Network


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

Hotspot


Dive into the research topics where Miriam C. Souroujon is active.

Publication


Featured researches published by Miriam C. Souroujon.


Journal of Immunology | 2001

Blockade of CD40 ligand suppresses chronic experimental myasthenia gravis by down-regulation of Th1 differentiation and up-regulation of CTLA-4.

Sin-Hyeog Im; Dora Barchan; Prasanta K. Maiti; Sara Fuchs; Miriam C. Souroujon

Myasthenia gravis (MG) and experimental autoimmune MG (EAMG) are T cell-dependent Ab-mediated autoimmune disorders, in which the nicotinic acetylcholine receptor (AChR) is the major autoantigen. Th1-type cells and costimulatory factors such as CD40 ligand (CD40L) contribute to disease pathogenesis by producing proinflammatory cytokines and by activating autoreactive B cells. In this study we demonstrate the capacity of CD40L blockade to modulate EAMG, and analyze the mechanism underlying this disease suppression. Anti-CD40L Abs given to rats at the chronic stage of EAMG suppress the clinical progression of the autoimmune process and lead to a decrease in the AChR-specific humoral response and delayed-type hypersensitivity. The cytokine profile of treated rats suggests that the underlying mechanism involves down-regulation of AChR-specific Th1-regulated responses with no significant effect on Th2- and Th3-regulated AChR-specific responses. EAMG suppression is also accompanied by a significant up-regulation of CTLA-4, whereas a series of costimulatory factors remain unchanged. Adoptive transfer of splenocytes from anti-CD40L-treated rats does not protect recipient rats against subsequently induced EAMG. Thus it seems that the suppressed progression of chronic EAMG by anti-CD40L treatment does not induce a switch from Th1 to Th2/Th3 regulation of the AChR-specific immune response and does not induce generation of regulatory cells. The ability of anti-CD40L treatment to suppress ongoing chronic EAMG suggests that blockade of CD40L may serve as a potential approach for the immunotherapy of MG and other Ab-mediated autoimmune diseases.


Journal of Clinical Investigation | 1999

Suppression of ongoing experimental myasthenia by oral treatment with an acetylcholine receptor recombinant fragment

Sin-Hyeog Im; Dora Barchan; Sara Fuchs; Miriam C. Souroujon

Myasthenia gravis (MG) is an autoimmune disorder in which the nicotinic acetylcholine receptor (AChR) is the major autoantigen. In an attempt to develop an antigen-specific therapy for MG, we administered a nonmyasthenogenic recombinant fragment of AChR orally to rats. This fragment, corresponding to the extracellular domain of the human AChR alpha-subunit (Halpha1-205), protected rats from subsequently induced experimental autoimmune myasthenia gravis (EAMG) and suppressed ongoing EAMG when treatment was initiated during either the acute or chronic phases of disease. Prevention and suppression of EAMG were accompanied by a significant decrease in AChR-specific humoral and cellular responses. The underlying mechanism for the Halpha1-205-induced oral tolerance seems to be active suppression, mediated by a shift from a T-helper 1 (Th1) to a Th2/Th3 response. This shift was assessed by changes in the cytokine profile, a deviation of anti-AChR IgG isotypes from IgG2 to IgG1, and a suppressed AChR-specific delayed-type hypersensitivity response. Our results in experimental myasthenia suggest that oral administration of AChR-specific recombinant fragments may be considered for antigen-specific immunotherapy of myasthenia gravis.


Journal of Immunology | 2008

Ex Vivo Generated Regulatory T Cells Modulate Experimental Autoimmune Myasthenia Gravis

Revital Aricha; Tali Feferman; Sara Fuchs; Miriam C. Souroujon

Naturally occurring CD4+CD25+ regulatory T (Treg) cells are key players in immune tolerance and have therefore been suggested as potential therapeutic tools for autoimmune diseases. In myasthenia gravis (MG), reduced numbers or functionally impaired Treg cells have been reported. We have observed that PBL from myasthenic rats contain decreased numbers of CD4+CD25highFoxp3+ cells as compared with PBL from healthy controls, and we have tested whether Treg cells from healthy donors can suppress experimental autoimmune MG in rats. Because the number of naturally occurring Treg cells is low, we used an approach for a large-scale ex vivo generation of functional Treg cells from CD4+ splenocytes of healthy donor rats. Treg cells were generated ex vivo from CD4+ cells by stimulation with anti-CD3 and anti-CD28 Abs in the presence of TGF-β and IL-2. The obtained cells expressed high levels of CD25, CTLA-4, and Foxp3, and they were capable of suppressing in vitro proliferation of T cells from myasthenic rats in response to acetylcholine receptor, the major autoantigen in myasthenia. Administration of ex vivo-generated Treg cells to myasthenic rats inhibited the progression of experimental autoimmune MG and led to down-regulation of humoral acetylcholine receptor-specific responses, and to decreased IL-18 and IL-10 expression. The number of CD4+CD25+ cells in the spleen of treated rats remained unchanged, but the subpopulation of CD4+CD25+ cells expressing Foxp3 was significantly elevated. Our findings imply that Treg cells play a critical role in the control of myasthenia and could thus be considered as potential agents for the treatment of MG patients.


Journal of Immunology | 2005

Overexpression of IFN-Induced Protein 10 and Its Receptor CXCR3 in Myasthenia Gravis

Tali Feferman; Prasanta K. Maiti; Sonia Berrih-Aknin; Jacky Bismuth; Jocelyne Bidault; Sara Fuchs; Miriam C. Souroujon

Myasthenia gravis (MG) and its animal model, experimental autoimmune MG (EAMG), are autoimmune disorders in which the acetylcholine receptor (AChR) is the major autoantigen. Microarray technology was used to identify new potential drug targets for treatment of myasthenia that would reduce the need for the currently used nonspecific immunosuppression. The chemokine IFN-γ-inducible protein 10 (IP-10; CXCL10), a CXC chemokine, and its receptor, CXCR3, were found to be overexpressed in lymph node cells of EAMG rats. Quantitative real-time PCR confirmed these findings and revealed up-regulated mRNA levels of another chemoattractant that activates CXCR3, monokine induced by IFN-γ (Mig; CXCL9). TNF-α and IL-1β, which act synergistically with IFN-γ to induce IP-10, were also up-regulated. These up-regulations were observed in immune response effector cells, namely, lymph node cells, and in the target organ of the autoimmune attack, the muscle of myasthenic rats, and were significantly reduced after suppression of EAMG by mucosal tolerance induction with an AChR fragment. The relevance of IP-10/CXCR3 signaling in myasthenia was validated by similar observations in MG patients. A significant increase in IP-10 and CXCR3 mRNA levels in both thymus and muscle was observed in myasthenic patients compared with age-matched controls. CXCR3 expression in PBMC of MG patients was markedly increased in CD4+, but not in CD8+, T cells or in CD19+ B cells. Our results demonstrate a positive association of IP-10/CXCR3 signaling with the pathogenesis of EAMG in rats as well as in human MG patients.


The FASEB Journal | 2001

Suppression of experimental myasthenia gravis, a B cell-mediated autoimmune disease, by blockade of IL-18

Sin-Hyeog Im; Dora Barchan; Prasanta K. Maiti; Lily Raveh; Miriam C. Souroujon; Sara Fuchs

Interleukin‐18 (IL‐18) is a pleiotropic proinflammatory cytokine that plays an important role in interferon gamma (IFN‐γ) production and IL‐12‐driven Th1 phenotype polarization. Increased expression of IL‐18 has been observed in several autoimmune diseases. In this study we have analyzed the role of IL‐18 in an antibody‐mediated autoimmune disease and elucidated the mechanisms involved in disease suppression mediated by blockade of IL‐18, using experimental autoimmune myasthenia gravis (EAMG) as a model. EAMG is a T cell‐regulated, antibody‐mediated autoimmune disease in which the nicotinic acetylcholine receptor (AChR) is the major autoantigen. Th1‐ and Th2‐type responses are both implicated in EAMG development. We show that treatment by anti‐IL‐18 during ongoing EAMG suppresses disease progression. The protective effect can be adoptively transferred to naive recipients and is mediated by increased levels of the immunosuppressive Th3‐type cytokine TGF‐β and decreased AChR‐specific Th1‐type cellular responses. Suppression of EAMG is accompanied by down‐regulation of the costimulatory factor CD40L and up‐regula‐tion of CTLA‐4, a key negative immunomodulator. Our results suggest that IL‐18 blockade may potentially be applied for immunointervention in myasthenia gravis.—Im, S.‐H., Barchan, D., Maiti, P. K., Raveh, L., Souroujon, M. C., Fuchs, S. Suppression of experimental myasthenia gravis, a B cell‐mediated autoimmune disease, by blockade of IL‐18. FASEB J. 15, 2140–2148 (2001)


Journal of Autoimmunity | 2011

Blocking of IL-6 suppresses experimental autoimmune myasthenia gravis

Revital Aricha; Keren Mizrachi; Sara Fuchs; Miriam C. Souroujon

Suppressive regulatory T cells (Treg) and pathogenic T helper 17 (Th17) cells are two lymphocyte subsets with opposing activities in autoimmune diseases. The proinflammatory cytokine IL-6 is a potent factor in switching immune responses in vivo from the induction of Treg to pathogenic Th17 cells. We studied the Treg and Th17 cell compartments in experimental autoimmune myasthenia gravis (EAMG) and healthy control rats in order to assess whether the equilibrium between Treg and Th17 cells is perturbed in the disease. We found that Th17 cell-related genes are upregulated and Treg-related genes are downregulated in EAMG. The shift in favor of Th17 cells in EAMG could be reversed by antibodies to IL-6. Administration of anti-IL-6 antibodies to myasthenic rats suppressed EAMG when treatment started at the acute or at the chronic phase of disease. Suppression of EAMG by anti-IL-6 antibodies was accompanied by a decrease in the overall rat anti-AChR antibody titer and by a reduced number of B cells as compared with control treatment. Administration of anti-IL-6 antibodies led to down-regulation of several Th17 related genes including IL-17, IL-17R, IL-23R and IL-21 but did not affect the number of Treg cells in the lymph nodes. These data identify IL-6 as an important target for modulation of autoimmune responses.


Journal of Neuroimmunology | 2004

Immunosuppression of rat myasthenia gravis by oral administration of a syngeneic acetylcholine receptor fragment

Prasanta K. Maiti; Tali Feferman; Sin-Hyeog Im; Miriam C. Souroujon; Sara Fuchs

A syngeneic rat recombinant fragment of the extracellular domain of the acetylcholine receptor (AChR) alpha-subunit (Ralpha1-205), administered orally, suppresses ongoing experimental autoimmune myasthenia gravis (EAMG) in rats. The underlying mechanism is a shift from Th1 to Th2 regulation as evidenced by downregulated mRNA expression levels of IFN-gamma and TNF-alpha, upregulated IL-10, changes in anti-AChR IgG isotypes and diminished Th1 signaling via CD28/CTLA-4:B7. Unlike the xenogeneic fragment, the syngeneic Ralpha1-205 does not induce elevation in TGF-beta and elicitation of autoregulatory cells. The ability to suppress EAMG by a non-immunogenic syngeneic fragment of AChR is encouraging and may in the future be applied for the treatment of myasthenia gravis in humans.


The FASEB Journal | 2005

Overexpression of phosphodiesterases in experimental autoimmune myasthenia gravis: suppression of disease by a phosphodiesterase inhibitor

Revital Aricha; Tali Feferman; Miriam C. Souroujon; Sara Fuchs

Myasthenia gravis (MG) and experimental autoimmune MG (EAMG) are T cell‐dependent antibody‐mediated autoimmune disorders, in which the nicotinic acetylcholine receptor (AChR) is the major autoantigen. DNA microarray analysis revealed increased levels of several phosphodiesterase (PDE) subtypes in lymph node cells (LNC) and muscles of EAMG rats compared with healthy controls. Quantitative real‐time PCR analysis indicated that EAMG is characterized by an increase of PDE subtypes 1, 3, 4, and 7 in LNC and of PDE subtypes 2, 3, 4, and 7 in muscles. Pentoxifylline (PTX), a general PDE inhibitor, inhibited the progression of EAMG when treatment started at either the acute or chronic stages of disease. This suppression was associated with down‐regulation of humoral and cellular AChR‐specific responses, as well as down‐regulation of PDE4, TNF‐α, IL‐18, IL‐12, and IL‐10 in LNC and of PDEs 1, 4, 7, and TNF‐α in muscles. The expression of Foxp3, a transcription factor essential for CD4+CD25+ regulatory T cell function, was increased in splenocytes although the number of these cells remained unchanged. PTX also reduced the expression of the endopeptidase cathepsin‐l, a marker of muscle damage, in EAMG muscles. This study demonstrates the involvement of PDE regulation in EAMG pathogenesis and suggests that PDE inhibitors may be considered for immunotherapy of MG.


European Journal of Immunology | 1998

Modulation of the anti-acetylcholine receptor response and experimental autoimmune myasthenia gravis by recombinant fragments of the acetylcholine receptor.

Dora Barchan; Orna Asher; Socrates J. Tzartos; Sara Fuchs; Miriam C. Souroujon

Myasthenia gravis (MG) is a neuromuscular disorder of man caused by a humoral response to the acetylcholine receptor (AChR). Most of the antibodies in MG and in experimental autoimmune myasthenia gravis (EAMG) are directed to the extracellular portion of the AChR α subunit, and within it, primarily to the main immunogenic region (MIR). We have cloned and expressed recombinant fragments, corresponding to the entire extracellular domain of the AChR α subunit (Hα1 – 210), and to portions of it that encompass either the MIR (Hα1 – 121) or the ligand binding site of AChR (Hα122 – 210), and studied their ability to interfere with the immunopathological anti‐AChR response in vitro and in vivo. All fragments were expressed as fusion proteins with glutathione S‐transferase. Fragments Hα1 – 121 and Hα1 – 210 protected AChR in TE671 cells against accelerated degradation induced by the anti‐MIR monoclonal antibody (mAb)198 in a dose‐dependent manner. Moreover, these fragments had a similar effect on the antigenic modulation of AChR by other anti‐MIR mAb and by polyclonal rat anti‐AChR antibodies. Fragments Hα1 – 121 and Hα1 – 210 were also able to modulate in vivo muscle AChR loss and development of clinical symptoms of EAMG, passively transferred to rats by mAb 198. Fragment Hα122 – 210 did not have such a protective activity. Our results suggest that the appropriate recombinant fragments of the human AChR may be employed in the future for antigen‐specific therapy of myasthenia.


Journal of Neuroimmunology | 2000

Mechanism of nasal tolerance induced by a recombinant fragment of acetylcholine receptor for treatment of experimental myasthenia gravis

Sin-Hyeog Im; Dora Barchan; Sara Fuchs; Miriam C. Souroujon

Acetylcholine receptor (AChR) is the major autoantigen in myasthenia gravis (MG) and experimental autoimmune MG (EAMG). Here we analyze the mechanisms involved in suppression of ongoing EAMG in rats by nasal administration of a recombinant fragment from the human AChR alpha-subunit. We demonstrate that such a fragment, expressed without a fusion partner, confers nasal tolerance that can be adoptively transferred. Our observations suggest that the underlying mechanism of this nasal tolerance is active suppression involving a shift from a Th1 to a Th2/Th3-regulated AChR-specific response which may be mediated by down regulation of costimulatory factors.

Collaboration


Dive into the Miriam C. Souroujon's collaboration.

Top Co-Authors

Avatar

Sara Fuchs

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Tali Feferman

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Revital Aricha

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Sin-Hyeog Im

Pohang University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Dora Barchan

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Prasanta K. Maiti

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Moshe Mittelman

Tel Aviv Sourasky Medical Center

View shared research outputs
Top Co-Authors

Avatar

Keren Mizrachi

Weizmann Institute of Science

View shared research outputs
Researchain Logo
Decentralizing Knowledge