mice treated with autoantigen peptide-loaded NPs were significantly higher than those in vehicle-treated mice. Moreover, NP-mediated disease control was abrogated after Treg depletion by repeated administration of Treg-depleting antibody. CONCLUSION: Our findings provide proof of principle that the selective delivery of autoantigen peptides to LSECs by NPs can induce antigen-specific Tregs and enable effective treatment of autoimmune disease. These findings highlight the importance of Treg induction by LSECs for immune tolerance.
Neutralizing IL-17 protects the optic nerve from autoimmune pathology and prevents retinal nerve fiber layer atrophy during experimental autoimmune encephalomyelitis
B. Knier, V. Rothhammer, S. Heink, O. Puk, J. Graw, B. Hemmer, and T. Korn
J Autoimmun, 56: 34-44, 2015
Neutralizing IL-17 protects the optic nerve from autoimmune pathology and prevents retinal nerve fiber layer atrophy during experimental autoimmune encephalomyelitis
B. Knier, V. Rothhammer, S. Heink, O. Puk, J. Graw, B. Hemmer, and T. Korn
Optic neuritis is a common inflammatory manifestation of multiple sclerosis (MS). In experimental autoimmune encephalomyelitis (EAE), the optic nerve is affected as well. Here, we investigated whether autoimmune inflammation in the optic nerve is distinct from inflammation in other parts of the central nervous system (CNS). In our study, inflammatory infiltrates in the optic nerve and the brain were characterized by a high fraction of Ly6G(+) granulocytes whereas in the spinal cord, macrophage infiltrates were predominant. At the peak of disease, IL-17 mRNA abundance was highest in the optic nerve as compared with other parts of the CNS. The ratio of IL-17 vs IFN-γ producing CD4(+) T cells was higher in the optic nerve and brain than in the spinal cord and more effector CD4(+) T cells were committed to the Th17 transcriptional program in the optic nerve than in the spinal cord. IL-17 producing γδ T cells but rather not Ly6G(+) granulocytes themselves contributed to IL-17 production. Optical coherence tomography (OCT) studies on murine eyes revealed a decline in thickness of the retinal nerve fiber layer (RNFL) and the common layer of ganglion cells and inner plexiform layer (GCL+) after the recovery from motor symptoms indicating that autoimmune inflammation induced a significant atrophy of optic nerve fibers during EAE. Neutralization of IL-17 by treatment with anti-IL-17 antibodies reduced but did not abrogate motor symptoms of EAE. However, RNFL and GCL+ atrophy were completely prevented by blocking IL-17. Thus, the optic nerve compartment is particularly prone to support IL-17 mediated inflammatory responses during CNS autoimmunity and structural integrity of the retina can be preserved by neutralizing IL-17.
2014
TGF-β-dependent induction of CD4(+)CD25(+)Foxp3(+) Tregs by liver sinusoidal endothelial cells
A. Carambia, B. Freund, D. Schwinge, M. Heine, A. Laschtowitz, S. Huber, D. C. Wraith, T. Korn, C. Schramm, A. W. Lohse, J. Heeren, and J. Herkel
J Hepatol, 61(3): 594-9, 2014
TGF-β-dependent induction of CD4(+)CD25(+)Foxp3(+) Tregs by liver sinusoidal endothelial cells
A. Carambia, B. Freund, D. Schwinge, M. Heine, A. Laschtowitz, S. Huber, D. C. Wraith, T. Korn, C. Schramm, A. W. Lohse, J. Heeren, and J. Herkel
BACKGROUND & AIMS: CD4(+) CD25(+) Foxp3(+) regulatory T cells (Tregs) have a profound ability to control immune responses. We have previously shown that the liver is a major source of peripherally induced Tregs. Here, we investigate the liver cell types and molecular mechanisms responsible for hepatic Treg induction. METHODS: To assess the Treg-inducing potential of liver resident antigen-presenting cell types, we studied the conversion of Foxp3(-) non-Tregs into Foxp3(+) Tregs induced by liver dendritic cells (DCs), liver sinusoidal endothelial cells (LSECs), or Kupffer cells (KCs). The dependency of Treg induction on TGF-β was tested in Treg conversion assays using T cells with reduced TGF-β sensitivity. The suppressive potential of liver cell-induced Tregs was assessed by an in vitro suppression assay and in vivo, in the model of experimental autoimmune encephalomyelitis (EAE). RESULTS: All tested liver cell types were capable of inducing Foxp3(+) Tregs; however, LSECs were most efficient in inducing Tregs. Treg-induction was antigen-specific and depended on TGF-β. LSECs featured membrane-bound LAP/TGF-β and the anchor molecule GARP, which is required for tethering LAP/TGF-β to the cell membrane. LSEC-induced Tregs suppressed proliferation and cytokine secretion of effector T cells in vitro. LSEC-induced Tregs were also functional suppressors in vivo, as neuroantigen-specific Tregs induced by LSECs were able to suppress EAE. CONCLUSIONS: We demonstrate that LSECs are the major liver cell type responsible for TGF-β dependent hepatic Treg induction. The extraordinary capacity of LSECs to induce Tregs was associated with their unique ability to tether TGF-β to their membrane.
Uncoupling Malt1 threshold function from paracaspase activity results in destructive autoimmune inflammation
A. Gewies, O. Gorka, H. Bergmann, K. Pechloff, F. Petermann, K. M. Jeltsch, M. Rudelius, M. Kriegsmann, W. Weichert, M. Horsch, J. Beckers, W. Wurst, M. Heikenwalder, T. Korn, V. Heissmeyer, and J. Ruland
Cell Rep, 9(4): 1292-305, 2014
Uncoupling Malt1 threshold function from paracaspase activity results in destructive autoimmune inflammation
A. Gewies, O. Gorka, H. Bergmann, K. Pechloff, F. Petermann, K. M. Jeltsch, M. Rudelius, M. Kriegsmann, W. Weichert, M. Horsch, J. Beckers, W. Wurst, M. Heikenwalder, T. Korn, V. Heissmeyer, and J. Ruland
The paracaspase Malt1 is a central regulator of antigen receptor signaling that is frequently mutated in human lymphoma. As a scaffold, it assembles protein complexes for NF-kappaB activation, and its proteolytic domain cleaves negative NF-kappaB regulators for signal enforcement. Still, the physiological functions of Malt1-protease are unknown. We demonstrate that targeted Malt1-paracaspase inactivation induces a lethal inflammatory syndrome with lymphocyte-dependent neurodegeneration in vivo. Paracaspase activity is essential for regulatory T cell (Treg) and innate-like B cell development, but it is largely dispensable for overcoming Malt1-dependent thresholds for lymphocyte activation. In addition to NF-kappaB inhibitors, Malt1 cleaves an entire set of mRNA stability regulators, including Roquin-1, Roquin-2, and Regnase-1, and paracaspase inactivation results in excessive interferon γ (IFNγ) production by effector lymphocytes that drive pathology. Together, our results reveal distinct threshold and modulatory functions of Malt1 that differentially control lymphocyte differentiation and activation pathways and demonstrate that selective paracaspase blockage skews systemic immunity toward destructive autoinflammation.
IL-27 and IL-12 oppose pro-inflammatory IL-23 in CD4+ T cells by inducing Blimp1
C. Heinemann, S. Heink, F. Petermann, A. Vasanthakumar, V. Rothhammer, E. Doorduijn, M. Mitsdoerffer, C. Sie, O. Prazeres da Costa, T. Buch, B. Hemmer, M. Oukka, A. Kallies, and T. Korn
Nat Commun, 5: 3770, 2014
IL-27 and IL-12 oppose pro-inflammatory IL-23 in CD4+ T cells by inducing Blimp1
C. Heinemann, S. Heink, F. Petermann, A. Vasanthakumar, V. Rothhammer, E. Doorduijn, M. Mitsdoerffer, C. Sie, O. Prazeres da Costa, T. Buch, B. Hemmer, M. Oukka, A. Kallies, and T. Korn
Central nervous system (CNS) autoimmunity is regulated by the balance of pro-inflammatory cytokines and IL-10. Here we identify the transcriptional regulator Blimp1 as crucial to induce IL-10 in inflammatory T helper cells. Pre-committed Th17 cells respond to IL-27 and IL-12 by upregulating Blimp1 and adopt a Tr-1-like phenotype characterized by IL-10 and IFN-γ production. Accordingly, Blimp1-deficient effector T cells fail to produce IL-10, and deficiency in Tr-1 cell function leads to uncontrolled Th17 cell-driven CNS pathology without the need to stabilize the Th17 phenotype with IL-23. IL-23 counteracts IL-27 and IL-12-mediated effects on Tr-1-development reinforcing the pro-inflammatory phenotype of Th17 cells. Thus, the balance of IL-23 vs IL-12/IL-27 signals into CD4(+) effector T cells determines whether tissue inflammation is perpetuated or resolves.
α4-integrins control viral meningoencephalitis through differential recruitment of T helper cell subsets
V. Rothhammer, A. Muschaweckh, G. Gasteiger, F. Petermann, S. Heink, D. H. Busch, M. Heikenwalder, B. Hemmer, I. Drexler, and T. Korn
Acta Neuropathol Commun, 2: 27, 2014
α4-integrins control viral meningoencephalitis through differential recruitment of T helper cell subsets
V. Rothhammer, A. Muschaweckh, G. Gasteiger, F. Petermann, S. Heink, D. H. Busch, M. Heikenwalder, B. Hemmer, I. Drexler, and T. Korn
INTRODUCTION: Natalizumab blocks α4-integrins and is a prototypic agent for a series of anti-inflammatory drugs that impair trafficking of immune cells into the CNS. However, modulation of the access of immune cells to the CNS is associated with impaired immune surveillance and detrimental viral infections of the CNS. Here, we explored the potency of cellular immune responses within the CNS to protect against viral encephalitis in mice with T cell conditional disruption of VLA-4 integrin (α4β1) expression. RESULTS: While VLA-4 expression in virus specific Th1 cells is non-redundant for their ability to access the CNS, α4-integrin deficient Th17 cells enter the CNS compartment and generate an inflammatory milieu upon intrathecal vaccinia virus (VV) infection. However, in contrast to Th1 cells that can adopt direct cytotoxic properties, Th17 cells fail to clear the virus due to insufficient Eomes induced perforin-1 expression. CONCLUSION: The quality of the intrathecal cellular antiviral response under conditions of impaired VLA-4 function jeopardizes host protection. Our functional in vivo data extend our mechanistic understanding of anti-viral immunity in the CNS and help to estimate the risk potential of upcoming therapeutic agents that target the trafficking of immune cells into distinct anatomical compartments.
Differential loss of KIR4.1 immunoreactivity in multiple sclerosis lesions
L. Schirmer, R. Srivastava, S. R. Kalluri, S. Bottinger, M. Herwerth, D. Carassiti, B. Srivastava, J. Gempt, J. Schlegel, T. Kuhlmann, T. Korn, R. Reynolds, and B. Hemmer
Ann Neurol, 75(6): 810-28, 2014
Differential loss of KIR4.1 immunoreactivity in multiple sclerosis lesions
L. Schirmer, R. Srivastava, S. R. Kalluri, S. Bottinger, M. Herwerth, D. Carassiti, B. Srivastava, J. Gempt, J. Schlegel, T. Kuhlmann, T. Korn, R. Reynolds, and B. Hemmer
OBJECTIVE: Serum antibodies against the glial potassium channel KIR4.1 are found in a subpopulation of multiple sclerosis (MS) patients. Little is known about the expression of KIR4.1 in human normal brain tissue and in MS lesions. METHODS: We analyzed the expression pattern of KIR4.1 in normal brain tissue and MS lesions of the subcortical white matter by immunohistochemistry. Markers of related glial proteins, myelin, and inflammatory cells were analyzed in parallel. RESULTS: KIR4.1 is expressed in oligodendrocytes and astrocytes in the adult human brain. In oligodendrocytes, KIR4.1 appears as a homotetramer channel, in astrocytes as homo- and heterotetramer channels together with KIR5.1. In acute MS lesions, KIR4.1 immunoreactivity (IR) was differentially lost on periplaque oligodendrocytes and perivascular astrocytes. In part of acute lesions, complement activation, apoptotic KIR4.1(+) glial cells, and phagocytes containing KIR4.1(+) fragments accompanied loss of glial KIR4.1 IR. Periplaque reactive astrocytes showed enhanced IR for both KIR4.1 and KIR5.1. In chronic active MS lesions, apart from a general loss of oligodendrocytes in the demyelinated area, we observed a decrease of astroglial KIR4.1 but not glial fibrillary acidic protein IR. In chronic inactive and remyelinating MS lesions, KIR4.1 IR was restored on astrocytes and found in a subset of presumably new myelinating oligodendrocytes. INTERPRETATION: The expression profile of KIR4.1 in glial cells and stage-dependent alterations of KIR4.1 IR in MS lesions are compatible with an immune response against KIR4.1 at least in a subset of MS patients.
Continuous T cell receptor signals maintain a functional regulatory T cell pool
J. C. Vahl, C. Drees, K. Heger, S. Heink, J. C. Fischer, J. Nedjic, N. Ohkura, H. Morikawa, H. Poeck, S. Schallenberg, D. Riess, M. Y. Hein, T. Buch, B. Polic, A. Schonle, R. Zeiser, A. Schmitt-Graff, K. Kretschmer, L. Klein, T. Korn, S. Sakaguchi, and M. Schmidt-Supprian
Immunity, 41(5): 722-36, 2014
Continuous T cell receptor signals maintain a functional regulatory T cell pool
J. C. Vahl, C. Drees, K. Heger, S. Heink, J. C. Fischer, J. Nedjic, N. Ohkura, H. Morikawa, H. Poeck, S. Schallenberg, D. Riess, M. Y. Hein, T. Buch, B. Polic, A. Schonle, R. Zeiser, A. Schmitt-Graff, K. Kretschmer, L. Klein, T. Korn, S. Sakaguchi, and M. Schmidt-Supprian
Regulatory T (Treg) cells maintain immune homeostasis and prevent inflammatory and autoimmune responses. During development, thymocytes bearing a moderately self-reactive T cell receptor (TCR) can be selected to become Treg cells. Several observations suggest that also in the periphery mature Treg cells continuously receive self-reactive TCR signals. However, the importance of this inherent autoreactivity for Treg cell biology remains poorly defined. To address this open question, we genetically ablated the TCR of mature Treg cells in vivo. These experiments revealed that TCR-induced Treg lineage-defining Foxp3 expression and gene hypomethylation were uncoupled from TCR input in mature Treg cells. However, Treg cell homeostasis, cell-type-specific gene expression and suppressive function critically depend on continuous triggering of their TCR.
2013
Analysis of immune-related loci identifies 48 new susceptibility variants for multiple sclerosis
Consortium International Multiple Sclerosis Genetics, A. H. Beecham, N. A. Patsopoulos, D. K. Xifara, M. F. Davis, A. Kemppinen, C. Cotsapas, T. S. Shah, C. Spencer, D. Booth, A. Goris, A. Oturai, J. Saarela, B. Fontaine, B. Hemmer, C. Martin, F. Zipp, S. D'Alfonso, F. Martinelli-Boneschi, B. Taylor, H. F. Harbo, I. Kockum, J. Hillert, T. Olsson, M. Ban, J. R. Oksenberg, R. Hintzen, L. F. Barcellos, Consortium Wellcome Trust Case Control, Genetics Consortium I. B. D. International, C. Agliardi, L. Alfredsson, M. Alizadeh, C. Anderson, R. Andrews, H. B. Sondergaard, A. Baker, G. Band, S. E. Baranzini, N. Barizzone, J. Barrett, C. Bellenguez, L. Bergamaschi, L. Bernardinelli, A. Berthele, V. Biberacher, T. M. Binder, H. Blackburn, I. L. Bomfim, P. Brambilla, S. Broadley, B. Brochet, L. Brundin, D. Buck, H. Butzkueven, S. J. Caillier, W. Camu, W. Carpentier, P. Cavalla, E. G. Celius, I. Coman, G. Comi, L. Corrado, L. Cosemans, I. Cournu-Rebeix, B. A. Cree, D. Cusi, V. Damotte, G. Defer, S. R. Delgado, P. Deloukas, A. di Sapio, A. T. Dilthey, P. Donnelly, B. Dubois, M. Duddy, S. Edkins, I. Elovaara, F. Esposito, N. Evangelou, B. Fiddes, J. Field, A. Franke, C. Freeman, I. Y. Frohlich, D. Galimberti, C. Gieger, P. A. Gourraud, C. Graetz, A. Graham, V. Grummel, C. Guaschino, A. Hadjixenofontos, H. Hakonarson, C. Halfpenny, G. Hall, P. Hall, A. Hamsten, J. Harley, T. Harrower, and others
Nat Genet, 45(11): 1353-60, 2013
Analysis of immune-related loci identifies 48 new susceptibility variants for multiple sclerosis
Consortium International Multiple Sclerosis Genetics, A. H. Beecham, N. A. Patsopoulos, D. K. Xifara, M. F. Davis, A. Kemppinen, C. Cotsapas, T. S. Shah, C. Spencer, D. Booth, A. Goris, A. Oturai, J. Saarela, B. Fontaine, B. Hemmer, C. Martin, F. Zipp, S. D'Alfonso, F. Martinelli-Boneschi, B. Taylor, H. F. Harbo, I. Kockum, J. Hillert, T. Olsson, M. Ban, J. R. Oksenberg, R. Hintzen, L. F. Barcellos, Consortium Wellcome Trust Case Control, Genetics Consortium I. B. D. International, C. Agliardi, L. Alfredsson, M. Alizadeh, C. Anderson, R. Andrews, H. B. Sondergaard, A. Baker, G. Band, S. E. Baranzini, N. Barizzone, J. Barrett, C. Bellenguez, L. Bergamaschi, L. Bernardinelli, A. Berthele, V. Biberacher, T. M. Binder, H. Blackburn, I. L. Bomfim, P. Brambilla, S. Broadley, B. Brochet, L. Brundin, D. Buck, H. Butzkueven, S. J. Caillier, W. Camu, W. Carpentier, P. Cavalla, E. G. Celius, I. Coman, G. Comi, L. Corrado, L. Cosemans, I. Cournu-Rebeix, B. A. Cree, D. Cusi, V. Damotte, G. Defer, S. R. Delgado, P. Deloukas, A. di Sapio, A. T. Dilthey, P. Donnelly, B. Dubois, M. Duddy, S. Edkins, I. Elovaara, F. Esposito, N. Evangelou, B. Fiddes, J. Field, A. Franke, C. Freeman, I. Y. Frohlich, D. Galimberti, C. Gieger, P. A. Gourraud, C. Graetz, A. Graham, V. Grummel, C. Guaschino, A. Hadjixenofontos, H. Hakonarson, C. Halfpenny, G. Hall, P. Hall, A. Hamsten, J. Harley, T. Harrower, and others
Using the ImmunoChip custom genotyping array, we analyzed 14,498 subjects with multiple sclerosis and 24,091 healthy controls for 161,311 autosomal variants and identified 135 potentially associated regions (P < 1.0 x 10(-4)). In a replication phase, we combined these data with previous genome-wide association study (GWAS) data from an independent 14,802 subjects with multiple sclerosis and 26,703 healthy controls. In these 80,094 individuals of European ancestry, we identified 48 new susceptibility variants (P < 5.0 x 10(-8)), 3 of which we found after conditioning on previously identified variants. Thus, there are now 110 established multiple sclerosis risk variants at 103 discrete loci outside of the major histocompatibility complex. With high-resolution Bayesian fine mapping, we identified five regions where one variant accounted for more than 50% of the posterior probability of association. This study enhances the catalog of multiple sclerosis risk variants and illustrates the value of fine mapping in the resolution of GWAS signals.
Enriched CD161high CCR6+ γδ T cells in the cerebrospinal fluid of patients with multiple sclerosis
L. Schirmer, V. Rothhammer, B. Hemmer, and T. Korn
JAMA Neurol, 70(3): 345-51, 2013
Enriched CD161high CCR6+ γδ T cells in the cerebrospinal fluid of patients with multiple sclerosis
L. Schirmer, V. Rothhammer, B. Hemmer, and T. Korn
OBJECTIVE: To investigate the expression of CD161 (KLRB1) and CCR6 on human γδ T cells in blood and cerebrospinal fluid (CSF) of patients with a clinically isolated syndrome (CIS) and multiple sclerosis (MS) in relapse. DESIGN: Flow cytometry analysis of CD161 and CCR6 expression and intracellular cytokine staining for interleukin 17 and interferon-γ on human γδ T cells in blood and CSF samples. SETTING: Department of Neurology, Klinikum rechts der Isar, Technische Universitat Munchen, a tertiary referral center. PATIENTS: Twenty-six patients with CIS/MS in active relapse, 10 patients with other autoimmune disorders, 12 patients with neuroinfectious diseases, and 15 patients with noninflammatory neurological diseases. MAIN OUTCOME MEASURES: Frequencies of CD161high and CCR6+ γδ T cells in blood and CSF samples of patients with CIS/MS in relapse and control patients. RESULTS: γδ T cells were increased in both blood and CSF of patients with CIS/MS in relapse as compared with controls with noninflammatory disease. The fraction of CD161high CCR6+ γδ T cells was significantly higher in the CSF of patients with CIS/MS in relapse than of those with systemic autoimmune disorders or controls with noninflammatory disease. The CD161high CCR6+ double-positive γδ T-cell population was further enriched in the CSF in relation to blood in patients with CIS/MS in relapse but not in patients with infectious disease or the other control groups. The CD161high CCR6+ γδ T-cell population was characterized by its capacity to produce interleukin 17. CONCLUSION: Interleukin 17-producing CD161high CCR6+ γδ T cells might contribute to the compartmentalized inflammatory process in the central nervous system of patients with MS.
2012
18F-FDG PET detects inflammatory infiltrates in spinal cord experimental autoimmune encephalomyelitis lesions
D. Buck, A. Forschler, C. Lapa, T. Schuster, P. Vollmar, T. Korn, S. Nessler, C. Stadelmann, A. Drzezga, A. K. Buck, H. J. Wester, C. Zimmer, B. J. Krause, and B. Hemmer
J Nucl Med, 53(8): 1269-76, 2012
18F-FDG PET detects inflammatory infiltrates in spinal cord experimental autoimmune encephalomyelitis lesions
D. Buck, A. Forschler, C. Lapa, T. Schuster, P. Vollmar, T. Korn, S. Nessler, C. Stadelmann, A. Drzezga, A. K. Buck, H. J. Wester, C. Zimmer, B. J. Krause, and B. Hemmer
{Multiple sclerosis (MS) is a heterogeneous disease with respect to lesion pathology, course of disease, and treatment response. Imaging modalities are needed that allow better definition of MS lesions in vivo. The aim of this study was to establish an MRI- and PET/CT-based imaging modality and to evaluate approved and promising PET tracers in experimental autoimmune encephalomyelitis (EAE), the animal model of MS. METHODS: MRI and PET/CT scans were obtained in Dark agouti rats with EAE and healthy control rats. The PET tracers 2-(18)F-fluoro-2-deoxy-d-glucose ((18)F-FDG), 3'-deoxy-3'-(18)F-fluorothymidine ((18)F-FLT), and O-(2-(18)F-fluoro-ethyl)-l-tyrosine ((18)F-FET) were used as surrogate markers of glucose utilization, proliferative activity, and amino acid transport and protein biosynthesis. Immediately after the PET/CT scan, animals were sacrificed for autoradiography, histologic work-up, or RNA expression analysis. RESULTS: EAE lesions were predominantly located in the spinal cord. With MRI, we were able to detect inflammatory lesions in diseased rats, which correlated well with inflammatory infiltrates as determined by histology. Increased (18)F-FDG uptake was observed in spinal cord lesions in all diseased rats. Further investigation by volume-of-interest analysis demonstrated a correlation between the density of histologically proven cellular infiltrates and the (18)F-FDG signal intensity in PET (F(DF=3) = 5.9
Neutralization of the IL-17 axis diminishes neutrophil invasion and protects from ischemic stroke
M. Gelderblom, A. Weymar, C. Bernreuther, J. Velden, P. Arunachalam, K. Steinbach, E. Orthey, T. V. Arumugam, F. Leypoldt, O. Simova, V. Thom, M. A. Friese, I. Prinz, C. Holscher, M. Glatzel, T. Korn, C. Gerloff, E. Tolosa, and T. Magnus
Blood, 120(18): 3793-802, 2012
Neutralization of the IL-17 axis diminishes neutrophil invasion and protects from ischemic stroke
M. Gelderblom, A. Weymar, C. Bernreuther, J. Velden, P. Arunachalam, K. Steinbach, E. Orthey, T. V. Arumugam, F. Leypoldt, O. Simova, V. Thom, M. A. Friese, I. Prinz, C. Holscher, M. Glatzel, T. Korn, C. Gerloff, E. Tolosa, and T. Magnus
The devastating effect of ischemic stroke is attenuated in mice lacking conventional and unconventional T cells, suggesting that inflammation enhances tissue damage in cerebral ischemia. We explored the functional role of αβ and γδ T cells in a murine model of stroke and distinguished 2 different T cell-dependent proinflammatory pathways in ischemia-reperfusion injury. IFN-γ produced by CD4(+) T cells induced TNF-α production in macrophages, whereas IL-17A secreted by γδ T cells led to neutrophil recruitment. The synergistic effect of TNF-α and IL-17A on astrocytes resulted in enhanced secretion of CXCL-1, a neutrophil chemoattractant. Application of an IL-17A-blocking antibody within 3 hours after stroke induction decreased infarct size and improved neurologic outcome in the murine model. In autoptic brain tissue of patients who had a stroke, we detected IL-17A-positive lymphocytes, suggesting that this aspect of the inflammatory cascade is also relevant in the human brain. We propose that selective targeting of IL-17A signaling might provide a new therapeutic option for the treatment of stroke.
CXCL13 is the major determinant for B cell recruitment to the CSF during neuroinflammation
M. C. Kowarik, S. Cepok, J. Sellner, V. Grummel, M. S. Weber, T. Korn, A. Berthele, and B. Hemmer
J Neuroinflammation, 9: 93, 2012
CXCL13 is the major determinant for B cell recruitment to the CSF during neuroinflammation
M. C. Kowarik, S. Cepok, J. Sellner, V. Grummel, M. S. Weber, T. Korn, A. Berthele, and B. Hemmer
{BACKGROUND: The chemokines and cytokines CXCL13, CXCL12, CCL19, CCL21, BAFF and APRIL are believed to play a role in the recruitment of B cells to the central nervous system (CNS) compartment during neuroinflammation. To determine which chemokines/cytokines show the strongest association with a humoral immune response in the cerebrospinal fluid (CSF), we measured their concentrations in the CSF and correlated them with immune cell subsets and antibody levels. METHODS: Cytokine/chemokine concentrations were measured in CSF and serum by ELISA in patients with non-inflammatory neurological diseases (NIND
Unlike αβ T cells, γδ T cells, LTi cells and NKT cells do not require IRF4 for the production of IL-17A and IL-22
H. Raifer, A. J. Mahiny, N. Bollig, F. Petermann, A. Hellhund, K. Kellner, A. Guralnik, K. Reinhard, E. Bothur, M. Huber, S. Bauer, M. Lohning, E. A. Kiss, S. C. Ganal, A. Diefenbach, T. Korn, and M. Lohoff
Eur J Immunol, 42(12): 3189-201, 2012
Unlike αβ T cells, γδ T cells, LTi cells and NKT cells do not require IRF4 for the production of IL-17A and IL-22
H. Raifer, A. J. Mahiny, N. Bollig, F. Petermann, A. Hellhund, K. Kellner, A. Guralnik, K. Reinhard, E. Bothur, M. Huber, S. Bauer, M. Lohning, E. A. Kiss, S. C. Ganal, A. Diefenbach, T. Korn, and M. Lohoff
Apart from conventional CD4(+) Th17 cells, the cytokines IL-17A and IL-22 can also be produced by γδ T cells, NK cells and lymphoid tissue inducer (LTi) cells. Th17 cells develop from precursor cells after T-cell receptor stimulation in the presence of TGF-β, IL-6 and IL-23. In contrast, a subset of γδ T cells ("γδT17") is committed for fast IL-17 production already in the thymus; however, γδ T cells can also produce IL-17 after prolonged in vitro stimulation via their γδ T-cell receptor plus IL-23. Here, we show that γδ T-, LTi- and NKT cells differ extensively from Th17 cells in their signalling requirements for the generation of IL-17A and IL-22. While production of these cytokines by Th17 cells totally depends on the transcription factor interferon regulatory factor 4 (IRF4), IRF4 is irrelevant in the other cell types. As for γδ T cells, this finding pertains to both thymic commitment and prolonged in vitro culture. Furthermore, IL-17A-producing γδ T cells accumulate in the central nervous system of IRF4 deficient (Irf4(-/-)) mice during experimental autoimmune encephalomyelitis. IL-17A-producing WT and Irf4(-/-) γδ T cells equally express CCR6 and lack CD27. The underlying IRF4-independent pathway partially involves STAT3 during in vitro stimulation.
Potassium channel KIR4.1 as an immune target in multiple sclerosis
R. Srivastava, M. Aslam, S. R. Kalluri, L. Schirmer, D. Buck, B. Tackenberg, V. Rothhammer, A. Chan, R. Gold, A. Berthele, J. L. Bennett, T. Korn, and B. Hemmer
N Engl J Med, 367(2): 115-23, 2012
Potassium channel KIR4.1 as an immune target in multiple sclerosis
R. Srivastava, M. Aslam, S. R. Kalluri, L. Schirmer, D. Buck, B. Tackenberg, V. Rothhammer, A. Chan, R. Gold, A. Berthele, J. L. Bennett, T. Korn, and B. Hemmer
BACKGROUND: Multiple sclerosis is a chronic inflammatory demyelinating disease of the central nervous system. Many findings suggest that the disease has an autoimmune pathogenesis; the target of the immune response is not yet known. METHODS: We screened serum IgG from persons with multiple sclerosis to identify antibodies that are capable of binding to brain tissue and observed specific binding of IgG to glial cells in a subgroup of patients. Using a proteomic approach focusing on membrane proteins, we identified the ATP-sensitive inward rectifying potassium channel KIR4.1 as the target of the IgG antibodies. We used a multifaceted validation strategy to confirm KIR4.1 as a target of the autoantibody response in multiple sclerosis and to show its potential pathogenicity in vivo. RESULTS: Serum levels of antibodies to KIR4.1 were higher in persons with multiple sclerosis than in persons with other neurologic diseases and healthy donors (P<0.001 for both comparisons). We replicated this finding in two independent groups of persons with multiple sclerosis or other neurologic diseases (P<0.001 for both comparisons). Analysis of the combined data sets indicated the presence of serum antibodies to KIR4.1 in 186 of 397 persons with multiple sclerosis (46.9%), in 3 of 329 persons with other neurologic diseases (0.9%), and in none of the 59 healthy donors. These antibodies bound to the first extracellular loop of KIR4.1. Injection of KIR4.1 serum IgG into the cisternae magnae of mice led to a profound loss of KIR4.1 expression, altered expression of glial fibrillary acidic protein in astrocytes, and activation of the complement cascade at sites of KIR4.1 expression in the cerebellum. CONCLUSIONS: KIR4.1 is a target of the autoantibody response in a subgroup of persons with multiple sclerosis. (Funded by the German Ministry for Education and Research and Deutsche Forschungsgemeinschaft.).
IL-17A production by renal γδ T cells promotes kidney injury in crescentic GN
J. E. Turner, C. Krebs, A. P. Tittel, H. J. Paust, C. Meyer-Schwesinger, S. B. Bennstein, O. M. Steinmetz, I. Prinz, T. Magnus, T. Korn, R. A. Stahl, C. Kurts, and U. Panzer
J Am Soc Nephrol, 23(9): 1486-95, 2012
IL-17A production by renal γδ T cells promotes kidney injury in crescentic GN
J. E. Turner, C. Krebs, A. P. Tittel, H. J. Paust, C. Meyer-Schwesinger, S. B. Bennstein, O. M. Steinmetz, I. Prinz, T. Magnus, T. Korn, R. A. Stahl, C. Kurts, and U. Panzer
The Th17 immune response appears to contribute to the pathogenesis of human and experimental crescentic GN, but the cell types that produce IL-17A in the kidney, the mechanisms involved in its induction, and the IL-17A-mediated effector functions that promote renal tissue injury are incompletely understood. Here, using a murine model of crescentic GN, we found that CD4(+) T cells, γδ T cells, and a population of CD3(+)CD4(-)CD8(-)γδT cell receptor(-)NK1.1(-) T cells all produce IL-17A in the kidney. A time course analysis identified γδ T cells as a major source of IL-17A in the early phase of disease, before the first CD4(+) Th17 cells arrived. The production of IL-17A by renal γδ T cells depended on IL-23p19 signaling and retinoic acid-related orphan receptor-γt but not on IL-1β or IL-6. In addition, depletion of dendritic cells, which produce IL-23 in the kidney, reduced IL-17A production by renal γδ T cells. Furthermore, the lack of IL-17A production in γδ T cells, as well as the absence of all γδ T cells, reduced neutrophil recruitment into the kidney and ameliorated renal injury. Taken together, these data suggest that γδ T cells produce IL-17A in the kidney, induced by IL-23, promoting neutrophil recruitment, and contributing to the immunopathogenesis of crescentic GN.
Dendritic cells ameliorate autoimmunity in the CNS by controlling the homeostasis of PD-1 receptor(+) regulatory T cells
N. Yogev, F. Frommer, D. Lukas, K. Kautz-Neu, K. Karram, D. Ielo, E. von Stebut, H. C. Probst, M. van den Broek, D. Riethmacher, T. Birnberg, T. Blank, B. Reizis, T. Korn, H. Wiendl, S. Jung, M. Prinz, F. C. Kurschus, and A. Waisman
Immunity, 37(2): 264-75, 2012
Dendritic cells ameliorate autoimmunity in the CNS by controlling the homeostasis of PD-1 receptor(+) regulatory T cells
N. Yogev, F. Frommer, D. Lukas, K. Kautz-Neu, K. Karram, D. Ielo, E. von Stebut, H. C. Probst, M. van den Broek, D. Riethmacher, T. Birnberg, T. Blank, B. Reizis, T. Korn, H. Wiendl, S. Jung, M. Prinz, F. C. Kurschus, and A. Waisman
Mature dendritic cells (DCs) are established as unrivaled antigen-presenting cells (APCs) in the initiation of immune responses, whereas steady-state DCs induce peripheral T cell tolerance. Using various genetic approaches, we depleted CD11c(+) DCs in mice and induced autoimmune CNS inflammation. Unexpectedly, mice lacking DCs developed aggravated disease compared to control mice. Furthermore, when we engineered DCs to present a CNS-associated autoantigen in an induced manner, we found robust tolerance that prevented disease, which coincided with an upregulation of the PD-1 receptor on antigen-specific T cells. Additionally, we showed that PD-1 was necessary for DC-mediated induction of regulatory T cells. Our results show that a reduction of DCs interferes with tolerance, resulting in a stronger inflammatory response, and that other APC populations could compensate for the loss of immunogenic APC function in DC-depleted mice.
2011
Expression of miRNAs miR-133b and miR-206 in the Il17a/f locus is co-regulated with IL-17 production in αβ and γδ T cells
J. D. Haas, K. Nistala, F. Petermann, N. Saran, V. Chennupati, S. Schmitz, T. Korn, L. R. Wedderburn, R. Forster, A. Krueger, and I. Prinz
PLoS One, 6(5): e20171, 2011
Expression of miRNAs miR-133b and miR-206 in the Il17a/f locus is co-regulated with IL-17 production in αβ and γδ T cells
J. D. Haas, K. Nistala, F. Petermann, N. Saran, V. Chennupati, S. Schmitz, T. Korn, L. R. Wedderburn, R. Forster, A. Krueger, and I. Prinz
Differentiation of T helper 17 cells (Th17) is a multistep process that involves the cytokines IL-6, TGF-β, and IL-23 as well as IL-1β, IL-21, and TNF-α. Thereby, robust induction of the capacity to produce IL-17 involves epigenetic modifications of the syntenic Il17a/f locus. Using inbred mouse strains, we identified co-regulation of gene transcription at the Il17a/f locus with the nearby microRNAs miR-133b and miR-206 that are clustered approximately 45 kb upstream of Il17a/f. Expression of these microRNAs was specific for Th17 as compared to other CD4(+) T cell subsets and this was equally valid for in vitro polarized and ex vivo derived cells. From all factors analyzed, IL-23 was the most important cytokine for the in vitro induction of miR-133b and miR-206 in naive CD4(+) T cells of wild type mice. However, analysis of IL-23R deficient mice revealed that IL-23R signaling was not essential for the induction of miR-133b and miR-206. Importantly, we found a similar co-regulation in CCR6(+) and other γδ T cell subsets that are predisposed to production of IL-17. Taken together, we discovered a novel feature of T cell differentiation towards an IL-17-producing phenotype that is shared between αβ and γδ T cells. Notably, the specific co-regulation of miR-133b and miR-206 with the Il17a/f locus also extended to human Th17 cells. This qualifies expression of miR-133b and miR-206 in T cells as novel biomarkers for Th17-type immune reactions.
Functional characterization of aquaporin-4 specific T cells: towards a model for neuromyelitis optica
S. R. Kalluri, V. Rothhammer, O. Staszewski, R. Srivastava, F. Petermann, M. Prinz, B. Hemmer, and T. Korn
PLoS One, 6(1): e16083, 2011
Functional characterization of aquaporin-4 specific T cells: towards a model for neuromyelitis optica
S. R. Kalluri, V. Rothhammer, O. Staszewski, R. Srivastava, F. Petermann, M. Prinz, B. Hemmer, and T. Korn
BACKGROUND: Antibodies to the water channel protein aquaporin-4 (AQP4), which is expressed in astrocytic endfeet at the blood brain barrier, have been identified in the serum of Neuromyelitis optica (NMO) patients and are believed to induce damage to astrocytes. However, AQP4 specific T helper cell responses that are required for the generation of anti-AQP4 antibodies and most likely also for the formation of intraparenchymal CNS lesions have not been characterized. METHODOLOGY/PRINCIPAL FINDINGS: Using overlapping 15-meric peptides of AQP4, we identified the immunogenic T cell epitopes of AQP4 that are restricted to murine major histocompatibility complex (MHC) I-A(b). The N-terminal region of AQP4 was highly immunogenic. More precisely, the intracellular epitope AQP4(22-36) was detected as major immunogenic determinant. AQP4(82-108) (located in the second transmembrane domain), AQP4(139-153) (located in the second extracellular loop), AQP4(211-225) (located in the fifth transmembrane domain), AQP4(235-249) (located in the sixth transmembrane domain), as well as AQP4(289-306) in the intracellular C-terminal region were also immunogenic epitopes. AQP4(22-36) and AQP4(289-303) specific T cells were present in the natural T cell repertoire of wild type C57BL/6 mice and T cell lines were raised. However, active immunization with these AQP4 peptides did not induce signs of spinal cord disease. Rather, sensitization with AQP4 peptides resulted in production of IFN-γ, but also IL-5 and IL-10 by antigen-specific T cells. Consistent with this cytokine profile, the AQP4 specific antibody response upon immunization with full length AQP4 included IgG1 and IgG2, which are associated with a mixed Th2/Th1 T cell response. CONCLUSIONS AND SIGNIFICANCE: AQP4 is able to induce an autoreactive T cell response. The identification of I-A(b) restricted AQP4 specific T cell epitopes will allow us to investigate how AQP4 specific autoimmune reactions are regulated and to establish faithful mouse models of NMO that include both cellular and humoral responses against AQP4.
Differential effects of fingolimod (FTY720) on immune cells in the CSF and blood of patients with MS
M. C. Kowarik, H. L. Pellkofer, S. Cepok, T. Korn, T. Kumpfel, D. Buck, R. Hohlfeld, A. Berthele, and B. Hemmer
Neurology, 76(14): 1214-21, 2011
Differential effects of fingolimod (FTY720) on immune cells in the CSF and blood of patients with MS
M. C. Kowarik, H. L. Pellkofer, S. Cepok, T. Korn, T. Kumpfel, D. Buck, R. Hohlfeld, A. Berthele, and B. Hemmer
BACKGROUND: The oral immunomodulator fingolimod (FTY720) has recently been shown to be highly effective in relapsing-remitting multiple sclerosis (MS). Fingolimod is a functional antagonist of the sphingosine-1-phosphate receptor 1 and thereby inhibits sphingosine-1-phosphate-dependent lymphocyte egress from secondary lymphoid tissues, resulting in a pronounced lymphopenia in the peripheral blood. The effects of fingolimod treatment on the CSF of patients with MS have not been studied so far. METHODS: We analyzed the leukocyte count, albumin quotient, immunoglobulin G (IgG) index, and oligoclonal bands in the CSF of fingolimod-treated patients with MS. Moreover, we performed immunophenotyping of CSF and peripheral blood leukocytes by flow cytometry. The results were compared to those from treatment-naive or natalizumab-treated patients with MS and patients with other inflammatory and noninflammatory neurologic diseases. RESULTS: Fingolimod therapy significantly decreased CSF leukocyte counts, but had little impact on the extent of intrathecal IgG synthesis and presence of oligoclonal bands in the CSF. Fingolimod decreased the proportion of CSF CD4+ T cells but to a lesser extent than in the peripheral blood. While fingolimod strongly reduced B cells in the periphery, it had little impact on B cells in the CSF. The percentage of CSF CD8+ T cells, NK cells, and monocytes increased compared to treatment-naive patients. The CD4+/CD8+ T-cell ratio in CSF reversed in most of the patients. CONCLUSION: Fingolimod treatment has a profound impact on CSF, which to some extent differs from the peripheral effects of the drug.
Antigen targeting to plasmacytoid dendritic cells via Siglec-H inhibits Th cell-dependent autoimmunity
J. Loschko, S. Heink, D. Hackl, D. Dudziak, W. Reindl, T. Korn, and A. B. Krug
J Immunol, 187(12): 6346-56, 2011
Antigen targeting to plasmacytoid dendritic cells via Siglec-H inhibits Th cell-dependent autoimmunity
J. Loschko, S. Heink, D. Hackl, D. Dudziak, W. Reindl, T. Korn, and A. B. Krug
Plasmacytoid dendritic cells (PDCs) have been shown to present Ags and to contribute to peripheral immune tolerance and to Ag-specific adaptive immunity. However, modulation of adaptive immune responses by selective Ag targeting to PDCs with the aim of preventing autoimmunity has not been investigated. In the current study, we demonstrate that in vivo Ag delivery to murine PDCs via the specifically expressed surface molecule sialic acid binding Ig-like lectin H (Siglec-H) inhibits Th cell and Ab responses in the presence of strong immune stimulation in an Ag-specific manner. Correlating with sustained low-level MHC class II-restricted Ag presentation on PDCs, Siglec-H-mediated Ag delivery induced a hyporesponsive state in CD4(+) T cells leading to reduced expansion and Th1/Th17 cell polarization without conversion to Foxp3(+) regulatory T cells or deviation to Th2 or Tr1 cells. Siglec-H-mediated delivery of a T cell epitope derived from the autoantigen myelin oligodendrocyte glycoprotein to PDCs effectively delayed onset and reduced disease severity in myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis by interfering with the priming phase without promoting the generation or expansion of myelin oligodendrocyte glycoprotein-specific Foxp3(+) regulatory T cells. We conclude that Ag delivery to PDCs can be harnessed to inhibit Ag-specific immune responses and prevent Th cell-dependent autoimmunity.
TNF-α-dependent loss of IKKβ-deficient myeloid progenitors triggers a cytokine loop culminating in granulocytosis
A. K. Mankan, O. Canli, S. Schwitalla, P. Ziegler, J. Tschopp, T. Korn, and F. R. Greten
Proc Natl Acad Sci U S A, 108(16): 6567-72, 2011
TNF-α-dependent loss of IKKβ-deficient myeloid progenitors triggers a cytokine loop culminating in granulocytosis
A. K. Mankan, O. Canli, S. Schwitalla, P. Ziegler, J. Tschopp, T. Korn, and F. R. Greten
Loss of IkappaB kinase (IKK) β-dependent NF-kappaB signaling in hematopoietic cells is associated with increased granulopoiesis. Here we identify a regulatory cytokine loop that causes neutrophilia in Ikkβ-deficient mice. TNF-α-dependent apoptosis of myeloid progenitor cells leads to the release of IL-1β, which promotes Th17 polarization of peripheral CD4(+) T cells. Although the elevation of IL-17 and the consecutive induction of granulocyte colony-stimulating factor compensate for the loss of myeloid progenitor cells, the facilitated induction of Th17 cells renders Ikkβ-deficient animals more susceptible to the development of experimental autoimmune encephalitis. These results unravel so far unanticipated direct and indirect functions for IKKβ in myeloid progenitor survival and maintenance of innate and Th17 immunity and raise concerns about long-term IKKβ inhibition in IL-17-mediated diseases.
Th17 lymphocytes traffic to the central nervous system independently of α4 integrin expression during EAE
V. Rothhammer, S. Heink, F. Petermann, R. Srivastava, M. C. Claussen, B. Hemmer, and T. Korn
J Exp Med, 208(12): 2465-76, 2011
Th17 lymphocytes traffic to the central nervous system independently of α4 integrin expression during EAE
V. Rothhammer, S. Heink, F. Petermann, R. Srivastava, M. C. Claussen, B. Hemmer, and T. Korn
The integrin α4β1 (VLA-4) is used by encephalitogenic T cells to enter the central nervous system (CNS). However, both Th1 and Th17 cells are capable of inducing experimental autoimmune encephalomyelitis (EAE), and the molecular cues mediating the infiltration of Th1 versus Th17 cells into the CNS have not yet been defined. We investigated how blocking of α4 integrins affected trafficking of Th1 and Th17 cells into the CNS during EAE. Although antibody-mediated inhibition of α4 integrins prevented EAE when MOG(35-55)-specific Th1 cells were adoptively transferred, Th17 cells entered the brain, but not the spinal cord parenchyma, irrespective of α4 blockade. Accordingly, T cell-conditional α4-deficient mice were not resistant to actively induced EAE but showed an ataxic syndrome with predominantly supraspinal infiltrates of IL-23R(+)CCR6(+)CD4(+) T cells. The entry of α4-deficient Th17 cells into the CNS was abolished by blockade of LFA-1 (αLβ2 integrin). Thus, Th1 cells preferentially infiltrate the spinal cord via an α4 integrin-mediated mechanism, whereas the entry of Th17 cells into the brain parenchyma occurs in the absence of α4 integrins but is dependent on the expression of αLβ2. These observations have implications for the understanding of lesion localization, immunosurveillance, and drug design in multiple sclerosis.
CCL17-expressing dendritic cells drive atherosclerosis by restraining regulatory T cell homeostasis in mice
C. Weber, S. Meiler, Y. Doring, M. Koch, M. Drechsler, R. T. Megens, Z. Rowinska, K. Bidzhekov, C. Fecher, E. Ribechini, M. A. van Zandvoort, C. J. Binder, I. Jelinek, M. Hristov, L. Boon, S. Jung, T. Korn, M. B. Lutz, I. Forster, M. Zenke, T. Hieronymus, T. Junt, and A. Zernecke
J Clin Invest, 121(7): 2898-910, 2011
CCL17-expressing dendritic cells drive atherosclerosis by restraining regulatory T cell homeostasis in mice
C. Weber, S. Meiler, Y. Doring, M. Koch, M. Drechsler, R. T. Megens, Z. Rowinska, K. Bidzhekov, C. Fecher, E. Ribechini, M. A. van Zandvoort, C. J. Binder, I. Jelinek, M. Hristov, L. Boon, S. Jung, T. Korn, M. B. Lutz, I. Forster, M. Zenke, T. Hieronymus, T. Junt, and A. Zernecke
Immune mechanisms are known to control the pathogenesis of atherosclerosis. However, the exact role of DCs, which are essential for priming of immune responses, remains elusive. We have shown here that the DC-derived chemokine CCL17 is present in advanced human and mouse atherosclerosis and that CCL17+ DCs accumulate in atherosclerotic lesions. In atherosclerosis-prone mice, Ccl17 deficiency entailed a reduction of atherosclerosis, which was dependent on Tregs. Expression of CCL17 by DCs limited the expansion of Tregs by restricting their maintenance and precipitated atherosclerosis in a mechanism conferred by T cells. Conversely, a blocking antibody specific for CCL17 expanded Tregs and reduced atheroprogression. Our data identify DC-derived CCL17 as a central regulator of Treg homeostasis, implicate DCs and their effector functions in atherogenesis, and suggest that CCL17 might be a target for vascular therapy.
2010
Proinflammatory T helper type 17 cells are effective B-cell helpers
M. Mitsdoerffer, Y. Lee, A. Jager, H. J. Kim, T. Korn, J. K. Kolls, H. Cantor, E. Bettelli, and V. K. Kuchroo
Proc Natl Acad Sci U S A, 107(32): 14292-7, 2010
Proinflammatory T helper type 17 cells are effective B-cell helpers
M. Mitsdoerffer, Y. Lee, A. Jager, H. J. Kim, T. Korn, J. K. Kolls, H. Cantor, E. Bettelli, and V. K. Kuchroo
T helper type 17 (TH17) cells are highly proinflammatory effector T cells that are characterized by the production of high amounts of IL-17A, IL-17F, IL-21, and IL-22. Furthermore, TH17 cells have been associated with a number of autoimmune diseases. However, it is not clear whether TH17 cells can also serve as effective helper cells. Here we show that TH17 cells can function as B-cell helpers in that they not only induce a strong proliferative response of B cells in vitro but also trigger antibody production with class switch recombination in vivo. Transfer of TH17 cells into WT or T-cell receptor α-deficient mice, which lack endogenous T cells, induces a pronounced antibody response with preferential isotype class switching to IgG1, IgG2a, IgG2b, and IgG3, as well as the formation of germinal centers. Conversely, blockade of IL-17 signaling results in a significant reduction in both number and size of germinal centers. Whereas IL-21 is known to help B cells, IL-17 on its own drives B cells to undergo preferential isotype class switching to IgG2a and IgG3 subtypes. These observations provide insights into the unappreciated role of TH17 cells and their signature cytokines in mediating B-cell differentiation and class switch recombination.
γδ T cells enhance autoimmunity by restraining regulatory T cell responses via an interleukin-23-dependent mechanism
F. Petermann, V. Rothhammer, M. C. Claussen, J. D. Haas, L. R. Blanco, S. Heink, I. Prinz, B. Hemmer, V. K. Kuchroo, M. Oukka, and T. Korn
Immunity, 33(3): 351-63, 2010
γδ T cells enhance autoimmunity by restraining regulatory T cell responses via an interleukin-23-dependent mechanism
F. Petermann, V. Rothhammer, M. C. Claussen, J. D. Haas, L. R. Blanco, S. Heink, I. Prinz, B. Hemmer, V. K. Kuchroo, M. Oukka, and T. Korn
Mice that lack interleukin-23 (IL-23) are resistant to T cell-mediated autoimmunity. Although IL-23 is a maturation factor for T helper 17 (Th17) cells, a subset of γδ T cells expresses the IL-23 receptor (IL-23R) constitutively. Using IL-23R reporter mice, we showed that γδ T cells were the first cells to respond to IL-23 during experimental autoimmune encephalomyelitis (EAE). Although γδ T cells produced Th17 cell-associated cytokines in response to IL-23, their major function was to prevent the development of regulatory T (Treg) cell responses. IL-23-activated γδ T cells rendered αβ effector T cells refractory to the suppressive activity of Treg cells and also prevented the conversion of conventional T cells into Foxp3(+) Treg cells in vivo. Thus, IL-23, which by itself has no direct effect on Treg cells, is able to disarm Treg cell responses and promote antigen-specific effector T cell responses via activating γδ T cells.
Active immunization with amyloid-β 1-42 impairs memory performance through TLR2/4-dependent activation of the innate immune system
P. Vollmar, J. S. Kullmann, B. Thilo, M. C. Claussen, V. Rothhammer, H. Jacobi, J. Sellner, S. Nessler, T. Korn, and B. Hemmer
J Immunol, 185(10): 6338-47, 2010
Active immunization with amyloid-β 1-42 impairs memory performance through TLR2/4-dependent activation of the innate immune system
P. Vollmar, J. S. Kullmann, B. Thilo, M. C. Claussen, V. Rothhammer, H. Jacobi, J. Sellner, S. Nessler, T. Korn, and B. Hemmer
Active immunization with amyloid-β (Aβ) peptide 1-42 reverses amyloid plaque deposition in the CNS of patients with Alzheimer's disease and in amyloid precursor protein transgenic mice. However, this treatment may also cause severe, life-threatening meningoencephalitis. Physiological responses to immunization with Aβ(1-42) are poorly understood. In this study, we characterized cognitive and immunological consequences of Aβ(1-42)/CFA immunization in C57BL/6 mice. In contrast to mice immunized with myelin oligodendrocyte glycoprotein (MOG)(35-55)/CFA or CFA alone, Aβ(1-42)/CFA immunization resulted in impaired exploratory activity, habituation learning, and spatial-learning abilities in the open field. As morphological substrate of this neurocognitive phenotype, we identified a disseminated, nonfocal immune cell infiltrate in the CNS of Aβ(1-42)/CFA-immunized animals. In contrast to MOG(35-55)/CFA and PBS/CFA controls, the majority of infiltrating cells in Aβ(1-42)/CFA-immunized mice were CD11b(+)CD14(+) and CD45(high), indicating their blood-borne monocyte/macrophage origin. Immunization with Aβ(1-42)/CFA was significantly more potent than immunization with MOG(35-55)/CFA or CFA alone in activating macrophages in the secondary lymphoid compartment and peripheral tissues. Studies with TLR2/4-deficient mice revealed that the TLR2/4 pathway mediated the Aβ(1-42)-dependent proinflammatory cytokine release from cells of the innate immune system. In line with this, TLR2/4 knockout mice were protected from cognitive impairment upon immunization with Aβ(1-42)/CFA. Thus, this study identifies adjuvant effects of Aβ(1-42), which result in a clinically relevant neurocognitive phenotype highlighting potential risks of Aβ immunotherapy.
2009
Cutting edge: IL-23 receptor gfp reporter mice reveal distinct populations of IL-17-producing cells
A. Awasthi, L. Riol-Blanco, A. Jager, T. Korn, C. Pot, G. Galileos, E. Bettelli, V. K. Kuchroo, and M. Oukka
J Immunol, 182(10): 5904-8, 2009
Cutting edge: IL-23 receptor gfp reporter mice reveal distinct populations of IL-17-producing cells
A. Awasthi, L. Riol-Blanco, A. Jager, T. Korn, C. Pot, G. Galileos, E. Bettelli, V. K. Kuchroo, and M. Oukka
IL-23, an IL-12 family member, has been implicated in the development of Th17 cells and the progression of autoimmune diseases. However, due to the lack of availability of sensitive Ab reagents specific for the IL-23 receptor (IL-23R), it has been difficult to characterize the cell types that express the IL-23R and are responsive to IL-23 in vivo. To address the role of IL-23 in vivo, we have generated a novel "knock-in" mouse in which we have replaced the intracellular domain of the IL-23R with the GFP. We show that in addition to Th17 cells, a subset of myeloid cells express IL-23R and respond to IL-23 by producing IL-17 and IL-22. Our studies further demonstrate that IL-23R expression is crucial for generation of encephalitogenic Th17 cells, but its expression on the innate immune system is dispensible in the development of experimental autoimmune encephalomyelitis.
2008
IL-6 controls Th17 immunity in vivo by inhibiting the conversion of conventional T cells into Foxp3+ regulatory T cells
T. Korn, M. Mitsdoerffer, A. L. Croxford, A. Awasthi, V. A. Dardalhon, G. Galileos, P. Vollmar, G. L. Stritesky, M. H. Kaplan, A. Waisman, V. K. Kuchroo, and M. Oukka
Proc Natl Acad Sci U S A, 105(47): 18460-5, 2008
IL-6 controls Th17 immunity in vivo by inhibiting the conversion of conventional T cells into Foxp3+ regulatory T cells
T. Korn, M. Mitsdoerffer, A. L. Croxford, A. Awasthi, V. A. Dardalhon, G. Galileos, P. Vollmar, G. L. Stritesky, M. H. Kaplan, A. Waisman, V. K. Kuchroo, and M. Oukka
The conditions leading to the induction of adaptive Foxp3(+) regulatory T cells (T-regs) from peripheral T cells in vivo are incompletely understood. Here, we show that unresponsiveness of T cells to IL-6 by T cell-selective deletion of gp130 or immunization of wild-type mice with antigen in incomplete Freund's adjuvant (IFA), which fails to induce IL-6, promotes the conversion of peripheral CD4(+) T cells into adaptive Foxp3(+) T-regs. Thus, both T cell-conditional gp130 knockout (KO) mice immunized with MOG35-55 in complete Freund's adjuvant (CFA) and wild-type mice immunized with MOG35-55 in IFA develop overwhelming antigen-specific T-reg responses and are protected from experimental autoimmune encephalomyelitis (EAE). Depletion of T-regs restores T helper (Th)17 responses and clinical EAE in MOG/CFA-immunized T cell-conditional gp130 KO mice, but not in MOG/IFA-immunized wild-type mice. We conclude that in the absence of T-regs, IL-6 signaling is dispensable for the induction of Th17 cells, and alternative pathways exist to induce Th17 cells and EAE in the absence of IL-6 signaling. However, IL-6 signaling is dominant in inhibiting the conversion of conventional T cells into Foxp3(+) T-regs in vivo, and in the absence of IL-6 signaling, no other cytokine can substitute in inhibiting T-reg conversion. These data identify IL-6 as an important target to modulate autoimmune responses and chronic inflammation.
Control of T(reg) and T(H)17 cell differentiation by the aryl hydrocarbon receptor
F. J. Quintana, A. S. Basso, A. H. Iglesias, T. Korn, M. F. Farez, E. Bettelli, M. Caccamo, M. Oukka, and H. L. Weiner
Nature, 453(7191): 65-71, 2008
Control of T(reg) and T(H)17 cell differentiation by the aryl hydrocarbon receptor
F. J. Quintana, A. S. Basso, A. H. Iglesias, T. Korn, M. F. Farez, E. Bettelli, M. Caccamo, M. Oukka, and H. L. Weiner
Regulatory T cells (T(reg)) expressing the transcription factor Foxp3 control the autoreactive components of the immune system. The development of T(reg) cells is reciprocally related to that of pro-inflammatory T cells producing interleukin-17 (T(H)17). Although T(reg) cell dysfunction and/or T(H)17 cell dysregulation are thought to contribute to the development of autoimmune disorders, little is known about the physiological pathways that control the generation of these cell lineages. Here we report the identification of the ligand-activated transcription factor aryl hydrocarbon receptor (AHR) as a regulator of T(reg) and T(H)17 cell differentiation in mice. AHR activation by its ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin induced functional T(reg) cells that suppressed experimental autoimmune encephalomyelitis. On the other hand, AHR activation by 6-formylindolo[3,2-b]carbazole interfered with T(reg) cell development, boosted T(H)17 cell differentiation and increased the severity of experimental autoimmune encephalomyelitis in mice. Thus, AHR regulates both T(reg) and T(H)17 cell differentiation in a ligand-specific fashion, constituting a unique target for therapeutic immunomodulation.
Retinoic acid increases Foxp3+ regulatory T cells and inhibits development of Th17 cells by enhancing TGF-β-driven Smad3 signaling and inhibiting IL-6 and IL-23 receptor expression
S. Xiao, H. Jin, T. Korn, S. M. Liu, M. Oukka, B. Lim, and V. K. Kuchroo
J Immunol, 181(4): 2277-84, 2008
Retinoic acid increases Foxp3+ regulatory T cells and inhibits development of Th17 cells by enhancing TGF-β-driven Smad3 signaling and inhibiting IL-6 and IL-23 receptor expression
S. Xiao, H. Jin, T. Korn, S. M. Liu, M. Oukka, B. Lim, and V. K. Kuchroo
The de novo generation of Foxp3+ regulatory T (Treg) cells in the peripheral immune compartment and the differentiation of Th17 cells both require TGF-β, and IL-6 and IL-21 are switch factors that drive the development of Th17 cells at the expense of Treg cell generation. The major vitamin A metabolite all-trans retinoic acid (RA) not only enforces the generation of Treg cells but also inhibits the differentiation of Th17 cells. Herein we show that RA enhances TGF-β signaling by increasing the expression and phosphorylation of Smad3, and this results in increased Foxp3 expression even in the presence of IL-6 or IL-21. RA also inhibits the expression of IL-6Rα, IRF-4, and IL-23R and thus inhibits Th17 development. In vitro, RA significantly promotes Treg cell conversion, but in vivo during the development of experimental autoimmune encephalomyelitis it does not increase the frequency of Treg cells in the face of an ongoing inflammation. However, RA suppresses the disease very efficiently by inhibiting proinflammatory T cell responses, especially pathogenic Th17 responses. These data not only identify the signaling mechanisms by which RA can affect both Treg cell and Th17 differentiation, but they also highlight that in vivo during an autoimmune reaction, RA suppresses autoimmunity mainly by inhibiting the generation of effector Th17 cells.
J Immunol, 181(4): 2277-84, 2008
2007
Anti-thymocyte globulin (ATG) prevents autoimmune encephalomyelitis by expanding myelin antigen-specific Foxp3+ regulatory T cells
D. T. Chung, T. Korn, J. Richard, M. Ruzek, A. P. Kohm, S. Miller, S. Nahill, and M. Oukka
Int Immunol, 19(8): 1003-10, 2007
Anti-thymocyte globulin (ATG) prevents autoimmune encephalomyelitis by expanding myelin antigen-specific Foxp3+ regulatory T cells
D. T. Chung, T. Korn, J. Richard, M. Ruzek, A. P. Kohm, S. Miller, S. Nahill, and M. Oukka
The T cell-depleting polyclonal antibody, anti-thymocyte globulin (ATG) has long been used in organ transplantation to treat acute rejection episodes. More recently, it is also being used as part of an induction regimen to protect allografts. It has been proposed that ATG might deplete effector T cells (T-effs) while sparing regulatory T cells (T-regs). In order to test whether ATG is effective in autoimmune disease, we used Foxp3gfp 'knock-in' mice in combination with a myelin oligodendrocyte glycoprotein (MOG)(35-55)/IA(b) tetramer to study more closely the effect of ATG treatment on antigen-specific T cell responses in vivo during MOG-induced experimental autoimmune encephalomyelitis (EAE), an animal model for Multiple Sclerosis. ATG treatment enhanced the expansion of MOG-specific T-regs (CD4(+)Foxp3(+)) in MOG-immunized mice. T-effs were depleted, but on a single-cell basis, the effector function of residual T-effs was not compromised by ATG. Thus, ATG tipped the balance of T-effs and T-regs and skewed an auto-antigen-specific immune reaction from a pathogenic T cell response to a potentially protective T-reg response. In both acute and relapsing remitting disease models, ATG treatment resulted in the attenuation from EAE, both in a preventive and early therapeutic setting. We conclude that ATG treatment enforces the development of a dominant immunoregulatory environment which may be advantageous for the treatment of T cell-driven autoimmune diseases.
IL-21 initiates an alternative pathway to induce proinflammatory T(H)17 cells
T. Korn, E. Bettelli, W. Gao, A. Awasthi, A. Jager, T. B. Strom, M. Oukka, and V. K. Kuchroo
Nature, 448(7152): 484-7, 2007
IL-21 initiates an alternative pathway to induce proinflammatory T(H)17 cells
T. Korn, E. Bettelli, W. Gao, A. Awasthi, A. Jager, T. B. Strom, M. Oukka, and V. K. Kuchroo
On activation, naive T cells differentiate into effector T-cell subsets with specific cytokine phenotypes and specialized effector functions. Recently a subset of T cells, distinct from T helper (T(H))1 and T(H)2 cells, producing interleukin (IL)-17 (T(H)17) was defined and seems to have a crucial role in mediating autoimmunity and inducing tissue inflammation. We and others have shown that transforming growth factor (TGF)-β and IL-6 together induce the differentiation of T(H)17 cells, in which IL-6 has a pivotal function in dictating whether T cells differentiate into Foxp3+ regulatory T cells (T(reg) cells) or T(H)17 cells. Whereas TGF-β induces Foxp3 and generates T(reg) cells, IL-6 inhibits the generation of T(reg) cells and induces the production of IL-17, suggesting a reciprocal developmental pathway for T(H)17 and T(reg) cells. Here we show that IL-6-deficient (Il6-/-) mice do not develop a T(H)17 response and their peripheral repertoire is dominated by Foxp3+ T(reg) cells. However, deletion of T(reg) cells leads to the reappearance of T(H)17 cells in Il6-/- mice, suggesting an additional pathway by which T(H)17 cells might be generated in vivo. We show that an IL-2 cytokine family member, IL-21, cooperates with TGF-β to induce T(H)17 cells in naive Il6-/- T cells and that IL-21-receptor-deficient T cells are defective in generating a T(H)17 response.
Myelin-specific regulatory T cells accumulate in the CNS but fail to control autoimmune inflammation
T. Korn, J. Reddy, W. Gao, E. Bettelli, A. Awasthi, T. R. Petersen, B. T. Backstrom, R. A. Sobel, K. W. Wucherpfennig, T. B. Strom, M. Oukka, and V. K. Kuchroo
Nat Med, 13(4): 423-31, 2007
Myelin-specific regulatory T cells accumulate in the CNS but fail to control autoimmune inflammation
T. Korn, J. Reddy, W. Gao, E. Bettelli, A. Awasthi, T. R. Petersen, B. T. Backstrom, R. A. Sobel, K. W. Wucherpfennig, T. B. Strom, M. Oukka, and V. K. Kuchroo
Treatment with ex vivo-generated regulatory T cells (T-reg) has been regarded as a potentially attractive therapeutic approach for autoimmune diseases. However, the dynamics and function of T-reg in autoimmunity are not well understood. Thus, we developed Foxp3gfp knock-in (Foxp3gfp.KI) mice and myelin oligodendrocyte glycoprotein (MOG)(35-55)/IA(b) (MHC class II) tetramers to track autoantigen-specific effector T cells (T-eff) and T-reg in vivo during experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis. MOG tetramer-reactive, Foxp3(+) T-reg expanded in the peripheral lymphoid compartment and readily accumulated in the central nervous system (CNS), but did not prevent the onset of disease. Foxp3(+) T cells isolated from the CNS were effective in suppressing naive MOG-specific T cells, but failed to control CNS-derived encephalitogenic T-eff that secreted interleukin (IL)-6 and tumor necrosis factor (TNF). Our data suggest that in order for CD4(+)Foxp3(+) T-reg to effectively control autoimmune reactions in the target organ, it may also be necessary to control tissue inflammation.
Evidence that nucleocytoplasmic Olig2 translocation mediates brain-injury-induced differentiation of glial precursors to astrocytes
T. Magnus, T. Coksaygan, T. Korn, H. Xue, T. V. Arumugam, M. R. Mughal, D. M. Eckley, S. C. Tang, L. Detolla, M. S. Rao, R. Cassiani-Ingoni, and M. P. Mattson
J Neurosci Res, 85(10): 2126-37, 2007
Evidence that nucleocytoplasmic Olig2 translocation mediates brain-injury-induced differentiation of glial precursors to astrocytes
T. Magnus, T. Coksaygan, T. Korn, H. Xue, T. V. Arumugam, M. R. Mughal, D. M. Eckley, S. C. Tang, L. Detolla, M. S. Rao, R. Cassiani-Ingoni, and M. P. Mattson
The mechanisms by which neural and glial progenitor cells in the adult brain respond to tissue injury are unknown. We studied the responses of these cells to stab wound injury in rats and in two transgenic mouse models in which Y/GFP is driven either by Sox2 (a neural stem cell marker) or by Tα-1 (which marks newly born neurons). The response of neural progenitors was low in all nonneurogenic regions, and no neurogenesis occurred at the injury site. Glial progenitors expressing Olig2 and NG2 showed the greatest response. The appearance of these progenitors preceded the appearance of reactive astrocytes. Surprisingly, we found evidence of the translocation of the transcription factor Olig2 into cytoplasm in the first week after injury, a mechanism that is known to mediate the differentiation of astrocytes during brain development. Translocation of Olig2, down-regulation of NG2, and increased glial fibrillary acidic protein expression were recapitulated in vitro after exposure of glial progenitors to serum components or bone morphogentic protein by up-regulation of Notch-1. The glial differentiation and Olig2 translocation could be blocked by inhibition of Notch-1 with the γ-secretase inhibitor DAPT. Together, these data indicate that the prompt maturation of numerous Olig2(+) glial progenitors to astrocytes underlies the repair process after a traumatic injury. In contrast, neural stem cells and neuronal progenitor cells appear to play only a minor role in the injured adult CNS.
Differential engagement of Tim-1 during activation can positively or negatively costimulate T cell expansion and effector function
S. Xiao, N. Najafian, J. Reddy, M. Albin, C. Zhu, E. Jensen, J. Imitola, T. Korn, A. C. Anderson, Z. Zhang, C. Gutierrez, T. Moll, R. A. Sobel, D. T. Umetsu, H. Yagita, H. Akiba, T. Strom, M. H. Sayegh, R. H. DeKruyff, S. J. Khoury, and V. K. Kuchroo
J Exp Med, 204(7): 1691-702, 2007
Differential engagement of Tim-1 during activation can positively or negatively costimulate T cell expansion and effector function
S. Xiao, N. Najafian, J. Reddy, M. Albin, C. Zhu, E. Jensen, J. Imitola, T. Korn, A. C. Anderson, Z. Zhang, C. Gutierrez, T. Moll, R. A. Sobel, D. T. Umetsu, H. Yagita, H. Akiba, T. Strom, M. H. Sayegh, R. H. DeKruyff, S. J. Khoury, and V. K. Kuchroo
It has been suggested that T cell immunoglobulin mucin (Tim)-1 expressed on T cells serves to positively costimulate T cell responses. However, crosslinking of Tim-1 by its ligand Tim-4 resulted in either activation or inhibition of T cell responses, thus raising the issue of whether Tim-1 can have a dual function as a costimulator. To resolve this issue, we tested a series of monoclonal antibodies specific for Tim-1 and identified two antibodies that showed opposite functional effects. One anti-Tim-1 antibody increased the frequency of antigen-specific T cells, the production of the proinflammatory cytokines IFN-γ and IL-17, and the severity of experimental autoimmune encephalomyelitis. In contrast, another anti-Tim-1 antibody inhibited the generation of antigen-specific T cells, production of IFN-γ and IL-17, and development of autoimmunity, and it caused a strong Th2 response. Both antibodies bound to closely related epitopes in the IgV domain of the Tim-1 molecule, but the activating antibody had an avidity for Tim-1 that was 17 times higher than the inhibitory antibody. Although both anti-Tim-1 antibodies induced CD3 capping, only the activating antibody caused strong cytoskeletal reorganization and motility. These data indicate that Tim-1 regulates T cell responses and that Tim-1 engagement can alter T cell function depending on the affinity/avidity with which it is engaged.
2006
Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells
E. Bettelli, Y. Carrier, W. Gao, T. Korn, T. B. Strom, M. Oukka, H. L. Weiner, and V. K. Kuchroo
Nature, 441(7090): 235-8, 2006
Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells
E. Bettelli, Y. Carrier, W. Gao, T. Korn, T. B. Strom, M. Oukka, H. L. Weiner, and V. K. Kuchroo
On activation, T cells undergo distinct developmental pathways, attaining specialized properties and effector functions. T-helper (T(H)) cells are traditionally thought to differentiate into T(H)1 and T(H)2 cell subsets. T(H)1 cells are necessary to clear intracellular pathogens and T(H)2 cells are important for clearing extracellular organisms. Recently, a subset of interleukin (IL)-17-producing T (T(H)17) cells distinct from T(H)1 or T(H)2 cells has been described and shown to have a crucial role in the induction of autoimmune tissue injury. In contrast, CD4+CD25+Foxp3+ regulatory T (T(reg)) cells inhibit autoimmunity and protect against tissue injury. Transforming growth factor-β (TGF-β) is a critical differentiation factor for the generation of T(reg) cells. Here we show, using mice with a reporter introduced into the endogenous Foxp3 locus, that IL-6, an acute phase protein induced during inflammation, completely inhibits the generation of Foxp3+ T(reg) cells induced by TGF-β. We also demonstrate that IL-23 is not the differentiation factor for the generation of T(H)17 cells. Instead, IL-6 and TGF-β together induce the differentiation of pathogenic T(H)17 cells from naive T cells. Our data demonstrate a dichotomy in the generation of pathogenic (T(H)17) T cells that induce autoimmunity and regulatory (Foxp3+) T cells that inhibit autoimmune tissue injury.
2005
Lipooligosaccharide of Campylobacter jejuni prevents myelin-specific enteral tolerance to autoimmune neuritis–a potential mechanism in Guillain-Barre syndrome?
S. Jung, S. Zimmer, E. Luneberg, M. Frosch, H. Karch, T. Korn, and K. V. Toyka
Neurosci Lett, 381(1-2): 175-8, 2005
Lipooligosaccharide of Campylobacter jejuni prevents myelin-specific enteral tolerance to autoimmune neuritis–a potential mechanism in Guillain-Barre syndrome?
S. Jung, S. Zimmer, E. Luneberg, M. Frosch, H. Karch, T. Korn, and K. V. Toyka
Campylobacter jejuni-induced enteritis is the most common infection preceding Guillain-Barre syndrome (GBS), an immune-mediated polyradiculoneuritis. The acute autoimmune attack is thought to be based on C. jejuni antigens which may mimick antigens of the peripheral nervous system. Additional pathomechanisms, like disturbance of natural T cell immunoregulation by C. jejuni, have not been evaluated so far. In experimental autoimmune neuritis (EAN), a T lymphocyte-mediated animal model of human GBS, tolerance to myelin-derived autoantigens can be induced by oral feeding of the respective antigen. Here we investigated whether the lipooligosaccharide (LOS) fraction of C. jejuni may directly alter immunologic tolerance through gastrointestinal pathways. While EAN, actively induced by immunization with bovine peripheral nerve myelin could be ameliorated by precedent feeding of myelin, feeding of C. jejuni LOS along with the myelin antigen not only prevented the tolerizing effects of oral myelin but even accelerated the onset of overt EAN and augmented the myelin-specific B cell response. These findings provide evidence that LOS of C. jejuni, as produced in the gut during C. jejuni-induced enteritis, can disturb natural tolerance to definite proteins which may be or may mimic peripheral nerve antigens. In human patients this may be one of the potential mechanisms to explain why C. jejuni enteritis is a common trigger of GBS.
Autoantigen specific T cells inhibit glutamate uptake in astrocytes by decreasing expression of astrocytic glutamate transporter GLAST: a mechanism mediated by tumor necrosis factor-α
T. Korn, T. Magnus, and S. Jung
FASEB J, 19(13): 1878-80, 2005
Autoantigen specific T cells inhibit glutamate uptake in astrocytes by decreasing expression of astrocytic glutamate transporter GLAST: a mechanism mediated by tumor necrosis factor-α
T. Korn, T. Magnus, and S. Jung
Glutamate excitotoxicity is increasingly being recognized as a pathogenic mechanism in autoimmune inflammatory disorders of the central nervous system (CNS). Astrocytes are the predominant players in clearing the extracellular space from glutamate and normally have extensive spare capacities in terms of glutamate uptake. We asked what might be the basis of glutamate accumulation in T cell triggered autoimmune inflammation. In vitro, coculture of primary rat astrocytes with activated myelin basic protein (MBP)-specific T cells resulted in a decrease of astrocytic glutamate uptake rates (Vmax). In parallel, the amount of the Na+-dependent glutamate transporter GLAST was reduced within 48-60 h. Significant decreases of GLAST protein were observed in astrocytes harvested after incubation with T cells activated by MBP during coculture or after incubation with T cell blasts preactivated in the presence of splenocytes beforehand. Since exposure of astrocytes to cell-free supernatant of MBP-activated T cells also resulted in reduced expression of GLAST, a humoral factor appeared to be the driving agent. In blocking experiments using neutralizing antibodies and by incubation of astrocytes with recombinant cytokines, tumor necrosis factor-α (TNF-α) was identified as being responsible for the down-modulation of GLAST. GLAST was also down-regulated in the CNS of autoimmune encephalomyelitic rats but not in animals suffering from systemic inflammation. Since the loss of GLAST was not confined to inflammatory infiltrates, here too, a humoral factor seemed to be causative. In conclusion, T cell derived TNF-α impairs glutamate clearance capacity of astrocytes in vitro and probably also in vivo providing a pathogenic link to glutamate excitotoxicity that may contribute to early axonal dysfunction remote from active autoimmune inflammatory demyelination.
Interaction with antigen-specific T cells regulates expression of the lactate transporter MCT1 in primary rat astrocytes: specific link between immunity and homeostasis
T. Korn, T. Magnus, and S. Jung
Glia, 49(1): 73-83, 2005
Interaction with antigen-specific T cells regulates expression of the lactate transporter MCT1 in primary rat astrocytes: specific link between immunity and homeostasis
T. Korn, T. Magnus, and S. Jung
Monocarboxylates like lactate are provided by astrocytes and can be used as fuel by neurons and oligodendrocytes. In an autoimmune inflammatory environment, homeostatic functions of astrocytes are incompletely understood. In primary Lewis rat astrocytes, co-culture with MHC class II-restricted myelin basic protein (MBP)-specific T cells in the presence of MBP resulted in a marked upregulation of the astrocytic lactate transporter MCT1 that is to export lactate into the extracellular space. It was evident that the increase in MCT1 was triggered by T cells in an antigen-dependent manner. The glial isoform of the glucose transporter GLUT1 was not regulated under these conditions. T-cell blasts that had been pre-activated by antigen and splenic antigen-presenting cells (APCs) beforehand also led to an increase in the expression of astrocytic MCT1 after co-culture. Resting T cells did not induce a relevant upregulation of MCT1 in astrocytes. However, resting T cells stimulated the expression of MCT1 when anti-MHC class II antibodies, but not when anti-MHC class I antibodies, were added to the co-culture. Therefore, even in the presence of inactive T cells, complexation of MHC class II molecules on astrocytes might lead to the regulation of certain astrocytic transport proteins. Consistent with the in vitro experiments, an upregulation of MCT1 was observed in the spinal cord of autoimmune encephalitic rats while GLUT1 expression appeared to be unchanged. This T-cell-mediated regulation of MCT1 might contribute to a compensatory or protective mechanism in order to guarantee substrate pools for neurons and oligodendrocytes under inflammatory conditions.
Microglial expression of the B7 family member B7 homolog 1 confers strong immune inhibition: implications for immune responses and autoimmunity in the CNS
T. Magnus, B. Schreiner, T. Korn, C. Jack, H. Guo, J. Antel, I. Ifergan, L. Chen, F. Bischof, A. Bar-Or, and H. Wiendl
J Neurosci, 25(10): 2537-46, 2005
Microglial expression of the B7 family member B7 homolog 1 confers strong immune inhibition: implications for immune responses and autoimmunity in the CNS
T. Magnus, B. Schreiner, T. Korn, C. Jack, H. Guo, J. Antel, I. Ifergan, L. Chen, F. Bischof, A. Bar-Or, and H. Wiendl
Inflammation of the CNS is usually locally limited to avoid devastating consequences. Critical players involved in this immune regulatory process are the resident immune cells of the brain, the microglia. Interactions between the growing family of B7 costimulatory ligands and their receptors are increasingly recognized as important pathways for costimulation and/or inhibition of immune responses. Human and mouse microglial cells constitutively express B7 homolog 1 (B7-H1) in vitro. However, under inflammatory conditions [presence of interferon-γ (IFN-γ) or T-helper 1 supernatants], a significant upregulation of B7-H1 was detectable. Expression levels of B7-H1 protein on microglial cells were substantially higher compared with astrocytes or splenocytes. Coculture experiments of major histocompatibility complex class II-positive antigen-presenting cells (APC) with syngeneic T cells in the presence of antigen demonstrated the functional consequences of B7-H1 expression on T-cell activation. In the presence of a neutralizing anti-B7-H1 antibody, both the production of inflammatory cytokines (IFN-γ and interleukin-2) and the upregulation of activation markers (inducible costimulatory signal) by T cells were markedly enhanced. Interestingly, this effect was clearly more pronounced when microglial cells were used as APC, compared with astrocytes or splenocytes. Furthermore, B7-H1 was highly upregulated during the course of myelin oligodendrocyte glycoprotein-induced and proteolipid protein-induced experimental allergic encephalomyelitis in vivo. Expression was predominantly localized to areas of strongest inflammation and could be colocalized with microglial cells/macrophages as well as T cells. Together, our data propose microglial B7-H1 as an important immune inhibitory molecule capable of downregulating T-cell activation in the CNS and thus confining immunopathological damage.
2004
Biphasic form of experimental autoimmune neuritis in dark Agouti rats and its oral therapy by antigen-specific tolerization
S. Jung, S. Gaupp, T. Korn, G. Kollner, H. P. Hartung, and K. V. Toyka
J Neurosci Res, 75(4): 524-35, 2004
Biphasic form of experimental autoimmune neuritis in dark Agouti rats and its oral therapy by antigen-specific tolerization
S. Jung, S. Gaupp, T. Korn, G. Kollner, H. P. Hartung, and K. V. Toyka
A new and biphasic form of experimental autoimmune neuritis (EAN) is described in dark agouti rats (DA rats) and is inducible by a single immunization with bovine peripheral nerve myelin (BPM) in complete Freund's adjuvant (DA-EAN). Animals develop a mild episode of disease; after recovery, 66-100% of the rats suffer from a more severe bout of EAN with paraparesis 25-30 days after immunization. By histology, DA-EAN is an inflammatory and demyelinating polyradiculoneuropathy virtually without axonal damage. Demyelination affects mainly spinal roots. This is also reflected by markedly increased F-wave latencies in nerve conduction studies of sciatic nerves. In sciatic nerves, inflammation and demyelination are found only focally and may be the histopathologic basis for conduction failure in some fibers. Immunologic investigations revealed stronger proliferative responses of DA than of Lewis rat lymph node cells to BPM and various peptides derived from the P2 protein. Proliferative and Th1-cytokine responses were particularly pronounced in spleen during the late phase of DA-EAN as compared to the monophasic EAN of Lewis rats. The data suggest that persistent lymphocyte proliferation with secretion of interferon (IFN)-γ may be relevant for the relapsing course of DA-EAN whereas epitope spreading may explain the increased severity of the second bout of disease. The extended Th1 response in DA rats did not go along with a lack of downregulatory mechanisms, because the second DA-EAN attack was self-limited and splenocytes from DA rats produced considerable amounts of interleukin (IL)-10 and transforming growth factor (TGF)-β. To substantiate further a functional immunoregulation in DA rats, we modulated DA-EAN by antigen-specific oral tolerization, which is known to involve active suppressor mechanisms. Preventive feeding of BPM in combination with cholera toxin (CT) induced a long-lasting resistance to DA-EAN. Even therapeutic administration of BPM or BPM/CT after onset of signs of disease significantly mitigated the further course of disease and prevented development of paraparesis. Because DA-EAN is easily inducible and leads consistently to relapses in most rats, it can be used for studies of immune factors that determine a relapsing course of autoimmunity. Furthermore, DA-EAN may serve as a model for relapsing inflammatory demyelinating polyneuropathies such as chronic inflammatory demyelinating polyneuropathy (CIDP) and for treatment studies. Our findings on effective prevention and therapy of DA-EAN by oral application of myelin/CT corroborate this form of immunomodulation as a treatment strategy for cell-mediated processes in chronic inflammatory neuropathies.
Modulation of effector cell functions in experimental autoimmune encephalomyelitis by leflunomide–mechanisms independent of pyrimidine depletion
T. Korn, T. Magnus, K. Toyka, and S. Jung
J Leukoc Biol, 76(5): 950-60, 2004
Modulation of effector cell functions in experimental autoimmune encephalomyelitis by leflunomide–mechanisms independent of pyrimidine depletion
T. Korn, T. Magnus, K. Toyka, and S. Jung
Leflunomide inhibits de novo pyrimidine synthesis and is a novel, immunosuppressive agent that has been successfully used to treat rheumatoid arthritis. Here, we investigated the efficacy of leflunomide and its mode of action in experimental autoimmune encephalomyelitis (EAE), which is a T helper cell type 1 cell-borne disease model to simulate inflammatory aspects of multiple sclerosis and was induced in Lewis rats by adoptive transfer of myelin basic protein (MBP)-specific T line cells. Given in vivo for 7 days after cell transfer, leflunomide suppressed clinical signs of disease even in uridine-substituted animals. MBP-specific T line cells that had been antigen-activated in vitro in the presence of A77 1726 (active metabolite of leflunomide) produced less interferon-γ, whereas interleukin (IL)-10 secretion had a tendency to be increased without changes in signal transducer and activator of transcription 6 trafficking. Furthermore, these T cells exhibited reduced chemotaxis and induced a significantly mitigated disease course upon transfer into naive rats. The effects of leflunomide on MBP-specific memory type T line cells in vitro may not be mediated by pyrimidine depletion, as they were not reversible by exogenous uridine. Moreover, A77 1726 led to increased expression of CD86 (B7-2) and secretion of IL-10 in cultured microglial cells in vitro, strengthening their down-modulatory impact on activated, autoantigen-specific T cells. In conclusion, our observations underline that the immunomodulatory potential of leflunomide in effector cells of EAE is clinically relevant and is not exclusively dependent on the depletion of cellular pyrimidine pools.
Impaired volitional closure of the left eyelid after right anterior cerebral artery infarction: apraxia due to interhemispheric disconnection?
T. Korn, W. Reith, and G. Becker
Arch Neurol, 61(2): 273-5, 2004
Impaired volitional closure of the left eyelid after right anterior cerebral artery infarction: apraxia due to interhemispheric disconnection?
T. Korn, W. Reith, and G. Becker
BACKGROUND: The inability of volitional unilateral eyelid closure is an uncommon symptom of a central nervous system disorder. When it occurs as the result of a localized brain lesion, it is debated to be a form of supranuclear facial palsy or an apraxic phenomenon. OBJECTIVES: To report and discuss a unilateral (left-sided) higher-order movement disorder of the facial periocular musculature bearing apraxic features. SETTING: University neurology department. PATIENT: A 78-year-old right-handed man was admitted to the hospital with a left-sided brachiofacial hemiparesis of sudden onset. After thrombolysis with intravenous recombinant tissue-type plasminogen activator, the hemiparesis, including the left-sided facial weakness, disappeared. Serial computed tomographic scans showed that the patient was left with a stroke in the right anterior cerebral artery territory, affecting the frontal commissural fibers of the corpus callosum. There were no signs of upper motor neuron facial paresis on the left side when gesturing in a natural context. Eyelid closure was complete during sleep. However, left eyelid closure and elevation of the left eyebrow were not possible on verbal command. In contrast, voluntary innervation of the perioral facial musculature was performed properly. CONCLUSIONS: The voluntary-automatic dissociation of our patient's eyelid closure was suggestive of an apraxic disorder. Disconnection from a praxis center caused by callosal damage may be assumed to be the underlying cause. The unilaterality of the symptom might imply that in a bilaterally organized corticonuclear system such as upper face innervation, it is the crossing fibers that are primarily involved in praxis tasks.
Chronically stimulated microglial cells do no longer alter their immune functions in response to the phagocytosis of apoptotic cells
T. Magnus, T. Korn, and S. Jung
J Neuroimmunol, 155(1-2): 64-72, 2004
Chronically stimulated microglial cells do no longer alter their immune functions in response to the phagocytosis of apoptotic cells
T. Magnus, T. Korn, and S. Jung
In an autoimmune inflammatory setting, ingestion of apoptotic T cells leads to a down-regulation of microglial immune functions. Recent studies have indicated that microglia can be matured by exposure to GM-CSF. GM-CSF stimulation led to a differentiated microglial phenotype and enhanced antigen-presenting capabilities. The secretion of TNF-α was significantly decreased by the uptake of apoptotic cells in unstimulated microglia, but not in GM-CSF-differentiated microglia. IL-10 secretion was unaffected. After ingestion of apoptotic cells, only previously unstimulated, but not GM-CSF-differentiated microglial cells decreased their T cell-activating potential as measured by IFN-γ secretion in antigen-activated MBP-specific T cells. Thus, GM-CSF stimulation reduces the immunomodulatory functions of microglial cells.
2003
Vav1-deficient mice are resistant to MOG-induced experimental autoimmune encephalomyelitis due to impaired antigen priming
T. Korn, K. D. Fischer, I. Girkontaite, G. Kollner, K. Toyka, and S. Jung
J Neuroimmunol, 139(1-2): 17-26, 2003
Vav1-deficient mice are resistant to MOG-induced experimental autoimmune encephalomyelitis due to impaired antigen priming
T. Korn, K. D. Fischer, I. Girkontaite, G. Kollner, K. Toyka, and S. Jung
Mice that lack the guanine nucleotide exchange factor (GEF) Vav1 exhibit particular defects in antigen-triggered T cell activation but may have an autoreactive T cell repertoire due to impaired intra-thymic negative selection. MOG(35-55)-induced experimental autoimmune encephalomyelitis (EAE) was used to test the susceptibility of Vav1(-/-) mice to organ-specific autoimmunity. Vav1(-/-) animals were found to be resistant to MOG(35-55)-EAE since the priming and in vivo expansion of myelin oligodendrocyte glycoprotein (MOG)-specific T cells was inefficient despite fully functional antigen presentation. Protection from cell-mediated autoimmunity was not due to a Th2 bias, to the lack of IL-2 or a failure of Vav1(-/-) T cells in terms of chemotactic mobility.
J Neuroimmunol, 139(1-2): 17-26, 2003
2001
The plasma membrane-associated protein RS1 decreases transcription of the transporter SGLT1 in confluent LLC-PK1 cells
T. Korn, T. Kuhlkamp, C. Track, I. Schatz, K. Baumgarten, V. Gorboulev, and H. Koepsell
J Biol Chem, 276(48): 45330-40, 2001
The plasma membrane-associated protein RS1 decreases transcription of the transporter SGLT1 in confluent LLC-PK1 cells
T. Korn, T. Kuhlkamp, C. Track, I. Schatz, K. Baumgarten, V. Gorboulev, and H. Koepsell
Previously we cloned RS1, a 67-kDa polypeptide that is associated with the intracellular side of the plasma membrane. Upon co-expression in Xenopus laevis oocytes, human RS1 decreased the concentration of the Na(+)-D-glucose co-transporter hSGLT1 in the plasma membrane (Valentin, M., Kuhlkamp, T., Wagner, K., Krohne, G., Arndt, P., Baumgarten, K., Weber, W.-M., Segal, A., Veyhl, M., and Koepsell, H. (2000) Biochim. Biophys. Acta 1468, 367-380). Here, the porcine renal epithelial cell line LLC-PK1 was used to investigate whether porcine RS1 (pRS1) plays a role in transcriptional up-regulation of SGLT1 after confluence and in down-regulation of SGLT1 by high extracellular D-glucose concentrations. Western blots indicated a dramatic decrease of endogenous pRS1 protein at the plasma membrane after confluence but no significant effect of D-glucose. In confluent LLC-PK1 cells overexpressing pRS1, SGLT1 mRNA, protein, and methyl-α-D-glucopyranoside uptakes were drastically decreased; however, the reduction of methyl-α-D-glucopyranoside uptake after cultivation with 25 mm D-glucose remained. In confluent pRS1 antisense cells, the expression of SGLT1 mRNA and protein was strongly increased, whereas the reduction of SGLT1 expression during cultivation with high D-glucose was not influenced. Nuclear run-on assays showed that the transcription of SGLT1 was 10-fold increased in the pRS1 antisense cells. The data suggest that RS1 participates in transcriptional up-regulation of SGLT1 after confluence but not in down-regulation by D-glucose.
Suppression of experimental autoimmune neuritis by leflunomide
T. Korn, K. Toyka, H. P. Hartung, and S. Jung
Brain, 124(Pt 9): 1791-802, 2001
Suppression of experimental autoimmune neuritis by leflunomide
T. Korn, K. Toyka, H. P. Hartung, and S. Jung
Leflunomide is a new immunosuppressive drug whose active metabolite, A77 1726, impairs cellular nucleotide metabolism by inhibiting the dihydroorotate dehydrogenase (DHODH), a rate-limiting enzyme of de novo pyrimidine synthesis. Furthermore, A77 1726 suppresses tyrosine kinases involved in signal transduction pathways. We investigated the immunosuppressive effects of leflunomide in experimental autoimmune neuritis (EAN) in rats, which is a model of immune-mediated neuropathies. In EAN that was actively induced by subcutaneous injection of peripheral nerve myelin, leflunomide completely prevented paraparesis if applied orally from the day of immunization. Leflunomide was much more effective than azathioprine, which did not mitigate EAN at all. Even when leflunomide was administered therapeutically after the appearance of the first neuropathical signs, it halted the progression and markedly reduced the severity and duration of EAN. Inflammatory infiltrates, demyelination and axonal degeneration in sciatic nerve sections of leflunomide-treated EAN rats were strongly reduced. Leflunomide-treated rats did not mount autoantibodies as specified by ELISA (enzyme-linked immunosorbent assay) with a mixture of peripheral myelin proteins, including P2 and myelin basic protein. In EAN that was adoptively transferred by injection of neuritogenic cells of a P2-specific T-helper line, application of leflunomide also clearly reduced signs of disease. Additional injection of uridine did not neutralize the effect of leflunomide. Similarly, transfer of neuritogenic P2-specific T cells, which were activated in the presence of A77 1726 plus uridine in vitro, still resulted in reduced severity of adoptive transfer EAN in vivo, although proliferation of these cells in vitro was identical to that of control cells. The T-cell receptor-mediated in vitro activatability of a P2-specific T-cell hybridoma was diminished by high concentrations of A77 1726, as evidenced by reduced Ca(2+) flux into the cytosol. Together with the findings in adoptive transfer EAN, this indicates that the antiproliferative effect is probably not the only mechanism of immunosuppressive action by leflunomide. In summary, leflunomide suppresses EAN efficiently and may constitute a promising therapy for immune-mediated neuropathies.
Brain, 124(Pt 9): 1791-802, 2001
Review Articles
2023
Foxp3(+) regulatory T cells in the central nervous system and other nonlymphoid tissues
T. Korn
Eur J Immunol, 53(8): e2250227, 2023
Foxp3(+) regulatory T cells in the central nervous system and other nonlymphoid tissues
T. Korn
Foxp3(+) regulatory T (Treg) cells are indispensable for the maintenance of immunologic self-tolerance as well as for the confinement of autoimmune inflammation after the breach of self-tolerance. In order to fulfill these tasks, Treg cells operate in secondary lymphoid tissues and nonlymphoid tissues. The conditions for Treg cell stability and for their modes of action are different according to their compartment of residence. In addition, Treg cells initiate residency programs to inhabit niches in nonlympoid tissues (NLT) in steady state and after re-establishment of previously deflected homeostasis for extended periods of time. These NLT Treg cells are different from lymphoid tissue residing Treg cells and are functionally specialized to subserve not only immune functions but support intrinsic functions of their tissue of residence. This review will highlight current ideas about the functional specialization of NLT Treg cells in particular in the central nervous system (CNS) and discuss challenges that we are facing in an effort to exploit the power of NLT Treg cells for maintenance of tissue homeostasis and perhaps also tissue regeneration.
2022
Fatigue, depression, and pain in multiple sclerosis: How neuroinflammation translates into dysfunctional reward processing and anhedonic symptoms
H. Heitmann, T. F. M. Andlauer, T. Korn, M. Muhlau, P. Henningsen, B. Hemmer, and M. Ploner
Mult Scler, 28(7): 1020-1027, 2022
Fatigue, depression, and pain in multiple sclerosis: How neuroinflammation translates into dysfunctional reward processing and anhedonic symptoms
H. Heitmann, T. F. M. Andlauer, T. Korn, M. Muhlau, P. Henningsen, B. Hemmer, and M. Ploner
Fatigue, depression, and pain affect the majority of multiple sclerosis (MS) patients, which causes a substantial burden to patients and society. The pathophysiology of these symptoms is not entirely clear, and current treatments are only partially effective. Clinically, these symptoms share signs of anhedonia, such as reduced motivation and a lack of positive affect. In the brain, they are associated with overlapping structural and functional alterations in areas involved in reward processing. Moreover, neuroinflammation has been shown to directly impede monoaminergic neurotransmission that plays a key role in reward processing. Here, we review recent neuroimaging and neuroimmunological findings, which indicate that dysfunctional reward processing might represent a shared functional mechanism fostering the symptom cluster of fatigue, depression, and pain in MS. We propose a framework that integrates these findings with a focus on monoaminergic neurotransmission and discuss its therapeutic implications, limitations, and perspectives.
The Role of OCT Criteria and Machine Learning in Multiple Sclerosis and Optic Neuritis Diagnosis
R. C. Kenney, M. Liu, L. Hasanaj, B. Joseph, A. A. Al-Hassan, L. J. Balk, R. Behbehani, A. Brandt, P. A. Calabresi, E. Frohman, T. C. Frohman, J. Havla, B. Hemmer, H. Jiang, B. Knier, T. Korn, L. Leocani, E. H. Martinez-Lapiscina, A. Papadopoulou, F. Paul, A. Petzold, M. Pisa, P. Villoslada, H. Zimmermann, L. E. Thorpe, H. Ishikawa, J. S. Schuman, G. Wollstein, Y. Chen, S. Saidha, S. Galetta, and L. J. Balcer
Neurology, 2022
The Role of OCT Criteria and Machine Learning in Multiple Sclerosis and Optic Neuritis Diagnosis
R. C. Kenney, M. Liu, L. Hasanaj, B. Joseph, A. A. Al-Hassan, L. J. Balk, R. Behbehani, A. Brandt, P. A. Calabresi, E. Frohman, T. C. Frohman, J. Havla, B. Hemmer, H. Jiang, B. Knier, T. Korn, L. Leocani, E. H. Martinez-Lapiscina, A. Papadopoulou, F. Paul, A. Petzold, M. Pisa, P. Villoslada, H. Zimmermann, L. E. Thorpe, H. Ishikawa, J. S. Schuman, G. Wollstein, Y. Chen, S. Saidha, S. Galetta, and L. J. Balcer
{BACKGROUND AND OBJECTIVES: Recent studies have suggested that inter-eye differences (IEDs) in peripapillary retinal nerve fiber layer (pRNFL) or ganglion cell+inner plexiform (GCIPL) thickness by spectral-domain optical coherence tomography (SD-OCT) may identify people with a history of unilateral optic neuritis (ON). However, this requires further validation. Machine learning classification may be useful for validating thresholds for OCT IEDs and for examining added utility for visual function tests, such as low-contrast letter acuity (LCLA), in the diagnosis of people with multiple sclerosis (PwMS) and for unilateral ON history. METHODS: Participants were from 11 sites within the International Multiple Sclerosis Visual System (IMSVISUAL) consortium. pRNFL and GCIPL thicknesses were measured using SD-OCT. A composite score combining OCT and visual measures was compared individual measurements to determine the best model to distinguish PwMS from controls. These methods were also used to distinguish those with history of ON among PwMS. ROC curve analysis was performed on a training dataset (2/3 of cohort), then applied to a testing dataset (1/3 of cohort). Support vector machine (SVM) analysis was used to assess whether machine learning models improved diagnostic capability of OCT. RESULTS: Among 1,568 PwMS and 552 controls, variable selection models identified GCIPL IED, average GCIPL thickness (both eyes), and binocular 2.5% LCLA as most important for classifying PwMS vs. controls. This composite score performed best, with AUC=0.89 (95% CI 0.85, 0.93)
2021
APOSTEL 2.0 Recommendations for Reporting Quantitative Optical Coherence Tomography Studies
A. Aytulun, A. Cruz-Herranz, O. Aktas, L. J. Balcer, L. Balk, P. Barboni, A. A. Blanco, P. A. Calabresi, F. Costello, B. Sanchez-Dalmau, D. C. DeBuc, N. Feltgen, R. P. Finger, J. L. Frederiksen, E. Frohman, T. Frohman, D. Garway-Heath, I. Gabilondo, J. S. Graves, A. J. Green, H. P. Hartung, J. Havla, F. G. Holz, J. Imitola, R. Kenney, A. Klistorner, B. Knier, T. Korn, S. Kolbe, J. Kramer, W. A. Lagreze, L. Leocani, O. Maier, E. H. Martinez-Lapiscina, S. Meuth, O. Outteryck, F. Paul, A. Petzold, G. Pihl-Jensen, J. L. Preiningerova, G. Rebolleda, M. Ringelstein, S. Saidha, S. Schippling, J. S. Schuman, R. C. Sergott, A. Toosy, P. Villoslada, S. Wolf, E. A. Yeh, P. Yu-Wai-Man, H. G. Zimmermann, A. U. Brandt, and P. Albrecht
Neurology, 97(2): 68-79, 2021
APOSTEL 2.0 Recommendations for Reporting Quantitative Optical Coherence Tomography Studies
A. Aytulun, A. Cruz-Herranz, O. Aktas, L. J. Balcer, L. Balk, P. Barboni, A. A. Blanco, P. A. Calabresi, F. Costello, B. Sanchez-Dalmau, D. C. DeBuc, N. Feltgen, R. P. Finger, J. L. Frederiksen, E. Frohman, T. Frohman, D. Garway-Heath, I. Gabilondo, J. S. Graves, A. J. Green, H. P. Hartung, J. Havla, F. G. Holz, J. Imitola, R. Kenney, A. Klistorner, B. Knier, T. Korn, S. Kolbe, J. Kramer, W. A. Lagreze, L. Leocani, O. Maier, E. H. Martinez-Lapiscina, S. Meuth, O. Outteryck, F. Paul, A. Petzold, G. Pihl-Jensen, J. L. Preiningerova, G. Rebolleda, M. Ringelstein, S. Saidha, S. Schippling, J. S. Schuman, R. C. Sergott, A. Toosy, P. Villoslada, S. Wolf, E. A. Yeh, P. Yu-Wai-Man, H. G. Zimmermann, A. U. Brandt, and P. Albrecht
OBJECTIVE: To update the consensus recommendations for reporting of quantitative optical coherence tomography (OCT) study results, thus revising the previously published Advised Protocol for OCT Study Terminology and Elements (APOSTEL) recommendations. METHODS: To identify studies reporting quantitative OCT results, we performed a PubMed search for the terms "quantitative" and "optical coherence tomography" from 2015 to 2017. Corresponding authors of the identified publications were invited to provide feedback on the initial APOSTEL recommendations via online surveys following the principle of a modified Delphi method. The results were evaluated and discussed by a panel of experts and changes to the initial recommendations were proposed. A final survey was recirculated among the corresponding authors to obtain a majority vote on the proposed changes. RESULTS: A total of 116 authors participated in the surveys, resulting in 15 suggestions, of which 12 were finally accepted and incorporated into an updated 9-point checklist. We harmonized the nomenclature of the outer retinal layers, added the exact area of measurement to the description of volume scans, and suggested reporting device-specific features. We advised to address potential bias in manual segmentation or manual correction of segmentation errors. References to specific reporting guidelines and room light conditions were removed. The participants' consensus with the recommendations increased from 80% for the previous APOSTEL version to greater than 90%. CONCLUSIONS: The modified Delphi method resulted in an expert-led guideline (evidence Class III; Grading of Recommendations, Assessment, Development and Evaluations [GRADE] criteria) concerning study protocol, acquisition device, acquisition settings, scanning protocol, funduscopic imaging, postacquisition data selection, postacquisition analysis, nomenclature and abbreviations, and statistical approach. It will be essential to update these recommendations to new research and practices regularly.
Role of IL-6 in the commitment of T cell subsets
T. Korn and M. Hiltensperger
Cytokine, 146: 155654, 2021
Role of IL-6 in the commitment of T cell subsets
T. Korn and M. Hiltensperger
IL-6 gained much attention with the discovery that this cytokine is a non-redundant differentiation factor for Th17 cells and T follicular helper cells. Adaptive immune responses to fungi and extracellular bacteria are impaired in the absence of IL-6. IL-6 is also required for the induction of ROR-gammat(+) Treg cells, which are gatekeepers of homeostasis in the gut lamina propria in the presence of commensal bacteria. Conversely, severe immunopathology in T cell-mediated autoimmunity is mediated by Th17 cells that rely on IL-6 for their generation and maintenance. Recently, it has been discovered that the differentiation of these distinct T helper cell subsets may be linked to distinct signaling modalities of IL-6. Here, we summarize the current knowledge on the mode of action of IL-6 in the differentiation and maintenance of T cell subsets and propose that a context-dependent understanding of the impact of IL-6 on T cell subsets might inform rational IL-6-directed interventions in autoimmunity and chronic inflammation.
Repositioning TH cell polarization from single cytokines to complex help
S. Tuzlak, A. S. Dejean, M. Iannacone, F. J. Quintana, A. Waisman, F. Ginhoux, T. Korn, and B. Becher
Nat Immunol, 22(10): 1210-1217, 2021
Repositioning TH cell polarization from single cytokines to complex help
S. Tuzlak, A. S. Dejean, M. Iannacone, F. J. Quintana, A. Waisman, F. Ginhoux, T. Korn, and B. Becher
When helper T (TH) cell polarization was initially described three decades ago, the TH cell universe grew dramatically. New subsets were described based on their expression of few specific cytokines. Beyond TH1 and TH2 cells, this led to the coining of various TH17 and regulatory (Treg) cell subsets as well as TH22, TH25, follicular helper (TFH), TH3, TH5 and TH9 cells. High-dimensional single-cell analysis revealed that a categorization of TH cells into a single-cytokine-based nomenclature fails to capture the complexity and diversity of TH cells. Similar to the simple nomenclature used to describe innate lymphoid cells (ILCs), we propose that TH cell polarization should be categorized in terms of the help they provide to phagocytes (type 1), to B cells, eosinophils and mast cells (type 2) and to non-immune tissue cells, including the stroma and epithelium (type 3). Studying TH cells based on their helper function and the cells they help, rather than phenotypic features such as individual analyzed cytokines or transcription factors, better captures TH cell plasticity and conversion as well as the breadth of immune responses in vivo.
2020
Fatigue, depression, and pain in multiple sclerosis: How neuroinflammation translates into dysfunctional reward processing and anhedonic symptoms
H. Heitmann, T. F. M. Andlauer, T. Korn, M. Muhlau, P. Henningsen, B. Hemmer, and M. Ploner
Mult Scler, 2020
Fatigue, depression, and pain in multiple sclerosis: How neuroinflammation translates into dysfunctional reward processing and anhedonic symptoms
H. Heitmann, T. F. M. Andlauer, T. Korn, M. Muhlau, P. Henningsen, B. Hemmer, and M. Ploner
Fatigue, depression, and pain affect the majority of multiple sclerosis (MS) patients, which causes a substantial burden to patients and society. The pathophysiology of these symptoms is not entirely clear, and current treatments are only partially effective. Clinically, these symptoms share signs of anhedonia, such as reduced motivation and a lack of positive affect. In the brain, they are associated with overlapping structural and functional alterations in areas involved in reward processing. Moreover, neuroinflammation has been shown to directly impede monoaminergic neurotransmission that plays a key role in reward processing. Here, we review recent neuroimaging and neuroimmunological findings, which indicate that dysfunctional reward processing might represent a shared functional mechanism fostering the symptom cluster of fatigue, depression, and pain in MS. We propose a framework that integrates these findings with a focus on monoaminergic neurotransmission and discuss its therapeutic implications, limitations, and perspectives.
2019
Stability and Maintenance of Foxp3(+) Treg Cells in Non-lymphoid Microenvironments
T. Korn and A. Muschaweckh
Front Immunol, 10: 2634, 2019
Stability and Maintenance of Foxp3(+) Treg Cells in Non-lymphoid Microenvironments
T. Korn and A. Muschaweckh
Foxp3(+) Treg cells are indispensable for maintaining self-tolerance in secondary lymphoid organs (SLOs). However, Treg cells are also recruited to non-lymphoid tissues (NLTs) during inflammation. Recent advances in the understanding of Treg cell biology provided us with molecular mechanisms-both transcriptional and epigenetic-that enable Treg cells to retain their identity in an inflammatory milieu that is per se hostile to sustained expression of high levels of Foxp3. While Treg cells are recruited to sites of inflammation in order to resolve inflammation and re-establish appropriate organ function, it is increasingly recognized that a series of inflammatory (but also non-inflammatory) perturbations of organ function lead to the constitution of relatively long lived populations of Treg cells in NLTs. NLT Treg cells are heterogeneous according to their respective site of residence and it will be an important goal of future investigations to determine how these NLT Treg cells are maintained, e.g., what the role of antigen recognition by NLT Treg cells is and which growth factors are responsible for their self-renewal in the relative deficiency of IL-2. Finally, it is an open question what functions NLT Treg cells have besides their role in maintaining immunologic tolerance. In this review, we will highlight and summarize major ideas on the biology of NLT Treg cells (in the central nervous system but also at other peripheral sites) during inflammation and in steady state.
2018
Interleukin-6: designing specific therapeutics for a complex cytokine
C. Garbers, S. Heink, T. Korn, and S. Rose-John
Nat Rev Drug Discov, 2018
Interleukin-6: designing specific therapeutics for a complex cytokine
C. Garbers, S. Heink, T. Korn, and S. Rose-John
Interleukin-6 (IL-6) is a pivotal cytokine with a diverse repertoire of physiological functions that include regulation of immune cell proliferation and differentiation. Dysregulation of IL-6 signalling is associated with inflammatory and lymphoproliferative disorders such as rheumatoid arthritis and Castleman disease, and several classes of therapeutics have been developed that target components of the IL-6 signalling pathway. So far, monoclonal antibodies against IL-6 or IL-6 receptor (IL-6R) and Janus kinases (JAK) inhibitors have been successfully developed for the treatment of autoimmune diseases such as rheumatoid arthritis. However, clinical trials of agents targeting IL-6 signalling have also raised questions about the diseases and patient populations for which such agents have an appropriate benefit-risk profile. Knowledge from clinical trials and advances in our understanding of the complexities of IL-6 signalling, including the potential to target an IL-6 trans-signalling pathway, are now indicating novel opportunities for therapeutic intervention. In this Review, we overview the roles of IL-6 in health and disease and analyse progress with several approaches of inhibiting IL-6-signalling, with the aim of illuminating when and how to apply IL-6 blockade.
The Interleukin (IL)-23/T helper (Th)17 Axis in Experimental Autoimmune Encephalomyelitis and Multiple Sclerosis
M. Hiltensperger and T. Korn
Cold Spring Harb Perspect Med, 8(1), 2018
The Interleukin (IL)-23/T helper (Th)17 Axis in Experimental Autoimmune Encephalomyelitis and Multiple Sclerosis
M. Hiltensperger and T. Korn
T helper (Th)17 cells are responsible for host defense against fungi and certain extracellular bacteria but have also been reported to play a role in a variety of autoimmune diseases. Th17 cells respond to environmental cues, are very plastic, and might also be involved in tissue homeostasis and regeneration. The imprinting of pathogenic properties in Th17 cells in autoimmunity seems highly dependent on interleukin (IL)-23. Since Th17 cells were first described in experimental autoimmune encephalomyelitis, they have been suggested to also promote tissue damage in multiple sclerosis (MS). Indeed, some studies linked Th17 cells to disease severity in MS, and the efficacy of anti-IL-17A therapy in MS supported this idea. In this review, we will summarize molecular features of Th17 cells and discuss the evidence for their function in experimental models of autoimmune diseases and MS.
2017
T cell responses in the central nervous system
T. Korn and A. Kallies
Nat Rev Immunol, 2017
T cell responses in the central nervous system
T. Korn and A. Kallies
T cells are required for immune surveillance of the central nervous system (CNS); however, they can also induce severe immunopathology in the context of both viral infections and autoimmunity. The mechanisms that are involved in the priming and recruitment of T cells to the CNS are only partially understood, but there has been renewed interest in this topic since the 'rediscovery' of lymphatic drainage from the CNS. Moreover, tissue-resident memory T cells have been detected in the CNS and are increasingly recognized as an autonomous line of host defence. In this Review, we highlight the main mechanisms that are involved in the priming and CNS recruitment of CD4+ T cells, CD8+ T cells and regulatory T cells. We also consider the plasticity of T cell responses in the CNS, with a focus on viral infection and autoimmunity.
2016
From Leflunomide to Teriflunomide: Drug development and Immunosuppressive Oral Drugs in the Treatment of Multiple Sclerosis
L. Aly, B. Hemmer, and T. Korn
Curr Neuropharmacol, 2016
From Leflunomide to Teriflunomide: Drug development and Immunosuppressive Oral Drugs in the Treatment of Multiple Sclerosis
L. Aly, B. Hemmer, and T. Korn
Immunosuppressive drugs have been used in the treatment of multiple sclerosis (MS) for a long time. Today, the increased number of approved substances and the possibility of an oral availability of some immunomodulators improve the therapeutic repertory and increase patient satisfaction and compliance. Teriflunomide is indicated as first line oral disease modifying therapy (DMT) in relapsing-remitting MS (RRMS). Its immunosuppressive capacity results from an inhibition of de novo pyrimidine synthesis in rapidly proliferating lymphocytes. While Teriflunomide has been approved for the treatment of RRMS only since 2012, there is substantial therapeutic experience with its prodrug Leflunomide used in the treatment of rheumatoid arthritis (RA). In MS, a daily dose of 14 mg Teriflunomide reduces the annualized relapse rate (ARR) by more than 30% and disability progression by 30% compared to placebo while it provides a reasonable safety profile. This review presents an overview on oral immunosuppressants used in the treatment of MS. With an emphasis on Teriflunomide it summarizes discovery, mechanism of action and clinical effectiveness in phase II and III trials as well as important aspects for treating physicians.
Curr Neuropharmacol, 2016
Dendritic cells in central nervous system autoimmunity
C. Sie and T. Korn
Semin Immunopathol, 2016
Dendritic cells in central nervous system autoimmunity
C. Sie and T. Korn
Dendritic cells (DCs) operate at the intersection of the innate and adaptive immune systems. DCs can promote or inhibit adaptive immune responses against neuroantigens. While DC intrinsic properties, i.e., their maturation state or the subset they belong to, are important determinants of the outcome of an autoimmune reaction, tissue-specific cues might also be relevant for the function of DCs. Thus, a better understanding of the performance of distinct DC subsets in specific anatomical niches, not only in lymphoid tissue but also in non-lymphoid tissues such as the meninges, the choroid plexus, and the inflamed CNS parenchyma, will be instrumental for the design of immune intervention strategies to chronic inflammatory diseases that do not put at risk basic surveillance functions of the immune system in the CNS. Here, we will review modern concepts of DC biology in steady state and during autoimmune neuroinflammation.
2015
Role of the innate and adaptive immune responses in the course of multiple sclerosis
B. Hemmer, M. Kerschensteiner, and T. Korn
Lancet Neurol, 14(4): 406-19, 2015
Role of the innate and adaptive immune responses in the course of multiple sclerosis
B. Hemmer, M. Kerschensteiner, and T. Korn
Multiple sclerosis is a chronic disease of the CNS that leads to substantial disability in most patients. The early phase is characterised by relapses and the later phase by progressive disability. Results from immunological, genetic, and histopathological studies and treatment trials have shown that the immune system plays a key part in the disease course. Findings from animal models and immunological studies of patients with multiple sclerosis suggest a change in the involvement of the immune system during disease initiation and progression. These findings suggest that a peripheral immune response targeting the CNS drives the disease process during the early phase, whereas immune reactions within the CNS dominate the progressive phase. These concepts for the differential involvement of immune responses in the early and progressive phase of this disease have important implications for future research in the pathogenesis and treatment of multiple sclerosis.
[Current aspects of therapy conversion for multiple sclerosis]
P. Kolber, F. Luessi, S. G. Meuth, L. Klotz, T. Korn, C. Trebst, B. Tackenberg, B. Kieseier, T. Kumpfel, V. Fleischer, H. Tumani, B. Wildemann, M. Lang, P. Flachenecker, U. Meier, W. Bruck, V. Limmroth, A. Haghikia, H. P. Hartung, M. Stangel, R. Hohlfeld, B. Hemmer, R. Gold, H. Wiendl, and F. Zipp
Nervenarzt, 86(10): 1236-47, 2015
[Current aspects of therapy conversion for multiple sclerosis]
P. Kolber, F. Luessi, S. G. Meuth, L. Klotz, T. Korn, C. Trebst, B. Tackenberg, B. Kieseier, T. Kumpfel, V. Fleischer, H. Tumani, B. Wildemann, M. Lang, P. Flachenecker, U. Meier, W. Bruck, V. Limmroth, A. Haghikia, H. P. Hartung, M. Stangel, R. Hohlfeld, B. Hemmer, R. Gold, H. Wiendl, and F. Zipp
In recent years the approval of new substances has led to a substantial increase in the number of course-modifying immunotherapies available for multiple sclerosis. Therapy conversion therefore represents an increasing challenge. The treatment options sometimes show complex adverse effect profiles and necessitate a long-term and comprehensive monitoring. This article presents an overview of therapy conversion of immunotherapies for multiple sclerosis in accordance with the recommendations of the Disease-Related Competence Network for Multiple Sclerosis and the German Multiple Sclerosis Society as well as the guidelines on diagnostics and therapy for multiple sclerosis of the German Society of Neurology and the latest research results. At the present point in time it should be noted that no studies have been carried out for most of the approaches for therapy conversion given here; however, the recommendations are based on theoretical considerations and therefore correspond to recommendations at the level of expert consensus, which is currently essential for the clinical daily routine.
B cells assume the command
A. Waisman and T. Korn
Sci Transl Med, 7(310): 310fs42, 2015
2014
Novel monoclonal antibodies for therapy of multiple sclerosis
B. Knier, B. Hemmer, and T. Korn
Expert Opin Biol Ther, 14(4): 503-13, 2014
Novel monoclonal antibodies for therapy of multiple sclerosis
B. Knier, B. Hemmer, and T. Korn
INTRODUCTION: Monoclonal antibodies play an important role in the therapy of different autoimmune diseases. With the introduction of natalizumab, the importance of monoclonal antibodies in the therapy of multiple sclerosis (MS) has dramatically increased during the past years. AREAS COVERED: In this review, we will focus on newly approved and emerging antibodies for MS therapy. Based on published original articles and citable meeting abstracts, we will discuss their mode of action as well as data on efficacy and safety. EXPERT OPINION: Natalizumab was a breakthrough in MS therapy. However, side effects of this monoclonal antibody limit its use. The risk/benefit ratios of new biologicals in MS therapy are not yet clear. High-yield process daclizumab might qualify as first-line MS therapy, unless hepatotoxicity becomes a relevant safety concern. Alemtuzumab has been approved for MS therapy in Europe but will be reserved for selected patients with highly active disease due to frequent induction of potentially dangerous secondary autoimmune phenomena. Ocrelizumab will likely also be licensed as a second-line therapy in highly active MS. Neutralizing antibodies to interleukin (IL)-17A and blocking antibodies to leucine rich repeat and Ig domain containing 1 might be the most interesting upcoming new antibodies as both offer a new and pathophysiologically relevant approach in MS therapy.
Th17 cells in central nervous system autoimmunity
C. Sie, T. Korn, and M. Mitsdoerffer
Exp Neurol, 262 Pt A: 18-27, 2014
Th17 cells in central nervous system autoimmunity
C. Sie, T. Korn, and M. Mitsdoerffer
Multiple sclerosis (MS) is the most important autoimmune disease of the central nervous system (CNS). Its animal model experimental autoimmune encephalomyelitis (EAE) has been instrumental in defining the features of the novel T helper cell subset Th17. Conversely, the broad characterization of Th17 immune responses has substantially advanced our understanding of organ-specific autoimmunity and inspired almost a decade of immunological research. Here, we review the current knowledge on Th17 cells and their contribution to the immunopathology in EAE and MS, covering recent proceedings in the induction, modulation and effector mechanisms of this versatile T lymphocyte subset. In particular, we discuss the emerging role of mucosal immunity in the regulation of Th17 cells and CNS autoimmunity, the accumulating evidence for extensive plasticity in the Th17 subset, and their molecular mode of action in promoting this debilitating disease.
2013
Immunology of neuromyelitis optica: a T cell-B cell collaboration
M. Mitsdoerffer, V. Kuchroo, and T. Korn
Ann N Y Acad Sci, 1283: 57-66, 2013
Immunology of neuromyelitis optica: a T cell-B cell collaboration
M. Mitsdoerffer, V. Kuchroo, and T. Korn
Neuromyelitis optica (NMO) is a debilitating autoimmune inflammatory disease of the central nervous system (CNS) that is distinct from multiple sclerosis (MS). The discovery of NMO-immunoglobulin G (IgG) in the serum of NMO-but not MS-patients was a breakthrough in defining diagnostic criteria for NMO. NMO-IgG is an antibody directed against the astrocytic water channel protein aquaporin-4 (AQP4). While there is evidence that NMO-IgG is also involved in mediating tissue damage in the CNS, many aspects of the pathogenic cascade in NMO remain to be determined. It is clear that antigen-specific T cells contribute to the generation of NMO-IgG in the peripheral immune compartment, as well as to the development of NMO lesions in the CNS. T helper 17 (Th17) cells, equipped both in providing B cell help and inducing tissue inflammation, may be involved in NMO development and pathogenesis. Here, we review immunologic aspects of NMO, placing recent findings in the biology of T-B cell cooperation into perspective with autoimmunity of the CNS.
2012
Immune mechanisms of new therapeutic strategies in MS: teriflunomide
M. C. Claussen and T. Korn
Clin Immunol, 142(1): 49-56, 2012
Immune mechanisms of new therapeutic strategies in MS: teriflunomide
M. C. Claussen and T. Korn
At present, a series of oral disease-modifying agents is being introduced for the treatment of multiple sclerosis. With the exception of laquinimod, the "new" oral compounds have already been approved for other indications such as organ transplantation (FTY720), psoriasis (dimethylfumarate), hairy cell leukemia (cladribine), and rheumatoid arthritis (leflunomide). Leflunomide is the prodrug of teriflunomide which is the latest compound that has successfully been tested in a large phase III clinical trial in relapsing MS. Due to its favorable safety profile and its efficacy in rheumatoid arthritis where the aberrant immune response is in various aspects similar to the autoimmune reaction in MS patients, teriflunomide is a promising treatment option for MS patients. Here, we review the most important cell biological and immunological modes of action of teriflunomide, report on the available data on its pharmacokinetics in humans, and summarize the recent clinical trials of teriflunomide in relapsing MS.
Development and function of interleukin 17-producing γδ T cells
T. Korn and F. Petermann
Ann N Y Acad Sci, 1247: 34-45, 2012
Development and function of interleukin 17-producing γδ T cells
T. Korn and F. Petermann
Interleukin (IL) 17 is a phylogenetically ancient cytokine that has been adopted by the adaptive immune system, and the investigation of adaptive T helper (Th) 17 cells has substantially contributed to our understanding of the molecular requirements for the induction, regulation, and function of IL-17. However, IL-17 is in fact produced by a large variety of innate immune cells and exerts its most significant biological functions at the interface of the organism with its environment, such as, for example, at epithelial surfaces, where γδ T cells are a prominent source of IL-17. In this review, we will give an overview on the concepts of commitment of γδ T cells to effector phenotypes, focusing on IL-17-producing γδ T cells (γδT17 cells). The role of γδT17 cells in animal models of autoimmunity will be discussed as well as the prerequisites for the development of human γδT17 cells and their potential importance for human disease conditions.
2011
Cytokines and effector T cell subsets causing autoimmune CNS disease
F. Petermann and T. Korn
FEBS Lett, 585(23): 3747-57, 2011
Cytokines and effector T cell subsets causing autoimmune CNS disease
F. Petermann and T. Korn
Although experimental autoimmune encephalomyelitis (EAE) is limited in its potency to reproduce the entirety of clinical and histopathologic features of multiple sclerosis (MS), this model has been successfully used to prove that MS like autoimmunity in the CNS is orchestrated by autoantigen specific T cells. EAE was also very useful to refute the idea that IFN-γ producing T helper type 1 (Th1) cells were the sole players within the pathogenic T cell response. Rather, "new" T cell lineages such as IL-17 producing Th17 cells or IL-9 producing Th9 cells have been first discovered in the context of EAE. Here, we will summarize new concepts of early and late T cell plasticity and the cytokine network that shapes T helper cell responses and lesion development in CNS specific autoimmunity.
2010
How T cells take developmental decisions by using the aryl hydrocarbon receptor to sense the environment
T. Korn
Proc Natl Acad Sci U S A, 107(48): 20597-8, 2010
Immunological basis for the development of tissue inflammation and organ-specific autoimmunity in animal models of multiple sclerosis
T. Korn, M. Mitsdoerffer, and V. K. Kuchroo
Results Probl Cell Differ, 51: 43-74, 2010
Immunological basis for the development of tissue inflammation and organ-specific autoimmunity in animal models of multiple sclerosis
T. Korn, M. Mitsdoerffer, and V. K. Kuchroo
Experimental autoimmune encephalomyelitis (EAE) is an animal model for multiple sclerosis (MS) that has shaped our understanding of autoimmune tissue inflammation in the central nervous system (CNS). Major therapeutic approaches to MS have been first validated in EAE. Nevertheless, EAE in all its modifications is not able to recapitulate the full range of clinical and histopathogenic aspects of MS. Furthermore, autoimmune reactions in EAE-prone rodent strains and MS patients may differ in terms of the relative involvement of various subsets of immune cells. However, the role of specific molecules that play a role in skewing the immune response towards pathogenic autoreactivity is very similar in mice and humans. Thus, in this chapter, we will focus on the identification of a novel subset of inflammatory T cells, called Th17 cells, in EAE and their interplay with other immune cells including protective regulatory T cells (T-regs). It is likely that the discovery of Th17 cells and their relationship with T-regs will change our understanding of organ-specific autoimmune diseases in the years to come.
2009
IL-17 and Th17 Cells
T. Korn, E. Bettelli, M. Oukka, and V. K. Kuchroo
Annu Rev Immunol, 27: 485-517, 2009
IL-17 and Th17 Cells
T. Korn, E. Bettelli, M. Oukka, and V. K. Kuchroo
CD4+ T cells, upon activation and expansion, develop into different T helper cell subsets with different cytokine profiles and distinct effector functions. Until recently, T cells were divided into Th1 or Th2 cells, depending on the cytokines they produce. A third subset of IL-17-producing effector T helper cells, called Th17 cells, has now been discovered and characterized. Here, we summarize the current information on the differentiation and effector functions of the Th17 lineage. Th17 cells produce IL-17, IL-17F, and IL-22, thereby inducing a massive tissue reaction owing to the broad distribution of the IL-17 and IL-22 receptors. Th17 cells also secrete IL-21 to communicate with the cells of the immune system. The differentiation factors (TGF-β plus IL-6 or IL-21), the growth and stabilization factor (IL-23), and the transcription factors (STAT3, RORγt, and RORα) involved in the development of Th17 cells have just been identified. The participation of TGF-β in the differentiation of Th17 cells places the Th17 lineage in close relationship with CD4+CD25+Foxp3+ regulatory T cells (Tregs), as TGF-β also induces differentiation of naive T cells into Foxp3+ Tregs in the peripheral immune compartment. The investigation of the differentiation, effector function, and regulation of Th17 cells has opened up a new framework for understanding T cell differentiation. Furthermore, we now appreciate the importance of Th17 cells in clearing pathogens during host defense reactions and in inducing tissue inflammation in autoimmune disease.
2008
Induction and effector functions of T(H)17 cells
E. Bettelli, T. Korn, M. Oukka, and V. K. Kuchroo
Nature, 453(7198): 1051-7, 2008
Induction and effector functions of T(H)17 cells
E. Bettelli, T. Korn, M. Oukka, and V. K. Kuchroo
T helper (T(H)) cells constitute an important arm of the adaptive immune system because they coordinate defence against specific pathogens, and their unique cytokines and effector functions mediate different types of tissue inflammation. The recently discovered T(H)17 cells, the third subset of effector T helper cells, have been the subject of intense research aimed at understanding their role in immunity and disease. Here we review emerging data suggesting that T(H)17 cells have an important role in host defence against specific pathogens and are potent inducers of autoimmunity and tissue inflammation. In addition, the differentiation factors responsible for their generation have revealed an interesting reciprocal relationship with regulatory T (T(reg)) cells, which prevent tissue inflammation and mediate self-tolerance.
Role of Th1 and Th17 cells in organ-specific autoimmunity
V. Dardalhon, T. Korn, V. K. Kuchroo, and A. C. Anderson
J Autoimmun, 31(3): 252-6, 2008
Role of Th1 and Th17 cells in organ-specific autoimmunity
V. Dardalhon, T. Korn, V. K. Kuchroo, and A. C. Anderson
CD4(+) IFN-γ-producing Th1 cells have long been associated with the pathogenesis of many organ-specific autoimmune diseases; however, the observation of disease in mice deficient in molecules involved in Th1 cell differentiation raised the possibility that other effector T cells were responsible for inducing autoimmunity. Recently, a new CD4(+) effector T cell subset that produces IL-17 (Th17) has emerged. The fact that Th17 cells are highly auto-pathogenic has fueled a debate as to what role, if any, Th1 cells play in the induction of tissue inflammation and autoimmune disease. This review will discuss the respective roles of the Th1 and Th17 subsets in organ-specific autoimmunity.
Pathophysiology of multiple sclerosis
T. Korn
J Neurol, 255 Suppl 6: 2-6, 2008
Pathophysiology of multiple sclerosis
T. Korn
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system (CNS). Both genetic and environmental causes for MS have been suggested. Recent genome-wide association studies revealed new susceptibility alleles for MS besides the HLA complex that are all related with immune functions. Whereas there is little evidence to support a purely environmental trigger for the disease in the sense of an infectious agent, the autoimmune hypothesis of MS is well established. Myelin antigen-specific CD4+ T cells become activated in the peripheral immune compartment, cross the blood-brain barrier and trigger the disease. Here, current concepts of the commitment of T cells to pro-inflammatory effector T helper cell lineages including Th17 cells that appear to be important inducers of organ-specific autoimmunity will be discussed.
2007
Th17: the third member of the effector T cell trilogy
E. Bettelli, T. Korn, and V. K. Kuchroo
Curr Opin Immunol, 19(6): 652-7, 2007
Th17: the third member of the effector T cell trilogy
E. Bettelli, T. Korn, and V. K. Kuchroo
T helper responses have now grown to include three T cell subsets: Th1, Th2 and Th17. Th17 cells have recently emerged as a third independent T cell subset that may play an essential role in protection against certain extracellular pathogens. However, Th17 cells with specificity for self-antigens are highly pathogenic and lead to the development of inflammation and severe autoimmunity. A combination of TGF-β plus IL-6 and the transcription factors STAT3 and RORγt were recently described to be essential for initial differentiation of Th17 cells and IL-23 for the later stabilization of the Th17 cell subset. Here, we introduce another player IL-21 produced by Th17 themselves, which plays an important role in the amplification of Th17 cells. Thus, Th17 cells may undergo three distinct steps of development: differentiation, amplification and stabilization in which distinct cytokines play a role.
The dynamics of effector T cells and Foxp3+ regulatory T cells in the promotion and regulation of autoimmune encephalomyelitis
T. Korn, A. C. Anderson, E. Bettelli, and M. Oukka
J Neuroimmunol, 191(1-2): 51-60, 2007
The dynamics of effector T cells and Foxp3+ regulatory T cells in the promotion and regulation of autoimmune encephalomyelitis
T. Korn, A. C. Anderson, E. Bettelli, and M. Oukka
The Th1/Th2 paradigm of T helper cell subsets had to be revised when IL-17 producing T cells (Th17) were identified as a distinct T helper cell lineage. Th17 cells are very efficient inducers of tissue inflammation and crucial initiators of organ-specific autoimmunity. Whereas Th17 cells promote autoimmune tissue inflammation, Foxp3+ regulatory T cells (T-reg) are necessary and sufficient to prevent autoimmunity throughout the life span of an individual. Here, we review recent findings of how responses of effector T cells and T-reg cells with a defined antigen-specificity develop in autoimmune encephalomyelitis. Moreover, Th17 cells and Foxp3+ T-reg seem to be dichotomously related in that TGF-β induces Foxp3 in naive T cells, but TGF-β and IL-6 together drive the generation of Th17 cells. Thus, we give an overview of how Th17 cells, induced Foxp3+ T-reg, as well as how naturally occurring T-reg cells might cooperate to promote and regulate autoimmune inflammation of the central nervous system (CNS). The monitoring of the population dynamics of these T cell subsets in reporter mice in vivo will enable us to revisit the pathogenic concept of autoimmune inflammation in the CNS and design rational and phase-specific therapeutic interventions.
Dynamics of antigen-specific regulatory T-cells in the context of autoimmunity
T. Korn and M. Oukka
Semin Immunol, 19(4): 272-8, 2007
Dynamics of antigen-specific regulatory T-cells in the context of autoimmunity
T. Korn and M. Oukka
Naturally occurring regulatory T-cells (nT-reg) are crucial in preventing generalized autoimmunity. However, antigen-specific T-reg responses during organ-specific autoimmunity have not been characterized until recently when novel tools including MHC class II-tetramers and Foxp3gfp "knock-in" reporter mice (Foxp3gfp.KI) became available. In this review, we introduce the concept of functional compartmentalisation in that T-reg may behave differently when they maintain peripheral tolerance in the secondary lymphoid tissue in a non-inflammatory environment as compared to when they are expanded and targeted to the site of inflammation following an autoantigen-specific, immunogenic stimulus.
Th17 cells: effector T cells with inflammatory properties
T. Korn, M. Oukka, V. Kuchroo, and E. Bettelli
Semin Immunol, 19(6): 362-71, 2007
Th17 cells: effector T cells with inflammatory properties
T. Korn, M. Oukka, V. Kuchroo, and E. Bettelli
Upon activation, naive CD4(+) T cells differentiate into effector T cells with specific effector functions and cytokine profiles. The Th1/Th2 paradigm has recently been reevaluated to include a third population of T helper cells, producing IL-17 and designated Th17. The differentiation of Th17 cells requires the coordinate and specific action of the proinflammatory cytokine IL-6 and the immunosuppressive cytokine TGF-β. In addition, the IL-12 family member IL-23 is involved in the maintenance of these cells. Analogous to other T helper cell subsets, Th17 commitment is initiated by sequential involvement of STAT molecules, i.e. STAT3 downstream of cytokine receptors, and specific transcription factors, i.e. ROR-γt. Recent data also support the existence of a complex network of cytokines regulating Th17 cells. Clearly, the specific effector functions of Th17 cells expand beyond previously described effects of Th1 and Th2 immunity, with specific roles in host defense against certain pathogens and in organ-specific autoimmunity. The potential dynamics of Th17 cell populations and their interplay with other inflammatory cells in the induction of tissue inflammation in host defense and organ-specific autoimmunity are discussed.
Autoimmune modulation of astrocyte-mediated homeostasis
T. Korn, M. Rao, and T. Magnus
Neuromolecular Med, 9(1): 1-16, 2007
Autoimmune modulation of astrocyte-mediated homeostasis
T. Korn, M. Rao, and T. Magnus
Astrocytes are principal mediators of homeostasis in the central nervous system (CNS). They supply neurons and oligodendrocytes with substrates for energy metabolism and clear the extracellular space of excess neurotransmitters. In neuroinflammation, astrocytes have classically been regarded as unimportant since their capacity to present antigen to T cells is limited and has been questioned in vivo. However, it is an evolving concept that autoimmunity in the CNS has a profound impact on astrocytes. In this review, we focus on the alterations in astrocyte functions that occur during an autoimmune attack of the CNS.
Neuromolecular Med, 9(1): 1-16, 2007