Network


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

Hotspot


Dive into the research topics where Viqar Syed is active.

Publication


Featured researches published by Viqar Syed.


Oncogene | 2005

Identification of ATF-3, caveolin-1, DLC-1, and NM23-H2 as putative antitumorigenic, progesterone-regulated genes for ovarian cancer cells by gene profiling

Viqar Syed; Kasturi Mukherjee; James Lyons-Weiler; Kin Mang Lau; Tetsuo Mashima; Takashi Tsuruo; Shuk-Mei Ho

Although progesterone (P4) has been implicated to offer protection against ovarian cancer (OCa), little is known of its mechanism of action. The goal of this study was to identify P4-regulated genes that have anti-OCa action. Three immortalized nontumorigenic human ovarian surface epithelial (HOSE) cell lines and three OCa (OVCA) cell lines were subjected to 5 days of P4 treatment. Transcriptional profiling with a cDNA microarray containing ≈2400 known genes was used to identify genes (1) whose expression was consistently downregulated in OVCA cell lines compared to HOSE cell lines, and (2) whose expression was restored in OCa cell lines by P4 treatment. From the candidates selected, activating transcription factor-3 (ATF-3), caveolin-1, deleted in liver cancer-1 (DLC-1), and nonmetastatic clone 23 (NM23-H2) were chosen for post hoc functional studies based on their previously reported action as tumor suppressors or apoptosis inducers. Semiquantitative RT–PCR analyses confirmed loss of or reduced transcription of these genes in OVCA cells when compared to HOSE cells and their upregulation following P4 treatment. Hormonal specificity was demonstrated by blockade experiments with a progestin antagonist RU 38486. Ectopic expression of caveolin-1, DLC-1, and NM23-H2 caused growth inhibition in OVCA cell cultures, but not in HOSE cell cultures, while forced expression of ATF-3 suppressed growth in both. Overexpression of AFT-3 also enhanced caspase-3 activity in both HOSE and OVCA cells, whereas ectopic expression of caveolin-1 and DLC-1 only activated this enzyme in OCa cells. In contrast, NM23-H2 overexpression was ineffective in activating caspase-3. Overexpression of any of the four genes in OCa cells reduced soft-agar colony formation and cell invasiveness. Taken together, we have identified four new P4-regulated, antitumor genes for OCa. However, their modes of action differ significantly; ATF-3 primarily functions as an apoptosis inducer, NM23-H2 as a suppressor of cell motility, and caveolin-1 and DLC-1 exhibiting features of classical tumor suppressors. To the best of our knowledge, except for NM23-H2, this is the first report linking P4 to the regulation of these tumor suppressor/proapoptotic genes, which could serve as future therapeutic targets.


Journal of Cellular Biochemistry | 2010

Curcumin suppresses constitutive activation of STAT‐3 by up‐regulating protein inhibitor of activated STAT‐3 (PIAS‐3) in ovarian and endometrial cancer cells

Manush Saydmohammed; D. Joseph; Viqar Syed

Signal transducer and activator of transcription‐3 (STAT‐3) is constitutively activated in ovarian and endometrial cancers and is implicated in uncontrolled cell growth. Thus, its disruption could be an effective approach to control tumorigenesis. Curcumin is a dihydroxyphenolic compound, with proven anti‐cancer efficacy in various cancer models. We examined the anti‐tumor mechanism of curcumin on STAT‐3 and on the negative regulators of STAT‐3, including suppressors of cytokine signaling proteins (SOCS‐1 and SOCS‐3), protein inhibitors of activated STAT (PIAS‐1 and PIAS‐3), and SH2 domain‐containing phosphatases (SHP‐1 and SHP‐2) in ovarian and endometrial cancer cell lines. Treatment of cancer cells with curcumin induced a dose‐ and time‐dependent decrease of constitutive IL‐6 expression and of constitutive and IL‐6‐induced STAT‐3 phosphorylation, which is associated with decreased cell viability and increased cleavage of caspase‐3. The inhibition of STAT‐3 activation by curcumin was reversible, and phosphorylated STAT‐3 levels returned to control levels 24 h after curcumin removal. Compared to normal cells baseline expression of SOCS‐3 was high in cancer cells and a marked decrease in SOCS‐3 expression was seen following curcumin treatment. Overexpression of SOCS‐3 in curcumin‐treated cells increased expression of phosphorylated STAT‐3 and resulted in increased cell viability. Normal ovarian and endometrial cells exhibited high expression of PIAS‐3 protein, whereas in cancer cells the expression was greatly reduced. Curcumin increased PIAS‐3 expression in cancer cells. Of significance, siRNA‐mediated knockdown of PIAS‐3 overcomes the inhibitory effect of curcumin on STAT‐3 phosphorylation and cell viability. In conclusion, curcumin suppresses JAK‐STAT signaling via activation of PIAS‐3, thus attenuating STAT‐3 phosphorylation and tumor cell growth. J. Cell. Biochem. 110: 447–456, 2010.


Molecular Cancer Research | 2011

Progesterone and 1,25-Dihydroxyvitamin D3 Inhibit Endometrial Cancer Cell Growth by Upregulating Semaphorin 3B and Semaphorin 3F

Huyen Nguyen; Vessela S. Ivanova; Leyla Kavandi; Gustavo C. Rodriguez; George Larry Maxwell; Viqar Syed

Class 3 semaphorins (SEMA), SEMA3B and SEMA3F, are secreted proteins that regulate angiogenesis, tumor growth, and metastasis by binding to their transmembrane receptor complex consisting of plexins and neuropilins (NP). Expression of SEMAs and their receptors was assessed in tissue microarrays by immunohistochemistry. SEMA3B, SEMA3F, and plexin A3 were expressed strongly in normal endometrial tissues, whereas grade-dependent decreases were found in endometrial carcinomas. No change was observed in the expression of plexin A1, NP1, and NP2 in normal versus endometrial cancer tissues. Endometrial cancer cells showed decreased expression of SEMA3B, SEMA3F, and plexin A3 compared with their normal counterparts. Treatment of cancer cells with progesterone (P4) and 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] for a period of 72 hours induced a significant upregulation of SEMA3B and SEMA3F as well as inhibited growth of cancer cells by increasing caspase-3 activity. Cotreatment of cell lines with P4 or 1,25(OH)2D3 and their respective antagonists confirmed the specificity of their actions. Transfection of siRNA-targeting SEMA3B and SEMA3F in endometrial cancer cells attenuated P4 or 1,25(OH)2D3-induced growth inhibition. Restoration of SEMA3B or SEMA3F expression in cancer cells caused growth inhibition, reduced soft agar colony formation, and cell invasiveness by inhibiting expression of matrix metalloproteinase-2 (MMP-2), MMP-9, integrin αvβ3, and proangiogenic genes and by upregulating antiangiogenic genes. Thus, we have identified two new P4 and 1,25(OH)2D3-regulated antitumor genes for endometrial cancer. These results suggest that the loss of SEMAs contribute to the malignant phenotype of endometrial cancer cells and that reexpression of SEMAs by ectopic expression or with anticancer agents P4 or 1,25(OH)2D3 can be a promising therapeutic treatment against endometrial cancer. Mol Cancer Res; 9(11); 1479–92. ©2011 AACR.


Molecular Cancer Therapeutics | 2010

Hormonal Regulation and Distinct Functions of Semaphorin-3B and Semaphorin-3F in Ovarian Cancer

D. Joseph; Shuk-Mei Ho; Viqar Syed

Semaphorins comprise a family of molecules that influence neuronal growth and guidance. Class-3 semaphorins, semaphorin-3B (SEMA3B) and semaphorin-3F (SEMA3F), illustrate their effects by forming a complex with neuropilins (NP-1 or NP-2) and plexins. We examined the status and regulation of semaphorins and their receptors in human ovarian cancer cells. A significantly reduced expression of SEMA3B (83 kDa), SEMA3F (90 kDa), and plexin-A3 was observed in ovarian cancer cell lines when compared with normal human ovarian surface epithelial cells. The expression of NP-1, NP-2, and plexin-A1 was not altered in human ovarian surface epithelial and ovarian cancer cells. The decreased expression of SEMA3B, SEMA3F, and plexin-A3 was confirmed in stage 3 ovarian tumors. The treatment of ovarian cancer cells with luteinizing hormone, follicle-stimulating hormone, and estrogen induced a significant upregulation of SEMA3B, whereas SEMA3F was upregulated only by estrogen. Cotreatment of cell lines with a hormone and its specific antagonist blocked the effect of the hormone. Ectopic expression of SEMA3B or SEMA3F reduced soft-agar colony formation, adhesion, and cell invasion of ovarian cancer cell cultures. Forced expression of SEMA3B, but not SEMA3F, inhibited viability of ovarian cancer cells. Overexpression of SEMA3B and SEMA3F reduced focal adhesion kinase phosphorylation and matrix metalloproteinase-2 and matrix metalloproteinase-9 expression in ovarian cancer cells. Forced expression of SEMA3F, but not SEMA3B in ovarian cancer cells, significantly inhibited endothelial cell tube formation. Collectively, our results suggest that the loss of SEMA3 expression could be a hallmark of cancer progression. Furthermore, gonadotropin- and/or estrogen-mediated maintenance of SEMA3 expression could control ovarian cancer angiogenesis and metastasis. Mol Cancer Ther; 9(2); 499–509


Journal of Cellular Biochemistry | 2012

Progesterone and calcitriol attenuate inflammatory cytokines CXCL1 and CXCL2 in ovarian and endometrial cancer cells

Leyla Kavandi; Michael A. Collier; Huyen Nguyen; Viqar Syed

Cytokines/chemokines are key players in cancer‐related inflammation. Increasing evidence suggests that chemokines produced by tumor cells are the mediators of metastasis. Thus, agents that can downregulate chemokines expression have potential against cancer metastasis. We have previously shown inhibition of ovarian and endometrial cancer cell growth with progesterone and calcitriol. In the present study, we evaluated the effect of these two agents on the expression of inflammatory genes. Using a RT‐PCR array of inflammatory cytokines/chemokines and their receptors, we found a marked attenuation of CXCL1 and CXCL2 (GRO‐α and ‐β) in cancer cells by both treatments. Knockdown of NFκB resulted in a reduced expression of CXCL1 and CXCL2 and the inhibitory effect of progesterone and calcitriol on the expression of chemokines was abrogated in NFκB‐silenced cancer cells. Silencing of IκBα increased the expression of CXCL1 and CXCL2 in cancer cells, which can be attributed to the increased activation of NFκB‐p65, caused by the lack of its inhibitor. Progesterone and calcitriol‐induced inhibition was abolished in IκBα‐knockdown cells. Our results demonstrate that suppression of IκBα phosphorylation by progesterone and calcitriol contributes to the reduced expression of CXCL1 and CXCL2. Downregulation of CXCL1 and CXCL2 was associated with a marked inhibition of metastasis‐promoting genes. Overall, our results indicate that progesterone and calcitriol inhibit IκBα phosphorylation, NFκB activation, and the expression of NFκB regulated metastasis promoting genes. These results provide attractive data for the possible use of progesterone and calcitriol in the management of endometrial and ovarian tumors. J. Cell. Biochem. 113: 3143–3152, 2012.


Biochemical and Biophysical Research Communications | 2008

Protein kinase D1 (PKD1) influences androgen receptor (AR) function in prostate cancer cells

Paul Mak; Meena Jaggi; Viqar Syed; Subhash C. Chauhan; Sazzad Hassan; Helal Uddin Biswas; K.C. Balaji

Protein kinase D1 (PKD1), founding member of PKD protein family, is down-regulated in advanced prostate cancer (PCa). We demonstrate that PKD1 and androgen receptor (AR) are present as a protein complex in PCa cells. PKD1 is associated with a transcriptional complex which contains AR and promoter sequence of the Prostate Specific Antigen (PSA) gene. Ectopic expression of wild type PKD1 and the kinase dead mutant PKD1 (K628W) attenuated the ligand-dependent transcriptional activation of AR in prostate cancer cells and yeast cells indicating that PKD1 can affect AR transcription activity, whereas knocking down PKD1 enhanced the ligand-dependent transcriptional activation of AR. Co-expression of kinase dead mutant with AR significantly inhibited androgen-mediated cell proliferation in both LNCaP and DU145 PC cells. Our data demonstrate for the first time that PKD1 can influence AR function in PCa cells.


Cancer Prevention Research | 2013

Progesterone enhances calcitriol antitumor activity by upregulating vitamin D receptor expression and promoting apoptosis in endometrial cancer cells.

Laura R. Lee; P.N. Teng; Huyen Nguyen; Brian L. Hood; Leyla Kavandi; Guisong Wang; Jane Turbov; Larry G. Thaete; Chad A. Hamilton; George Larry Maxwell; Gustavo C. Rodriguez; Thomas P. Conrads; Viqar Syed

Human studies suggest that progesterone and calcitriol may prove beneficial in preventing or inhibiting oncogenesis, but the underlying mechanism is not fully understood. The current study investigates the effects of progesterone, calcitriol, and their combination on immortalized human endometrial epithelial cells and endometrial cancer cells and identifies their targets of action. Combination treatment with both agents enhanced vitamin D receptor expression and inhibited cell proliferation through caspase-3 activation and induction of G0–G1 cell-cycle arrest with associated downregulation of cyclins D1 and D3 and p27 induction. We used mass spectrometry–based proteomics to measure protein abundance differences between calcitriol-, progesterone-, or combination-exposed endometrial cells. A total of 117 proteins showed differential expression among these three treatments. Four proteins were then selected for validation studies: histone H1.4 (HIST1H1E), histidine triad nucleotide-binding protein 2 (HINT2), IFN-induced, double-stranded RNA-activated protein kinase (EIF2AK2), and Bcl-2–associated X protein (BAX). Abundance levels of selected candidates were low in endometrial cancer cell lines versus the immortalized endometrial epithelial cell line. All four proteins displayed elevated expression in cancer cells upon exposure to calcitriol, progesterone, or the combination. Further BAX analysis through gain- or loss-of-function experiments revealed that upregulation of BAX decreased cell proliferation by changing the BAX:BCL-2 ratio. Knockdown of BAX attenuated progesterone- and calcitriol-induced cell growth inhibition. Our results showed that progesterone and calcitriol upregulate the expression of BAX along with other apoptosis-related proteins, which induce inhibition of endometrial cancer cell growth by apoptosis and cell-cycle arrest. Cancer Prev Res; 6(7); 731–43. ©2013 AACR.


Journal of Cellular Biochemistry | 2016

TGF-β Signaling in Cancer.

Viqar Syed

The transforming growth factor‐β (TGF‐β) is a family of structurally related proteins that comprises of TGF‐β, activins/inhibins, and bone morphogenic proteins (BMPs). Members of the TGF‐β family control numerous cellular functions including proliferation, apoptosis, differentiation, epithelial‐mesenchymal transition (EMT), and migration. The first identified member, TGF‐β is implicated in several human diseases, such as vascular diseases, autoimmune disorders, and carcinogenesis. Activation of the TGF‐β receptor by its ligands induces the phosphorylation of serine/threonine residues and triggers phosphorylation of the intracellular effectors, SMADs. Upon activation, SMAD proteins translocate to the nucleus and induce transcription of their target genes, regulating several cellular functions. TGF‐β dysregulation has been implicated in carcinogenesis. In early stages of cancer, TGF‐β exhibits tumor suppressive effects by inhibiting cell cycle progression and promoting apoptosis. However, in late stages TGF‐β exerts tumor promoting effects, increasing tumor invasiveness, and metastasis. Furthermore, the TGF‐β signaling pathway communicates with other signaling pathways in a synergistic or antagonistic manner and regulates cellular functions. Elevated TGF‐β activity has been associated with poor clinical outcome. Given the pivotal role of TGF‐β in tumor progression, this pathway is an attractive target for cancer therapy. Several therapeutic tools such as TGF‐β antibodies, antisense oligonucleotides, and small molecules inhibitors of TGF‐β receptor‐1 (TGF‐βR1) have shown immense potential to inhibit TGF‐β signaling. Finally, in the interest of developing future therapies, further studies are warranted to identify novel points of convergence of TGF‐β with other signaling pathways and oncogenic factors in the tumor microenvironment. J. Cell. Biochem. 117: 1279–1287, 2016.


Gynecological Endocrinology | 2011

Progesterone inhibits growth and induces apoptosis in cancer cells through modulation of reactive oxygen species

Huyen Nguyen; Viqar Syed

Objective. Progesterone (P4) has been implicated as a protective factor for ovarian and endometrial cancers, yet little is known about its mechanism of action. We have shown apoptosis in ovarian and endometrial cancer cells with high doses of P4. Increased generation of reactive oxygen species (ROS) and an altered redox status have long been observed in cancer cells. The goal of this study was to assess the effect of P4 on cell growth, ROS generation, oxidative stress markers, and the expression of antioxidant proteins. Methods. All experiments were performed in vitro using cancer cell lines. Cell proliferation was determined using MTS proliferation assay. Production of ROS in cells was measured with the ROS indicator dye, aminophenyl fluorescein. Alterations in expression of antioxidant and apoptotic proteins were assessed by Western blotting. Results. The exposure of ovarian and endometrial cancer cell cultures to various doses of P4 caused a dose-dependent decrease in cell viability and the activation of caspase-3. Levels of ROS, markers of oxidative stress, and antioxidant proteins were elevated in cancer cells compared to normal cells and a marked decrease in their expression was seen following P4 treatment. In cancer cells, ROS was elevated while p-53 expression was low. P4 exposure of cells resulted in increased p-53 and BAX and decreased BCL-2 expression. Conclusions. The data indicates that P4 has antioxidant effects. It alleviates ROS stress and causes apoptosis by upregulating proapoptotic (p-53 and BAX) and decreasing antiapoptotic (BCL-2) gene expression in cancer cells. These findings could have potential therapeutic implications.


Cancer Prevention Research | 2014

Progesterone Inhibits Endometrial Cancer Invasiveness by Inhibiting the TGFβ Pathway

Amber A. Bokhari; Laura R. Lee; Dewayne Raboteau; Chad A. Hamilton; G.L. Maxwell; Gustavo C. Rodriguez; Viqar Syed

Increased expression of TGFβ isoforms in human endometrial cancer correlates with decreased survival and poor prognosis. Progesterone has been shown to exert a chemoprotective effect against endometrial cancer, and previous animal models have suggested that these effects are accompanied by changes in TGFβ. The goal of this study was to characterize the effect of progesterone on TGFβ signaling pathway components and on TGFβ-induced protumorigenic activities in endometrial cancer cell lines. Progesterone significantly decreased expression of three TGFβ isoforms at 72 hours after treatment except for TGFβ2 in HEC-1B and TGFβ3 in Ishikawa cells. Progesterone treatment for 120 hours attenuated expression of the three isoforms in all cell lines. Progesterone exposure for 72 hours reduced expression of TGFβ receptors in HEC-1B cells and all but TGFβR1 in Ishikawa cells. Progesterone reduced TGFβR3 expression in RL-95 cells at 72 hours, but TGFβR1 and βR2 expression levels were not affected by progesterone at any time point. SMAD2/3 and pSMAD2/3 were substantially reduced at 72 hours in all cell lines. SMAD4 expression was reduced in RL-95 cells at 24 hours and in HEC-1B and Ishikawa cells at 72 hours following progesterone treatment. Furthermore, progesterone effectively inhibited basal and TGFβ1-induced cancer cell viability and invasion, which was accompanied by increased E-cadherin and decreased vimentin expression. An inhibitor of TGFβRI blocked TGFβ1-induced effects on cell viability and invasion and attenuated antitumor effects of progesterone. These results suggest that downregulation of TGFβ signaling is a key mechanism underlying progesterone inhibition of endometrial cancer growth. Cancer Prev Res; 7(10); 1045–55. ©2014 AACR.

Collaboration


Dive into the Viqar Syed's collaboration.

Top Co-Authors

Avatar

Chad A. Hamilton

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Gustavo C. Rodriguez

NorthShore University HealthSystem

View shared research outputs
Top Co-Authors

Avatar

Amber A. Bokhari

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Huyen Nguyen

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Jane Turbov

NorthShore University HealthSystem

View shared research outputs
Top Co-Authors

Avatar

Larry G. Thaete

NorthShore University HealthSystem

View shared research outputs
Top Co-Authors

Avatar

Laura R. Lee

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Leyla Kavandi

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

George Larry Maxwell

Walter Reed Army Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge