hosted by
publicationslist.org
    
suryasarathi dasgupta
Channing Laboratory, 6th Floor
Brigham and Women's Hospital,
Harvard Medical School,
181 Longwood Avenue,
Boston 02115,
MA, USA
sdasgupta@rics.bwh.harvard.edu

Journal articles

2009
 
PMID 
S André, Y Meslier, J D Dimitrov, Y Repessé, S V Kaveri, S Lacroix-Desmazes, S Dasgupta (2009)  A Cellular Viewpoint of Anti-FVIII Immune Response in Hemophilia A.   Clin Rev Allergy Immunol. Jan  
Abstract: A large proportion of hemophilia A patients who receive replacement therapy, develop an immune response toward the infused factor VIII (FVIII). In this review, we discuss recent progress in several aspects of the anti-FVIII immune response, focusing on the sites of FVIII endocytosis (marginal zone of the spleen and bleeding site), the type of antigen-presenting cells (dendritic cells, macrophages and B cells) and endocytic receptors, implicated in FVIII presentation to T cells during primary and secondary immune response. Although it is becoming increasingly clear that regulatory T cells are involved in FVIII tolerance in healthy subjects and potentially in patients without inhibitors, we would like to demonstrate that little is known about the different T cells subsets and the cytokines network, which are also crucial for the development of allo- and autoimmune diseases. As more information on these issues becomes available, a better understanding of the role of each immune cells compartment in promoting FVIII tolerance or inhibitors development might lead to new strategies to promote FVIII tolerance in hemophilia A patients.
Notes:
2008
 
DOI   
PMID 
S Lacroix-Desmazes, A M Navarrete, S André, J Bayry, S V Kaveri, S Dasgupta (2008)  Dynamics of factor VIII interactions determine its immunological fate in hemophilia A.   Blood 112: 2. 240-249 May  
Abstract: Pro-coagulant factor VIII (FVIII) is either produced endogenously under physiological conditions, or administered exogenously as a therapeutic hemostatic drug in patients with hemophilia A. In the circulation, FVIII interacts with a multitude of glycoproteins, and may be used for coagulation at the sites of bleeding, eliminated by scavenger cells or be processed by the immune system, either as a self-constituent or as a foreign antigen. The fate of FVIII is dictated by the immune status of the individual, the location of FVIII in the body at a given time-point, and the inflammatory microenvironment. It also depends on the local concentration of FVIII and of each interacting partner, and on the affinity of the respective interactions. FVIII, by virtue of its promiscuity, thus constitutes the core of a dynamic network that links the coagulation cascade, cells of the immune system and, presumably, the inflammatory compartment. We describe the different interactions that FVIII is prone to establish during its life cycle, with a special focus on players of the innate and adaptive immune response. Lessons can be learned from understanding the dynamics of FVIII interactions; lessons that should pave the way to the conception of long-lasting hemostatic drugs devoid of iatrogenic immunogenicity.
Notes:
 
PMID 
Bharath Wootla, Suryasarathi Dasgupta, Jordan D Dimitrov, Jagadeesh Bayry, Hervé Lévesque, Jeanne-Yvonne Borg, Annie Borel-Derlon, Desirazu N Rao, Alain Friboulet, Srinivas V Kaveri, Sébastien Lacroix-Desmazes (2008)  Factor VIII Hydrolysis Mediated by Anti-Factor VIII Autoantibodies in Acquired Hemophilia.   J Immunol 180: 11. 7714-7720 Jun  
Abstract: Acquired hemophilia is a rare hemorrhagic disorder caused by the spontaneous appearance of inhibitory autoantibodies directed against endogenous coagulation factor VIII (FVIII). Inhibitory Abs also arise in patients with congenital hemophilia A as alloantibodies directed to therapeutic FVIII. Both autoimmune and alloimmune inhibitors neutralize FVIII by steric hindrance. We have described FVIII-hydrolyzing IgG in 50% of inhibitor-positive patients with severe hemophilia A that inactivate therapeutic FVIII. In this study, we investigated the presence of autoimmune FVIII-hydrolyzing IgG in patients with acquired hemophilia. Pooled IgG from healthy donors demonstrated moderate FVIII-hydrolyzing activity (56 +/- 26 mumol/min/mol). Purified IgG from 21 of 45 patients with acquired hemophilia demonstrated FVIII hydrolysis rates (mean 219 +/- 94 mumol/min/mol) significantly greater than that of control IgG. Three of four patients followed over the course of the disease had rates of FVIII hydrolysis that co-evolved with inhibitory titers in plasma, suggesting that IgG-mediated FVIII hydrolysis participates, in part, in FVIII inactivation. The present work extends the scope of the diseases associated with FVIII proteolysis and points toward the importance of FVIII as a key target substrate for hydrolytic immunoglobulins. Our data suggest that elevated levels of FVIII-hydrolyzing IgG in acquired hemophilia result from the exacerbation of a physiological catalytic immune response.
Notes:
 
PMID 
S Lacroix-Desmazes, Y Repessé, S V Kaveri, S Dasgupta (2008)  The role of VWF in the immunogenicity of FVIII.   Thromb Res 122: Supp 2. S3-6  
Abstract: Up to 33% of patients with severe haemophilia A develop inhibitory antibodies to factor VIM (FVIII) that can significantly impair treatment with FVIII. The plasma protein von Willebrand factor (VWF) binds to FVIII and is known to be important for the functioning of FVIII. Accumulating data suggest that VWF may also be important for reducing the immunogenicity of therapeutically administered FVIII in patients with haemophilia A. Although contradictory results have been reported for studies in patients, studies in mice have shown that the levels of FVIII-binding antibodies induced following treatment with recombinant FVIII (rFVIII) are higher than those following treatment with plasma-derived FVIII preparations containing VWF, and that the addition of VWF to rFVIII reduces the levels of FVIII-binding antibodies induced. In in vitro studies, VWF has been shown to inhibit both the uptake of FVIII by immature dendritic cells and the activation of FVIII-specificT-cells in a dose-dependent manner. However, recombinant VWF (rVWF) lacking the FVIII-binding domain did not inhibit T-cell activation. These data suggest that VWF may reduce the immunogenicity of FVIII by inhibiting the uptake of FVIII by antigen presenting cells, the first step in the development of an immune response against a foreign antigen. Further studies are required to confirm the applicability of these results to patients with haemophilia. If confirmed, these data would encourage the use of VWF in conjunction with FVIII in the management of patients with haemophilia A.
Notes:
 
DOI   
PMID 
Suryasarathi Dasgupta, Ana Maria Navarrete, Sebastien André, Bharath Wootla, Sandrine Delignat, Yohann Repessé, Jagadeesh Bayry, Antonino Nicoletti, Evgueni L Saenko, d'Oiron Roseline, Marc Jacquemin, Jean-Marie Saint-Remy, Srini V Kaveri, Sebastien Lacroix-Desmazes (2008)  Factor VIII bypasses CD91/LRP for endocytosis by dendritic cells leading to T-cell activation.   Haematologica 93: 1. 83-89 Jan  
Abstract: BACKGROUND: The development of factor VIII (FVIII) inhibitors remains the major hurdle in the clinical management of patients with hemophilia A. FVIII uptake by professional antigen-presenting cells (APC) is the first step involved in initiation of immune responses to FVIII. Studies on FVIII catabolism have highlighted the role played by CD91/LRP as a potential target for increasing FVIII half-life in patients and prolonging treatment efficiency. We investigated the involvement of CD91 in FVIII endocytosis by human dendritic cells (DC), a model of professional APC. DESIGN AND METHODS: Immature DC were generated from circulating monocytes from healthy donors. Surface expression of CD91 was assessed by flow cytometry. Uptake of fluorescein isothiocyanate-conjugated ligands by immature DC was studied in the presence of various blocking agents. RESULTS: CD91 was expressed on approximately 20% of DC and mediated the internalization of its model ligand, alpha2-macroglobulin. DC internalized FVIII and activated a human FVIII-specific T-cell clone in a dose-dependent manner. FVIII uptake by DC and subsequent T-cell activation were not inhibited by receptor-associated protein. CONCLUSIONS: Our results indicate that CD91 and other members of the LDL receptor family are not strongly implicated in FVIII internalization by monocyte-derived DC, and suggest the involvement of alternative divalent ion-dependent endocytic receptors.
Notes:
2007
 
DOI   
PMID 
Suryasarathi Dasgupta, Ana-Maria Navarrete, Jagadeesh Bayry, Sandrine Delignat, Bharath Wootla, Sébastien André, Olivier Christophe, Michelina Nascimbeni, Marc Jacquemin, Luisa Martinez-Pomares, Teunis B H Geijtenbeek, Arnaud Moris, Jean-Marie Saint-Remy, Michel D Kazatchkine, Srinivas V Kaveri, Sébastien Lacroix-Desmazes (2007)  A role for exposed mannosylations in presentation of human therapeutic self-proteins to CD4+ T lymphocytes.   Proc Natl Acad Sci U S A 104: 21. 8965-8970 May  
Abstract: Several therapeutic self-proteins elicit immune responses when administered to patients. Such adverse immune responses reduce drug efficacy. To induce an immune response, a protein must interact with different immune cells, including antigen-presenting cells, T cells, and B cells. Each cell type recognizes distinct immunogenic patterns on antigens. Mannose-terminating glycans have been identified as pathogen-associated molecular patterns that are essential for internalization of microbes by antigen-presenting cells, leading to presentation. Here, we have investigated the importance of exposed mannosylation on an immunogenic therapeutic self-protein, procoagulant human factor VIII (FVIII). Administration of therapeutic FVIII to hemophilia A patients induces inhibitory anti-FVIII antibodies in up to 30% of the cases. We demonstrate that entry of FVIII into human dendritic cells (DC) leading to T cell activation, is mediated by mannose-terminating glycans on FVIII. Further, we identified macrophage mannose receptor (CD206) as a candidate endocytic receptor for FVIII on DC. Saturation of mannose receptors on DC with mannan, and enzymatic removal of mannosylated glycans from FVIII lead to reduced T cell activation. The interaction between FVIII and CD206 was blocked by VWF, suggesting that, under physiological conditions, the intrinsic mannose-dependent immunogenicity of FVIII is quenched by endogenous immunochaperones. These data provide a link between the mannosylation of therapeutic self-proteins and their iatrogenic immunogenicity. Such a link would be of special relevance in the context of replacement therapy where mechanisms of central and peripheral tolerance have not been established during ontogeny because of the absence of the antigen.
Notes:
 
PMID 
S Dasgupta, A M Navarrete, S Delignat, B Wootla, S Andre, V Nagaraja, S Lacroix-Desmazes, S V Kaveri (2007)  Immune response against therapeutic factor VIII in hemophilia A patients--a survey of probable risk factors.   Immunol Lett. 110: 1. 23-28 Apr  
Abstract: A number of diseases are treated by passive administration of human proteins. Human coagulation factor VIII (FVIII) is one such protein which is administered to hemophilia A patients in order to manage and treat hemorrhagic incidences. This mode of therapy suffers from the side effect of generating anti-FVIII antibodies (inhibitors) which neutralizes the function of the infused FVIII. At a time when efficient viral screening procedures are at place, development of inhibitors poses the greatest threat to such a therapy. Various predisposing factors, both patient and product-related, are responsible for the development of inhibitory antibodies. A proper understanding of these "risk-factors" would eventually help to better design therapeutic regimen to tackle hemophilia A.
Notes:
 
DOI   
PMID 
Suryasarathi Dasgupta, Yohann Repessé, Jagadeesh Bayry, Ana-Maria Navarrete, Bharath Wootla, Sandrine Delignat, Theano Irinopoulou, Caroline Kamaté, Jean-Marie Saint-Remy, Marc Jacquemin, Peter J Lenting, Annie Borel-Derlon, Srinivas V Kaveri, Sébastien Lacroix-Desmazes (2007)  VWF protects FVIII from endocytosis by dendritic cells and subsequent presentation to immune effectors.   Blood 109: 2. 610-612 Jan  
Abstract: Von Willebrand factor (VWF) is a chaperone molecule for procoagulant factor VIII (FVIII). Its role in the reduction of the immunogenicity of therapeutic FVIII in patients with hemophilia A has been evoked but lacks clear cellular and molecular rationale. Here, we demonstrate that VWF protects FVIII from being endocytosed by human dendritic cells (DCs) and subsequently presented to FVIII-specific T cells. The immunoprotective effect of VWF requires a physical interaction with FVIII because the endocytosis of FVIII was significantly restored on hindering the formation of the VWF-FVIII complex. Interestingly, VWF had no direct inhibitory effect either on the ability of DCs to present antigenic peptides or on the activation potency of CD4+ T cells. We thus propose that VWF may reduce the immunogenicity of FVIII by preventing, upstream from the activation of immune effectors, the entry of FVIII in professional antigen-presenting cells.
Notes:
Suryasarathi Dasgupta, Jagadeesh Bayry, Sebastien Lacroix-Desmazes, Srinivas V Kaveri (2007)  Human Mannose Receptor (CD206) in Immune Response: Novel Insights into Vaccination Strategies Using a Humanized Mouse Model   Expert Rev Clin Immunol. 3: 05. 677-681  
Abstract: Induction of adaptive immune response requires, in most cases, internalization of the antigen by professional antigen-presenting cells. Various endocytic receptors may mediate such interaction. However, little is known regarding the exact participation of such receptors in the context of immune response in vivo. This paper evaluates a recent work that utilizes one such molecule, the human CD206, transgenically incorporated into mouse, in order to establish its role in immune response. Such a study benefits from the fact that the function of the human form of the receptor is elaborated and thus would help to define new targets for future vaccine strategies.
Notes:
 
DOI   
PMID 
Sandrine Delignat, Suryasarathi Dasgupta, Sébastien André, Ana-Maria Navarrete, Srinivas V Kaveri, Jagadeesh Bayry, Marie-Hélène André, Sami Chtourou, Zéra Tellier, Sébastien Lacroix-Desmazes (2007)  Comparison of the immunogenicity of different therapeutic preparations of human factor VIII in the murine model of hemophilia A.   Haematologica 92: 10. 1423-1426 Oct  
Abstract: Von Willebrand factor (VWF) has been proposed to reduce the immunogenicity of therapeutic factor VIII (FVIII) in patients with hemophilia A. Using FVIII-deficient mice, we compared the immunogenicity of different preparations of plasma-derived (pd) and recombinant (r) FVIII. Treatment of mice with pdFVIII induced significantly lower titers of FVIII inhibitors, as measured by ELISA and in vitro coagulation assays, compared with rFVIII. Furthermore, pre-incubation of rFVIII with excess VWF significantly reduced rFVIII immunogenicity. Our data confirm that pdFVIII induces lower levels of inhibitors than rFVIII, and that VWF is an immuno-chaperone molecule for FVIII.
Notes:
 
PMID 
S V Kaveri, S Dasgupta, S Andre, A M Navarrete, Y Repessé, B Wootla, S Lacroix-Desmazes (2007)  Factor VIII inhibitors: role of von Willebrand factor on the uptake of factor VIII by dendritic cells.   Haemophilia 13: Supp 5. 61-64 Dec  
Abstract: In patients with haemophilia A, factor VIII (FVIII) therapy leads to the development of anti-FVIII alloantibodies that inhibit FVIII pro-coagulant activity, in up to 25% of the cases. At a time when efficient viral screening procedures are at place, development of inhibitors poses the greatest threat to haemophilia A patients. Various risk factors, both patient and product-related, are responsible for the development of inhibitory antibodies. The role of FVIII-specific CD4+ T lymphocytes in the initiation of the humoral immune response to exogenous FVIII has been well. In view of their capacity to stimulate naïve T cells, dendritic cells (DCs) play a central role in the initiation of the primary immune response. Thus, in the context of a primary alloimmunization against FVIII, i.e. when FVIII-specific B lymphocytes are not there to take up FVIII from the circulation and to serve as antigen presenting cells (APCs), DCs are the only cell type that internalize FVIII, leading to activation of FVIII-specific CD4+ T lymphocytes. von Willebrand factor (VWF) present in plasma-derived FVIII therapeutic concentrates, is known to act as a chaperone molecule for procoagulant FVIII. In addition to its role in reducing the 'antigenicity' of FVIII, the role of VWF in the reduction of the 'immunogenicity' of therapeutic FVIII in patients with haemophilia A has also been suggested. We have recently demonstrated that VWF protects FVIII from being endocytosed by human DCs and subsequently being presented to FVIII-specific T cells. We propose that VWF may reduce the immunogenicity of FVIII by preventing, upstream from the activation of immune effectors, the entry of FVIII in professional antigen presenting cells.
Notes:
2006
 
PMID 
S Lacroix-Desmazes, B Wootla, S Delignat, S Dasgupta, V Nagaraja, M D Kazatchkine, S V Kaveri (2006)  Pathophysiology of catalytic antibodies.   Immunol Lett. 103: 1. 3-7 Feb  
Abstract: Immunoglobulins have initially been illustrated as proteins produced by the immune system for binding and neutralizing foreign molecules potentially harmful to the organism. The number of V(H), D(H), J(H), V(L) and J(L) genes that encode the variable regions of immunoglobulins and the junctional diversity that occurs at the time of somatic rearrangement determine the extent of the repertoire of antibodies that may be potentially produced by an organism. This potential repertoire includes antibodies the antigen binding site of which may recognize external as well as autologous antigens, or may structurally resemble the active site of enzymes and be endowed with enzymatic activity. Under physiological conditions, B cell clones that produce antibodies naturally endowed with catalytic activity are negatively regulated and subjected to apoptosis. Catalytic antibodies are expressed only following active immunization, or if the physiological regulatory mechanisms that control the expression of catalytic antibody-producing B cell clones are perturbed, e.g. in the context of pregnancy or in the course of autoimmune diseases.
Notes:
 
PMID 
Sébastien Lacroix-Desmazes, Bharath Wootla, Suryasarathi Dasgupta, Sandrine Delignat, Jagadeesh Bayry, Joseph Reinbolt, Johan Hoebeke, Evgueni Saenko, Michel D Kazatchkine, Alain Friboulet, Olivier Christophe, Valakunja Nagaraja, Srini V Kaveri (2006)  Catalytic IgG from patients with hemophilia A inactivate therapeutic factor VIII.   J Immunol 177: 2. 1355-1363 Jul  
Abstract: Factor VIII (FVIII) inhibitors are anti-FVIII IgG that arise in up to 50% of the patients with hemophilia A, upon therapeutic administration of exogenous FVIII. Factor VIII inhibitors neutralize the activity of the administered FVIII by sterically hindering its interaction with molecules of the coagulation cascade, or by forming immune complexes with FVIII and accelerating its clearance from the circulation. We have shown previously that a subset of anti-factor VIII IgG hydrolyzes FVIII. FVIII-hydrolyzing IgG are detected in over 50% of inhibitor-positive patients with severe hemophilia A, and are not found in inhibitor-negative patients. Although human proficient catalytic Abs have been described in a number of inflammatory and autoimmune disorders, their pathological relevance remains elusive. We demonstrate here that the kinetics of FVIII degradation by FVIII-hydrolyzing IgG are compatible with a pathogenic role for IgG catalysts. We also report that FVIII-hydrolyzing IgG from each patient exhibit multiple cleavage sites on FVIII and that, while the specificity of cleavage varies from one patient to another, catalytic IgG preferentially hydrolyze peptide bonds containing basic amino acids.
Notes:
 
PMID 
J Bayary, S Dasgupta, N Misra, A Ephrem, JP Van Huyen, S Delignat, G Hassan, G Caligiuri, A Nicoletti, S Lacroix-Desmazes, M D Kazatchkine, S Kaveri (2006)  Intravenous immunoglobulin in autoimmune disorders: an insight into the immunoregulatory mechanisms.   Int Immunopharmacol. 6: 4. 528-534 Dec  
Abstract: Intravenous immunoglobulin (IGIV) has increasingly been used for the treatment of autoimmune and systemic inflammatory diseases in addition to supportive therapy of immunodeficient patients. IGIV is beneficial in several diseases, including acute and chronic/relapsing diseases, autoimmune diseases and inflammatory disorders. Therapeutic efficacy of IGIV has also been established in a number of dermatologic diseases. Although a considerable progress has been made in understanding the mechanisms by which IGIV exerts immunomodulatory functions in autoimmune diseases, they remain not fully elucidated. The mode of action of IGIV is complex, involving modulation of expression and function of Fc receptors, interference with activation of complement and the cytokine network, modulation of idiotype network, regulation of cell growth, alteration of cellular adhesion process, and effects on the activation differentiation and effector functions of T and B cells and of antigen-presenting cells. The therapeutic effects of IGIV most likely reflect the functions of natural antibodies in maintaining immune homeostasis in healthy people. The ability of IGIV to interact through V regions with complementary V regions of antibodies and antigen receptors as well as with relevant soluble and surface molecules provides the basis for inducing the selection of immune repertoires. Since IGIV is frequently used to treat autoimmune and inflammatory diseases for which evidence of its efficacy is insufficiently documented, controlled trials, particularly of some neurologic and dermatologic diseases, are imperative.
Notes:
 
PMID 
B Wootla, S Dasgupta, V Mallet, M D Kazatchkine, V Nagaraja, A Friboulet, S V Kaveri, S Lacroix-Desmazes (2006)  Physiopathology of catalytic antibodies: the case for factor VIII-hydrolyzing immunoglobulin G.   Blood Coagul Fibrinolysis. 17: 4. 229-234  
Abstract: Antibodies that are able to catalyze the antigen for which they are specific are produced spontaneously by the immune system. Catalytic immunoglobulins (Igs) both of the IgM and IgG isotypes have been detected in the serum of healthy donors, where they have been proposed to participate in the removal of metabolic waste and in the defense of the organism against invading pathogens. Conversely, antigen-specific hydrolytic IgG have been reported in a number of inflammatory, autoimmune and neoplastic disorders: their pathogenic effects have been demonstrated occasionally. The pathophysiological relevance of catalytic antibodies thus remains an elusive issue. Through the description of the pro-coagulation factor VIII as a model target antigen for catalytic antibodies, we propose that catalytic antibodies have either a beneficial or a deleterious role depending on the physiopathological context. Physiology thus relies on a delicate equilibrium between the levels of soluble target antigen and that of antigen-specific hydrolyzing immunoglobulins. Indeed, in patients with hemophilia A, in whom endogenous factor VIII is deficient or missing and exogenous factor VIII needs to be administered to treat hemorrhagic events, the development of factor VIII-hydrolyzing IgG that inactivate the therapeutically administered factor VIII, may reveal deleterious. In contrast, in a situation in which excess factor VIII may be detrimental and lead to excessive coagulation, disseminated thrombosis and organ ischemia, as seen in severe sepsis, our recent data suggest that the presence of factor VIII-hydrolyzing IgG may be beneficial to the patient.
Notes:
2005
 
DOI   
PMID 
Namita Misra, Jagadeesh Bayry, Amal Ephrem, Suryasarathi Dasgupta, Sandrine Delignat, Jean-Paul Duong Van Huyen, Fabienne Prost, Sebastien Lacroix-Desmazes, Antonino Nicoletti, Michel D Kazatchkine, Srini V Kaveri (2005)  Intravenous immunoglobulin in neurological disorders: a mechanistic perspective.   J Neurol 252 Suppl 1: I1-I6 May  
Abstract: Intravenous immunoglobulin (IVIg) has been used in the treatment of primary and secondary antibody deficiencies for over 25 years. It is a safe preparation with no long-term side effects. IVIg was first demonstrated to be effective in autoimmune disorders, two decades ago, in the treatment of acute immune thrombocytopenia. Since then, the therapeutic efficacy of IVIg has been established in Guillain Barré syndrome (GBS), chronic inflammatory demyelinating polyneuropathy (CIDP), myasthenia gravis (MG), dermatomyositis (DM), Kawasaki syndrome and the prevention of graft-versus-host disease in recipients of allogeneic bone marrow transplants and reported in a large number of other autoimmune and systemic inflammatory conditions.
Notes:
 
PMID 
N Misra, J Bayary, S Dasgupta, A Ephrem, J P Huyen, S Delignat, G Hassan, G Caligiuri, A Nicoletti, S Lacroix-Desmazes, M D Kazatchkine, S V Kaveri (2005)  Intravenous immunoglobulin and dendritic cells.   Clin Rev Allergy Immunol. 29: 3. 201-205 Dec  
Abstract: Intravenous immunoglobulin (IVIg) has increasingly been used for the treatment of autoimmune and systemic inflammatory diseases, and in supportive therapy of immunodeficient patients. Available clinical and experimental evidence suggests, however, that a wide spectrum of immune-mediated conditions could benefit from IVIg, including acute and chronic/relapsing diseases and autoimmune diseases mediated by pathogenic autoantibodies or by autoaggressive T-cells. Dendritic cells (DCs) are professional antigen-presenting cells and because of their capacity to stimulate naïve T-cells, they play a central role in the initiation of primary immune responses. Several immunomodulatory agents have been shown to inhibit DC activation. Recently, we examined the effects of IVIg on differentiation, maturation, and functions of DCs. We demonstrate that DCs are one of the targets for the immunomodulatory effects of IVIg.
Notes:
2004
 
PMID 
S Dasgupta, V Chandran, A Bhinge, S Sewlikar, A Nimbkar, D Datta (2004)  Role of L-lysine HCl in adoptive immune therapy towards development of suitable tuberculosis vaccination.   Indian J Exp Biol. 42: 8. 758-765 Aug  
Abstract: L-Lysine HCI is being proposed to be a possible biocompatible adjuvant to enhance immune response by virtue of its probable non-specific bridging action and cellular proliferation properties. This proposal has been tried to be substantiated by designing an in vitro culture protocol, varying the concentration of L-lysine HCI and its further in vivo application. Splenic lymphocyte population has been extracted from mice and co-cultured with extracted mice macrophage population in presence of either Bacille Calmette Guerrin (BCG) or Hepatitis B surface antigen (HbsAg) and added L-lysine hydrochloride in culture media. Post incubation of these cultures, "taught" cell population has been adoptively transferred in naïve mice. These mice were then challenged by respective antigen dose, Change in Immune response with this challenge was noted. Antibody titre was followed in all the experiments as a measure of immune response. In adoptive immune transfer experiment of with HbsAg (AIT-HbsAg), similar to that with BCG (AIT-BCG), after the incubation period, antibody titre was higher in added lysine containing cultures in comparison with the control ones. Post transfer followed by antigen challenge, in AIT-BCG the expected augmentation in immune response was hardly visible. But in AIT-HbsAg, with the help of lysine booster, the animals responded better as far as the antibody titre is concerned.
Notes:
2003
 
PMID 
S Dasgupta, A Bhinge, V Chandran, S Sewlikar, A Nimbkar, D Datta (2003)  Role of L-lysine HCl in immunopotentiation towards development of suitable tuberculosis vaccination   Vaccine 21: 32. 4722-4727 Dec  
Abstract: L-Lysine HCl is being proposed to be a possible biocompatible adjuvant to enhance immune response by virtue of its probable non-specific bridging action and cellular proliferation properties. This proposal has been tried to be substantiated by carrying out experimentation where L-lysine HCl has been used as an adjuvant (various groups based on mode of application and frequency of booster dose were designed) in tuberculosis vaccination experiments with heat killed Mycobacterium tuberculosis (MTB) and Bacille Calmette Guerin (BCG). Antibody titre has been followed in all the experiments as a measure of immune response. Amongst the various groups designed, group 1A (L-lysine HCl was given at a separate site as that of the antigen; lysine booster was given to this group intermittently, i.e. lysine given on 0th, 7th, 14th, 21st days of immunization) came out as the stand-alone leader. This mode and frequency of application was then compared with a group which received a standard adjuvant, viz. alhydrogel. Results were obtained which showed the following order in terms of decreasing antibody titre: alhydrogel group > lysine group > control group. Considering the biocompatible nature of lysine in comparison with the reportedly hazardous nature of alum adjuvants, we propose L-lysine HCl as a probable adjuvant in vaccination.
Notes:
 
PMID 
J Giri, A Ray, S Dasgupta, D Datta, D Bahadur (2003)  Investigation on Tc tuned nano particles of magnetic oxides for hyperthermia applications.   Biomed Mater Eng. 13: 4. 387-399  
Abstract: Superparamagnetic as well as fine ferrimagnetic particles such as Fe3O4, have been extensively used in magnetic field induced localized hyperthermia for the treatment of cancer. The magnetic materials with Curie temperature (Tc) between 42 and 50 degrees C, with sufficient biocompatibility are the best candidates for effective treatment such that during therapy it acts as in vivo temperature control switch and thus over heating could be avoided. Ultrafine particles of substituted ferrite Co(1-a)Zn(a)Fe2O4 and substituted yttrium-iron garnet Y3Fe(5-x)Al(x)O12 have been prepared through microwave refluxing and citrate-gel route respectively. Single-phase compounds were obtained with particle size below 100 nm. In order to make these magnetic nano particles biocompatible, we have attempted to coat these above said composition by alumina. The coating of alumina was done by hydrolysis method. The coating of hydrous aluminium oxide has been done over the magnetic particles by aging the preformed solid particles in the solution of aluminium sulfate and formamide at elevated temperatures. In vitro study is carried out to verify the innocuousness of coated materials towards cells. In vitro biocompatibility study has been carried out by cell culture method for a period of three days using human WBC cell lines. Study of cell counts and SEM images indicates the cells viability/growth. The in vitro experiments show that the coated materials are biocompatible.
Notes:
2001
 
PMID 
D Datta, P K Kundu, S Dasgupta, K Fulzele, S Sewlikar (2001)  Effect of addition of proton carriers in culture medium on growth and secretion of hybridoma cell line OKT3.   Indian J Physiol Pharmacol. 45: 3. 367-372 Jul  
Abstract: Monoclonal antibodies (MAb) constitute the centre of all in-vitro diagnostic measures and almost all in-vivo therapeutic manoeuvres now. Production emphasis for these antibodies is having a current shift from animal-based large-scale culture to in-vitro bioreactor-based high-density culture. One of the major difficulties in high-density culture is end-metabolite accumulation in batch and fed-batch cultures in the forms of H+, NH4+ etc.. thereby reducing cellular growth and secretions. In the present study, effects of added proton carries--NAD and NADP--over and above the metabolic pools of the molecules, were examined on the cellular growth and secretion kinetics. Although NADP fortification showed a remarkable improvement in cellular growth (time dependent 200-300% improvements compared to controls) and size, cumulative MAb titre was better with NAD fortification. Combined additional loads of the proton carriers would be interesting to study in high density culture conditions.
Notes:
2000
 
PMID 
D Datta, P K Kundu, S Biswas, S Dasgupta, A Bhinge, V Chandran (2000)  Effect of cationic amino acid, L-lysine and its polymers on the growth and secretion of hybridoma cell line OKT-3.   Hybridoma 19: 4. 339-346 Aug  
Abstract: Apart from their pivotal roles in anabolic protein synthesis, cationic amino acids, particularly, L-lysine HCl and its oligomers, up to molecular weight 1000, showed a remarkable property of cellular growth stimulation both in vitro and in vivo. L- and D-Lysine HCl, at a maximal stimulatory concentration of 7 microg/mL of added load of the amino acid, supported a characteristic time-scaled cellular expansion in vitro, and L-lysine-mediated cell expansion in batch cultures always showed a stimulation index (S.I.) ranging up to approximately 35, compared with the matched control populations. Variable S.I. was possibly due to factors such as seeding density, type of media additives, number of passages the cells have undergone before being stimulated, etc. Beyond and before maximal stimulatory concentration of the amino acid, there is a sharp decline in the cellular growth-promoting activity of monomeric L-lysine HCl in vitro, thereby showing a clear concentration window for maximum cellular growth promotion. While the essential amino acid does not have any dedicated cell surface receptor, the monomeric and oligomeric amino acid molecule(s) possibly mediates the serum-derived growth factor-receptor binding on the cell membrane by having two cationic charge centres at two ends of the molecule. Beyond a cutoff molecular weight of 1000, oligomeric lysines did not show any positive effects on either cell division and secretion.
Notes:
1999
Powered by publicationslist.org.