Network


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

Hotspot


Dive into the research topics where Subhra Mohapatra is active.

Publication


Featured researches published by Subhra Mohapatra.


Nature Medicine | 2005

Inhibition of respiratory syncytial virus infection with intranasal siRNA nanoparticles targeting the viral NS1 gene

W. Zhang; Hong Yang; Xiaoyuan Kong; Subhra Mohapatra; Homero San Juan-Vergara; Gary Hellermann; Sumita Behera; Rajeswari Singam; Richard F. Lockey; Shyam S. Mohapatra

Respiratory syncytial virus (RSV) infection is one of the major causes of respiratory tract infection for which no vaccine or antiviral treatment is available. The RSV NS1 protein seems to antagonize the host interferon (IFN) response; however, its mechanism is unknown. Here, we used a plasmid-borne small interfering RNA targeting the NS1 gene (siNS1) to examine the role of NS1 in modulating RSV infection. RSV replication was reduced in A549 cells, but not IFN–deficient Vero cells, transfected with siNS1. siNS1 induced upregulated expression of IFN-β and IFN-inducible genes in A549 cells. siNS1-transfected human dendritic cells, upon RSV infection, produced elevated type-1 IFN and induced differentiation of naive CD4+ T cells to T helper type 1 (TH1) cells. Mice treated intranasally with siNS1 nanoparticles before or after infection with RSV showed substantially decreased virus titers in the lung and decreased inflammation and airway reactivity compared to controls. Thus, siNS1 nanoparticles may provide an effective inhibition of RSV infection in humans.


Nanomedicine: Nanotechnology, Biology and Medicine | 2010

Multifunctional magnetic nanoparticles for targeted delivery

Arun Kumar; P.K. Jena; Sumita Behera; Richard F. Lockey; Subhra Mohapatra; Shyam S. Mohapatra

UNLABELLED A major problem associated with drug therapy is the inability to deliver pharmaceuticals to a specific site of the body without causing nonspecific toxicity. Development of magnetic nanoparticles and techniques for their safe transport and concentration in specific sites in the body would constitute a powerful tool for gene/drug therapy in vivo. Furthermore, drug delivery in vitro could improve further if the drugs were modified with antibodies, proteins, or ligands. For in vivo experiments, magnetic nanoparticles were conjugated with plasmid DNA expressing enhanced green fluorescent protein (EGFP) and then coated with chitosan. These particles were injected into mice through the tail vein and directed to the heart and kidneys by means of external magnets of 25 gauss or 2kA-kA/m. These particles were concentrated in the lungs, heart, and kidneys of mice, and the expression of EGFP in these sites were monitored. The expression of EGFP in specific locations was visualized by whole-body fluorescent imaging, and the concentration of these particles in the designated body locations was confirmed by transmission electron microscopy. In another model system, we used atrial natriuretic peptide and carcinoembryonic antigen antibodies coupled to the chitosan-coated magnetic nanoparticles to target cells in vitro. The present work demonstrates that a simple external magnetic field is all that is necessary to target a drug to a specific site inside the body without the need to functionalize the nanoparticles. However, the option to use magnetic targeting with external magnets on functionalized nanoparticles could prove as a more efficient means of drug delivery. FROM THE CLINICAL EDITOR This paper addresses targeted drug delivery with magnetic nanoparticles. The authors demonstrate that a simple external magnetic field is sufficient to target a drug to specific sites in the body without the need for functionalized nanoparticles, at least in selected organs and diseases.


Journal of Neuroinflammation | 2012

New perspectives on central and peripheral immune responses to acute traumatic brain injury

Mahasweta Das; Subhra Mohapatra; Shyam S. Mohapatra

Traumatic injury to the brain (TBI) results in a complex set of responses involving various symptoms and long-term consequences. TBI of any form can cause cognitive, behavioral and immunologic changes in later life, which underscores the problem of underdiagnosis of mild TBI that can cause long-term neurological deficits. TBI disrupts the blood–brain barrier (BBB) leading to infiltration of immune cells into the brain and subsequent inflammation and neurodegeneration. TBI-induced peripheral immune responses can also result in multiorgan damage. Despite worldwide research efforts, the methods of diagnosis, monitoring and treatment for TBI are still relatively ineffective. In this review, we delve into the mechanism of how TBI-induced central and peripheral immune responses affect the disease outcome and discuss recent developments in the continuing effort to combat the consequences of TBI and new ways to enhance repair of the damaged brain.


Journal of Biological Chemistry | 2005

Activation of p27Kip1 Expression by E2F1 A NEGATIVE FEEDBACK MECHANISM

Chuangui Wang; Xinghua Hou; Subhra Mohapatra; Yihong Ma; W. Douglas Cress; W. Jack Pledger; Jiandong Chen

The E2F1 transcription factor is a critical regulator of cell cycle due to its ability to promote S phase entry. However, E2F1 overexpression also sensitizes cells to apoptosis and E2F1-null mice are predisposed to tumor development, suggesting that it also has properties of a growth suppressor. E2F1 transcription function is regulated by interaction with hypophosphorylated pRb. Cdk inhibitors such as p16INK4a and p27Kip1 inhibit pRb phosphorylation by the cyclin D/Cdk4 and cyclin E/Cdk2 complexes, thus keeping E2F1 in an inactive state. We found that E2F1 binds to the p27 promoter in vivo and activates p27 mRNA and protein expression. Depletion of endogenous E2F1 by siRNA causes a reduction in basal p27 expression level. Inhibition of endogenous p27 expression by siRNA increases E2F1 transcriptional activity and permits accelerated cell cycle progression by exogenous E2F1. These observations suggest that induction of p27 acts as a negative feedback mechanism for E2F1 and may also contribute to other functions of E2F1.


Molecular and Cellular Biology | 2003

p27Kip1 and p21Cip1 Are Not Required for the Formation of Active D Cyclin-cdk4 Complexes

Tapan K. Bagui; Subhra Mohapatra; Eric B. Haura; W. J. Pledger

ABSTRACT Our studies address questions pertaining to the regulation of D cyclin-cdk4 activity, and the following results were obtained. Conditions that increased the abundance of the D cyclins also increased the abundance of enzymatically active D cyclin-cdk4 complexes in mouse embryo fibroblasts (MEFs) lacking both p27Kip1 and p21Cip1 (p27/p21−/−). Such conditions included ectopic expression of cyclin D1 and inhibition of D cyclin degradation by the proteasome inhibitor MG132. However, as determined by treatment of wild-type MEFs with MG132, maximal accumulation of D cyclin-cdk4 complexes required p27Kip1 and p21Cip1 and coincided with the formation of inactive D cyclin-cdk4-p27Kip1 or -p21Cip1 complexes. p27Kip1 or p21Cip1 also increased the abundance of D cyclin-cdk4 complexes and reduced amounts of cdk4 activity when ectopically expressed in p27/p21−/− MEFs. Lastly, increases in the stability of the D cyclins accounted for their greater abundance in wild-type MEFs than in p27/p21−/− MEFs. We conclude that (i) D cyclin-cdk4 complexes are formed and become active in the absence of p27Kip1 and p21Cip1 and (ii) p27Kip1 and p21Cip1 maximize the accumulation but inhibit the activity of D cyclin-cdk4 complexes. We suggest that D cyclin-cdk4 complexes are more stable when bound to p27Kip1 or p21Cip1 and that formation of ternary complexes also stabilizes the D cyclins.


Journal of Materials Chemistry B | 2013

Multifunctional chitosan magnetic-graphene (CMG) nanoparticles: a theranostic platform for tumor-targeted co-delivery of drugs, genes and MRI contrast agents

Chunyan Wang; Sowndharya Ravi; Ujjwala Sree Garapati; Mahasweta Das; Mark Howell; Jaya Mallela; Subbiah Alwarappan; Shyam S. Mohapatra; Subhra Mohapatra

Combing chemotherapy with gene therapy has been one of the most promising strategies for the treatment of cancer. The noninvasive MRI with superparamagnetic iron oxide (SPIO) as contrast agent is one of the most effecitve techniques for evaluating the antitumor therapy. However, to construct a single system that can deliver efficiently gene, drug and SPIO to the cancer site remains a challenge. Herein, we report a chitosan functionalized magnetic graphene nanoparticle (CMG) platform for simultaneous gene/drug and SPIO delivery to tumor. The phantom and ex vivo MRI images suggest CMG as a strong T2 contrast-enhancing agent. The CMGs are biocompatible as evaluated by the WST assay and predominantly accumulate in tumors as shown by biodistribution studies and MRI. The anticancer drug doxorubicin (DOX) loaded CMGs (DOX-CMGs) release DOX faster at pH 5.1 than at pH 7.4, and more effective (IC50 = 2 μM) in killing A549 lung cancer cells than free DOX (IC50 = 4 μM). CMGs efficiently deliver DNA into A549 lung cancer cells and C42b prostate cancer cells. In addition, i.v. administration of GFP-plasmid encapsulated within DOX-CMGs into tumor-bearing mice has showed both GFP expression and DOX accumulation at the tumor site at 24 and 48 hrs after administration. These results indicate CMGs provide a robust and safe theranostic platform, which integrates targeted delivery of both gene medicine and chemotherapeutic drug(s), and enhanced MR imaging of tumors. The integrated chemo- and gene- therapeutic and diagnostic design of CMG nanoparticles shows promise for simultaneous targeted imaging, drug delivery and real -time monitoring of therapeutic effect for cancer.


Cancer Research | 2008

Natriuretic Peptide Receptor A as a Novel Anticancer Target

Xiaoyuan Kong; Xiaoqin Wang; Weidong Xu; Sumita Behera; Gary Hellermann; Arun Kumar; Richard F. Lockey; Subhra Mohapatra; Shyam S. Mohapatra

The receptor for atrial natriuretic peptide (ANP), natriuretic peptide receptor A (NPRA), is expressed in cancer cells, and natriuretic peptides have been implicated in cancers. However, the direct role of NPRA signaling in tumorigenesis remains elusive. Here, we report that NPRA expression and signaling is important for tumor growth. NPRA-deficient mice showed significantly reduced antigen-induced pulmonary inflammation. NPRA deficiency also substantially protected C57BL/6 mice from lung, skin, and ovarian cancers. Furthermore, a nanoparticle-formulated interfering RNA for NPRA attenuated B16 melanoma tumors in mice. Ectopic expression of a plasmid encoding NP73-102, the NH(2)-terminal peptide of the ANP prohormone, which down-regulates NPRA expression, also suppressed lung metastasis of A549 cells in nude mice and tumorigenesis of Line 1 cells in immunocompetent BALB/c mice. The antitumor activity of NP73-102 was in part attributed to apoptosis of tumor cells. Western blot and immunohistochemistry staining indicated that the transcription factor, nuclear factor-kappaB, was inactivated, whereas the level of tumor suppressor retinoblastoma protein was up-regulated in the lungs of NPRA-deficient mice. Furthermore, expression of vascular endothelial growth factor was down-regulated in the lungs of NPRA-deficient mice compared with that in wild-type mice. These results suggest that NPRA is involved in tumor angiogenesis and represents a new target for cancer therapy.


Nanomedicine: Nanotechnology, Biology and Medicine | 2013

A chitosan-modified graphene nanogel for noninvasive controlled drug release

Chunyan Wang; Jaya Mallela; Ujjwala Sree Garapati; Sowndharya Ravi; Vignesh Chinnasamy; Yvonne K. Girard; Mark Howell; Subhra Mohapatra

UNLABELLED A near infrared (NIR) triggered drug delivery platform based on the chitosan-modified chemically reduced graphene oxide (CRGO) incorporated into a thermosensitive nanogel (CGN) was developed. CGN exhibited an NIR-induced thermal effect similar to that of CRGO, reversible thermo-responsive characteristics at 37-42 °C and high doxorubicin hydrochloride (DOX) loading capacity (48 wt%). The DOX loaded CGN (DOX-CGN) released DOX faster at 42 °C than at 37 °C. The fluorescence images revealed DOX expression in the cytoplasm of cancer cells when incubated with DOX-CGN at 37 °C but in the nucleus at 42 °C. Upon irradiation with NIR light (808 nm), a rapid, repetitive DOX release from the DOX-CGN was observed. Furthermore, the cancer cells incubated with DOX-CGN and irradiated with NIR light displayed significantly greater cytotoxicity than without irradiation owing to NIR-triggered increase in temperature leading to nuclear DOX release. These results demonstrate CGNs promising application for on-demand drug release by NIR light. FROM THE CLINICAL EDITOR These investigators report the successful development of a novel near infrared triggered drug delivery platform based on chitosan-modified chemically reduced graphene oxide (CRGO) incorporated into a thermosensitive nanogel (CGN).


Cancer Research | 2005

Accumulation of p53 and reductions in XIAP abundance promote the apoptosis of prostate cancer cells.

Subhra Mohapatra; Baoky Chu; Xiuhua Zhao; W. J. Pledger

Toward the goal of developing effective treatments for prostate cancers, we examined the effects of cyclin-dependent kinase inhibitors on the survival of prostate cancer cells. We show that roscovitine, R-roscovitine, and CGP74514A (collectively referred to as CKIs) induce the apoptosis of LNCaP and LNCaP-Rf cells, both of which express wild-type p53. Apoptosis required caspase-9 and caspase-3 activity, and cytochrome c accumulated in the cytosol of CKI-treated cells. Amounts of p53 increased substantially in CKI-treated cells, whereas amounts of the endogenous caspase inhibitor XIAP decreased. CKIs did not appreciably induce the apoptosis of LNCaP cells treated with pifithrin-alpha, which prevents p53 accumulation, or of prostate cancer cells that lack p53 function (PC3 and DU145). Ectopic expression of p53 in PC3 cells for 44 hours did not reduce XIAP abundance or induce apoptosis. However, p53-expressing PC3 cells readily apoptosed when exposed to CKIs or when depleted of XIAP by RNA interference. These findings show that CKIs induce the mitochondria-mediated apoptosis of prostate cancer cells by a dual mechanism: p53 accumulation and XIAP depletion. They suggest that these events in combination may prove useful in the treatment of advanced prostate cancers.


PLOS ONE | 2012

Respiratory syncytial virus NS1 protein colocalizes with mitochondrial antiviral signaling protein MAVS following infection.

Sandhya Boyapalle; Terianne Wong; Julio Garay; Michael N. Teng; Homero San Juan-Vergara; Subhra Mohapatra; Shyam S. Mohapatra

Respiratory syncytial virus (RSV) nonstructural protein 1(NS1) attenuates type-I interferon (IFN) production during RSV infection; however the precise role of RSV NS1 protein in orchestrating the early host-virus interaction during infection is poorly understood. Since NS1 constitutes the first RSV gene transcribed and the production of IFN depends upon RLR (RIG-I-like receptor) signaling, we reasoned that NS1 may interfere with this signaling. Herein, we report that NS1 is localized to mitochondria and binds to mitochondrial antiviral signaling protein (MAVS). Live-cell imaging of rgRSV-infected A549 human epithelial cells showed that RSV replication and transcription occurs in proximity to mitochondria. NS1 localization to mitochondria was directly visualized by confocal microscopy using a cell-permeable chemical probe for His6-NS1. Further, NS1 colocalization with MAVS in A549 cells infected with RSV was shown by confocal laser microscopy and immuno-electron microscopy. NS1 protein is present in the mitochondrial fraction and co-immunoprecipitates with MAVS in total cell lysatesof A549 cells transfected with the plasmid pNS1-Flag. By immunoprecipitation with anti-RIG-I antibody, RSV NS1 was shown to associate with MAVS at an early stage of RSV infection, and to disrupt MAVS interaction with RIG-I (retinoic acid inducible gene) and the downstream IFN antiviral and inflammatory response. Together, these results demonstrate that NS1 binds to MAVS and that this binding inhibits the MAVS-RIG-I interaction required for IFN production.

Collaboration


Dive into the Subhra Mohapatra's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chunyan Wang

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Gary Hellermann

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Mark Howell

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Richard F. Lockey

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Xiaoyuan Kong

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Mahasweta Das

University of South Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ryan Green

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Sandhya Boyapalle

University of South Florida

View shared research outputs
Researchain Logo
Decentralizing Knowledge