Network


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

Hotspot


Dive into the research topics where Moufida Ben Nasr is active.

Publication


Featured researches published by Moufida Ben Nasr.


Diabetes | 2014

Autologous Nonmyeloablative Hematopoietic Stem Cell Transplantation in New-Onset Type 1 Diabetes: A Multicenter Analysis

Francesca D'Addio; Alessandro Valderrama Vasquez; Moufida Ben Nasr; Edward Franek; Dalong Zhu; Lirong Li; Guang Ning; Emilian Snarski; Paolo Fiorina

Type 1 diabetes (T1D) is one of the major autoimmune diseases affecting children and young adults worldwide. To date, the different immunotherapies tested have achieved insulin independence in <5% of treated individuals. Recently, a novel hematopoietic stem cell (HSC)–based strategy has been tested in individuals with new-onset T1D. The aim of this study was to determine the effects of autologous nonmyeloablative HSC transplantation in 65 individuals with new-onset T1D who were enrolled in two Chinese centers and one Polish center, pooled, and followed up for 48 months. A total of 59% of individuals with T1D achieved insulin independence within the first 6 months after receiving conditioning immunosuppression therapy (with antithymocyte globulin and cyclophosphamide) and a single infusion of autologous HSCs, and 32% remained insulin independent at the last time point of their follow-up. All treated subjects showed a decrease in HbA1c levels and an increase in C-peptide levels compared with pretreatment. Despite a complete immune system recovery (i.e., leukocyte count) after treatment, 52% of treated individuals experienced adverse effects. Our study suggests the following: 1) that remission of T1D is possible by combining HSC transplantation and immunosuppression; 2) that autologous nonmyeloablative HSC transplantation represents an effective treatment for selected individuals with T1D; and 3) that safer HSC-based therapeutic options are required.


Journal of The American Society of Nephrology | 2014

Role of Podocyte B7-1 in Diabetic Nephropathy

Paolo Fiorina; Andrea Vergani; Roberto Bassi; Monika A. Niewczas; Mehmet M. Altintas; Marcus G. Pezzolesi; Francesca D’Addio; Melissa Chin; Sara Tezza; Moufida Ben Nasr; Deborah Mattinzoli; Masami Ikehata; Domenico Corradi; Valérie Schumacher; Lisa Buvall; Chih-Chuan Yu; Jer-Ming Chang; Stefano La Rosa; Giovanna Finzi; Anna Solini; Flavio Vincenti; Maria Pia Rastaldi; Jochen Reiser; Andrzej S. Krolewski; Peter Mundel; Mohamed H. Sayegh

Podocyte injury and resulting albuminuria are hallmarks of diabetic nephropathy, but targeted therapies to halt or prevent these complications are currently not available. Here, we show that the immune-related molecule B7-1/CD80 is a critical mediator of podocyte injury in type 2 diabetic nephropathy. We report the induction of podocyte B7-1 in kidney biopsy specimens from patients with type 2 diabetes. Genetic and epidemiologic studies revealed the association of two single nucleotide polymorphisms at the B7-1 gene with diabetic nephropathy. Furthermore, increased levels of the soluble isoform of the B7-1 ligand CD28 correlated with the progression to ESRD in individuals with type 2 diabetes. In vitro, high glucose conditions prompted the phosphatidylinositol 3 kinase-dependent upregulation of B7-1 in podocytes, and the ectopic expression of B7-1 in podocytes increased apoptosis and induced disruption of the cytoskeleton that were reversed by the B7-1 inhibitor CTLA4-Ig. Podocyte expression of B7-1 was also induced in vivo in two murine models of diabetic nephropathy, and treatment with CTLA4-Ig prevented increased urinary albumin excretion and improved kidney pathology in these animals. Taken together, these results identify B7-1 inhibition as a potential therapeutic strategy for the prevention or treatment of diabetic nephropathy.


Science Translational Medicine | 2017

PD-L1 genetic overexpression or pharmacological restoration in hematopoietic stem and progenitor cells reverses autoimmune diabetes

Moufida Ben Nasr; Sara Tezza; Francesca D’Addio; Chiara Mameli; Vera Usuelli; Anna Maestroni; Domenico Corradi; Silvana Belletti; Luca Albarello; Gabriella Becchi; Gian Paolo Fadini; Christian Schuetz; James F. Markmann; Clive Wasserfall; Leonard I. Zon; Gian Vincenzo Zuccotti; Paolo Fiorina

Restoration of a PD-L1 defect in HSPCs reverses diabetes in NOD mice and thus may represent a potential cure for T1D. Stemming attacks on the pancreas In type 1 diabetes, autoreactive CD4 T cells attack and kill pancreatic β cells, disrupting insulin production. Many approaches have been taken to inhibit this process, but few have translated into real benefit for diabetic patients. Ben Nasr et al. demonstrate that hematopoietic stem and progenitor cells from NOD mice and diabetic patients express less PD-L1, which is a T cell inhibitory molecule. Induction of PD-L1 expression on stem cells reversed diabetes in NOD mice and inhibited human autoimmune responses in vitro. Either gene therapy or pharmacological modulation of PD-L1 on stem cells could be brought into the clinic, providing a new way to interrupt the autoimmune response and help people with diabetes. Immunologically based clinical trials performed thus far have failed to cure type 1 diabetes (T1D), in part because these approaches were nonspecific. Because the disease is driven by autoreactive CD4 T cells, which destroy β cells, transplantation of hematopoietic stem and progenitor cells (HSPCs) has been recently offered as a therapy for T1D. Our transcriptomic profiling of HSPCs revealed that these cells are deficient in programmed death ligand 1 (PD-L1), an important immune checkpoint, in the T1D nonobese diabetic (NOD) mouse model. Notably, the immunoregulatory molecule PD-L1 plays a determinant role in controlling/inhibiting activated T cells and thus maintains immune tolerance. Furthermore, our genome-wide and bioinformatic analysis revealed the existence of a network of microRNAs (miRNAs) controlling PD-L1 expression, and silencing one of key altered miRNAs restored PD-L1 expression in HSPCs. We therefore sought to determine whether restoration of this defect would cure T1D as an alternative to immunosuppression. Genetically engineered or pharmacologically modulated HSPCs overexpressing PD-L1 inhibited the autoimmune response in vitro, reverted diabetes in newly hyperglycemic NOD mice in vivo, and homed to the pancreas of hyperglycemic NOD mice. The PD-L1 expression defect was confirmed in human HSPCs in T1D patients as well, and pharmacologically modulated human HSPCs also inhibited the autoimmune response in vitro. Targeting a specific immune checkpoint defect in HSPCs thus may contribute to establishing a cure for T1D.


Cell Stem Cell | 2015

Circulating IGF-I and IGFBP3 Levels Control Human Colonic Stem Cell Function and Are Disrupted in Diabetic Enteropathy

Francesca D’Addio; Stefano La Rosa; Anna Maestroni; Peter Jung; Elena Orsenigo; Moufida Ben Nasr; Sara Tezza; Roberto Bassi; Giovanna Finzi; Alessandro Marando; Andrea Vergani; Roberto Frego; Luca Albarello; Annapaola Andolfo; Roberta Manuguerra; Edi Viale; Carlo Staudacher; Domenico Corradi; Eduard Batlle; David T. Breault; Antonio Secchi; Franco Folli; Paolo Fiorina

The role of circulating factors in regulating colonic stem cells (CoSCs) and colonic epithelial homeostasis is unclear. Individuals with long-standing type 1 diabetes (T1D) frequently have intestinal symptoms, termed diabetic enteropathy (DE), though its etiology is unknown. Here, we report that T1D patients with DE exhibit abnormalities in their intestinal mucosa and CoSCs, which fail to generate in vitro mini-guts. Proteomic profiling of T1D+DE patient serum revealed altered levels of insulin-like growth factor 1 (IGF-I) and its binding protein 3 (IGFBP3). IGFBP3 prevented in vitro growth of patient-derived organoids via binding its receptor TMEM219, in an IGF-I-independent manner, and disrupted in vivo CoSC function in a preclinical DE model. Restoration of normoglycemia in patients with long-standing T1D via kidney-pancreas transplantation or in diabetic mice by treatment with an ecto-TMEM219 recombinant protein normalized circulating IGF-I/IGFBP3 levels and reestablished CoSC homeostasis. These findings demonstrate that peripheral IGF-I/IGFBP3 controls CoSCs and their dysfunction in DE.


Diabetes | 2015

Interleukin-10+ Regulatory B Cells Arise Within Antigen-Experienced CD40+ B Cells to Maintain Tolerance to Islet Autoantigens

Sonja Kleffel; Andrea Vergani; Sara Tezza; Moufida Ben Nasr; Monika A. Niewczas; Susan Wong; Roberto Bassi; Francesca D’Addio; Tobias Schatton; Reza Abdi; Mark A. Atkinson; Mohamed H. Sayegh; Li Wen; Clive Wasserfall; Kevin C. O’Connor; Paolo Fiorina

Impaired regulatory B cell (Breg) responses are associated with several autoimmune diseases in humans; however, the role of Bregs in type 1 diabetes (T1D) remains unclear. We hypothesized that naturally occurring, interleukin-10 (IL-10)–producing Bregs maintain tolerance to islet autoantigens, and that hyperglycemic nonobese diabetic (NOD) mice and T1D patients lack these potent negative regulators. IgVH transcriptome analysis revealed that islet-infiltrating B cells in long-term normoglycemic (Lnglc) NOD, which are naturally protected from diabetes, are more antigen-experienced and possess more diverse B-cell receptor repertoires compared to those of hyperglycemic (Hglc) mice. Importantly, increased levels of Breg-promoting CD40+ B cells and IL-10–producing B cells were found within islets of Lnglc compared to Hglc NOD. Likewise, healthy individuals showed increased frequencies of both CD40+ and IL-10+ B cells compared to T1D patients. Rituximab-mediated B-cell depletion followed by adoptive transfer of B cells from Hglc mice induced hyperglycemia in Lnglc human CD20 transgenic NOD mouse models. Importantly, both murine and human IL-10+ B cells significantly abrogated T-cell–mediated responses to self- or islet-specific peptides ex vivo. Together, our data suggest that antigen-matured Bregs may maintain tolerance to islet autoantigens by selectively suppressing autoreactive T-cell responses, and that Hglc mice and individuals with T1D lack this population of Bregs.


American Journal of Respiratory Cell and Molecular Biology | 2014

Inhibition of the Purinergic Pathway Prolongs Mouse Lung Allograft Survival

Kaifeng Liu; Andrea Vergani; Picheng Zhao; Moufida Ben Nasr; Xiao Wu; Khadija Iken; Dawei Jiang; Xiaofeng Su; Carmen Fotino; Paolo Fiorina; Gary A. Visner

Lung transplantation has limited survival with current immunosuppression. ATP is released from activated T cells, which act as costimulatory molecules through binding to the purinergic receptor P2XR7. We investigated the role of blocking the ATP/purinergic pathway, primarily P2XR7, using its inhibitor oxidized ATP (oATP) in modulating rejection of mouse lung allografts. Mouse lung transplants were performed using mice with major histocompatibility complex mismatch, BALB/c to C57BL6. Recipients received suramin or oATP, and lung allografts were evaluated 15 to ≥ 60 days after transplantation. Recipients were also treated with oATP after the onset of moderate to severe rejection to determine its ability to rescue lung allografts. Outcomes measures included lung function, histology, thoracic imaging, and allo-immune responses. Blocking purinergic receptors with the nonselective inhibitor suramin or with the P2XR7-selective inhibitor oATP reduced acute rejection and prolonged lung allograft survival for ≥ 60 days with no progression in severity. There were fewer inflammatory cells within lung allografts, less rejection, and improved lung function, which was maintained over time. CD4 and CD8 T cells were reduced within lung allografts with impaired activation with prolonged impairment of CD8 responses. In vitro, oATP reduced CD8 activation of Th1 inflammatory cytokines IFN-γ and TNF-α and cytolytic machinery, granzyme B. Cotreatment with immunosuppressive agents, cyclosporine, rapamycin, or CTLA-4Ig resulted in no additive benefits, and oATP alone resulted in better outcomes than cyclosporine alone. This study illustrates a potential new pathway to target in hopes of prolonging survival of lung transplant recipients.


Experimental Diabetes Research | 2012

Regenerative Therapies for Diabetic Microangiopathy

Roberto Bassi; Alessio Trevisani; Sara Tezza; Moufida Ben Nasr; Francesca Gatti; Andrea Vergani; Antonio Farina; Paolo Fiorina

Hyperglycaemia occurring in diabetes is responsible for accelerated arterial remodeling and atherosclerosis, affecting the macro- and the microcirculatory system. Vessel injury is mainly related to deregulation of glucose homeostasis and insulin/insulin-precursors production, generation of advanced glycation end-products, reduction in nitric oxide synthesis, and oxidative and reductive stress. It occurs both at extracellular level with increased calcium and matrix proteins deposition and at intracellular level, with abnormalities of intracellular pathways and increased cell death. Peripheral arterial disease, coronary heart disease, and ischemic stroke are the main causes of morbidity/mortality in diabetic patients representing a major clinical and economic issue. Pharmacological therapies, administration of growth factors, and stem cellular strategies are the most effective approaches and will be discussed in depth in this comprehensive review covering the regenerative therapies of diabetic microangiopathy.


Journal of Endocrinological Investigation | 2017

Immunotherapy for type 1 diabetes

Davide Frumento; Moufida Ben Nasr; Basset El Essawy; Francesca D’Addio; Gian Vincenzo Zuccotti; Paolo Fiorina

IntroductionAlthough many approaches have been tested to overcome the insulin dependence caused by the pancreatic β-cells destruction observed in individuals affected by type 1 diabetes (T1D), medical research has largely failed to halt the onset or to reverse T1D.MethodsIn this work, the state of the art of immunotherapy will be examined, and the most important achievement in the field will be critically discussed. Particularly, we will focus on the clinical aspect, thus avoiding the tedious preclinical work done in NOD mice, which has been so poorly translated to the bedside.ConclusionsStem cell therapies achieved thus this far the most promising results, while immune ablation and standard immunosuppressants did not maintain the premises of preclinical results. The next step will be to generate a feasible and safe clinical approach in order to cure the thousands of patients affected by T1D.


PLOS ONE | 2017

Metabolomic Profiling in Individuals with a Failing Kidney Allograft

Roberto Bassi; Monika A. Niewczas; Luigi Biancone; Stefania Bussolino; Sai Merugumala; Sara Tezza; Francesca D’Addio; Moufida Ben Nasr; Alessandro Valderrama-Vasquez; Vera Usuelli; Valentina De Zan; Basset El Essawy; Massimo Venturini; Antonio Secchi; Francesco De Cobelli; Alexander Lin; Anil Chandraker; Paolo Fiorina

Background Alteration of certain metabolites may play a role in the pathophysiology of renal allograft disease. Methods To explore metabolomic abnormalities in individuals with a failing kidney allograft, we analyzed by liquid chromatography-mass spectrometry (LC-MS/MS; for ex vivo profiling of serum and urine) and two dimensional correlated spectroscopy (2D COSY; for in vivo study of the kidney graft) 40 subjects with varying degrees of chronic allograft dysfunction stratified by tertiles of glomerular filtration rate (GFR; T1, T2, T3). Ten healthy non-allograft individuals were chosen as controls. Results LC-MS/MS analysis revealed a dose-response association between GFR and serum concentration of tryptophan, glutamine, dimethylarginine isomers (asymmetric [A]DMA and symmetric [S]DMA) and short-chain acylcarnitines (C4 and C12), (test for trend: T1-T3 = p<0.05; p = 0.01; p<0.001; p = 0.01; p = 0.01; p<0.05, respectively). The same association was found between GFR and urinary levels of histidine, DOPA, dopamine, carnosine, SDMA and ADMA (test for trend: T1-T3 = p<0.05; p<0.01; p = 0.001; p<0.05; p = 0.001; p<0.001; p<0.01, respectively). In vivo 2D COSY of the kidney allograft revealed significant reduction in the parenchymal content of choline, creatine, taurine and threonine (all: p<0.05) in individuals with lower GFR levels. Conclusions We report an association between renal function and altered metabolomic profile in renal transplant individuals with different degrees of kidney graft function.


Cell Transplantation | 2015

TIM4 Regulates the Anti-Islet Th2 Alloimmune Response.

Andrea Vergani; Francesca Gatti; Kang M. Lee; Francesca D’Addio; Sara Tezza; Melissa Chin; Roberto Bassi; Ze Tian; Erxi Wu; Paola Maffi; Moufida Ben Nasr; James I. Kim; Antonio Secchi; James F. Markmann; David M. Rothstein; Laurence A. Turka; Mohamed H. Sayegh; Paolo Fiorina

The role of the novel costimulatory molecule TIM4 in anti-islet response is unknown. We explored TIM4 expression and targeting in Th1 (BALB/c islets into C57BL/6 mice) and Th2 (BALB/c islets into Tbet-/- C57BL/6 mice) models of anti-islet alloimmune response and in a model of anti-islet autoimmune response (diabetes onset in NOD mice). The targeting of TIM4, using the monoclonal antibody RMT4-53, promotes islet graft survival in a Th1 model, with 30% of the graft surviving in the long term; islet graft protection appears to be mediated by a Th1 to Th2 skewing of the immune response. Differently, in the Th2 model, TIM4 targeting precipitates graft rejection by further enhancing the Th2 response. The effect of anti-TIM4 treatment in preventing autoimmune diabetes was marginal with only minor Th1 to Th2 skewing. B-Cell depletion abolished the effect of TIM4 targeting. TIM4 is expressed on human B-cells and is upregulated in diabetic and islet-transplanted patients. Our data suggest a model in which TIM4 targeting promotes Th2 response over Th1 via B-cells. The targeting of TIM4 could become a component of an immunoregulatory protocol in clinical islet transplantation, aiming at redirecting the immune system toward a Th2 response.

Collaboration


Dive into the Moufida Ben Nasr's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Francesca D’Addio

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Sara Tezza

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Vera Usuelli

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Andrea Vergani

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Roberto Bassi

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anna Maestroni

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Reza Abdi

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge