Network


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

Hotspot


Dive into the research topics where Supurna Ghosh is active.

Publication


Featured researches published by Supurna Ghosh.


Cancer Research | 2007

Engagement of Collagen-Binding Integrins Promotes Matrix Metalloproteinase-9–Dependent E-Cadherin Ectodomain Shedding in Ovarian Carcinoma Cells

Jaime Symowicz; Brian P. Adley; Kara J. Gleason; Jeffrey J. Johnson; Supurna Ghosh; David A. Fishman; Laurie G. Hudson; M. Sharon Stack

Reversible modulation of cell-cell adhesion, cell-matrix adhesion, and proteolytic activity plays a critical role in remodeling of the neoplastic ovarian epithelium during metastasis, implicating cadherins, integrins, and proteinases in i.p. metastatic dissemination of epithelial ovarian carcinoma (EOC). Aberrant epithelial differentiation is an early event in ovarian carcinogenesis; thus, in contrast to most carcinomas that lose E-cadherin expression with progression, E-cadherin is abundant in primary EOC. Metastasizing EOCs engage in integrin-mediated adhesion to submesothelial interstitial collagens and express matrix metalloproteinases (MMP) that facilitate collagen invasion, thereby anchoring secondary lesions in the submesothelial matrix. As metalloproteinases have also been implicated in E-cadherin ectodomain shedding, the current study was undertaken to model the effects of matrix-induced integrin clustering on proteinase-catalyzed E-cadherin ectodomain shedding. Aggregation of collagen-binding integrins induced shedding of an 80-kDa E-cadherin ectodomain [soluble E-cadherin (sE-cad)] in a MMP- and Src kinase-dependent manner, and sE-cad was prevalent in ascites from ovarian cancer patients. Expression of MMP-9 was elevated by integrin aggregation, integrin-mediated ectodomain shedding was inhibited by a MMP-9 function blocking antibody, and incubation of cells with exogenous MMP-9 catalyzed E-cadherin ectodomain shedding. In contrast to other tumors wherein sE-cad is released into the circulation, EOC tumors maintain direct contact with sE-cad-rich ascites at high concentration, and incubation of EOC cells with physiologically relevant concentrations of recombinant sE-cad disrupted adherens junctions. These data support a novel mechanism for posttranslational modification of E-cadherin function via MMP-9 induction initiated by cell-matrix contact and suggest a mechanism for promotion of EOC metastatic dissemination.


Journal of Biological Chemistry | 2000

Urinary-type Plasminogen Activator (uPA) Expression and uPA Receptor Localization Are Regulated by α3β1Integrin in Oral Keratinocytes

Supurna Ghosh; Renee Brown; Jonathan C. R. Jones; Shawn M. Ellerbroek; M. Sharon Stack

Expression of urinary-type plasminogen activator (uPA) and its receptor (uPAR) is correlated with matrix proteolysis, cell adhesion, motility, and invasion. To evaluate the functional link between adhesion and proteolysis in gingival keratinocytes (pp126), cells were treated with immobilized integrin antibodies to induce integrin clustering. Clustering of α3 and β1 integrin subunits, but not α2, α5, α6, or β4, enhanced uPA secretion. Bead-immobilized laminin-5 and collagen I, two major α3β1 ligands, also induced uPA expression. Coordinate regulation of the serpin plasminogen activator inhibitor 1 was also apparent; however, a net increase in uPA activity was predominant. α3β1 integrin clustering induced extracellular signal-regulated kinase 1/2 phosphorylation, and both uPA induction and extracellular signal-regulated kinase activation were blocked by the mitogen-activated protein kinase/extracellular signal-regulated kinase kinase inhibitor PD98059. Integrin aggregation also promoted a dramatic redistribution of uPAR on the cell surface to sites of clustered α3β1 integrins. Co-immunoprecipitation of β1 integrin with uPAR provided further evidence that protein-protein interactions between uPAR and β1 integrin control uPAR distribution. As a functional consequence of uPA up-regulation and uPA-mediated plasminogen activation, the globular domain of the laminin-5 α3subunit, a major pp126 matrix protein, was proteolytically processed from a 190-kDa form to a 160-kDa species. Laminin-5 containing the 160-kDa α3 subunit efficiently nucleates hemidesmosome formation and reduces cell motility. Together, these data suggest that multivalent aggregation of the α3β1integrin regulates proteinase expression, matrix proteolysis, and subsequent cellular behavior.


Journal of Biological Chemistry | 2012

Suppression of Nuclear Factor-κB Activation and Inflammation in Microglia by Physically Modified Saline

Saurabh Khasnavis; Arundhati Jana; Avik Roy; Monalisa Mazumder; Bharat Bhushan; Tony Wood; Supurna Ghosh; Richard T. Watson; Kalipada Pahan

Background: Microglial activation plays an important role in the pathogenesis of neurodegenerative disorders. Results: Taylor-Couette-Poiseuille flow-modified saline (RNS60) inhibits microglial inflammation via type 1A phosphatidylinositol 3-kinase-Akt-CREB-mediated up-regulation of IκBα and inhibition of NF-κB activation. Conclusion: These results delineate a novel biological function of a physically modified saline. Significance: RNS60 may be of therapeutic benefit in neurodegenerative disorders. Chronic inflammation involving activated microglia and astroglia is becoming a hallmark of many human diseases, including neurodegenerative disorders. Although NF-κB is a multifunctional transcription factor, it is an important target for controlling inflammation as the transcription of many proinflammatory molecules depends on the activation of NF-κB. Here, we have undertaken a novel approach to attenuate NF-κB activation and associated inflammation in activated glial cells. RNS60 is a 0.9% saline solution containing charge-stabilized nanostructures that are generated by subjecting normal saline to Taylor-Couette-Poiseuille (TCP) flow under elevated oxygen pressure. RNS60, but not normal saline, RNS10.3 (TCP-modified saline without excess oxygen), and PNS60 (saline containing excess oxygen without TCP modification) were found to inhibit the production of nitric oxide (NO) and the expression of inducible NO synthase in activated microglia. Similarly, RNS60 also inhibited the expression of inducible NO synthase in activated astroglia. Inhibition of NF-κB activation by RNS60 suggests that RNS60 exerts its anti-inflammatory effect through the inhibition of NF-κB. Interestingly, RNS60 induced the activation of type IA phosphatidylinositol (PI) 3-kinase and Akt and rapidly up-regulated IκBα, a specific endogenous inhibitor of NF-κB. Inhibition of PI 3-kinase and Akt by either chemical inhibitors or dominant-negative mutants abrogated the RNS60-mediated up-regulation of IκBα. Furthermore, we demonstrate that RNS60 induced the activation of cAMP-response element-binding protein (CREB) via the PI 3-kinase-Akt pathway and that RNS60 up-regulated IκBα via CREB. These results describe a novel anti-inflammatory property of RNS60 via type IA PI 3-kinase-Akt-CREB-mediated up-regulation of IκBα, which may be of therapeutic benefit in neurodegenerative disorders.


Microscopy Research and Technique | 2000

Proteolytic modification of laminins: functional consequences.

Supurna Ghosh; M. Sharon Stack

The laminin family contains a number of complex, multi‐domain proteins that participate in a large variety of biologic processes. Limited proteolysis has been utilized extensively as a tool with which to determine laminin structure/function relationships. In addition, proteolytic modification of laminins may occur as a component of heterotrimer assembly and secretion, or may follow incorporation of mature laminin into the extracellular matrix. Conversely, laminin binding to cellular receptors may also influence proteinase expression. This review will highlight specific examples to demonstrate the functional interplay between laminins and proteinases in the regulation of laminin structure and function as well as in the subsequent control of proteinase expression. Microsc. Res. Tech. 51:238–246, 2000.


Journal of Biological Chemistry | 2006

Functional relevance of urinary-type plasminogen activator receptor-α3β1 integrin association in proteinase regulatory pathways

Supurna Ghosh; Jeffrey J. Johnson; Ratna Sen; Subhendu Mukhopadhyay; Yueying Liu; Feng Zhang; Ying Wei; Harold A. Chapman; M. Sharon Stack

Squamous cell carcinoma of the oral cavity is characterized by persistent, disorganized expression of integrin α3β1 and enhanced production of urinary-type plasminogen activator (uPA) and its receptor (uPAR) relative to normal oral mucosa. Because multivalent aggregation of α3β1 integrin up-regulates uPA and induces a dramatic co-clustering of uPAR, we explored the hypothesis that lateral ligation of α3β1 integrin by uPAR contributes to uPA regulation in oral mucosal cells. To investigate mechanisms by which uPAR/α3β1 binding enhances uPA expression, integrin-dependent signal activation was assessed. Both Src and ERK1/2 were phosphorylated in response to integrin aggregation, and blocking Src kinase activity completely abrogated ERK1/2 activation and uPA induction, whereas inhibition of epidermal growth factor receptor tyrosine kinase activity did not alter uPA expression. Proteinase up-regulation occurred at the transcriptional level and mutation of the AP1 (–1967) site in the uPA promoter blocked the uPAR/integrin-mediated transcriptional activation. Because uPAR is redistributed to clustered α3β1 integrins, the requirement for uPAR/α3β1 interaction in uPA regulation was assessed. Clustering of α3β1 in the presence of a peptide (α325) that disrupts uPAR/α3β1 integrin binding prevented uPA induction. Depletion of cell surface uPAR using small interfering RNA also blocked uPA induction following integrin α3β1 clustering. These results were confirmed using a genetic strategy in which α3 null epithelial cells reconstituted with wild type α3 integrin, but not a mutant α3 unable to bind uPAR, induced uPA expression upon integrin clustering, confirming the critical role of uPAR in integrin-regulated proteinase expression. Disruption of uPAR/α3β1 binding using peptide α325 or small interfering RNA blocked filopodia formation and matrix invasion, indicating that this interaction stimulates invasive behavior. Together these data support a model wherein matrix-induced clustering ofα3β1 integrin promotes uPAR/α3β1 interaction, thereby potentiating cellular signal transduction pathways culminating in activation of uPA expression and enhanced uPA-dependent invasive behavior.


Journal of Biological Chemistry | 2008

Polypyrimidine Tract-binding Protein (PTB) Differentially Affects Malignancy in a Cell Line-dependent Manner

Chen Wang; John Norton; Supurna Ghosh; Julie Kim; Kazuo Fushimi; Jane Y. Wu; M. Sharon Stack; Sui Huang

RNA processing is altered during malignant transformation, and expression of the polypyrimidine tract-binding protein (PTB) is often increased in cancer cells. Although some data support that PTB promotes cancer, the functional contribution of PTB to the malignant phenotype remains to be clarified. Here we report that although PTB levels are generally increased in cancer cell lines from multiple origins and in endometrial adenocarcinoma tumors, there appears to be no correlation between PTB levels and disease severity or metastatic capacity. The three isoforms of PTB increase heterogeneously among different tumor cells. PTB knockdown in transformed cells by small interfering RNA decreases cellular growth in monolayer culture and to a greater extent in semi-solid media without inducing apoptosis. Down-regulation of PTB expression in a normal cell line reduces proliferation even more significantly. Reduction of PTB inhibits the invasive behavior of two cancer cell lines in Matrigel invasion assays but enhances the invasive behavior of another. At the molecular level, PTB in various cell lines differentially affects the alternative splicing pattern of the same substrates, such as caspase 2. Furthermore, overexpression of PTB does not enhance proliferation, anchorage-independent growth, or invasion in immortalized or normal cells. These data demonstrate that PTB is not oncogenic and can either promote or antagonize a malignant trait dependent upon the specific intra-cellular environment.


Anti-Cancer Drugs | 2008

Synthesis and anticancer activities of 6-amino amonafide derivatives

John Norton; Mark A. Witschi; Lynn Luong; Akane Kawamura; Supurna Ghosh; M. Sharon Stack; Edith Sim; Michael J. Avram; Daniel H. Appella; Sui Huang

Amonafide is a DNA intercalator and topoisomerase II inhibitor in clinical development for the treatment of neoplastic diseases. Amonafide contains a free arylamine, which causes it to be metabolized in humans by N-acetyl transferase-2 (NAT2) into a toxic form. To eliminate the NAT2 acetylation of amonafide while retaining the anticancer properties, we have synthesized nine derivatives that are structurally similar to amonafide that should not be acetylated. Eight derivatives have arylamines at the 6-position (vs. 5-position of amonafide) and one derivative completely lacks the arylamine. The derivative with a free amine in the 6-position and one with a substituted amine in the 6-position are not acetylated, whereas amonafide is extensively acetylated as determined by an NAT2 assay. The biological activities of these compounds were evaluated to determine whether they behaved similarly to amonafide in purified systems and in vitro. We found that three compounds had similar cancer cell-selective growth inhibition to amonafide, while retaining similar subcellular localization, DNA intercalation and topoisomerase II inhibition activities. In addition, these compounds were able to eliminate a marker of metastatic potential, the perinucleolar compartment. These three compounds (named numonafides) might thus allow for better patient management than those treated with amonafide; hence, they should be developed further as potential clinical replacements for amonafide or as novel anticancer drugs.


PLOS ONE | 2012

Protection of Tregs, suppression of Th1 and Th17 cells, and amelioration of experimental allergic encephalomyelitis by a physically-modified saline.

Susanta Mondal; Jeffrey Martinson; Supurna Ghosh; Richard T. Watson; Kalipada Pahan

In multiple sclerosis (MS) and other autoimmune diseases, the autoreactive T cells overcome the resistance provided by the regulatory T cells (Tregs) due to a decrease in the number of Foxp3-expressing Tregs. Therefore, upregulation and/or maintenance of Tregs during an autoimmune insult may have therapeutic efficacy in autoimmune diseases. Although several immunomodulatory drugs and molecules are available, most present significant side effects over long-term use. Here we have undertaken an innovative approach to upregulate Tregs and achieve immunomodulation. RNS60 is a 0.9% saline solution generated by subjecting normal saline to Taylor-Couette-Poiseuille (TCP) flow under elevated oxygen pressure. RNS60, but not NS (normal saline), RNS10.3 (TCP-modified saline without excess oxygen) and PNS60 (saline containing excess oxygen without TCP modification), was found to upregulate Foxp3 and enrich Tregs in MBP-primed T cells. Moreover, RNS60, but not NS, RNS10.3 and PNS60, inhibited the production of nitric oxide (NO) and the expression of iNOS in MBP-primed splenocytes. Incubation of the cells with an NO donor abrogated the RNS60-mediated upregulation of Foxp3. These results suggest that RNS60 boosts Tregs via suppression of NO production. Consistent to the suppressive activity of Tregs towards autoreactive T cells, RNS60, but not NS, RNS10.3, or PNS60, suppressed the differentiation of Th17 and Th1 cells and shifted the balance towards a Th2 response. Finally, RNS60 treatment exhibited immunomodulation and ameliorated adoptive transfer of experimental allergic encephalomyelitis, an animal model of MS, via Tregs. These results describe a novel immunomodulatory property of RNS60 and suggest its exploration for therapeutic intervention in MS and other autoimmune disorders.


Cancer | 2002

Loss of adhesion-regulated proteinase production is correlated with invasive activity in oral squamous cell carcinoma.

Supurna Ghosh; Hidayatullah G. Munshi; Ratna Sen; Laura A. Linz-McGillem; Robert D. Goldman; Jochen Lorch; Kathleen J. Green; Jonathan C. R. Jones; M. Sharon Stack

Oral squamous cell carcinoma (OSCC) is the most common malignancy of the oral cavity. However, the cellular and biochemical factors that underlie locoregional and distant spread of the disease are poorly understood. Invasion of OSCC requires multiple cellular events including dissolution of cell‐cell junctions, basement membrane attachment, extracellular matrix proteolysis, and migration.


PLOS ONE | 2014

A physically-modified saline suppresses neuronal apoptosis, attenuates tau phosphorylation and protects memory in an animal model of Alzheimer's disease

Khushbu K. Modi; Arundhati Jana; Supurna Ghosh; Richard T. Watson; Kalipada Pahan

Alzheimers disease (AD), the leading cause of dementia in the aging population, is characterized by the presence of neuritic plaques, neurofibrillary tangles and extensive neuronal apoptosis. Neuritic plaques are mainly composed of aggregates of amyloid-β (Aβ) protein while neurofibrillary tangles are composed of the hyperphosphorylated tau protein. Despite intense investigations, no effective therapy is currently available to halt the progression of this disease. Here, we have undertaken a novel approach to attenuate apoptosis and tau phosphorylation in cultured neuronal cells and in a transgenic animal model of AD. RNS60 is a 0.9% saline solution containing oxygenated nanobubbles that is generated by subjecting normal saline to Taylor-Couette-Poiseuille (TCP) flow under elevated oxygen pressure. In our experiments, fibrillar Aβ1-42, but not the reverse peptide Aβ42-1, induced apoptosis and cell death in human SHSY5Y neuronal cells. RNS60, but not NS (normal saline), RNS10.3 (TCP-modified saline without excess oxygen) or PNS60 (saline containing excess oxygen without TCP modification), attenuated Aβ(1–42)-induced cell death. RNS60 inhibited neuronal cell death via activation of the type 1A phosphatidylinositol-3 (PI-3) kinase – Akt – BAD pathway. Furthermore, RNS60 also decreased Aβ(1–42)-induced tau phosphorylation via (PI-3 kinase – Akt)-mediated inhibition of GSK-3β. Similarly, RNS60 treatment suppressed neuronal apoptosis, attenuated Tau phosphorylation, inhibited glial activation, and reduced the burden of Aβ in the hippocampus and protected memory and learning in 5XFAD transgenic mouse model of AD. Therefore, RNS60 may be a promising pharmaceutical candidate in halting or delaying the progression of AD.

Collaboration


Dive into the Supurna Ghosh's collaboration.

Top Co-Authors

Avatar

Kalipada Pahan

Rush University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Avik Roy

Rush University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Saurabh Khasnavis

Rush University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Susanta Mondal

Rush University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Arundhati Jana

Rush University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ratna Sen

Northwestern University

View shared research outputs
Researchain Logo
Decentralizing Knowledge