Network


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

Hotspot


Dive into the research topics where Smriti Kundu-Raychaudhuri is active.

Publication


Featured researches published by Smriti Kundu-Raychaudhuri.


Journal of Investigative Dermatology | 2008

FR255734, a Humanized, Fc-Silent, Anti-CD28 Antibody, Improves Psoriasis in the SCID Mouse-Psoriasis Xenograft Model

Siba P. Raychaudhuri; Smriti Kundu-Raychaudhuri; Kouichi Tamura; Taro Masunaga; Kaori Kubo; Kaori Hanaoka; Wen Yue Jiang; Leonore A. Herzenberg; Leonard A. Herzenberg

In psoriasis, CD28/B7 costimulatory molecules are well characterized. Here, using the severe combined immunodeficient (SCID) mouse-psoriasis xenograft model, we report therapeutic efficacy of a humanized anti-CD28 monoclonal antibody (FR255734; Astellas Pharmaceuticals Inc., Tokyo, Japan). Transplanted psoriasis plaques on the SCID mouse were treated weekly for 4 weeks with intraperitoneal injections of FR255734 at 10, 3, and 1-mg kg(-1) doses. Groups treated with doses of 10 and 3 mg kg(-1) had significant thinning of the epidermis and reduced HLA-DR-positive lymphocytic infiltrates. The length of the rete pegs changed from 415.2+/-59.6 to 231.4+/-40.4 microm (P<0.005) in the 10-mg kg(-1) group, and from 323.4+/-69.6 to 237.5+/-73.6 microm in the 3-mg kg(-1) group (P=0.002). Positive controls treated with CTLA4-Ig and cyclosporine had significant histological improvement, whereas plaques treated with saline and isotype controls (human and mouse IgG2) remained unchanged. In vitro studies have shown that FR255734 effectively blocked T-cell proliferation and proinflammatory cytokine production. These observations warrant studies to evaluate the efficacy of FR255734 in human autoimmune diseases.


Journal of Autoimmunity | 2014

Kv1.3 in psoriatic disease: PAP-1, a small molecule inhibitor of Kv1.3 is effective in the SCID mouse psoriasis – Xenograft model

Smriti Kundu-Raychaudhuri; Yi Je Chen; Heike Wulff; Siba P. Raychaudhuri

Kv1.3 channels regulate the activation/proliferation of effector memory T cells and thus play a critical role in the pathogenesis of autoimmune diseases. Using a combination of immunohistochemistry, confocal microscopy, flow cytometry and electrophysiology methods we observed a significant enrichment of activated Kv1.3(+) memory T cells in psoriasis plaques and synovial fluid from patients with psoriasis/psoriatic arthritis (PsA) compared to non-lesional psoriatic skin, normal skin or peripheral blood lympho-mononuclear cells. In in vitro studies performed with lesional mononuclear cells or T cells derived from skin and joints of psoriatic disease, the small molecule Kv1.3 blocker PAP-1 dose-dependently inhibited proliferation and suppressed IL-2 and IFN-γ production. To further substantiate the pathologic role of Kv1.3 high TEM cells in psoriatic disease we tested whether PAP-1 is able to improve psoriatic disease pathology in the SCID mouse-psoriasis skin xenograft model. Following four weeks of daily treatment with 2% PAP-1 ointment we noticed about 50% reduction in the epidermal thickness (rete peg length) and the number of CD3(+) lymphocytes/mm(2) of dermis decreased by 85%. Vehicle treated and untreated plaques in contrast remained unchanged and showed no reduction in epidermis thickness and infiltrating CD3(+) T cells and HLA-DR(+) T cells. Based on these results we propose the development of Kv1.3 targeted topical immunotherapy for psoriasis and possibly for other inflammatory skin conditions, where effector memory T cells are involved in the pathogenesis.


Journal of Investigative Dermatology | 2013

1α,25-Dihydroxyvitamin-D3-3-bromoacetate regulates AKT/mTOR signaling cascades: a therapeutic agent for psoriasis.

Ananya Datta Mitra; Siba P. Raychaudhuri; Christine Abria; Anupam Mitra; Rebecca Wright; Rahul Ray; Smriti Kundu-Raychaudhuri

The efficacy of 1α,25-dihydroxyvitamin D3 (Vit-D) limits its topical use despite its profound effects on cellular differentiation, proliferation, and immunomodulation. Therefore, in search for a more effective analog of Vit-D, in this study we have evaluated the antiproliferative and proapoptotic effects of 1α,25-dihydroxyvitamin D3-3-bromoacetate (BE). Proliferation and apoptosis studies in normal human epidermal keratinocytes (NHEKs) were conducted by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide), CFSE (carboxy fluorescein succinimidyl ester) dilution, and Annexin V assays. Western blot analysis and real-time PCR were performed to determine its effect on signal transduction. A reconstructed human epidermis (RHE) model was used to further validate the therapeutic role of BE in psoriasis. BE was significantly more potent than an equivalent concentration of Vit-D in inhibiting growth and survival of human keratinocytes. The antimitotic effect was found to be due to the inhibition of phosphorylation of serine/threonine protein kinase (AKT) and its downstream target, mammalian target of rapamycin (mTOR). In the RHE model, BE reversed IL-22-induced psoriasiform changes more effectively than Vit-D. Interestingly, BE inhibited the IL-22-induced gene expression of AKT1, MTOR, chemokines [IL-8 and RANTES (regulated upon activation, normal T-cell expressed and secreted)], and psoriasin (S100A7) more significantly than Vit-D. These results suggest the potential of BE as a prospective therapeutic agent for psoriasis.


Cytokine | 2016

IL-9, a local growth factor for synovial T cells in inflammatory arthritis

Smriti Kundu-Raychaudhuri; Christine Abria; Siba P. Raychaudhuri

OBJECTIVEnThe regulatory role of the Th9 cells along with its signature cytokine IL-9 in human immune system and its aberrant activation in autoimmune diseases is currently under investigation. We are reporting the functional significance of IL-9 in the pathogenesis of autoimmune inflammatory arthritis.nnnMETHODSnCD3(+) T cells were obtained from peripheral blood (PB) and synovial fluid (SF) of psoriatic arthritis (PsA), rheumatoid arthritis (RA), and osteoarthritis (OA) patients. MTT, FACS based CFSE dilution assay and apoptosis assay (Annexin-V) were performed to determine the pro-growth/survival effect of human recombinant IL-9 on activated CD3(+) T cells. Immunoblots were performed to determine the signaling proteins responsible for the progrowth/survival effect of IL-9.nnnRESULTSnSF of PsA and RA was enriched with IL-9 producing CD3(+) T cells compared to the SF in OA. IL-9 level measured by ELISA was significantly elevated in PsA and RA patients compared to SF in OA (<.001). Activated T cells of PsA and RA had higher levels of IL-9 receptors. IL-9 promoted proliferation and survival of the CD3(+) T cells of PB and SF of PsA and RA and compared to untreated (media) controls (p<.005, t-test). IL-9 induced proliferation of T cells was dependent on PI3K/Akt/mTOR signaling pathway.nnnCONCLUSIONnIL-9 is functionally active, and is a pro-growth/survival factor for the localized pathologic T cells in the synovium of inflammatory arthritis. The pro-growth/survival effect is mediated by the activation of mTOR kinase cascade. To our knowledge, this is the first report of a functional role of IL-9 in human autoimmune arthritis.


Journal of Investigative Dermatology | 2015

Dual mTOR Inhibition Is Required to Prevent TGF-β-Mediated Fibrosis: Implications for Scleroderma

Anupam Mitra; Jesus I. Luna; Alina I. Marusina; Alexander A. Merleev; Smriti Kundu-Raychaudhuri; David Fiorentino; Siba P. Raychaudhuri; Emanual Maverakis

TO THE EDITOR n nTransforming growth factor-β (TGF-β) and platelet-derived growth factor (PDGF) are central mediators of fibrosis, and their overexpression contributes to the pathophysiology of scleroderma, chiefly by inducing the overproduction of extracellular matrix proteins (ECM) by dermal fibroblasts (Gay et al., 1989; Sargent et al., 2010; Bhattacharyya et al., 2012). TGF-β also promotes the differentiation of dermal fibroblasts into myofibroblasts, which are key mediators of scleroderma (Abraham et al., 2007). Thus, targeting this pathway is a reasonable strategy to treat a variety of fibrotic diseases including scleroderma, for which current treatment options are limited. Herein we explore the potential of novel mTOR inhibition as a means to block the pro-fibrotic effects of TGF-β. Recent studies have suggested a functional role of mTOR in fibrotic diseases and autoimmunity (Ong et al., 2007; Fried et al., 2008; Su et al., 2009; Raychaudhuri and Raychaudhuri, 2014). This pathway is initiated by cytokines and growth factors that induce phosphorylation of Akt(Thr308) and its downstream mediators mTORC1, p70S6K1, and 4E-BP1. In addition to mTORC1, mTOR is now known to comprise a second multi-protein complex, mTORC2, which positively regulates the activity of mTOR through phosphorylation of AktSer473 (Bhagwat et al., 2011). Blockade of mTORC1 has already been attempted as a treatment for scleroderma without much understanding about its molecular mechanism (Fried et al., 2008; Su et al., 2009; Yoshizaki et al., 2010). Such strategies focused only on mTORC1 and did not account for the contributions of mTORC2 (Bhagwat et al., 2011). To increase the effectiveness of mTOR blockade, dual inhibitors targeting both mTORC1 and mTORC2 have now been developed (Bhagwat et al., 2011). Herein, we demonstrate that dual mTOR blockade can more effectively inhibit the pro-fibrotic effects of TGF-β and PDGF. These have therapeutic implications for scleroderma and other TGF-β dominant fibrotic diseases.


PLOS ONE | 2015

Cross Talk between Neuroregulatory Molecule and Monocyte: Nerve Growth Factor Activates the Inflammasome

Ananya Datta-Mitra; Smriti Kundu-Raychaudhuri; Anupam Mitra; Siba P. Raychaudhuri

Background Increasing evidence points to a role for the extra-neuronal nerve growth factor (NGF) in acquired immune responses. However, very little information is available about its role and underlying mechanism in innate immunity. The role of innate immunity in autoimmune diseases is becoming increasingly important. In this study, we explored the contribution of pleiotropic NGF in the innate immune response along with its underlying molecular mechanism with respect to IL-1β secretion. Methods Human monocytes, null and NLRP3 deficient THP-1 cell lines were used for this purpose. We determined the effect of NGF on secretion of IL-1β at the protein and mRNA levels. To determine the underlying molecular mechanism, the effect of NGF on NLRP1/NLRP3 inflammasomes and its downstream key protein, activated caspase-1, were evaluated by ELISA, immunoflorescence, flow cytometry, and real-time PCR. Results In human monocytes and null THP-1 cell line, NGF significantly upregulates IL-1β at protein and mRNA levels in a caspase-1 dependent manner through its receptor, TrkA. Furthermore, we observed that NGF induces caspase-1 activation through NLRP1/NLRP3 inflammasomes, and it is dependent on the master transcription factor, NF-κB. Conclusions To best of our knowledge, this is the first report shedding light on the mechanistic aspect of a neuroregulatory molecule, NGF, in innate immune response, and thus enriches our understanding regarding its pathogenic role in inflammation. These observations add further evidence in favor of anti-NGF therapy in autoimmune diseases and also unlock a new area of research about the role of NGF in IL-1β mediated diseases.


International Immunopharmacology | 2013

1,25-Dihydroxyvitamin D3-3-bromoacetate, a novel vitamin D analog induces immunosuppression through PI3K/Akt/mTOR signaling cascade.

Ananya Datta-Mitra; Anupam Mitra; Rahul Ray; Siba P. Raychaudhuri; Smriti Kundu-Raychaudhuri

PURPOSEnThe molecular mechanism responsible for the immunomodulatory effect of 1,25-dihydroxyvitamin D3 (Vit-D) is still not well elucidated. Unavoidable systemic toxicity of Vit-D has encouraged to develop more potent and less toxic Vit-D analogs, such as 1,25-dihydroxyvitamin D3-3-bromoacetate (BE). Our aim was to explore the immunosuppressive effect of BE and its molecular mechanism in autoimmune diseases.nnnMETHODnMagnetically sorted CD3(+) T cells (T cells) from PBMCs of psoriasis and autoimmune arthritis patients were cultured with/without BE and Vit-D followed by proliferation (MTT, CFSE dilution assays) and apoptosis assays (annexin V). Immunoblot was performed to determine the signaling cascade responsible for the antiproliferative effect.nnnRESULTSnIn MTT assay, BE (OD: 0.64±0.08) markedly inhibited the anti-CD3/CD28 stimulated proliferation of T cells (OD: 1.8±0.30, p<0.001) and at equivalent doses, the inhibitory effect was more than that of Vit-D (OD: 0.91±0.11, p<0.05). The antiproliferative effect of BE was extended to activated CD4(+) and CD8(+) memory T cells (CD45RA(-)CD11a(+)) without much effect on the naïve T cells. BE induced more apoptosis of T cells (45.01±4.27%, p<0.01) compared to untreated cells (3.45±1.8%), and the proapoptotic effect was markedly more than that of Vit-D (26.1±2.05%, p<0.05). BE effectively inhibited the anti-CD3/CD28-induced phosphorylation of Akt and mTOR and in both, BE showed more potency than Vit-D (p<0.05).nnnCONCLUSIONnTopical Vit-D is being used successfully in psoriasis for years. However, its potency is less compared to topical corticosteroids. The de novo BE showed significantly more immunosuppression than conventional Vit-D and the immunosuppressive effect is PI3K/Akt/mTOR dependent. Our results indicate that BE could be an effective therapeutic agent for psoriasis and other T-cell-mediated autoimmune diseases.


International Journal of Rheumatic Diseases | 2016

In vivo quantification of mouse autoimmune arthritis by PET/CT.

Smriti Kundu-Raychaudhuri; Anupam Mitra; Ananya Datta-Mitra; Abhijit J. Chaudhari; Siba P. Raychaudhuri

To quantify the progression and severity of mouse collagen‐induced arthritis (CIA) using an in vivo imaging tool, 18F‐fluorodeoxyglucose (18F‐FDG) PET/CT and validate it against gold standard ‘histopathological’ evaluation.


AIDS Research and Human Retroviruses | 2001

Effect of Therapeutic Immunization with HIV Type 1 Recombinant Glycoprotein 160 ImmunoAG Vaccine in HIV-Infected Individuals with CD4+ T Cell Counts of ≥500 and 200-400/mm3 (AIDS Clinical Trials Group Study 246/946)

Smriti Kundu-Raychaudhuri; Anne Sevin; Patrick Kilgo; Mostafa Nokta; Richard B. Pollard; Thomas C. Merigan

AIDS Clinical Trials Group (ACTG) 246/946 was a double-blinded, randomized, controlled trial of HIV-1 MN rgp160 ImmunoAG vaccine in HIV-infected patients with CD4(+) T cell counts >or=500 and 200-400/mm(3). The main objectives were to study the safety and immunogenicity of this vaccine and to study the persistence of the immune responses after vaccination over a longer period of time. Fifteen patients with CD4(+) T cell counts of >or=500/mm(3) were enrolled in the ACTG 246 study. ACTG 246 patients received a monthly injection of vaccine or control for 6 months and then injections every 2 months. After completion of this study, seven new patients with CD4(+) T cell counts of 200-400/mm(3) entered into the ACTG 946 study. These study patients received highly active antiretroviral therapy (HAART) (ritonavir, didanosine, and stavudine) for 9 weeks to stabilize their viral load and then each patient received a monthly injection of vaccine or control substance for 6 months with HAART. The study of these two relatively small populations showed that the vaccine was safe without any adverse effect both in the patients with CD4(+) T cell counts of >or=500 and 200-400/mm(3). The vaccine was also immunogenic in patients with CD4(+) T cell counts of >or=500/mm(3) as measured by gp160-specific lymphocyte proliferative responses, and it persisted after they had received more than six vaccine injections, for a longer period of time.


Indian Journal of Dermatology, Venereology and Leprology | 2014

Severe combined immunodeficiency mouse-psoriatic human skin xenograft model: A modern tool connecting bench to bedside

Smriti Kundu-Raychaudhuri; Ananya Datta-Mitra; Christine Abria; John H. Peters; Siba P. Raychaudhuri

Psoriasis is a multifactorial chronic inflammatory disease. Research into the pathogenesis of this disease is hindered by the lack of a proper animal model. Over the past two decades, many scientists were involved in the development of animal models that nearly mirror the immunopathogenesis of psoriasis. One such model, which has opened doors to the study of molecular complexities of psoriasis as well as its treatment, is the severe combined immunodeficiency (SCID) mouse-human skin chimera model. This model not only mirrors the clinical and histopathological features of psoriasis but also help in the study of cell proliferation, angiogenesis, function of T cells, neurogenic inflammation and cytokines involved in inflammatory reactions. In this article, we have reviewed the prospects and the limitations of the SCID mouse model of psoriasis.

Collaboration


Dive into the Smriti Kundu-Raychaudhuri's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anupam Mitra

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rebecca Wright

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge