Network


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

Hotspot


Dive into the research topics where Shilpa Choudhary is active.

Publication


Featured researches published by Shilpa Choudhary.


Principles of Bone Biology (Third Edition) | 2008

Prostaglandins and Bone Metabolism

Carol C. Pilbeam; Shilpa Choudhary; Katherine Blackwell; Lawrence G. Raisz

Publisher Summary This chapter focuses on the effects of prostaglandins (PG) and other eicosanoids on bone resorption and formation. Eicosanoids are oxygenated 20-carbon fatty acids derived from polyunsaturated eicosatrienoic, eicosatetranoic (arachidonic), and eicosapentanoic fatty acids. The production of PGs involves three major steps which include hormone-or stress-activated mobilization of AA, conversion of AA to prostaglandin endoperoxide H 2 (PGH 2 ), and conversion of diffusible PGH 2 by tissue-specific isomerases and reductases to PGE 2 , PGD 2 , PGF 2α , prostacyclin (PGI 2 ), and thromboxane. The committed step in the conversion of AA to PGs is catalyzed by a bifunctional enzyme, which converts free AA to PGG 2 in a cyclooxgenase reaction followed by reduction of PGG 2 to PGH 2 in a peroxidase reaction. This enzyme, formally named prostaglandin endoperoxide H synthase or prostaglandin G/H synthase (PGHS), is popularly called cyclooxgygenase (COX) in reference to its first function. The individual PG synthases are differentially distributed in tissues and are believed to influence PG production largely by determining the predominant type of prostanoid synthesized in a particular tissue. PGE 1 and PGE 2 stimulate osteoclast formation in marrow cultures. The PG enhancement of stimulated osteoclast formation in marrow culture may reflect the increased formation of new osteoclastic precursors, while stimulated resorption in organ culture may be more dependent on activation of a pool of available osteoclastic precursors. When added to isolated osteoclasts in vitro, PGE 2 transiently inhibits bone resorption. PGs can have both stimulatory and inhibitory effects on bone formation. PGs can increase both periosteal and endosteal bone formation in the rat and produce substantial increases in bone mass, similar to the effects of PTH. At high concentrations, PGs can inhibit collagen synthesis in cell and organ culture. This inhibitory effect occurs largely via transcriptional inhibition of collagen and are mediated by the FP receptor rather than an EP receptor.


Journal of Bone and Mineral Research | 2007

Strontium Ranelate Promotes Osteoblastic Differentiation and Mineralization of Murine Bone Marrow Stromal Cells: Involvement of Prostaglandins

Shilpa Choudhary; Philippe Halbout; Cynthia B. Alander; Lawrence G. Raisz; Carol C. Pilbeam

Strontium ranelate is a new anti‐osteoporosis treatment. This study showed that strontium ranelate stimulated PGE2 production and osteoblastic differentiation in murine marrow stromal cells, which was markedly reduced by inhibition of COX‐2 activity or disruption of COX‐2 gene expression. Hence, some anabolic effects of strontium ranelate may be mediated by the induction of COX‐2 and PGE2 production.


Biochemical and Biophysical Research Communications | 2002

Fluid flow induces COX-2 expression in MC3T3-E1 osteoblasts via a PKA signaling pathway.

Sunil Wadhwa; Shilpa Choudhary; Maria Voznesensky; Mary Anne F. Epstein; Lawrence G. Raisz; Carol C. Pilbeam

Mechanical loading of bone generates fluid flow within the mineralized matrix which can exert fluid shear stress (FSS) at cell membranes. FSS induces new transcription of cyclooxygenase-2 (COX-2) in MC3T3-E1 osteoblasts, with peak effects at 4-5h. Using MC3T3-E1 cells stably transfected with the COX-2 promoter fused to a luciferase reporter, we examined involvement of the protein kinase A (PKA) and protein kinase C (PKC) signaling pathways in the peak COX-2 mRNA and luciferase responses to FSS (10dyn/cm(2)). Neither inhibition nor down-regulation of the PKC pathway affected the FSS stimulation of COX-2 mRNA or luciferase activity. In contrast, inhibitors of the PKA pathway, used at doses which inhibited forskolin-stimulated luciferase activity by 70-80%, reduced FSS-stimulated COX-2 mRNA expression and luciferase activity by 50-80%. Hence, peak FSS induction of COX-2 expression in MC3T3-E1 osteoblastic cells is largely dependent on the PKA signaling pathway.


Journal of Bone and Mineral Research | 2003

Extracellular Calcium Is a Potent Inducer of Cyclo‐oxygenase‐2 in Murine Osteoblasts Through an ERK Signaling Pathway

Shilpa Choudhary; Sunil Wadhwa; Lawrence G. Raisz; Cynthia B. Alander; Carol C. Pilbeam

[Ca2+]e may be important in bone turnover. We found [Ca2+]e induces COX‐2 transcription and PGE2 production in primary calvarial osteoblasts through an ERK signaling pathway. Inhibition of PGE2 production inhibited the [Ca2+]e stimulation of osteoblastic differentiation but not the increase in cell number. Hence, some effects of [Ca2+]e on bone may be mediated by COX‐2.


Journal of Bone and Mineral Research | 2004

Biphasic Effect of Prostaglandin E2 on Osteoclast Formation in Spleen Cell Cultures: Role of the EP2 Receptor†

Katsuhiro Ono; Hironori Kaneko; Shilpa Choudhary; Carol C. Pilbeam; Joseph A. Lorenzo; Takuhiko Akatsu; Nobuo Kugai; Lawrence G. Raisz

We examined the effect of PGE2 on OC formation from spleen cells treated with M‐CSF and RANKL. PGE2 decreased OC number at 5–6 days of culture and increased OC number, size, and resorptive activity at 7–8 days. A selective EP2 receptor agonist mimicked these effects. Deletion of the EP2 receptor or depletion of T‐cells abrogated the increase in OC number.


Cancer Research | 2006

Overexpression of COX-2 in human osteosarcoma cells decreases proliferation and increases apoptosis.

Zheng Xu; Shilpa Choudhary; Olga Voznesensky; Meenal Mehrotra; Monica Woodard; Marc F. Hansen; Harvey R. Herschman; Carol C. Pilbeam

Overexpression of cyclooxygenase-2 (COX-2) is generally considered to promote tumorigenesis. To investigate a potential role of COX-2 in osteosarcoma, we overexpressed COX-2 in human osteosarcoma cells. Saos-2 cells deficient in COX-2 expression were retrovirally transduced or stably transfected with murine COX-2 cDNA. Functional expression of COX-2 was confirmed by Northern and Western analyses and prostaglandin production. Overexpression of COX-2 reduced cell numbers by 50% to 70% compared with controls. Decreased proliferation in COX-2-overexpressing cells was associated with cell cycle prolongation in G(2)-M. Apoptosis, measured by both Annexin V binding assay and terminal deoxyribonucleotidyl transferase-mediated dUTP nick end labeling staining, was increased in cells overexpressing COX-2, and the increase was not reversed by treatment with NS-398, indicating that the effects were not mediated by prostaglandins. Retroviral COX-2 overexpression in two other human osteosarcoma cell lines, U2OS and TE85, also decreased cell viability. However, in the human colon carcinoma HCT-116 cell line, which is deficient in COX-2, retroviral overexpression of COX-2, at similar efficiency as in Saos-2 cells, increased resistance to apoptosis. Reactive oxygen species (ROS), measured by flow cytometry, were increased by COX-2 overexpression in Saos-2 cells but not in HCT-116 cells. Inhibition of peroxidase activity, but not of COX activity, blocked the ROS increase. Antioxidants blocked the increase in ROS and the increase in apoptosis due to COX-2 overexpression in Saos-2 cells. Our results suggest that (a) COX-2 overexpression in osteosarcoma cells may increase resistance to tumorigenesis by increasing ROS to levels that decrease cell viability and (b) the effects of COX-2 overexpression are cell type/tissue dependent.


Prostaglandins & Other Lipid Mediators | 2009

Effects of prostaglandin E2 on bone in mice in vivo

Qi Gao; Manshan Xu; Cynthia B. Alander; Shilpa Choudhary; Carol C. Pilbeam; Lawrence G. Raisz

We have examined the effects of varying doses, schedules and routes of administration of prostaglandin E(2) (PGE(2)) on bone in mice. Male C57BL/6 mice treated with a high dose of PGE(2) (6 mg/kg/d) showed decreased trabecular bone volume (BV/TV) by 14 d, indicating increased bone resorption. However, there was also stimulation of bone formation at 14 d after 3 d treatment with PGE(2,) since mineral apposition rate (MAR) and bone formation rate (BFR/BS) were increased. In CD-1 male and female mice, PGE(2) (3mg/kg, 2/wk for 4 wk) increased MAR by 50% and BFR/BS by 100%, but there was no significant change in BV/TV. Tibial mRNA showed an increase in BMP-2 and RUNX-2 expression with PGE(2). Additional experiments using a higher dose or longer exposure did not increase bone mass. We conclude that exposure to high doses of PGE(2) in mice may be anabolic but is balanced by catabolic effects. Studies of PGE(2) in combination with an inhibitor of resorption could lead to development of a true anabolic model and permit assessment of the roles of specific PGE(2) receptors and signal transduction pathways.


Carcinogenesis | 2013

Macrophage migratory inhibitory factor promotes bladder cancer progression via increasing proliferation and angiogenesis

Shilpa Choudhary; Poornima Hegde; James Pruitt; Thais Sielecki; Dharamainder Choudhary; Kristen Scarpato; David J. DeGraff; Carol C. Pilbeam; John A. Taylor

Macrophage migratory inhibitory factor (MIF) is a proinflammatory cytokine shown to promote tumorigenesis. Using the N-butyl-N-(4-hydroxybutyl)-nitrosamine (BBN) model of bladder cancer, we previously showed that MIF knockout mice display decreased angiogenesis and invasion compared with wild-type. This study examines the role of MIF in bladder cancer via use of oral inhibitors of MIF. In vitro, high-grade bladder cancer cells were treated with recombinant human MIF +/- (rhMIF+/-) inhibitor. Measurements included cell counts, proliferation by (3)H-thymidine incorporation (TdR), extracellular signal-regulated kinase (ERK) phosphorylation by western blot analysis, messenger RNA (mRNA) expression by quantitative PCR and protein secretion by enzyme-linked immunosorbent assay. Treatment with rhMIF increased ERK phosphorylation, cell counts, TdR and mRNA expression and protein secretion of vascular endothelial growth factor, which were blocked by specific inhibitors of ERK and MIF. In vivo, 3-month-old male C57Bl/6 mice were given BBN for 22 and 16 weeks in study 1 and study 2, respectively. Mice (n = 8-10 per group) were gavaged with vehicle or doses of MIF inhibitors daily from weeks 16-22 in both studies. Average bladder weights, reflecting tumor mass, tumor stage/burden, mitotic rate and proliferation indices, and microvessel densities were reduced in inhibitor groups versus controls. In summary, MIF promotes bladder cancer via increasing cell proliferation and angiogenesis and oral inhibitors of MIF may prove useful in treatment of this disease.


Prostaglandins & Other Lipid Mediators | 2008

Effect of deletion of the prostaglandin EP2 receptor on the anabolic response to prostaglandin E2 and a selective EP2 receptor agonist

Shilpa Choudhary; Cynthia B. Alander; Peili Zhan; Qi Gao; Carol C. Pilbeam; Lawrence G. Raisz

Studies using prostaglandin E receptor (EP) agonists indicate that prostaglandin (PG) E(2) can have anabolic effects through both EP4 and EP2 receptors. We previously found that the anabolic response to a selective EP4 receptor agonist (EP4A, Ono Pharmaceutical) was substantially greater than to a selective EP2 receptor agonist (EP2A) in cultured murine calvarial osteoblastic cells. To further define the role of the EP2 receptor in PG-mediated effects on bone cells, we examined the effects of EP2A and PGE(2) on both calvarial primary osteoblasts (POB) and marrow stromal cells (MSC) cultured from mice with deletion of one (Het) or both (KO) alleles of the EP2 receptor compared to their wild-type (WT) littermates. Deletion of EP2 receptor was confirmed by quantitative real-time PCR, Western blot and immunohistochemistry. The 1 month-old mice used to provide cells in these studies did not show any significant differences in their femurs by static histomorphometry. EP2A was found to enhance osteoblastic differentiation as measured by alkaline phosphatase mRNA expression and activity as well as osteocalcin mRNA expression and mineralization in the WT cell cultures from both marrow and calvariae. The effects were somewhat diminished in cultures from Het mice and abrogated in cultures from KO mice. PGE(2) effects were greater than those of EP2A, particularly in POB cultures and were only moderately diminished in Het and KO cell cultures. We conclude that activation of the EP2 receptor is able to enhance differentiation of osteoblasts, that EP2A is a true selective agonist for this receptor and that PGE(2) has an additional anabolic effect likely mediated by the EP4 receptor.


Biochemical and Biophysical Research Communications | 2008

Anabolic effects of PTH in cyclooxygenase-2 knockout osteoblasts in vitro.

Shilpa Choudhary; Hechang Huang; Lawrence G. Raisz; Carol C. Pilbeam

PTH is a potent bone anabolic agent in vivo but anabolic effects on osteoblast differentiation in vitro are difficult to demonstrate. This study examined the role of cyclooxygenase (COX)-2 and prostaglandin (PG) production in the effects of PTH on osteoblast differentiation in vitro using marrow stromal cell (MSC) and calvarial osteoblast (COB) cultures from COX-2 knockout (KO) and wild type (WT) mice. Cells were treated with PTH (10 nM) or vehicle throughout culture. Alkaline phosphatase (ALP) and osteocalcin (OCN) mRNA levels were measured at days 14 and 21, respectively, and mineralization at day 21. cAMP concentrations were measured in the presence of a phosphodiesterase inhibitor. PTH did not stimulate differentiation in cultures from WT mice but significantly increased ALP and OCN mRNA expression 6- to 7-fold in KO MSC cultures and 2- to 4-fold in KO COB cultures. PTH also increased mineralization in both KO MSC and COB cultures. Effects in KO cells were mimicked in WT MSC cultures treated with NS-398, an inhibitor of COX-2 activity. PTH increased cAMP concentrations similarly in WT and KO COBs. Differential gene responses to PTH in COX-2 KO COBs relative to WT COBs included greater fold-increases in the cAMP-mediated early response genes, c-fos and Nr4a2; increased IGF-1 mRNA expression; and decreased mRNA expression of MAP kinase phosphatase-1. PTH inhibited SOST mRNA expression 91% in COX-2 KO MSC cultures compared to 67% in WT cultures. We conclude that endogenous PGs inhibit the anabolic responses to PTH in vitro, possibly by desensitizing cAMP pathways.

Collaboration


Dive into the Shilpa Choudhary's collaboration.

Top Co-Authors

Avatar

Carol C. Pilbeam

University of Connecticut Health Center

View shared research outputs
Top Co-Authors

Avatar

Lawrence G. Raisz

University of Connecticut Health Center

View shared research outputs
Top Co-Authors

Avatar

John A. Taylor

University of Connecticut Health Center

View shared research outputs
Top Co-Authors

Avatar

Olga Voznesensky

University of Connecticut Health Center

View shared research outputs
Top Co-Authors

Avatar

Poornima Hegde

University of Connecticut Health Center

View shared research outputs
Top Co-Authors

Avatar

Cynthia B. Alander

University of Connecticut Health Center

View shared research outputs
Top Co-Authors

Avatar

Dharamainder Choudhary

University of Connecticut Health Center

View shared research outputs
Top Co-Authors

Avatar

Douglas J. Adams

University of Connecticut Health Center

View shared research outputs
Top Co-Authors

Avatar

James Pruitt

West Chester University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Joseph A. Lorenzo

University of Connecticut Health Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge