Network


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

Hotspot


Dive into the research topics where DeLisa Fairweather is active.

Publication


Featured researches published by DeLisa Fairweather.


Psychosomatic Medicine | 2009

Cumulative Childhood Stress and Autoimmune Diseases in Adults

Shanta R. Dube; DeLisa Fairweather; William S. Pearson; Vincent J. Felitti; Robert F. Anda; Janet B. Croft

Objective: To examine whether childhood traumatic stress increased the risk of developing autoimmune diseases as an adult. Methods: Retrospective cohort study of 15,357 adult health maintenance organization members enrolled in the Adverse Childhood Experiences (ACEs) Study from 1995 to 1997 in San Diego, California, and eligible for follow-up through 2005. ACEs included childhood physical, emotional, or sexual abuse; witnessing domestic violence; growing up with household substance abuse, mental illness, parental divorce, and/or an incarcerated household member. The total number of ACEs (ACE Score range = 0-8) was used as a measure of cumulative childhood stress. The outcome was hospitalizations for any of 21 selected autoimmune diseases and 4 immunopathology groupings: T- helper 1 (Th1) (e.g., idiopathic myocarditis); T-helper 2 (Th2) (e.g., myasthenia gravis); Th2 rheumatic (e.g., rheumatoid arthritis); and mixed Th1/Th2 (e.g., autoimmune hemolytic anemia). Results: Sixty-four percent reported at least one ACE. The event rate (per 10,000 person-years) for a first hospitalization with any autoimmune disease was 31.4 in women and 34.4 in men. First hospitalizations for any autoimmune disease increased with increasing number of ACEs (p < .05). Compared with persons with no ACEs, persons with ≥2 ACEs were at a 70% increased risk for hospitalizations with Th1, 80% increased risk for Th2, and 100% increased risk for rheumatic diseases (p < .05). Conclusions: Childhood traumatic stress increased the likelihood of hospitalization with a diagnosed autoimmune disease decades into adulthood. These findings are consistent with recent biological studies on the impact of early life stress on subsequent inflammatory responses. ACE = adverse childhood experience; AD = autoimmune disease; Th1 = T-helper 1; Th2 = T-helper 2; CRP = C-reactive protein; CRH = corticoid releasing hormone.


Journal of Autoimmunity | 2009

Alternatively activated macrophages in infection and autoimmunity

DeLisa Fairweather; Daniela Cihakova

Macrophages are innate immune cells that play an important role in activation of the immune response and wound healing. Pathogens that require T helper-type 2 (Th2) responses for effective clearance, such as parasitic worms, are strong inducers of alternatively activated or M2 macrophages. However, infections such as bacteria and viruses that require Th1-type responses may induce M2 as a strategy to evade the immune system. M2 are particularly efficient at scavenging self tissues following injury through receptors like the mannose receptor and scavenger receptor-A. Thus, M2 may increase autoimmune disease by presenting self tissue to T cells. M2 may also exacerbate immune complex (IC)-mediated pathology and fibrosis, a hallmark of autoimmune disease in women, due to the release of profibrotic factors such as interleukin-1beta, transforming growth factor-beta, fibronectin and matrix metalloproteinases. We have found that M2 comprise anywhere from 30% to 70% of the infiltrate during acute viral or experimental autoimmune myocarditis, and shifts in M2 populations correlate with increased IC deposition, fibrosis and chronic autoimmune pathology. Thus, women may be at an increased risk of M2-mediated autoimmunity due to estrogens ability to increase Th2 responses.


Nature Immunology | 2012

The composition and signaling of the IL-35 receptor are unconventional

Lauren W. Collison; Greg M. Delgoffe; Clifford S. Guy; Kate M. Vignali; Vandana Chaturvedi; DeLisa Fairweather; Abhay R. Satoskar; K. Christopher Garcia; Christopher A. Hunter; Charles G. Drake; Peter J. Murray; Dario A. A. Vignali

Interleukin 35 (IL-35) belongs to the IL-12 family of heterodimeric cytokines but has a distinct functional profile. IL-35 suppresses T cell proliferation and converts naive T cells into IL-35-producing induced regulatory T cells (iTr35 cells). Here we found that IL-35 signaled through a unique heterodimer of receptor chains IL-12Rβ2 and gp130 or homodimers of each chain. Conventional T cells were sensitive to IL-35-mediated suppression in the absence of one receptor chain but not both receptor chains, whereas signaling through both chains was required for IL-35 expression and conversion into iTr35 cells. Signaling through the IL-35 receptor required the transcription factors STAT1 and STAT4, which formed a unique heterodimer that bound to distinct sites in the promoters of the genes encoding the IL-12 subunits p35 and Ebi3. This unconventional mode of signaling, distinct from that of other members of the IL-12 family, may broaden the spectrum and specificity of IL-35-mediated suppression.


Journal of Immunology | 2003

IL-12 Receptor β1 and Toll-Like Receptor 4 Increase IL-1β- and IL-18-Associated Myocarditis and Coxsackievirus Replication

DeLisa Fairweather; Susan Yusung; Sylvia Frisancho; Masheka A. Barrett; Shannon J. L. Gatewood; Ronelle Steele; Noel R. Rose

Th1-type immune responses, mediated by IL-12-induced IFN-γ, protect the host from most viral infections. To investigate the role of IL-12 and IFN-γ on the development of Coxsackievirus B3 (CB3)-induced myocarditis, we examined the level of inflammation, viral replication, and cytokine production in IL-12Rβ1- and IFN-γ-deficient mice following CB3 infection. We report that IL-12Rβ1 deficiency results in decreased viral replication and inflammation in the heart, while IFN-γ deficiency exacerbates CB3 replication. Importantly, decreased IL-1β and IL-18 levels in IL-12Rβ1-deficient hearts correlated directly with decreased myocardial inflammation. Because IL-1β and IL-18 were associated with myocardial inflammation, we examined the effect of TLR4 deficiency on CB3 infection and myocarditis. We found that TLR4-deficient mice also had significantly reduced levels of myocarditis, viral replication, and IL-1β/IL-18, just as we had observed in IL-12Rβ1-deficient mice. This is the first report that TLR4 influences CB3 replication. These results show that IL-12Rβ1 and TLR4 exacerbate CB3 infection and myocarditis while IFN-γ protects against viral replication. The remarkable similarities between the effects of IL-12Rβ1 and TLR4 suggest that these receptors share common downstream pathways that directly influence IL-1β and IL-18 production, and confirm that IL-1β and IL-18 play a significant role in the pathogenesis of CB3-induced myocarditis. These findings have important implications not only for the pathogenesis of myocarditis, but for other autoimmune diseases triggered by viral infections.


Nature Immunology | 2001

Contribution of the innate immune system to autoimmune myocarditis: a role for complement

Ziya Kaya; Marina Afanasyeva; Yan Wang; K. Malte Dohmen; Jens Schlichting; Theresa Tretter; DeLisa Fairweather; V. Michael Holers; Noel R. Rose

Myocarditis is a principal cause of heart disease among young adults and is often a precursor of heart failure due to dilated cardiomyopathy. We show here that complement is critical for the induction of experimental autoimmune myocarditis and that it acts through complement receptor type 1 (CR1) and type 2 (CR2). We also found a subset of CD44hiCD62Llo T cells that expresses CR1 and CR2 and propose that both receptors are involved in the expression of B and T cell activation markers, T cell proliferation and cytokine production. These findings provide a mechanism by which activated complement, a key product of the innate immune response, modulates the induction of an autoimmune disease.


American Journal of Pathology | 2004

Interferon-γ Protects against Chronic Viral Myocarditis by Reducing Mast Cell Degranulation, Fibrosis, and the Profibrotic Cytokines Transforming Growth Factor-β1, Interleukin-1β, and Interleukin-4 in the Heart

DeLisa Fairweather; Sylvia Frisancho-Kiss; Susy A. Yusung; Masheka A. Barrett; Sarah E. Davis; Shannon J. L. Gatewood; Dolores B. Njoku; Noel R. Rose

Inflammatory fibrosis is a characteristic feature of myocarditis, dilated cardiomyopathy (DCM), and congestive heart failure. Th1-type immune responses, mediated by interleukin (IL)-12-induced interferon (IFN)-γ, are believed to exacerbate autoimmune diseases including myocarditis. In this study, we examined the effect of IL-12Rβ1 and IFN-γ deficiency on the development of chronic CB3-induced myocarditis using knockout mice. We found increased chronic CB3-induced myocarditis (14.1 to 43.1%, P < 0.001); pericarditis (1.5 to 7.6%, P < 0.001); fibrosis (9.7 to 27.4%, P < 0.05); and the profibrotic cytokines transforming growth factor-β1, IL-1β, and IL-4 in the hearts of IFN-γ-deficient mice. All mice infected with CB3 developed DCM, but IFN-γ-deficient mice developed a fibrous, adhesive pericarditis associated with increased numbers of degranulating mast cells (MCs) in the pericardium (26.6 to 45.9%, P < 0.01), increased histamine levels (716 to 1930 ng/g of heart, P < 0.01), and reduced survival (100 to 43%). In contrast, IL-12Rβ1 deficiency did not significantly alter the development of chronic myocarditis. Thus, IFN-γ protects against the development of severe chronic myocarditis, pericarditis, and DCM after CB3 infection by reducing MC degranulation, fibrosis, and the profibrotic cytokines transforming growth factor-β1, IL-1β, and IL-4 in the heart.


Journal of Immunology | 2007

Cutting Edge: Cross-Regulation by TLR4 and T cell Ig Mucin-3 Determines Sex Differences in Inflammatory Heart Disease

Sylvia Frisancho-Kiss; Sarah E. Davis; Jennifer F. Nyland; J. Augusto Frisancho; Daniela Cihakova; Masheka A. Barrett; Noel R. Rose; DeLisa Fairweather

Recent clinical studies have reinforced the importance of sex-related differences in the pathogenesis of cardiovascular diseases, with an increased incidence and mortality in men. Similar to humans, male BALB/c mice infected with coxsackievirus B3 (CVB3) develop more severe inflammation in the heart even though viral replication is no greater than in females. We show that TLR4 and IFN-γ levels are significantly elevated and regulatory T cell (Treg) populations significantly reduced in the heart of males following CVB3 infection, whereas females have significantly increased T cell Ig mucin (Tim)-3, IL-4 and Treg. Blocking Tim-3 in males significantly increases inflammation and TLR4 expression while reducing Treg. In contrast, defective TLR4 signaling significantly reduces inflammation while increasing Tim-3 expression. Cross-regulation of TLR4 and Tim-3 occurs during the innate and adaptive immune response. This novel mechanism may help explain why inflammatory heart disease is more severe in males.


Journal of Immunology | 2006

Cutting Edge: T Cell Ig Mucin-3 Reduces Inflammatory Heart Disease by Increasing CTLA-4 during Innate Immunity

Sylvia Frisancho-Kiss; Jennifer F. Nyland; Sarah E. Davis; Masheka A. Barrett; Shannon J. L. Gatewood; Dolores B. Njoku; Daniela Cihakova; Ellen K. Silbergeld; Noel R. Rose; DeLisa Fairweather

Autoimmune diseases can be reduced or even prevented if proinflammatory immune responses are appropriately down-regulated. Receptors (such as CTLA-4), cytokines (such as TGF-β), and specialized cells (such as CD4+CD25+ T regulatory cells) work together to keep immune responses in check. T cell Ig mucin (Tim) family proteins are key regulators of inflammation, providing an inhibitory signal that dampens proinflammatory responses and thereby reducing autoimmune and allergic responses. We show in this study that reducing Tim-3 signaling during the innate immune response to viral infection in BALB/c mice reduces CD80 costimulatory molecule expression on mast cells and macrophages and reduces innate CTLA-4 levels in CD4+ T cells, resulting in decreased T regulatory cell populations and increased inflammatory heart disease. These results indicate that regulation of inflammation in the heart begins during innate immunity and that Tim-3 signaling on cells of the innate immune system critically influences regulation of the adaptive immune response.


Emerging Infectious Diseases | 2004

Women and Autoimmune Diseases

DeLisa Fairweather; Noel R. Rose

Recent evidence indicates that sex hormones may exacerbate autoimmune diseases, particularly in women, by increasing the adjuvant effect of infections.


American Journal of Pathology | 2008

Interleukin-13 Protects Against Experimental Autoimmune Myocarditis by Regulating Macrophage Differentiation

Daniela Cihakova; Jobert G. Barin; Marina Afanasyeva; Miho Kimura; DeLisa Fairweather; Michael Berg; Monica V. Talor; G. Christian Baldeviano; Sylvia Frisancho; Kathleen L. Gabrielson; Djahida Bedja; Noel R. Rose

We report here that interleukin (IL)-13 protects BALB/c mice from myocarditis, whether induced by peptide immunization or by viral infection. In contrast to mild disease in IL-4 knockout (KO) BALB/c mice, IL-13 KO BALB/c mice developed severe coxsackievirus B3 (CVB3)-induced autoimmune myocarditis and myocarditogenic peptide-induced experimental autoimmune myocarditis. Such severe disease was characterized by increased cardiac inflammation, increased total intracardiac CD45(+) leukocytes, elevated anti-cardiac myosin autoantibodies, and increased cardiac fibrosis. Echocardiography revealed that IL-13 KO mice developed severe dilated cardiomyopathy with impaired cardiac function and heart failure. Hearts of IL-13 KO mice had increased levels of the proinflammatory and profibrotic cytokines IL-1beta, IL-18, interferon-gamma, transforming growth factor-beta1, and IL-4 as well as histamine. The hallmark of the disease in IL-13 KO mice was the up-regulation of T-cell responses. CD4(+) T cells were increased in IL-13 KO hearts both proportionally and in absolute number. Splenic T cells from IL-13 KO mice were highly activated, and myosin stimulation additionally increased T-cell proliferation. CD4(+)CD25(+)Foxp3(+) regulatory T-cell numbers were decreased in the spleens of IL-13 KO mice. IL-13 deficiency led to decreased levels of alternatively activated CD206(+) and CD204(+) macrophages and increased levels of classically activated macrophages. IL-13 KO mice had increased caspase-1 activation, leading to increased production of both IL-1beta and IL-18. Therefore, IL-13 protects against myocarditis by modulating monocyte/macrophage populations and by regulating their function.

Collaboration


Dive into the DeLisa Fairweather's collaboration.

Top Co-Authors

Avatar

Noel R. Rose

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Djahida Bedja

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Kathleen L. Gabrielson

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Adriana Bucek

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Eric Abston

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge