Network


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

Hotspot


Dive into the research topics where Shirine Dada is active.

Publication


Featured researches published by Shirine Dada.


Journal of Immunology | 2009

Immunomodulatory Function of Bone Marrow-Derived Mesenchymal Stem Cells in Experimental Autoimmune Type 1 Diabetes

Paolo Fiorina; Mollie Jurewicz; Andrea Augello; Andrea Vergani; Shirine Dada; Stefano La Rosa; Martin K. Selig; Jonathan G. Godwin; Kenneth Law; Claudia Placidi; R. Neal Smith; Carlo Capella; Scott J. Rodig; Chaker N. Adra; Mark A. Atkinson; Mohamed H. Sayegh; Reza Abdi

Human clinical trials in type 1 diabetes (T1D) patients using mesenchymal stem cells (MSC) are presently underway without prior validation in a mouse model for the disease. In response to this void, we characterized bone marrow-derived murine MSC for their ability to modulate immune responses in the context of T1D, as represented in NOD mice. In comparison to NOD mice, BALB/c-MSC mice were found to express higher levels of the negative costimulatory molecule PD-L1 and to promote a shift toward Th2-like responses in treated NOD mice. In addition, transfer of MSC from resistant strains (i.e., nonobese resistant mice or BALB/c), but not from NOD mice, delayed the onset of diabetes when administered to prediabetic NOD mice. The number of BALB/c-MSC trafficking to the pancreatic lymph nodes of NOD mice was higher than in NOD mice provided autologous NOD-MSC. Administration of BALB/c-MSC temporarily resulted in reversal of hyperglycemia in 90% of NOD mice (p = 0.002). Transfer of autologous NOD-MSC imparted no such therapeutic benefit. We also noted soft tissue and visceral tumors in NOD-MSC-treated mice, which were uniquely observed in this setting (i.e., no tumors were present with BALB/c- or nonobese resistant mice-MSC transfer). The importance of this observation remains to be explored in humans, as inbred mice such as NOD may be more susceptible to tumor formation. These data provide important preclinical data supporting the basis for further development of allogeneic MSC-based therapies for T1D and, potentially, for other autoimmune disorders.


Diabetes | 2008

Targeting CD22 Reprograms B-Cells and Reverses Autoimmune Diabetes

Paolo Fiorina; Andrea Vergani; Shirine Dada; Mollie Jurewicz; Masie Wong; Kenneth Law; Erxi Wu; Ze Tian; Reza Abdi; Indira Guleria; Scott J. Rodig; Kyri Dunussi-Joannopoulos; Jeffrey A. Bluestone; Mohamed H. Sayegh

OBJECTIVES—To investigate a B-cell–depleting strategy to reverse diabetes in naïve NOD mice. RESEARCH DESIGN AND METHODS—We targeted the CD22 receptor on B-cells of naïve NOD mice to deplete and reprogram B-cells to effectively reverse autoimmune diabetes. RESULTS—Anti-CD22/cal monoclonal antibody (mAb) therapy resulted in early and prolonged B-cell depletion and delayed disease in pre-diabetic mice. Importantly, when new-onset hyperglycemic mice were treated with the anti-CD22/cal mAb, 100% of B-cell–depleted mice became normoglycemic by 2 days, and 70% of them maintained a state of long-term normoglycemia. Early therapy after onset of hyperglycemia and complete B-cell depletion are essential for optimal efficacy. Treated mice showed an increase in percentage of regulatory T-cells in islets and pancreatic lymph nodes and a diminished immune response to islet peptides in vitro. Transcriptome analysis of reemerging B-cells showed significant changes of a set of proinflammatory genes. Functionally, reemerging B-cells failed to present autoantigen and prevented diabetes when cotransferred with autoreactive CD4+ T-cells into NOD.SCID hosts. CONCLUSIONS—Targeting CD22 depletes and reprograms B-cells and reverses autoimmune diabetes, thereby providing a blueprint for development of novel therapies to cure autoimmune diabetes.


Journal of Immunology | 2007

A Link between PDL1 and T Regulatory Cells in Fetomaternal Tolerance

Antje Habicht; Shirine Dada; Mollie Jurewicz; Brian T. Fife; Hideo Yagita; Miyuki Azuma; Mohamed H. Sayegh; Indira Guleria

Acceptance of the fetus expressing allogeneic paternal Ags by the mother is a physiologic model of transplantation tolerance. Various mechanisms contribute to fetal evasion from immune attack by maternal leukocytes. We have recently demonstrated that the inhibitory costimulatory molecule PDL1 plays a critical role in fetomaternal tolerance in that PDL1 blockade or deficiency resulted in decreased allogeneic fetal survival rates. CD4+CD25+ T regulatory cells (Tregs) have also been demonstrated to play an important role in fetomaternal tolerance. Since PDL1 is expressed on Tregs, we explored the interactions between PDL1 and Tregs in vivo in a mouse model of fetomaternal tolerance. Depletion of CD25+ T cells abrogated the effect of anti-PDL1 Ab indicating that the effect of PDL1 is possibly mediated by CD25+ Tregs. Adoptive transfer of Tregs from wild-type but not PDL1-deficient mice into PDL1-deficient recipients significantly improved fetal survival. The frequency, phenotype and placental trafficking of Tregs from PDL1-deficient mice were similar to those of wild-type controls, but were defective in inhibiting alloreactive Th1 cells in vitro. This is the first report providing evidence for a link between PDL1 and T regulatory cells in mediating fetomaternal tolerance.


Circulation | 2008

Critical Role of Donor Tissue Expression of Programmed Death Ligand-1 in Regulating Cardiac Allograft Rejection and Vasculopathy

Jun Yang; Joyce Popoola; Shakila Khandwala; Nidyanandh Vadivel; Vijay K. Vanguri; Xueli Yuan; Shirine Dada; Indira Guleria; Chaorui Tian; M. Javeed Ansari; Tahiro Shin; Hideo Yagita; Miyuki Azuma; Mohamed H. Sayegh; Anil Chandraker

Background— Allograft vasculopathy is a major limiting factor in the long-term success of cardiac transplantation. T cells play a critical role in initiation of cardiac allograft rejection and allograft vasculopathy. The negative T-cell costimulatory pathway PD-1:PDL1/PDL2 (programmed death-1:programmed death ligand-1/2) plays an important role in regulating alloimmune responses. We investigated the role of recipient versus donor PD-1 ligands in the pathogenesis of allograft rejection with emphasis on the role of tissue expression in regulating this alloimmune response in vivo. Methods and Results— We used established major histocompatibility complex class II– and class I–mismatched models of vascularized cardiac allograft rejection, blocking anti-PDL1 and anti-PDL2 antibodies, and PDL1- and PDL2-deficient mice (as donors or recipients) to study the role of the PD-1:PDL1/PDL2 pathway in chronic rejection. We also used PDL1-deficient and wild-type mice and bone marrow transplantation to generate chimeric animals that express PDL1 exclusively on either hematopoietic or parenchymal cells. PDL1 but not PDL2 blockade significantly accelerated cardiac allograft rejection in the bm12-into-B6 and B6-into-bm12 models. Although wild-type cardiac allografts survived long term, PDL1−/− donor hearts transplanted into wild-type bm12 mice exhibited accelerated rejection and vasculopathy associated with enhanced recipient T-cell alloreactivity. Interestingly, PDL1−/− recipients did not exhibit an accelerated tempo of cardiac allograft rejection. Using chimeric animals as donors, we show that PDL1 expression on cardiac tissue alone significantly prolonged graft survival compared with full PDL1−/− donor grafts in transplanted wild-type recipients. Conclusions— This is the first report to demonstrate that expression of the negative costimulatory molecule PDL1 on donor cardiac tissue regulates recipient alloimmune responses, allograft rejection, and vasculopathy.


Clinical Immunology | 2011

Anti-CD3 mAb treatment cures PDL1-/-.NOD mice of diabetes but precipitates fatal myocarditis.

Bechara Mfarrej; Mary E. Keir; Shirine Dada; Subbulaxmi Trikudanathan; Mohamed H. Sayegh; Arlene H. Sharpe; Indira Guleria

Anti-CD3 mAb is an effective therapy that can reverse diabetes in NOD mice and has therapeutic potential in patients with type 1 diabetes (T1D). We administered anti-CD3 to PDL1-/-.NOD mice in order to determine whether this treatment would reverse the development of diabetes in these mice. Mice injected with anti-CD3 mAb neonatally were protected from T1D. However, all of these anti-CD3 mAb treated PDL1-/-.NOD mice developed a wasting disease between 12 and 20 weeks of age with sudden deterioration and weight loss, leading to death within 3-5 days of development of illness. Histology revealed severe inflammation in the heart and skeletal muscles. These results suggest that deficiency of PDL1 in NOD background has the potential to lead to immune-mediated tissue damage in organs other than the pancreas, but this cannot be appreciated in PDL1-/-.NOD mice as the mice develop T1D at an early age and die from diabetes prior to manifesting other autoimmune diseases.


Clinical Immunology | 2007

Mechanisms of PDL1-mediated regulation of autoimmune diabetes

Indira Guleria; Melanie Gubbels Bupp; Shirine Dada; Brian T. Fife; Qizhi Tang; Mohammed Javeed Ansari; Subbulaxmi Trikudanathan; Nidyanandh Vadivel; Paolo Fiorina; Hideo Yagita; Miyuki Azuma; Mark A. Atkinson; Jeffrey A. Bluestone; Mohamed H. Sayegh


Clinical Immunology | 2008

Role of ICOS pathway in autoimmune and alloimmune responses in NOD mice

Mohammed Javeed Ansari; Paolo Fiorina; Shirine Dada; Indira Guleria; Takuya Ueno; Xueli Yuan; Subbulaxmi Trikudanathan; R. Neal Smith; Gordon J. Freeman; Mohamed H. Sayegh


American Journal of Pathology | 2013

B7h (ICOS-L) maintains tolerance at the fetomaternal interface.

Leonardo V. Riella; Shirine Dada; Lola Chabtini; Brian R. Smith; Lei Huang; Pranal J. Dakle; Bechara Mfarrej; Francesca D'Addio; La Tonya Adams; Nora M. Kochupurakkal; Andrea Vergani; Paolo Fiorina; Andrew L. Mellor; Arlene H. Sharpe; Hideo Yagita; Indira Guleria


Clinical Immunology | 2014

Corrigendum to 'Mechanisms of PDL1-mediated regulation of autoimmune diabetes' [Clin. Immunol. 125 (2007) 16-25]

Indira Guleria; Melanie Gubbels Bupp; Shirine Dada; Brian T. Fife; Qizhi Tang; Mohammed Javeed Ansari; Subbulaxmi Trikudanathan; Nidyanandh Vadivel; Paolo Fiorina; Hideo Yagita; Miyuki Azuma; Mark A. Atkinson; Jeffrey A. Bluestone; Mohamed H. Sayegh


Clinical Immunology | 2008

OR.65. Anti-B Cells Strategy Prevent and Cure Diabetes in NOD Mice

Mollie Jurewicz; Andrea Vergani; Shirine Dada; Kyriaki Dunussi-Joannopoulos

Collaboration


Dive into the Shirine Dada's collaboration.

Top Co-Authors

Avatar

Indira Guleria

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Mohamed H. Sayegh

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

Miyuki Azuma

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Andrea Vergani

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge