Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Marc S. Horwitz is active.

Publication


Featured researches published by Marc S. Horwitz.


Nature Medicine | 1997

Primary demyelination in transgenic mice expressing interferon-γ

Marc S. Horwitz; Claire F. Evans; Dorian B. McGavern; Moses Rodriguez; Michael B. A. Oldstone

Ever since the use of interferon-γ to treat patients with multiple sclerosis resulted in enhanced disease, the role of IFN-γ in demyelination has been under question. To address this issue directly, transgenic mice were generated that expressed the cDNA of murine IFN-γ in the central nervous system by using an oligodendrocyte-specific promoter. Expression of the transgene occurred after 8 weeks of age, at which time the murine immune and central nervous systems are both fully developed. Directly associated with transgene expression, primary demyelination occurred and was accompanied by clinical abnormalities consistent with CNS disorders. Additionally, multiple hallmarks of immune-mediated CNS disease were observed including upregulation of MHC molecules, gliosis and lymphocytic Infiltration. These results demonstrate a direct role for IFN-γ as an inducer of CNS demyelination leading to disease and provide new opportunities for dissecting the mechanism of demyelination.


Nature Medicine | 2000

Pancreatic expression of interferon-gamma protects mice from lethal coxsackievirus B3 infection and subsequent myocarditis.

Marc S. Horwitz; Antonio La Cava; Cody Fine; Enrique Rodriguez; Alex Ilic; Nora Sarvetnick

Cardiovascular disease is one of the leading causes of death worldwide, and has been associated with many environmental risk factors. Recent evidence has indicated the involvement of pathogens such as viruses as causative agents, and specifically identified the coxsackievirus B serogroup as the leading culprit. Not only has coxsackievirus B3 (CB3) been identified from patients with cardiovascular disease, but also infection of mice with CB3 strains can reproduce human clinical heart disease in rodents. Several mechanisms have been proposed in an attempt to distinguish between pathology mediated by direct viral destruction of cardiac muscle cells or by the virus-induced immune response directed at infected myocytes or at ‘mimicked’ epitopes shared between viral and cardiac antigens. To distinguish between these mechanisms, we infected a unique mouse that diminishes the extent of infection and spread of the virus, but allows complete immunity to the virus. Transgenic mice expressing interferon-γ in their pancreatic β cells failed to develop CB-3-induced myocarditis. This work challenges the idea of the function of the immune response and ‘molecular mimicry’ in the CB-3-induced autoimmune myocarditis model, and instead favors the idea of virus-mediated damage. These results emphasize the benefit of reducing the level of viremia early during infection, thereby reducing the incidence of virus-mediated heart damage and autoimmunity.


Journal of Clinical Investigation | 2002

Presented antigen from damaged pancreatic β cells activates autoreactive T cells in virus-mediated autoimmune diabetes

Marc S. Horwitz; Alex Ilic; Cody Fine; Enrique Rodriguez; Nora Sarvetnick

The induction of autoimmunity by viruses has been attributed to numerous mechanisms. In mice, coxsackievirus B4 (CB4) induces insulin-dependent diabetes mellitus (IDDM) resembling the final step of disease progression in humans. The immune response following the viral insult clearly precipitates IDDM. However, the molecular pathway between viral infection and the subsequent activation of T cells specific for islet antigen has not been elucidated. These T cells could become activated through exposure to sequestered antigens released by damaged beta cells, or they could have responded to factors secreted by the inflammatory response itself. To distinguish between these possibilities, we treated mice harboring a diabetogenic T cell repertoire with either the islet-damaging agent streptozotocin (STZ) or poly I:C, which nonspecifically activates T cells. Significantly, only treatment of mice with STZ resulted in IDDM and mimicked the effects observed following CB4 infection. Furthermore, antigen-presenting cells from STZ-treated mice were shown to directly activate autoreactive T cells and induce diabetes. Therefore, the primary role of CB4 in the precipitation of IDDM is to damage tissue, causing release and presentation of sequestered islet antigen. These events stimulate autoreactive T cells and thereby initiate disease.


PLOS ONE | 2009

Toll-Like Receptor 3 Signaling on Macrophages Is Required for Survival Following Coxsackievirus B4 Infection

Martin J. Richer; Danielle J. Lavallée; Iryna Shanina; Marc S. Horwitz

Toll-like receptor 3 (TLR3) has been proposed to play a central role in the early recognition of viruses by sensing double stranded RNA, a common intermediate of viral replication. However, several reports have demonstrated that TLR3 signaling is either dispensable or even harmful following infection with certain viruses. Here, we asked whether TLR3 plays a role in the response to coxsackievirus B4 (CB4), a prevalent human pathogen that has been associated with pancreatitis, myocarditis and diabetes. We demonstrate that TLR3 signaling on macrophages is critical to establish protective immunity to CB4. TLR3 deficient mice produced reduced pro-inflammatory mediators and are unable to control viral replication at the early stages of infection resulting in severe cardiac damage. Intriguingly, the absence of TLR3 did not affect the activation of several key innate and adaptive cellular effectors. This suggests that in the absence of TLR3 signaling on macrophages, viral replication outpaces the developing adaptive immune response. We further demonstrate that the MyD88-dependent signaling pathways are not only unable to compensate for the loss of TLR3, they are also dispensable in the response to this RNA virus. Our results demonstrate that TLR3 is not simply part of a redundant system of viral recognition, but rather TLR3 plays an essential role in recognizing the molecular signatures associated with specific viruses including CB4.


Immunological Reviews | 1999

Viruses, host responses, and autoimmunity.

Marc S. Horwitz; Nora Sarvetnick

Summary: Conceptually, the initiation of autoimmune disease can be described as a three‐stage process involving both genetic and environmental influences. This process begins with the development of an autoimmune cellular repertoire, followed by activation of these autoreactive cells in response to a localized target and, finally, the immune systems failure to regulate these self reactive constituents. Viruses have long been associated with inciting autoimmune disorders. Two mechanisms have been proposed to explain how a viral infection can overcome immunological tolerance to self components and initiate an organ specific autoreactive process, these mechanisms arc molecular mimicry and bystander activation. Both pathways, as discussed here, could play pivotal roles in the development of autoimmunity without necessarily excluding each other. Transgene technology has allowed us and others to examine more closely the roles of these mechanisms in mice and to dissect the requirements for initiating disease. These results demonstrate that bystander activation is the must likely explanation fur disease development. Additional evidence suggests a further role for viruses in the reactivation and chronicity of autoimmune diseases. In this scenario, a second invasion by a previously infecting virus may restimulate already existing autoreactive lymphocytes and thereby contribute to the diversity of the immune response.


Autoimmunity Reviews | 2009

Coxsackievirus infection as an environmental factor in the etiology of type 1 diabetes

Martin J. Richer; Marc S. Horwitz

Susceptibility to type 1 diabetes (T1D) is dictated by a complex interplay between genetic determinants and environmental influences. Accumulating evidence strongly supports viral infection as an important factor in the etiology of T1D. To this effect, several viruses have been associated with the capacity to induce or exacerbate T1D in both humans and mice. The most convincing evidence linking viral infection and autoimmunity comes from studies on enteroviruses, particularly coxsackievirus. In this review we will discuss the evidence associating coxsackievirus infection to T1D and present the current state of knowledge on the potential mechanism of coxsackievirus-mediated T1D.


Immunological Reviews | 1996

Using Transgenic Mouse Models to Dissect the Pathogenesis of Virus‐Induced Autoimmune Disorders of the Islets of Langerhans and the Central Nervous System

Matthias G. Herrath; Claire F. Evans; Marc S. Horwitz; Michael B. A. Oldstone

Viruses have often been associated with autoimmune diseases. One mechanism by which self-destruction can be triggered is molecular mimicry. Many examples of cross-reactive immune responses between pathogens and self-antigens have been described. This review presents two transgenic models of autoimmune disease induced by a virus through activation of anti-self lymphocytes. Viral antigens are expressed as transgenes either in beta-cells of the pancreas or in the oligodendrocytes of the CNS. Infection by a virus encoding the same gene activated autoreactive T cells that cleared the viral infection, and as a consequence of transgene expression resulted in organ-specific autoimmune disease. In both transgenic mouse models, autoreactive lymphocytes that escaped thymic negative selection were present in the periphery. Several factors are described that play a role in the regulation of the self-reactive process precipitated by a viral infection. These include the quantity of activated autoreactive T cells, the affinity of these T cells, the number of memory T cells generated following primary infection, costimulation by accessory molecules, and the types and locations of cytokines produced. In addition, unique barriers exist in target tissues that prevent or suppress autoreactive responses and define to a large extent the outcome of disease. Restimulation of autoreactive memory lymphocytes may be required to bypass these barriers and enhance autoimmune disease. Therapy directed at modifying these factors can reduce and even prevent autoimmune disease after it has been initiated.


Diabetes | 2015

Reduced expression of the MDA5 Gene IFIH1 prevents autoimmune diabetes.

Pamela J. Lincez; Iryna Shanina; Marc S. Horwitz

Although it is widely accepted that type 1 diabetes (T1D) is the result of the autoimmune destruction of insulin-producing β-cells in the pancreas, little is known about the events leading to islet autoimmunity. Epidemiological and genetic data have associated virus infections and antiviral type I interferon (IFN-I) response genes with T1D. Genetic variants in the T1D risk locus interferon induced with helicase C domain 1 (IFIH1) have been identified by genome-wide association studies to confer resistance to T1D and result in the reduction in expression of the intracellular RNA virus sensor known as melanoma differentiation–associated protein 5 (MDA5). Here, we translate the reduction in IFIH1 gene expression that results in protection from T1D. Our functional studies demonstrate that mice heterozygous at the Ifih1 gene express less than half the level of MDA5 protein, which leads to a unique antiviral IFN-I signature and adaptive response after virus infection that protects from T1D. IFIH1 heterozygous mice have a regulatory rather than effector T-cell response at the site of autoimmunity, supporting IFIH1 expression as an essential regulator of the diabetogenic T-cell response and providing a potential mechanism for patients carrying IFIH1 protective polymorphisms.


PLOS Pathogens | 2012

Gammaherpesvirus latency accentuates EAE pathogenesis: relevance to Epstein-Barr virus and multiple sclerosis.

Costanza Casiraghi; Iryna Shanina; Sehyun Cho; Michael L. Freeman; Marcia A. Blackman; Marc S. Horwitz

Epstein-Barr virus (EBV) has been identified as a putative environmental trigger of multiple sclerosis (MS), yet EBVs role in MS remains elusive. We utilized murine gamma herpesvirus 68 (γHV-68), the murine homolog to EBV, to examine how infection by a virus like EBV could enhance CNS autoimmunity. Mice latently infected with γHV-68 developed more severe EAE including heightened paralysis and mortality. Similar to MS, γHV-68EAE mice developed lesions composed of CD4 and CD8 T cells, macrophages and loss of myelin in the brain and spinal cord. Further, T cells from the CNS of γHV-68 EAE mice were primarily Th1, producing heightened levels of IFN-γ and T-bet accompanied by IL-17 suppression, whereas a Th17 response was observed in uninfected EAE mice. Clearly, γHV-68 latency polarizes the adaptive immune response, directs a heightened CNS pathology following EAE induction reminiscent of human MS and portrays a novel mechanism by which EBV likely influences MS and other autoimmune diseases.


Diabetes | 2008

Regulatory T-Cells Protect From Type 1 Diabetes After Induction by Coxsackievirus Infection in the Context of Transforming Growth Factor-β

Martin J. Richer; Nadine Straka; Dianne Fang; Iryna Shanina; Marc S. Horwitz

OBJECTIVE—Coxsackievirus infections have long been associated with the induction of type 1 diabetes. Infection with coxsackievirus B4 (CB4) enhances type 1 diabetes onset in NOD mice by accelerating the presentation of β-cell antigen to autoreactive T-cells. It has been reported that a progressive defect in regulatory T-cell (Treg) function is, in part, responsible for type 1 diabetes onset in NOD mice. This defect may contribute to susceptibility to viral-induced type 1 diabetes. We asked whether the immune response after CB4 infection could be manipulated to reestablish peripheral tolerance while maintaining the immune response to virus. RESEARCH DESIGN AND METHODS—NOD mice expressing transforming growth factor-β (TGF-β) specifically in the β-cells were infected with CB4, and the functional role of Tregs in disease protection was measured. Systemic treatments with TGF-β were used to assess its therapeutic potential. RESULTS—Here, we report that Tregs induced after CB4 infection in the presence of TGF-β prevented type 1 diabetes. The capacity to directly infect pancreatic β-cells correlated with increased numbers of pancreatic Tregs, suggesting that presentation of β-cell antigen is integral to induction of diabetogenic protective Tregs. Furthermore, the presence of these viral induced Tregs correlated with protection from type 1 diabetes without altering the antiviral response. Finally, when TGF-β was administered systemically to NOD mice after infection, the incidence of type 1 diabetes was reduced, thereby signifying a potential therapeutic role for TGF-β. CONCLUSIONS—We demonstrate manipulations of the immune response that result in Treg-mediated protection from type 1 diabetes without concomitant loss of the capacity to control viral infection.

Collaboration


Dive into the Marc S. Horwitz's collaboration.

Top Co-Authors

Avatar

Iryna Shanina

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Nora Sarvetnick

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Martin J. Richer

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Maya C. Poffenberger

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Dianne Fang

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Cody Fine

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Costanza Casiraghi

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nadine Straka

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Alex Ilic

Scripps Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge