Network


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

Hotspot


Dive into the research topics where Sowbarnika Sachithanantham is active.

Publication


Featured researches published by Sowbarnika Sachithanantham.


Journal of Autoimmunity | 2014

CD8 T-cell reactivity to islet antigens is unique to type 1 while CD4 T-cell reactivity exists in both type 1 and type 2 diabetes.

Ghanashyam Sarikonda; Jeremy Pettus; Sonal Phatak; Sowbarnika Sachithanantham; Jacqueline Miller; Johnna D. Wesley; Eithon Cadag; Ji Chae; Lakshmi Ganesan; Ronna Mallios; Steve Edelman; Bjoern Peters; Matthias von Herrath

Previous cross-sectional analyses demonstrated that CD8(+) and CD4(+) T-cell reactivity to islet-specific antigens was more prevalent in T1D subjects than in healthy donors (HD). Here, we examined T1D-associated epitope-specific CD4(+) T-cell cytokine production and autoreactive CD8(+) T-cell frequency on a monthly basis for one year in 10 HD, 33 subjects with T1D, and 15 subjects with T2D. Autoreactive CD4(+) T-cells from both T1D and T2D subjects produced more IFN-γ when stimulated than cells from HD. In contrast, higher frequencies of islet antigen-specific CD8(+) T-cells were detected only in T1D. These observations support the hypothesis that general beta-cell stress drives autoreactive CD4(+) T-cell activity while islet over-expression of MHC class I commonly seen in T1D mediates amplification of CD8(+) T-cells and more rapid beta-cell loss. In conclusion, CD4(+) T-cell autoreactivity appears to be present in both T1D and T2D while autoreactive CD8(+) T-cells are unique to T1D. Thus, autoreactive CD8(+) cells may serve as a more T1D-specific biomarker.


PLOS ONE | 2013

Transient B-Cell Depletion with Anti-CD20 in Combination with Proinsulin DNA Vaccine or Oral Insulin: Immunologic Effects and Efficacy in NOD Mice

Ghanashyam Sarikonda; Sowbarnika Sachithanantham; Yulia Manenkova; Tinalyn Kupfer; Amanda Posgai; Clive Wasserfall; Philip Bernstein; Laura Straub; Philippe P. Pagni; Darius Schneider; Teresa Rodriguez Calvo; Marilyne Coulombe; Kevan C. Herold; Ronald G. Gill; Mark A. Atkinson; Gerald T. Nepom; Mario R. Ehlers; Teodora Staeva; Hideki Garren; Lawrence Steinman; Andrew C. Chan; Matthias von Herrath

A recent type 1 diabetes (T1D) clinical trial of rituximab (a B cell-depleting anti-CD20 antibody) achieved some therapeutic benefit in preserving C-peptide for a period of approximately nine months in patients with recently diagnosed diabetes. Our previous data in the NOD mouse demonstrated that co-administration of antigen (insulin) with anti-CD3 antibody (a T cell-directed immunomodulator) offers better protection than either entity alone, indicating that novel combination therapies that include a T1D-related autoantigen are possible. To accelerate the identification and development of novel combination therapies that can be advanced into the clinic, we have evaluated the combination of a mouse anti-CD20 antibody with either oral insulin or a proinsulin-expressing DNA vaccine. Anti-CD20 alone, given once or on 4 consecutive days, produced transient B cell depletion but did not prevent or reverse T1D in the NOD mouse. Oral insulin alone (twice weekly for 6 weeks) was also ineffective, while proinsulin DNA (weekly for up to 12 weeks) showed a trend toward modest efficacy. Combination of anti-CD20 with oral insulin was ineffective in reversing diabetes in NOD mice whose glycemia was controlled with SC insulin pellets; these experiments were performed in three independent labs. Combination of anti-CD20 with proinsulin DNA was also ineffective in diabetes reversal, but did show modest efficacy in diabetes prevention (p = 0.04). In the prevention studies, anti-CD20 plus proinsulin resulted in modest increases in Tregs in pancreatic lymph nodes and elevated levels of proinsulin-specific CD4+ T-cells that produced IL-4. Thus, combination therapy with anti-CD20 and either oral insulin or proinsulin does not protect hyperglycemic NOD mice, but the combination with proinsulin offers limited efficacy in T1D prevention, potentially by augmentation of proinsulin-specific IL-4 production.


Diabetes | 2014

Combination therapy with an anti-IL-1β antibody and GAD65 DNA vaccine can reverse recent-onset diabetes in the RIP-GP mouse model

Philippe P. Pagni; Damien Bresson; Teresa Rodriguez-Calvo; Amira Bel Hani; Yulia Manenkova; Natalie Amirian; Alecia Blaszczak; Sina Faton; Sowbarnika Sachithanantham; Matthias von Herrath

Type 1 diabetes is thought to be an autoimmune condition in which self-reactive T cells attack insulin-secreting pancreatic β-cells. As a proinflammatory cytokine produced by β-cells or macrophages, interleukin-1β (IL-1β) represents a potential therapeutic target in diabetes. We reasoned IL-1β blockade could be combined with islet antigen–specific approaches involving GAD of 65 kDa (GAD65)-expressing plasmids, as previously shown in combination therapies (CTs) with anti-CD3. Thus, we investigated whether anti–IL-1β antibody alone or combined with GAD65 vaccine could reverse diabetes development in a virus-induced mouse model. Given alone, anti–IL-1β had no effect on diabetes, while GAD65 plasmid resulted in 33% disease reversal after a 5-week observation. However, CTs cured 53% of animals and prevented worsening of glycemic control in nonprotected individuals for up to 12 weeks. While the GAD65 vaccine arm of the CT was associated with increased forkhead box p3+ regulatory T-cell frequency in pancreatic lymph nodes, islet infiltration by CD11b+/high cells was less frequent upon CT, and its extent correlated with treatment success or failure. Altogether, our CTs provided prolonged improvement of clinical and immunological features. Despite unsuccessful clinical trials using anti–IL-1β monotherapy, these data hold promise for treatment of type 1 diabetic patients with IL-1β blockade combined with antigen-specific vaccines.


Clinical Immunology | 2014

Regulatory T cells control diabetes without compromising acute anti-viral defense

Carmen Baca Jones; Philippe P. Pagni; Georgia Fousteri; Sowbarnika Sachithanantham; Amy Dave; Teresa Rodriguez-Calvo; Jacqueline Miller; Matthias von Herrath

While previous reports have demonstrated the efficacy of regulatory T cell therapy in the prevention of diabetes, systemic immunocompromise and Treg instability remain key safety concerns. Here we examined the influence of induced Treg (iTreg) cell therapy on anti-viral host defense and autoimmune T cell responses during acute viral infection in a murine model of autoimmune diabetes. Protective transfers of iTregs maintained IL-10 expression, expanded in vivo and controlled diabetes, despite losing FoxP3 expression. Adoptive transfer of iTregs affected neither the primary anti-viral CD8 T cell response nor viral clearance, although a significant and sustained suppression of CD4 T cell responses was observed. Following acute viral clearance, iTregs transferred early suppressed both CD4 and CD8 T cell responses, which resulted in the reversion of diabetes. These observations indicate that iTregs suppress local autoimmune processes while preserving the immunocompetent hosts ability to combat acute viral infection.


PLOS ONE | 2014

Direct Infection of Dendritic Cells during Chronic Viral Infection Suppresses Antiviral T Cell Proliferation and Induces IL-10 Expression in CD4 T Cells

Carmen Baca Jones; Christophe M. Filippi; Sowbarnika Sachithanantham; Teresa Rodriguez-Calvo; Katrin Ehrhardt; Matthias von Herrath

Elevated levels of systemic IL-10 have been associated with several chronic viral infections, including HCV, EBV, HCMV and LCMV. In the chronic LCMV infection model, both elevated IL-10 and enhanced infection of dendritic cells (DCs) are important for viral persistence. This report highlights the relationship between enhanced viral tropism for DCs and the induction of IL-10 in CD4 T cells, which we identify as the most frequent IL-10-expressing cell type in chronic LCMV infection. Here we report that infected CD8αneg DCs express elevated IL-10, induce IL-10 expression in LCMV specific CD4 T cells, and suppress LCMV-specific T cell proliferation. DCs exposed in vivo to persistent LCMV retain the capacity to stimulate CD4 T cell proliferation but induce IL-10 production by both polyclonal and LCMV-specific CD4 T cells. Our study delineates the unique effects of direct infection versus viral exposure on DCs. Collectively these data point to enhanced infection of DCs as a key trigger of the IL-10 induction cascade resulting in maintenance of elevated IL-10 expression in CD4 T cells and inhibition of LCMV-specific CD4 and CD8 T cell proliferation.


Journal of Autoimmunity | 2014

Beta-cell-specific production of IL6 in conjunction with a mainly intracellular but not mainly surface viral protein causes diabetes

Tom Van Belle; Philippe P. Pagni; Jeanette Liao; Sowbarnika Sachithanantham; Amy Dave; Amira Bel Hani; Yulia Manenkova; Natalie Amirian; Cheng Yang; Bret Morin; Haiqing Zhang; Iain L. Campbell; Matthias von Herrath

Inflammatory mechanisms play a key role in the pathogenesis of type 1 and type 2 diabetes. IL6, a pleiotropic cytokine with impact on immune and non-immune cell types, has been proposed to be involved in the events causing both forms of diabetes and to play a key role in experimental insulin-dependent diabetes development. The aim of this study was to investigate how beta-cell specific overexpression of IL-6 influences diabetes development. We developed two lines of rat insulin promoter (RIP)-lymphocytic choriomeningitis virus (LCMV) mice that also co-express IL6 in their beta-cells. Expression of the viral nucleoprotein (NP), which has a predominantly intracellular localization, together with IL6 led to hyperglycemia, which was associated with a loss of GLUT-2 expression in the pancreatic beta-cells and infiltration of CD11b(+) cells, but not T cells, in the pancreas. In contrast, overexpression of the LCMV glycoprotein (GP), which can localize to the surface, with IL-6 did not lead to spontaneous diabetes, but accelerated virus-induced diabetes by increasing autoantigen-specific CD8(+) T cell responses and reducing the regulatory T cell fraction, leading to increased pancreatic infiltration by CD4(+) and CD8(+) T cells as well as CD11b(+) and CD11c(+) cells. The production of IL-6 in beta-cells acts prodiabetic, underscoring the potential benefit of targeting IL6 in diabetes.


PLOS ONE | 2014

BDC12-4.1 T-cell receptor transgenic insulin-specific CD4 T cells are resistant to in vitro differentiation into functional Foxp3+ T regulatory cells.

Ghanashyam Sarikonda; Georgia Fousteri; Sowbarnika Sachithanantham; Jacqueline Miller; Amy Dave; Therese Juntti; Ken Coppieters; Matthias von Herrath

The infusion of ex vivo-expanded autologous T regulatory (Treg) cells is potentially an effective immunotherapeutic strategy against graft-versus-host disease (GvHD) and several autoimmune diseases, such as type 1 diabetes (T1D). However, in vitro differentiation of antigen-specific T cells into functional and stable Treg (iTreg) cells has proved challenging. As insulin is the major autoantigen leading to T1D, we tested the capacity of insulin-specific T-cell receptor (TCR) transgenic CD4+ T cells of the BDC12-4.1 clone to convert into Foxp3+ iTreg cells. We found that in vitro polarization toward Foxp3+ iTreg was effective with a majority (>70%) of expanded cells expressing Foxp3. However, adoptive transfer of Foxp3+ BDC12-4.1 cells did not prevent diabetes onset in immunocompetent NOD mice. Thus, in vitro polarization of insulin-specific BDC12-4.1 TCR transgenic CD4+ T cells toward Foxp3+ cells did not provide dominant tolerance in recipient mice. These results highlight the disconnect between an in vitro acquired Foxp3+ cell phenotype and its associated in vivo regulatory potential.


PLOS ONE | 2013

Temporal Intra-Individual Variation of Immunological Biomarkers in Type 1 Diabetes Patients: Implications for Future Use in Cross-Sectional Assessment

Ghanashyam Sarikonda; Jeremy Pettus; Sowbarnika Sachithanantham; Sonal Phatak; Jacqueline Miller; Lakshmi Ganesan; Ji Chae; Ronna Mallios; Steve Edelman; Bjoern Peters; Matthias von Herrath

Multiple immune parameters such as frequencies of autoreactive CD4+, CD8+ T-cells and CD4+CD25+Foxp3+ T-cells have been explored as biomarkers in human T1D. However, intra-individual temporal variation of these parameters has not been assessed systematically over time. We determined the variation in each of these parameters in a cohort of T1D and healthy donors (HDs), at monthly intervals for one year. Despite low intra- and inter-assay co-efficient of variation (CV), mean CVs for each of the immune parameters were 119.1% for CD4+ T-cell-derived IFN-γ, 50.44% for autoreactive CD8+ T-cells, and 31.24% for CD4+CD25+Foxp3+ T-cells. Further, both HDs and T1D donors had similar CVs. The variation neither correlated with BMI, age, disease duration or insulin usage, nor were there detectable cyclical patterns of variation. However, averaging results from multiple visits for an individual provided a better estimate of the CV between visits. Based on our data we predict that by averaging values from three visits a treatment effect on these parameters with a 50% effect size could be detected with the same power using 1.8–4-fold fewer patients within a trial compared to using values from a single visit. Thus, our present data contribute to a more robust, accurate endpoint design for future clinical trials in T1D and aid in the identification of truly efficacious therapies.


Journal of Autoimmunity | 2015

The Hsp60 peptide p277 enhances anti-CD3 mediated diabetes remission in non-obese diabetic mice

Ghanashyam Sarikonda; Sowbarnika Sachithanantham; Jacqueline Miller; Philippe P. Pagni; Ken Coppieters; Matthias von Herrath


Clinical Immunology | 2014

Higher proinflammatory cytokine production and spontaneous activation in some type 1 and type 2 diabetic subjects.

Ghanashyam Sarikonda; Sowbarnika Sachithanantham; Jeremy Pettus; Teresa Rodriguez-Calvo; Sonal Phatak; Steve Edelman; Matthias von Herrath

Collaboration


Dive into the Sowbarnika Sachithanantham's collaboration.

Top Co-Authors

Avatar

Ghanashyam Sarikonda

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Jacqueline Miller

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Philippe P. Pagni

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Teresa Rodriguez-Calvo

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Amy Dave

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar

Jeremy Pettus

University of California

View shared research outputs
Top Co-Authors

Avatar

Sonal Phatak

University of California

View shared research outputs
Top Co-Authors

Avatar

Steve Edelman

University of California

View shared research outputs
Top Co-Authors

Avatar

Yulia Manenkova

La Jolla Institute for Allergy and Immunology

View shared research outputs
Researchain Logo
Decentralizing Knowledge