1
|
Nelson EA, Tyler AL, Lakusta-Wong T, Lahue KG, Hankes KC, Teuscher C, Lynch RM, Ferris MT, Mahoney JM, Krementsov DN. Analysis of CNS autoimmunity in genetically diverse mice reveals unique phenotypes and mechanisms. JCI Insight 2024; 9:e184138. [PMID: 39325545 PMCID: PMC11601571 DOI: 10.1172/jci.insight.184138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024] Open
Abstract
Multiple sclerosis (MS) is a complex disease with significant heterogeneity in disease course and progression. Genetic studies have identified numerous loci associated with MS risk, but the genetic basis of disease progression remains elusive. To address this, we leveraged the Collaborative Cross (CC), a genetically diverse mouse strain panel, and experimental autoimmune encephalomyelitis (EAE). The 32 CC strains studied captured a wide spectrum of EAE severity, trajectory, and presentation, including severe-progressive, monophasic, relapsing remitting, and axial rotary-EAE (AR-EAE), accompanied by distinct immunopathology. Sex differences in EAE severity were observed in 6 strains. Quantitative trait locus analysis revealed distinct genetic linkage patterns for different EAE phenotypes, including EAE severity and incidence of AR-EAE. Machine learning-based approaches prioritized candidate genes for loci underlying EAE severity (Abcc4 and Gpc6) and AR-EAE (Yap1 and Dync2h1). This work expands the EAE phenotypic repertoire and identifies potentially novel loci controlling unique EAE phenotypes, supporting the hypothesis that heterogeneity in MS disease course is driven by genetic variation.
Collapse
Affiliation(s)
- Emily A. Nelson
- Department of Biomedical and Health Sciences, University of Vermont (UVM), Burlington, Vermont, USA
| | | | | | - Karolyn G. Lahue
- Department of Biomedical and Health Sciences, University of Vermont (UVM), Burlington, Vermont, USA
| | - Katherine C. Hankes
- Department of Biomedical and Health Sciences, University of Vermont (UVM), Burlington, Vermont, USA
| | - Cory Teuscher
- Department of Medicine, UVM, Larner College of Medicine, Burlington, Vermont, USA
| | - Rachel M. Lynch
- Department of Genetics, University of North Carolina at Chapel Hill (UNC), Chapel Hill, North Carolina, USA
| | - Martin T. Ferris
- Department of Genetics, University of North Carolina at Chapel Hill (UNC), Chapel Hill, North Carolina, USA
| | - J. Matthew Mahoney
- The Jackson Laboratory, Bar Harbor, Maine, USA
- Department of Neurological Sciences and
| | - Dimitry N. Krementsov
- Department of Biomedical and Health Sciences, University of Vermont (UVM), Burlington, Vermont, USA
| |
Collapse
|
2
|
Holt EA, Tyler A, Lakusta-Wong T, Lahue KG, Hankes KC, Teuscher C, Lynch RM, Ferris MT, Mahoney JM, Krementsov DN. Probing the basis of disease heterogeneity in multiple sclerosis using genetically diverse mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.03.597205. [PMID: 38895248 PMCID: PMC11185616 DOI: 10.1101/2024.06.03.597205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Multiple sclerosis (MS) is a complex disease with significant heterogeneity in disease course and progression. Genetic studies have identified numerous loci associated with MS risk, but the genetic basis of disease progression remains elusive. To address this, we leveraged the Collaborative Cross (CC), a genetically diverse mouse strain panel, and experimental autoimmune encephalomyelitis (EAE). The thirty-two CC strains studied captured a wide spectrum of EAE severity, trajectory, and presentation, including severe-progressive, monophasic, relapsing remitting, and axial rotary (AR)-EAE, accompanied by distinct immunopathology. Sex differences in EAE severity were observed in six strains. Quantitative trait locus analysis revealed distinct genetic linkage patterns for different EAE phenotypes, including EAE severity and incidence of AR-EAE. Machine learning-based approaches prioritized candidate genes for loci underlying EAE severity ( Abcc4 and Gpc6 ) and AR-EAE ( Yap1 and Dync2h1 ). This work expands the EAE phenotypic repertoire and identifies novel loci controlling unique EAE phenotypes, supporting the hypothesis that heterogeneity in MS disease course is driven by genetic variation. Summary The genetic basis of disease heterogeneity in multiple sclerosis (MS) remains elusive. We leveraged the Collaborative Cross to expand the phenotypic repertoire of the experimental autoimmune encephalomyelitis (EAE) model of MS and identify loci controlling EAE severity, trajectory, and presentation.
Collapse
|
3
|
Bontempi P, Piccolantonio G, Busato A, Conti A, Angelini G, Lopez N, Bani A, Constantin G, Marzola P. Resting-state functional magnetic resonance imaging reveals functional connectivity alteration in the experimental autoimmune encephalomyelitis model of multiple sclerosis. NMR IN BIOMEDICINE 2024; 37:e5127. [PMID: 38450807 DOI: 10.1002/nbm.5127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 01/08/2024] [Accepted: 01/25/2024] [Indexed: 03/08/2024]
Abstract
Multiple sclerosis (MS) is an autoimmune degenerative disease targeting white matter in the central nervous system. The most common animal model that mimics MS is experimental autoimmune encephalomyelitis (EAE) and it plays a crucial role in pharmacological research, from the identification of a therapeutic target to the in vivo validation of efficacy. Magnetic resonance imaging (MRI) is largely used to detect MS lesions, and resting-state functional MRI (rsfMRI) to investigate alterations in the brain functional connectivity (FC). MRI was mainly used in EAE studies to detect lesions in the spinal cord and brain. The current longitudinal MRI study aims to validate rsfMRI as a biomarker of the disease progression in the myelin oligodendrocyte glycoprotein 35-55 induced EAE animal model of MS. MR images were acquired 14, 25, and 50 days postimmunization. Seed-based analysis was used to investigate the whole-brain FC with some predefined areas, such as the thalamic regions, cerebellum, motor and somatosensory cortex. When compared with the control group, the EAE group exhibited a slightly altered FC and a decreasing trend in the total number of activated voxels along the disease progression. The most interesting result regards the whole-brain FC with the cerebellum. A hyperconnectivity behavior was found at an early phase and a significant reduced connectivity at a late phase. Moreover, we found a negative correlation between the total number of activated voxels during the late phase and the cumulative disease index. The results obtained provide a clinically relevant experimental platform that may be pivotal for the elucidation of the key mechanisms of accumulation of irreversible disability, as well as the development of innovative therapies for MS. Moreover, the negative correlation between the disease severity and the size of the activated area suggests a possible research pathway to follow for the resolution of the clinico-radiological paradox.
Collapse
Affiliation(s)
- Pietro Bontempi
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Giusi Piccolantonio
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Alice Busato
- Department of Computer Science, University of Verona, Verona, Italy
- Evotec Company, Verona, Italy
| | - Anita Conti
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | | | - Nicola Lopez
- Department of Medicine, University of Verona, Verona, Italy
| | | | | | - Pasquina Marzola
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| |
Collapse
|
4
|
Holt EA, Waytashek CM, Sessions KJ, Asarian L, Lahue KG, Usherwood EJ, Teuscher C, Krementsov DN. Host Genetic Variation Has a Profound Impact on Immune Responses Mediating Control of Viral Load in Chronic Gammaherpesvirus Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1526-1539. [PMID: 37819784 PMCID: PMC10841120 DOI: 10.4049/jimmunol.2300294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 09/14/2023] [Indexed: 10/13/2023]
Abstract
Chronic infection with the gammaherpesvirus EBV is a risk factor for several autoimmune diseases, and poor control of EBV viral load and enhanced anti-EBV responses elevate this risk further. However, the role of host genetic variation in the regulation of immune responses to chronic gammaherpesvirus infection and control of viral replication remains unclear. To address this question, we infected C57BL/6J (B6) and genetically divergent wild-derived inbred PWD/PhJ (PWD) mice with murine gammaherpesvirus-68 (MHV-68), a gammaherpesvirus similar to EBV, and determined the effect of latent gammaherpesvirus infection on the CD4 T cell transcriptome. Chronic MHV-68 infection of B6 mice resulted in a dramatic upregulation of genes characteristic of a cytotoxic Th cell phenotype, including Gzmb, Cx3cr1, Klrg1, and Nkg7, a response that was highly muted in PWD mice. Flow cytometric analyses revealed an expansion of CX3CR1+KLRG1+ cytotoxic Th cell-like cells in B6 but not PWD mice. Analysis of MHV-68 replication demonstrated that in spite of muted adaptive responses, PWD mice had superior control of viral load in lymphoid tissue, despite an absence of a defect in MHV-68 in vitro replication in PWD macrophages. Depletion of NK cells in PWD mice, but not B6 mice, resulted in elevated viral load, suggesting genotype-dependent NK cell involvement in MHV-68 control. Taken together, our findings demonstrate that host genetic variation can regulate control of gammaherpesvirus replication through disparate immunological mechanisms, resulting in divergent long-term immunological sequelae during chronic infection.
Collapse
Affiliation(s)
- Emily A. Holt
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05405, USA
| | - Courtney M. Waytashek
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05405, USA
| | - Katherine J. Sessions
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05405, USA
| | - Loredana Asarian
- Department of Medicine, Vermont Center for Immunology and Infectious Diseases, Larner College of Medicine, The University of Vermont, Burlington, VT 05405, USA
| | - Karolyn G Lahue
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05405, USA
| | - Edward J. Usherwood
- Department of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth College, Lebanon, NH 03756, USA
| | - Cory Teuscher
- Department of Medicine, Vermont Center for Immunology and Infectious Diseases, Larner College of Medicine, The University of Vermont, Burlington, VT 05405, USA
| | - Dimitry N. Krementsov
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05405, USA
| |
Collapse
|
5
|
Gomi M, Nakayama Y, Sakurai Y, Oyama R, Iwasaki K, Doi M, Liu Y, Hori M, Watanabe H, Hashimoto K, Tanaka H, Tange K, Nakai Y, Akita H. Tolerogenic Lipid Nanoparticles for Delivering Self-Antigen mRNA for the Treatment of Experimental Autoimmune Encephalomyelitis. Pharmaceuticals (Basel) 2023; 16:1270. [PMID: 37765078 PMCID: PMC10537621 DOI: 10.3390/ph16091270] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Multiple sclerosis is a disease caused by autoantigen-responsive immune cells that disrupt the myelin in the central nervous system (CNS). Although immunosuppressive drugs are used to suppress symptoms, no definitive therapy exists. As in the experimental autoimmune encephalitis (EAE) model of multiple sclerosis, a partial sequence of the myelin oligodendrocyte glycoprotein (MOG35-55) was identified as a causative autoantigen. This suggests that the induction of immune tolerance that is specific to MOG35-55 would be a fundamental treatment for EAE. We previously reported that lipid nanoparticles (LNPs) containing an anionic phospholipid, phosphatidylserine (PS), in their lipid composition, can be used to deliver mRNA and that this leads to proteins of interest to be expressed in the spleen. In addition to the targeting capability of PS, PS molecules avoid activating the immune system. Physiologically, the recognition of PS on apoptotic cells suppresses immune activation against these cells by releasing cytokines, such as interleukin-10 (IL-10) and transforming growth factor (TGF)-β that negatively regulate immunity. In this study, we tested whether mRNA delivery of autoantigens to the spleen by PS-LNPs causes the expression of MOG35-55 antigens with minimal immune stimulation and whether this could be used to treat an EAE model by inducing immune tolerance.
Collapse
Affiliation(s)
- Masaki Gomi
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba 260-0856, Japan
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Yuka Nakayama
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Yu Sakurai
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Ryotaro Oyama
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba 260-0856, Japan
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Koki Iwasaki
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba 260-0856, Japan
| | - Mizuki Doi
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba 260-0856, Japan
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Yi Liu
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba 260-0856, Japan
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Mizuho Hori
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Himeka Watanabe
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Kohei Hashimoto
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Hiroki Tanaka
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba 260-0856, Japan
| | - Kota Tange
- Life Science Research Laboratory, NOF CORPORATION, 3-3, Chidoricho, Kawasaki-ku, Kawasaki 210-0865, Japan
| | - Yuta Nakai
- Life Science Research Laboratory, NOF CORPORATION, 3-3, Chidoricho, Kawasaki-ku, Kawasaki 210-0865, Japan
| | - Hidetaka Akita
- Laboratory of DDS Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| |
Collapse
|
6
|
McGill MM, Richman AR, Boyd JR, Sabikunnahar B, Lahue KG, Montgomery TL, Caldwell S, Varnum S, Frietze S, Krementsov DN. p38 MAP Kinase Signaling in Microglia Plays a Sex-Specific Protective Role in CNS Autoimmunity and Regulates Microglial Transcriptional States. Front Immunol 2021; 12:715311. [PMID: 34707603 PMCID: PMC8542909 DOI: 10.3389/fimmu.2021.715311] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/17/2021] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune demyelinating disease of the central nervous system, representing the leading cause of non-traumatic neurologic disease in young adults. This disease is three times more common in women, yet more severe in men, but the mechanisms underlying these sex differences remain largely unknown. MS is initiated by autoreactive T helper cells, but CNS-resident and CNS-infiltrating myeloid cells are the key proximal effector cells regulating disease pathology. We have previously shown that genetic ablation of p38α MAP kinase broadly in the myeloid lineage is protective in the autoimmune model of MS, experimental autoimmune encephalomyelitis (EAE), but only in females, and not males. To precisely define the mechanisms responsible, we used multiple genetic approaches and bone marrow chimeras to ablate p38α in microglial cells, peripheral myeloid cells, or both. Deletion of p38α in both cell types recapitulated the previous sex difference, with reduced EAE severity in females. Unexpectedly, deletion of p38α in the periphery was protective in both sexes. In contrast, deletion of p38α in microglia exacerbated EAE in males only, revealing opposing roles of p38α in microglia vs. periphery. Bulk transcriptional profiling revealed that p38α regulated the expression of distinct gene modules in male vs. female microglia. Single-cell transcriptional analysis of WT and p38α-deficient microglia isolated from the inflamed CNS revealed a diversity of complex microglial states, connected by distinct convergent transcriptional trajectories. In males, microglial p38α deficiency resulted in enhanced transition from homeostatic to disease-associated microglial states, with the downregulation of regulatory genes such as Atf3, Rgs1, Socs3, and Btg2, and increased expression of inflammatory genes such as Cd74, Trem2, and MHC class I and II genes. In females, the effect of p38α deficiency was divergent, exhibiting a unique transcriptional profile that included an upregulation of tissue protective genes, and a small subset of inflammatory genes that were also upregulated in males. Taken together, these results reveal a p38α-dependent sex-specific molecular pathway in microglia that is protective in CNS autoimmunity in males, suggesting that autoimmunity in males and females is driven by distinct cellular and molecular pathways, thus suggesting design of future sex-specific therapeutic approaches.
Collapse
Affiliation(s)
- Mahalia M McGill
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Alyssa R Richman
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Joseph R Boyd
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Bristy Sabikunnahar
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Karolyn G Lahue
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Theresa L Montgomery
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Sydney Caldwell
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Stella Varnum
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Seth Frietze
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| | - Dimitry N Krementsov
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States
| |
Collapse
|
7
|
Lahue KG, Lara MK, Linton AA, Lavoie B, Fang Q, McGill MM, Crothers JW, Teuscher C, Mawe GM, Tyler AL, Mahoney JM, Krementsov DN. Identification of novel loci controlling inflammatory bowel disease susceptibility utilizing the genetic diversity of wild-derived mice. Genes Immun 2020; 21:311-325. [PMID: 32848229 PMCID: PMC7657953 DOI: 10.1038/s41435-020-00110-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 08/06/2020] [Accepted: 08/13/2020] [Indexed: 12/14/2022]
Abstract
Inflammatory bowel disease (IBD) is a complex disorder that imposes a growing health burden. Multiple genetic associations have been identified in IBD, but the mechanisms underlying many of these associations are poorly understood. Animal models are needed to bridge this gap, but conventional laboratory mouse strains lack the genetic diversity of human populations. To more accurately model human genetic diversity, we utilized a panel of chromosome (Chr) substitution strains, carrying chromosomes from the wild-derived and genetically divergent PWD/PhJ (PWD) strain on the commonly used C57BL/6J (B6) background, as well as their parental B6 and PWD strains. Two models of IBD were used, TNBS- and DSS-induced colitis. Compared with B6 mice, PWD mice were highly susceptible to TNBS-induced colitis, but resistant to DSS-induced colitis. Using consomic mice, we identified several PWD-derived loci that exhibited profound effects on IBD susceptibility. The most pronounced of these were loci on Chr1 and Chr2, which yielded high susceptibility in both IBD models, each acting at distinct phases of the disease. Leveraging transcriptomic data from B6 and PWD immune cells, together with a machine learning approach incorporating human IBD genetic associations, we identified lead candidate genes, including Itga4, Pip4k2a, Lcn10, Lgmn, and Gpr65.
Collapse
Affiliation(s)
- Karolyn G Lahue
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, 05405, USA
| | - Montana K Lara
- Department of Neurological Sciences, University of Vermont, Burlington, VT, 05405, USA
| | - Alisha A Linton
- Department of Neurological Sciences, University of Vermont, Burlington, VT, 05405, USA
| | - Brigitte Lavoie
- Department of Neurological Sciences, University of Vermont, Burlington, VT, 05405, USA
| | - Qian Fang
- Department of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Mahalia M McGill
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, 05405, USA
| | - Jessica W Crothers
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Cory Teuscher
- Department of Medicine, University of Vermont, Burlington, VT, 05405, USA
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Gary M Mawe
- Department of Neurological Sciences, University of Vermont, Burlington, VT, 05405, USA
| | - Anna L Tyler
- The Jackson Laboratory, 600 Main St., Bar Harbor, ME, 04609, USA
| | - J Matthew Mahoney
- Department of Neurological Sciences, University of Vermont, Burlington, VT, 05405, USA
- Department of Computer Science University of Vermont, Burlington, VT, 05405, USA
| | - Dimitry N Krementsov
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, 05405, USA.
| |
Collapse
|
8
|
Faber H, Kurtoic D, Krishnamoorthy G, Weber P, Pütz B, Müller-Myhsok B, Weber F, Andlauer TFM. Gene Expression in Spontaneous Experimental Autoimmune Encephalomyelitis Is Linked to Human Multiple Sclerosis Risk Genes. Front Immunol 2020; 11:2165. [PMID: 33072080 PMCID: PMC7531036 DOI: 10.3389/fimmu.2020.02165] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/10/2020] [Indexed: 12/23/2022] Open
Abstract
Recent genome-wide association studies have identified over 230 genetic risk loci for multiple sclerosis. Current experimental autoimmune encephalomyelitis (EAE) models requiring active induction of disease may not be optimally suited for the characterization of the function of these genes. We have thus used gene expression profiling to study whether spontaneous opticospinal EAE (OSE) or MOG-induced EAE mirrors the genetic contribution to the pathogenesis of multiple sclerosis more faithfully. To this end, we compared gene expression in OSE and MOG EAE models and analyzed the relationship of both models to human multiple sclerosis risk genes and T helper cell biology. We observed stronger gene expression changes and an involvement of more pathways of the adaptive immune system in OSE than MOG EAE. Furthermore, we demonstrated a more extensive enrichment of human MS risk genes among transcripts differentially expressed in OSE than was the case for MOG EAE. Transcripts differentially expressed only in diseased OSE mice but not in MOG EAE were significantly enriched for T helper cell-specific transcripts. These transcripts are part of immune-regulatory pathways. The activation of the adaptive immune system and the enrichment of both human multiple sclerosis risk genes and T helper cell-specific transcripts were also observed in OSE mice showing only mild disease signs. These expression changes may, therefore, be indicative of processes at disease onset. In summary, more human multiple sclerosis risk genes were differentially expressed in OSE than was observed for MOG EAE, especially in TH1 cells. When studying the functional role of multiple sclerosis risk genes and pathways during disease onset and their interactions with the environment, spontaneous OSE may thus show advantages over MOG-induced EAE.
Collapse
Affiliation(s)
- Hans Faber
- Max Planck Institute of Psychiatry, Munich, Germany
| | | | | | - Peter Weber
- Max Planck Institute of Psychiatry, Munich, Germany
| | - Benno Pütz
- Max Planck Institute of Psychiatry, Munich, Germany
| | - Bertram Müller-Myhsok
- Max Planck Institute of Psychiatry, Munich, Germany.,Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Frank Weber
- Max Planck Institute of Psychiatry, Munich, Germany
| | - Till F M Andlauer
- Max Planck Institute of Psychiatry, Munich, Germany.,Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
9
|
Saligrama N, Zhao F, Sikora MJ, Serratelli WS, Fernandes RA, Louis DM, Yao W, Ji X, Idoyaga J, Mahajan VB, Steinmetz LM, Chien YH, Hauser SL, Oksenberg JR, Garcia KC, Davis MM. Opposing T cell responses in experimental autoimmune encephalomyelitis. Nature 2019; 572:481-487. [PMID: 31391585 PMCID: PMC7145319 DOI: 10.1038/s41586-019-1467-x] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 07/05/2019] [Indexed: 11/30/2022]
Abstract
Experimental autoimmune encephalomyelitis is a model for multiple sclerosis. Here we show that induction generates successive waves of clonally expanded CD4+, CD8+ and γδ+ T cells in the blood and central nervous system, similar to gluten-challenge studies of patients with coeliac disease. We also find major expansions of CD8+ T cells in patients with multiple sclerosis. In autoimmune encephalomyelitis, we find that most expanded CD4+ T cells are specific for the inducing myelin peptide MOG35-55. By contrast, surrogate peptides derived from a yeast peptide major histocompatibility complex library of some of the clonally expanded CD8+ T cells inhibit disease by suppressing the proliferation of MOG-specific CD4+ T cells. These results suggest that the induction of autoreactive CD4+ T cells triggers an opposing mobilization of regulatory CD8+ T cells.
Collapse
Affiliation(s)
- Naresha Saligrama
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Institute of Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Fan Zhao
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael J Sikora
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - William S Serratelli
- Institute of Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Ricardo A Fernandes
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - David M Louis
- Institute of Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Winnie Yao
- Institute of Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Xuhuai Ji
- Human Immune Monitoring Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Juliana Idoyaga
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Vinit B Mahajan
- Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care, Palo Alto, CA, USA
| | - Lars M Steinmetz
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Genome Technology Center, Stanford University, Palo Alto, CA, USA
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Yueh-Hsiu Chien
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Institute of Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
- Program in Immunology, Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Stephen L Hauser
- Department of Neurology and UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Jorge R Oksenberg
- Department of Neurology and UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - K Christopher Garcia
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
- The Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Mark M Davis
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA.
- Institute of Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA.
- The Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
10
|
Guo Z, Tao Y, Yin S, Song Y, Lu X, Li X, Fan Y, Fan X, Xu S, Yang J, Yu Y. The transcription factor Foxp1 regulates the differentiation and function of dendritic cells. Mech Dev 2019; 158:103554. [PMID: 31077741 DOI: 10.1016/j.mod.2019.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/01/2019] [Accepted: 05/07/2019] [Indexed: 01/14/2023]
Abstract
Dendritic cells (DCs) are the sentinels of the immune system and play a critical role in initiating adaptive immune responses against pathogens. As the most powerful antigen presenting cells, DCs are also important in maintaining immune homeostasis and participating in the development of autoimmune diseases. How the maturation and function of DCs is regulated in these conditions and what is the function of various transcription factors is still unclear. In this study, we found that the expression of the transcription factor Foxp1 gradually increased during the maturation of DCs. Then, we constructed a recombinant adenovirus carrying Foxp1-interfering RNA (Ad-simFoxp1) and transfected murine bone marrow-derived DCs in vitro. DCs transfected with Ad-simFoxp1 exhibited markedly lower costimulatory molecules, and decreased cytokines. And Ad-simFoxp1 greatly inhibited mature DC-induced T cell responses. Moreover, in vivo infusion with Ad-simFoxp1-modified DCs significantly delayed the onset of experimental autoimmune encephalomyelitis (EAE). Therefore, adoptive transfection of Ad-simFoxp1 in DCs may be a potential treatment strategy against autoimmune diseases.
Collapse
Affiliation(s)
- Ziyi Guo
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, China; Department of Endocrine, Minhang Hospital, Fudan University, Shanghai, China
| | - Yijie Tao
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Shulei Yin
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Yuping Song
- Department of Endocrine, Minhang Hospital, Fudan University, Shanghai, China
| | - Xiaomin Lu
- Department of Endocrine, Minhang Hospital, Fudan University, Shanghai, China
| | - Xuesong Li
- Department of Endocrine, Minhang Hospital, Fudan University, Shanghai, China
| | - Yujuan Fan
- Department of Endocrine, Minhang Hospital, Fudan University, Shanghai, China
| | - Xiaofang Fan
- Department of Endocrine, Minhang Hospital, Fudan University, Shanghai, China
| | - Sheng Xu
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, China.
| | - Jialin Yang
- Department of Endocrine, Minhang Hospital, Fudan University, Shanghai, China.
| | - Yizhi Yu
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, China.
| |
Collapse
|
11
|
Extracellular vesicle fibrinogen induces encephalitogenic CD8+ T cells in a mouse model of multiple sclerosis. Proc Natl Acad Sci U S A 2019; 116:10488-10493. [PMID: 31068461 PMCID: PMC6535008 DOI: 10.1073/pnas.1816911116] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In this report, we show that fibrinogen, identified by proteomics to be present in blood plasma extracellular vesicles (EVs), is sufficient and required for autoimmune-mediated spontaneous relapsing disease activity in a murine model of multiple sclerosis (MS). Unique to this model is that plasma EVs induced CD8-mediated disease. Analysis of human plasma EVs identified fibrinogen in MS patient samples, thereby providing a compelling translational association between our experimental findings and the perpetuation of CD8-mediated autoimmunity in human MS. Hence, these findings provide evidence for EVs as means by which to model an important aspect of spontaneous CD8+ T cell development related to autoimmunity in MS. Extracellular vesicles (EVs) are emerging as potent mediators of intercellular communication with roles in inflammation and disease. In this study, we examined the role of EVs from blood plasma (pEVs) in an experimental autoimmune encephalomyelitis mouse model of central nervous system demyelination. We determined that pEVs induced a spontaneous relapsing−remitting disease phenotype in MOG35–55-immunized C57BL/6 mice. This modified disease phenotype was found to be driven by CD8+ T cells and required fibrinogen in pEVs. Analysis of pEVs from relapsing−remitting multiple sclerosis patients also identified fibrinogen as a significant portion of pEV cargo. Together, these data suggest that fibrinogen in pEVs contributes to the perpetuation of neuroinflammation and relapses in disease.
Collapse
|
12
|
Wang L, Winnewisser J, Federle C, Jessberger G, Nave KA, Werner HB, Kyewski B, Klein L, Hinterberger M. Epitope-Specific Tolerance Modes Differentially Specify Susceptibility to Proteolipid Protein-Induced Experimental Autoimmune Encephalomyelitis. Front Immunol 2017; 8:1511. [PMID: 29170668 PMCID: PMC5684123 DOI: 10.3389/fimmu.2017.01511] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 10/25/2017] [Indexed: 12/21/2022] Open
Abstract
Immunization with myelin components can elicit experimental autoimmune encephalomyelitis (EAE). EAE susceptibility varies between mouse strains, depending on the antigen employed. BL/6 mice are largely resistant to EAE induction with proteolipid protein (PLP), probably a reflection of antigen-specific tolerance. However, the extent and mechanism(s) of tolerance to PLP remain unclear. Here, we identified three PLP epitopes in PLP-deficient BL/6 mice. PLP-sufficient mice did not respond against two of these, whereas tolerance was “leaky” for an epitope with weak predicted MHCII binding, and only this epitope was encephalitogenic. In TCR transgenic mice, the “EAE-susceptibility-associated” epitope was “ignored” by specific CD4 T cells, whereas the “resistance-associated” epitope induced clonal deletion and Treg induction in the thymus. Central tolerance was autoimmune regulator dependent and required expression and presentation of PLP by thymic epithelial cells (TECs). TEC-specific ablation of PLP revealed that peripheral tolerance, mediated by dendritic cells through recessive tolerance mechanisms (deletion and anergy), could largely compensate for a lack of central tolerance. However, adoptive EAE was exacerbated in mice lacking PLP in TECs, pointing toward a non-redundant role of the thymus in dominant tolerance to PLP. Our findings reveal multiple layers of tolerance to a central nervous system autoantigen that vary among epitopes and thereby specify disease susceptibility. Understanding how different modalities of tolerance apply to distinct T cell epitopes of a target in autoimmunity has implications for antigen-specific strategies to therapeutically interfere with unwanted immune reactions against self.
Collapse
Affiliation(s)
- Lei Wang
- Institute for Immunology, Biomedical Center (BMC) Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Julia Winnewisser
- Institute for Immunology, Biomedical Center (BMC) Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Christine Federle
- Institute for Immunology, Biomedical Center (BMC) Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Gregor Jessberger
- Institute for Immunology, Biomedical Center (BMC) Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Goettingen, Germany
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Goettingen, Germany
| | - Bruno Kyewski
- Division of Developmental Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ludger Klein
- Institute for Immunology, Biomedical Center (BMC) Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Maria Hinterberger
- Institute for Immunology, Biomedical Center (BMC) Munich, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
13
|
Ma K, Chen X, Chen JC, Wang Y, Zhang XM, Huang F, Zheng JJ, Chen X, Yu W, Cheng KL, Feng YQ, Gu HY. Rifampicin attenuates experimental autoimmune encephalomyelitis by inhibiting pathogenic Th17 cells responses. J Neurochem 2016; 139:1151-1162. [PMID: 27774592 PMCID: PMC6680363 DOI: 10.1111/jnc.13871] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Revised: 09/15/2016] [Accepted: 10/07/2016] [Indexed: 02/02/2023]
Abstract
Rifampicin, a broad‐spectrum antibiotic, has neuroprotective, immunosuppressive, and anti‐inflammatory properties. However, the effect of rifampicin on autoimmune disorders of the nervous system is not clear. In this study, we investigated whether rifampicin was beneficial to myelin oligodendrocyte glycoprotein peptide (MOG33–35)‐induced female C57BL/6 experimental autoimmune encephalomyelitis (EAE) mice, the well‐established animal model of multiple sclerosis. Rifampicin treatment (daily from the first day after EAE immunization) remarkably attenuated clinical signs and loss of body weight, which are associated with suppression of inflammatory infiltration and demyelination in spinal cords of EAE mice. Furthermore, rifampicin dramatically reduced the disruption of blood–brain barrier integrity, down‐regulated serum concentration of IL‐6 and IL‐17A, inhibited pathological Th17 cell differentiation, and modulated the expression of p‐STAT3 and p‐p65. These results suggest that rifampicin is effective for attenuating the clinical severity of EAE mice, which may be related to its inhibitive ability in differentiation of Th17 cell and secretion of its key effector molecule IL‐17A via regulation of excessive activation of the key signaling molecules of JAK/STAT pathway. Our findings may be helpful for developing therapeutic and preventive strategies for multiple sclerosis. ![]()
Collapse
Affiliation(s)
- Ke Ma
- Department of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xi Chen
- Department of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jia-Cheng Chen
- Department of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ying Wang
- Department of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xi-Meng Zhang
- Department of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Fan Huang
- Department of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jun-Jiong Zheng
- Department of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiong Chen
- Department of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wei Yu
- Department of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ke-Ling Cheng
- Department of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yan-Qing Feng
- Department of Neurology, Guangdong Key Laboratory for Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Huai-Yu Gu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
14
|
Heiman-Patterson TD, Blankenhorn EP, Sher RB, Jiang J, Welsh P, Dixon MC, Jeffrey JI, Wong P, Cox GA, Alexander GM. Genetic background effects on disease onset and lifespan of the mutant dynactin p150Glued mouse model of motor neuron disease. PLoS One 2015; 10:e0117848. [PMID: 25763819 PMCID: PMC4357475 DOI: 10.1371/journal.pone.0117848] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 01/02/2015] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease primarily affecting motor neurons in the central nervous system. Although most cases of ALS are sporadic, about 5–10% of cases are familial (FALS) with approximately 20% of FALS caused by mutations in the Cu/Zn superoxide dismutase (SOD1) gene. We have reported that hSOD1-G93A transgenic mice modeling this disease show a more severe phenotype when the transgene is bred on a pure SJL background and a milder phenotype when bred on a pure B6 background and that these phenotype differences link to a region on mouse Chromosome 17.To examine whether other models of motor neuron degeneration are affected by genetic background, we bred the mutant human dynactin p150Glued (G59S-hDCTN1) transgene onto inbred SJL and B6 congenic lines. This model is based on an autosomal dominant lower motor neuron disease in humans linked to a mutation in the p150Glued subunit of the dynactin complex. As seen in hSOD1-G93A mice, we observed a more severe phenotype with earlier disease onset (p<0.001) and decreased survival (p<0.00001) when the G59S-hDCTN1 transgene was bred onto the SJL background and delayed onset (p<0.0001) with increased survival (p<0.00001) when bred onto the B6 background. Furthermore, B6 mice with an SJL derived chromosome 17 interval previously shown to delay disease onset in hSOD1-G93A mice also showed delays onset in G59S-hDCTN1 mice suggesting that at least some genetic modifiers are shared. We have shown that genetic background influences phenotype in G59S-hDCTN1 mice, in part through a region of chromosome 17 similar to the G93-hSOD1 ALS mouse model. These results support the presence of genetic modifiers in both these models some of which may be shared. Identification of these modifiers will highlight intracellular pathways involved in motor neuron disease and provide new therapeutic targets that may be applicable to motor neuron degeneration.
Collapse
Affiliation(s)
- Terry D Heiman-Patterson
- Department of Neurology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Elizabeth P Blankenhorn
- Department of Microbiology Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Roger B Sher
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, Maine, United States of America
| | - Juliann Jiang
- Department of Neurology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Priscilla Welsh
- Department of Neurology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Meredith C Dixon
- Department of Neurology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Jeremy I Jeffrey
- Department of Neurology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Philip Wong
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Gregory A Cox
- The Jackson Laboratory, Bar Harbour, Maine, United States of America
| | - Guillermo M Alexander
- Department of Neurology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
15
|
Bearoff F, Case LK, Krementsov DN, Wall EH, Saligrama N, Blankenhorn EP, Teuscher C. Identification of genetic determinants of the sexual dimorphism in CNS autoimmunity. PLoS One 2015; 10:e0117993. [PMID: 25671658 PMCID: PMC4324900 DOI: 10.1371/journal.pone.0117993] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 01/03/2015] [Indexed: 12/02/2022] Open
Abstract
Multiple sclerosis (MS) is a debilitating chronic inflammatory disease of the nervous system that affects approximately 2.3 million individuals worldwide, with higher prevalence in females, and a strong genetic component. While over 200 MS susceptibility loci have been identified in GWAS, the underlying mechanisms whereby they contribute to disease susceptibility remains ill-defined. Forward genetics approaches using conventional laboratory mouse strains are useful in identifying and functionally dissecting genes controlling disease-relevant phenotypes, but are hindered by the limited genetic diversity represented in such strains. To address this, we have combined the powerful chromosome substitution (consomic) strain approach with the genetic diversity of a wild-derived inbred mouse strain. Using experimental allergic encephalomyelitis (EAE), a mouse model of MS, we evaluated genetic control of disease course among a panel of 26 consomic strains of mice inheriting chromosomes from the wild-derived PWD strain on the C57BL/6J background, which models the genetic diversity seen in human populations. Nineteen linkages on 18 chromosomes were found to harbor loci controlling EAE. Of these 19 linkages, six were male-specific, four were female-specific, and nine were non-sex-specific, consistent with a differential genetic control of disease course between males and females. An MS-GWAS candidate-driven bioinformatic analysis using orthologous genes linked to EAE course identified sex-specific and non-sex-specific gene networks underlying disease pathogenesis. An analysis of sex hormone regulation of genes within these networks identified several key molecules, prominently including the MAP kinase family, known hormone-dependent regulators of sex differences in EAE course. Importantly, our results provide the framework by which consomic mouse strains with overall genome-wide genetic diversity, approximating that seen in humans, can be used as a rapid and powerful tool for modeling the genetic architecture of MS. Moreover, our data represent the first step towards mechanistic dissection of genetic control of sexual dimorphism in CNS autoimmunity.
Collapse
Affiliation(s)
- Frank Bearoff
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, 19129, United States of America
| | - Laure K. Case
- Department of Medicine, University of Vermont, Burlington, Vermont, 05405, United States of America
| | - Dimitry N. Krementsov
- Department of Medicine, University of Vermont, Burlington, Vermont, 05405, United States of America
| | - Emma H. Wall
- Department of Medicine, University of Vermont, Burlington, Vermont, 05405, United States of America
| | - Naresha Saligrama
- Department of Medicine, University of Vermont, Burlington, Vermont, 05405, United States of America
| | - Elizabeth P. Blankenhorn
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, 19129, United States of America
| | - Cory Teuscher
- Department of Medicine, University of Vermont, Burlington, Vermont, 05405, United States of America
- Department of Pathology, University of Vermont, Burlington, Vermont, 05405, United States of America
- * E-mail:
| |
Collapse
|
16
|
Case LK, Wall EH, Osmanski EE, Dragon JA, Saligrama N, Zachary JF, Lemos B, Blankenhorn EP, Teuscher C. Copy number variation in Y chromosome multicopy genes is linked to a paternal parent-of-origin effect on CNS autoimmune disease in female offspring. Genome Biol 2015; 16:28. [PMID: 25886764 PMCID: PMC4396973 DOI: 10.1186/s13059-015-0591-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 01/20/2015] [Indexed: 12/11/2022] Open
Abstract
Background The prevalence of some autoimmune diseases is greater in females compared with males, although disease severity is often greater in males. The reason for this sexual dimorphism is unknown, but it may reflect negative selection of Y chromosome-bearing sperm during spermatogenesis or male fetuses early in the course of conception/pregnancy. Previously, we showed that the sexual dimorphism in experimental autoimmune encephalomyelitis (EAE) is associated with copy number variation (CNV) in Y chromosome multicopy genes. Here, we test the hypothesis that CNV in Y chromosome multicopy genes influences the paternal parent-of-origin effect on EAE susceptibility in female mice. Results We show that C57BL/6 J consomic strains of mice possessing an identical X chromosome and CNV in Y chromosome multicopy genes exhibit sperm head abnormalities and female-biased sex ratio. This is consistent with X-Y intragenomic conflict arising from an imbalance in CNV between homologous X:Y chromosome multicopy genes. These males also display paternal transmission of EAE to female offspring and differential loading of microRNAs within the sperm nucleus. Furthermore, in humans, families of probands with multiple sclerosis similarly exhibit a female-biased sex ratio, whereas families of probands affected with non-sexually dimorphic autoimmune diseases exhibit unbiased sex ratios. Conclusions These findings provide evidence for a mechanism at the level of the male gamete that contributes to the sexual dimorphism in EAE and paternal parent-of-origin effects in female mice, raising the possibility that a similar mechanism may contribute to the sexual dimorphism in multiple sclerosis. Electronic supplementary material The online version of this article (doi:10.1186/s13059-015-0591-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Laure K Case
- Department of Medicine, University of Vermont, Given Medical Building C317, Burlington, VT, 05405, USA.
| | - Emma H Wall
- Department of Medicine, University of Vermont, Given Medical Building C317, Burlington, VT, 05405, USA.
| | - Erin E Osmanski
- Department of Medicine, University of Vermont, Given Medical Building C317, Burlington, VT, 05405, USA.
| | - Julie A Dragon
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT, 05405, USA.
| | - Naresha Saligrama
- Department of Medicine, University of Vermont, Given Medical Building C317, Burlington, VT, 05405, USA. .,Current address: Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - James F Zachary
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA.
| | - Bernardo Lemos
- Department of Environmental Health, Harvard School of Public Health, Boston, MA, 02115, USA.
| | - Elizabeth P Blankenhorn
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, 19129, USA.
| | - Cory Teuscher
- Department of Medicine, University of Vermont, Given Medical Building C317, Burlington, VT, 05405, USA. .,Department of Pathology, University of Vermont, Burlington, VT, 05405, USA.
| |
Collapse
|
17
|
Didonna A, Oksenberg JR. Genetic determinants of risk and progression in multiple sclerosis. Clin Chim Acta 2015; 449:16-22. [PMID: 25661088 DOI: 10.1016/j.cca.2015.01.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 01/21/2015] [Indexed: 01/07/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune disease that represents a primary cause of neurological disability in the young adult population. Converging evidence supports the importance of genetic determinants for MS etiology. However, with the exception of the major histocompatibility complex, their nature has been elusive for more than 20 years. In the last decade, the advent of large genome-wide association studies has significantly improved our understanding of the disease, leading to the golden era of MS genetic research. To date more than 110 genetic variants have been firmly associated to an increased risk of developing MS. A large part of these variants tag genes involved in the regulation of immune response and several of them are shared with other autoimmune diseases, suggesting a common etiological root for this class of disorders. Despite the impressive body of data obtained in the last years, we are still far from fully decoding MS genetic complexity. For example, we ignore how these genetic factors interact with each other and with the environment. Thus, the biggest challenge for the next era of MS research will consist in identifying and characterizing the molecular mechanisms and the cellular pathways in which these risk variants play a role.
Collapse
Affiliation(s)
- Alessandro Didonna
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Jorge R Oksenberg
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
18
|
Abstract
Living on a sun-drenched planet has necessitated adaption to and protection from the harmful effects of solar ultraviolet (UV) radiation, particularly skin cancer. However, convincing epidemiological and recent empirical evidence also supports a protective effect of UV against a range of diseases including multiple sclerosis, asthma and cardiovascular disease. Despite years of research attention into the biological effects of sunlight exposure, we are still far from being able to fully answer the question: How much sunlight is enough? This is probably because the answer is dependent on many complex and interacting variables. Many talented researchers are focused on exploring whether UV-induced vitamin D explains some of these effects. This perspectives article proposes an alternative hypothesis, namely that targeting UV-induced immune suppression by affecting the activation of regulatory cells and molecules will be of therapeutic benefit.
Collapse
Affiliation(s)
- Scott N Byrne
- Cellular Photoimmunology Group, Discipline of Infectious Diseases and Immunology, Sydney Medical School, University of Sydney, Australia.
| |
Collapse
|
19
|
Sher RB, Heiman-Patterson TD, Blankenhorn EA, Jiang J, Alexander G, Deitch JS, Cox GA. A major QTL on mouse chromosome 17 resulting in lifespan variability in SOD1-G93A transgenic mouse models of amyotrophic lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener 2014; 15:588-600. [PMID: 25008789 DOI: 10.3109/21678421.2014.932381] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis is a late-onset degenerative disease affecting motor neurons in the spinal cord, brainstem, and motor cortex. There is great variation in the expression of ALS symptoms even between siblings who both carry the same Cu/Zn superoxide dismutase (SOD1) mutations. One important use of transgenic mouse models of SOD1-ALS is the study of genetic influences on ALS severity. We utilized multiple inbred mouse strains containing the SOD1-G93A transgene to demonstrate a major quantitative trait locus (QTL) on mouse chromosome 17 resulting in a significant shift in lifespan. Reciprocal crosses between long- and short-lived strains identified critical regions, and we have narrowed the area for potential genetic modifier(s) to < 2Mb of the genome. Results showed that resequencing of this region resulted in 28 candidate genes with potentially functional differences between strains. In conclusion, these studies provide the first major modifier locus affecting lifespan in this model of FALS and, once identified, these candidate modifier genes may provide insight into modifiers of human disease and, most importantly, define new targets for the development of therapies.
Collapse
|
20
|
Berer K, Boziki M, Krishnamoorthy G. Selective accumulation of pro-inflammatory T cells in the intestine contributes to the resistance to autoimmune demyelinating disease. PLoS One 2014; 9:e87876. [PMID: 24504092 PMCID: PMC3913661 DOI: 10.1371/journal.pone.0087876] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 12/30/2013] [Indexed: 12/17/2022] Open
Abstract
Myelin-specific, pro-inflammatory TH17 cells are widely regarded as the drivers of experimental autoimmune encephalomyelitis (EAE), an animal model for Multiple sclerosis (MS). The factors, responsible for the generation and maintenance of TH17 cells as well as their participation in the pathogenic cascade leading to the demyelinating disease, have been studied extensively. However, how these harmful autoreactive cells are controlled in vivo remains unclear. By comparing TCR transgenic mice on a disease susceptible and a disease resistant genetic background, we show here that pathogenic TH17 cells are sequestered within the intestine of spontaneous EAE resistant B10.S mice. Disease resistant B10.S mice harbored higher frequencies of TH17 cells in the intestine compared to EAE susceptible SJL/J mice. Moreover, transferred TH17 cells selectively migrated to intestinal lymphoid organs of B10.S mice. The sequestration of TH17 cells in the gut was partially dependent on the gut homing receptor α4β7-mediated adhesion to the intestine. Administration of α4β7 blocking-antibodies increased the peripheral availability of TH17 cells, resulting in increased EAE severity after immunization in B10.S mice. Together, these results support the concept that the intestine is a check-point for controlling pathogenic, organ-specific T cells.
Collapse
Affiliation(s)
- Kerstin Berer
- Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Marina Boziki
- Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany
| | | |
Collapse
|
21
|
G proteins Gαi1/3 are critical targets for Bordetella pertussis toxin-induced vasoactive amine sensitization. Infect Immun 2013; 82:773-82. [PMID: 24478091 DOI: 10.1128/iai.00971-13] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Pertussis toxin (PTX) is an AB5-type exotoxin produced by the bacterium Bordetella pertussis, the causative agent of whooping cough. In vivo intoxication with PTX elicits a variety of immunologic and inflammatory responses, including vasoactive amine sensitization (VAAS) to histamine (HA), serotonin (5-HT), and bradykinin (BDK). Previously, by using a forward genetic approach, we identified the HA H1 receptor (Hrh1/H1R) as the gene in mice that controls differential susceptibility to B. pertussis PTX-induced HA sensitization (Bphs). Here we show, by using inbred strains of mice, F1 hybrids, and segregating populations, that, unlike Bphs, PTX-induced 5-HT sensitivity (Bpss) and BDK sensitivity (Bpbs) are recessive traits and are separately controlled by multiple loci unlinked to 5-HT and BDK receptors, respectively. Furthermore, we found that PTX sensitizes mice to HA independently of Toll-like receptor 4, a purported receptor for PTX, and that the VAAS properties of PTX are not dependent upon endothelial caveolae or endothelial nitric oxide synthase. Finally, by using mice deficient in individual Gαi/o G-protein subunits, we demonstrate that Gαi1 and Gαi3 are the critical in vivo targets of ADP-ribosylation underlying VAAS elicited by PTX exposure.
Collapse
|
22
|
Krementsov DN, Teuscher C. Environmental factors acting during development to influence MS risk: insights from animal studies. Mult Scler 2013; 19:1684-9. [PMID: 24077054 DOI: 10.1177/1352458513506954] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune demyelinating disease of the central nervous system with an increasing incidence in females. Epidemiological data strongly implicate environmental factors acting at the population level during gestation, childhood and adulthood in the increasing incidence of MS. Several such factors are implicated in disease risk, but their causality remains unproven, while other factors remain unknown. An understanding of the risk factors acting during development is particularly limited. Animal studies could potentially bridge the gap between observational epidemiology and clinical intervention, providing not only direct evidence of causality for a given environmental agent, but also an opportunity to dissect the underlying molecular mechanisms. Given a rodent's short gestational and developmental period, the effects of developmental exposure can also be readily addressed. Nonetheless, studies in this area so far are few. In this review, we summarize the insights gleaned from studies that test environmental influences in animal models of MS, with a particular focus on gestational and early life exposures.
Collapse
|
23
|
The emerging role of p38 mitogen-activated protein kinase in multiple sclerosis and its models. Mol Cell Biol 2013; 33:3728-34. [PMID: 23897428 DOI: 10.1128/mcb.00688-13] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Multiple sclerosis (MS), the most common disabling neurologic disease of young adults, is considered a classical T cell-mediated disease and is characterized by demyelination, axonal damage, and progressive neurological dysfunction. The currently available disease-modifying therapies are limited in their efficacy, and improved understanding of new pathways contributing to disease pathogenesis could reveal additional novel therapeutic targets. The p38 mitogen-activated protein kinase (MAPK) signaling pathway is known to be triggered by stress stimuli and to contribute to inflammatory responses. Importantly, a number of recent studies have identified this signaling pathway as a central player in MS and its principal animal model, experimental allergic encephalomyelitis. Here, we review the evidence from mouse and human studies supporting the role of p38 MAPK in regulating key immunopathogenic mechanisms underlying autoimmune inflammatory disease of the central nervous system and the potential of targeting this pathway as a disease-modifying therapy in MS.
Collapse
|
24
|
Passani MB, Ballerini C. Histamine and neuroinflammation: insights from murine experimental autoimmune encephalomyelitis. Front Syst Neurosci 2012; 6:32. [PMID: 22563309 PMCID: PMC3342557 DOI: 10.3389/fnsys.2012.00032] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 04/14/2012] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory, neurodegenerative disease of the CNS whose pathogenesis remains largely unknown, and available therapies are rarely successful in reversing neurological deficits or stopping disease progression. Ongoing studies on MS and the widely used murine model of experimental autoimmune encephalomyelitis (EAE) are focused on the many components of this complex and heterogeneous neurodegenerative disease in the hope of providing a mechanism-based characterization of MS that will afford successful strategies to limit and repair the neuronal damage. Recently, histamine has been postulated to have a key regulatory role in EAE and MS pathogenesis. Histamine is a mediator of inflammation and immune responses, exerting its many actions through four G protein-coupled receptors (H1,2,3,4R) that signal through distinct intracellular pathways and have different therapeutic potentials as they vary in expression, isoform distribution, signaling properties, and function. Immune cells involved in MS/EAE, including dendritic cells (DCs) and T lymphocytes, express H1R, H2R and H4R, and histamine may have varying and counteracting effects on a particular cell type, depending on the receptor subtypes being activated. Here, we review evidence of the complex and controversial role of histamine in the pathogenesis of MS and EAE and evaluate the therapeutic potential of histaminergic ligands in the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Maria B Passani
- Dipartimento di Farmacologia Preclinica e Clinica, Universita' di Firenze Firenze, Italy
| | | |
Collapse
|
25
|
|