Network


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

Hotspot


Dive into the research topics where Ayesha Siddiq is active.

Publication


Featured researches published by Ayesha Siddiq.


Hepatology | 2012

Astrocyte elevated gene-1 promotes hepatocarcinogenesis: Novel insights from a mouse model†‡

Jyoti Srivastava; Ayesha Siddiq; Luni Emdad; Prasanna K. Santhekadur; Dong Chen; Rachel Gredler; Xue-Ning Shen; Chadia L. Robertson; Catherine I. Dumur; Phillip B. Hylemon; Nitai D. Mukhopadhyay; Deepak Bhere; Khalid Shah; Rushdy Ahmad; Shah Giashuddin; Jillian E. Stafflinger; Mark A. Subler; Jolene J. Windle; Paul B. Fisher; Devanand Sarkar

Astrocyte elevated gene‐1 (AEG‐1) is a key contributor to hepatocellular carcinoma (HCC) development and progression. To enhance our understanding of the role of AEG‐1 in hepatocarcinogenesis, a transgenic mouse with hepatocyte‐specific expression of AEG‐1 (Alb/AEG1) was developed. Treating Alb/AEG‐1, but not wild‐type (WT) mice, with N‐nitrosodiethylamine resulted in multinodular HCC with steatotic features and associated modulation of expression of genes regulating invasion, metastasis, angiogenesis, and fatty acid synthesis. Hepatocytes isolated from Alb/AEG‐1 mice displayed profound resistance to chemotherapeutics and growth factor deprivation with activation of prosurvival signaling pathways. Alb/AEG‐1 hepatocytes also exhibited marked resistance toward senescence, which correlated with abrogation of activation of a DNA damage response. Conditioned media from Alb/AEG‐1 hepatocytes induced marked angiogenesis with elevation in several coagulation factors. Among these factors, AEG‐1 facilitated the association of factor XII (FXII) messenger RNA with polysomes, resulting in increased translation. Short interfering RNA–mediated knockdown of FXII resulted in profound inhibition of AEG‐1‐induced angiogenesis. Conclusion: We uncovered novel aspects of AEG‐1 functions, including induction of steatosis, inhibition of senescence, and activation of the coagulation pathway to augment aggressive hepatocarcinogenesis. The Alb/AEG‐1 mouse provides an appropriate model to scrutinize the molecular mechanism of hepatocarcinogenesis and to evaluate the efficacy of novel therapeutic strategies targeting HCC. (HEPATOLOGY 2012;56:1782–1791)


Molecular Therapy | 2013

Insulin-like growth factor-binding protein-7 (IGFBP7): a promising gene therapeutic for hepatocellular carcinoma (HCC).

Dong Chen; Ayesha Siddiq; Luni Emdad; Devaraja Rajasekaran; Rachel Gredler; Xue-Ning Shen; Prasanna K. Santhekadur; Jyoti Srivastava; Chadia L. Robertson; Igor Dmitriev; Elena A. Kashentseva; David T. Curiel; Paul B. Fisher; Devanand Sarkar

Hepatocellular carcinoma (HCC) is a highly fatal disease mandating development of novel, targeted therapies to elicit prolonged survival benefit to the patients. Insulin-like growth factor-binding protein-7 (IGFBP7), a secreted protein belonging to the IGFBP family, functions as a potential tumor suppressor for HCC. In the present study, we evaluated the therapeutic efficacy of a replication-incompetent adenovirus expressing IGFBP7 (Ad.IGFBP7) in human HCC. Ad.IGFBP7 profoundly inhibited viability and induced apoptosis in multiple human HCC cell lines by inducing reactive oxygen species (ROS) and activating a DNA damage response (DDR) and p38 MAPK. In orthotopic xenograft models of human HCC in athymic nude mice, intravenous administration of Ad.IGFBP7 profoundly inhibited primary tumor growth and intrahepatic metastasis. In a nude mice subcutaneous model, xenografts from human HCC cells were established in both flanks and only left-sided tumors received intratumoral injection of Ad.IGFBP7. Growth of both left-sided injected tumors and right-sided uninjected tumors were markedly inhibited by Ad.IGFBP7 with profound suppression of angiogenesis. These findings indicate that Ad.IGFBP7 might be a potent therapeutic eradicating both primary HCC and distant metastasis and might be an effective treatment option for terminal HCC patients.Hepatocellular carcinoma (HCC) is a highly fatal disease mandating development of novel, targeted therapies to elicit prolonged survival benefit to the patients. Insulin-like growth factor-binding protein-7 (IGFBP7), a secreted protein belonging to the IGFBP family, functions as a potential tumor suppressor for HCC. In the present study, we evaluated the therapeutic efficacy of a replication-incompetent adenovirus expressing IGFBP7 (Ad.IGFBP7) in human HCC. Ad.IGFBP7 profoundly inhibited viability and induced apoptosis in multiple human HCC cell lines by inducing reactive oxygen species (ROS) and activating a DNA damage response (DDR) and p38 MAPK. In orthotopic xenograft models of human HCC in athymic nude mice, intravenous administration of Ad.IGFBP7 profoundly inhibited primary tumor growth and intrahepatic metastasis. In a nude mice subcutaneous model, xenografts from human HCC cells were established in both flanks and only left-sided tumors received intratumoral injection of Ad.IGFBP7. Growth of both left-sided injected tumors and right-sided uninjected tumors were markedly inhibited by Ad.IGFBP7 with profound suppression of angiogenesis. These findings indicate that Ad.IGFBP7 might be a potent therapeutic eradicating both primary HCC and distant metastasis and might be an effective treatment option for terminal HCC patients.


Cancer Research | 2014

AEG-1 Regulates Retinoid X Receptor and Inhibits Retinoid Signaling

Jyoti Srivastava; Chadia L. Robertson; Devaraja Rajasekaran; Rachel Gredler; Ayesha Siddiq; Luni Emdad; Nitai D. Mukhopadhyay; Shobha Ghosh; Phillip B. Hylemon; Gregorio Gil; Khalid Shah; Deepak Bhere; Mark A. Subler; Jolene J. Windle; Paul B. Fisher; Devanand Sarkar

Retinoid X receptor (RXR) regulates key cellular responses such as cell growth and development, and this regulation is frequently perturbed in various malignancies, including hepatocellular carcinoma (HCC). However, the molecule(s) that physically govern this deregulation are mostly unknown. Here, we identified RXR as an interacting partner of astrocyte-elevated gene-1 (AEG-1)/metadherin (MTDH), an oncogene upregulated in all cancers. Upon interaction, AEG-1 profoundly inhibited RXR/retinoic acid receptor (RAR)-mediated transcriptional activation. Consequently, AEG-1 markedly protected HCC and acute myelogenous leukemia (AML) cells from retinoid- and rexinoid-induced cell death. In nontumorigenic cells and primary hepatocytes, AEG-1/RXR colocalizes in the nucleus in which AEG-1 interferes with recruitment of transcriptional coactivators to RXR, preventing transcription of target genes. In tumor cells and AEG-1 transgenic hepatocytes, overexpressed AEG-1 entraps RXR in cytoplasm, precluding its nuclear translocation. In addition, ERK, activated by AEG-1, phosphorylates RXR that leads to its functional inactivation and attenuation of ligand-dependent transactivation. In nude mice models, combination of all-trans retinoic acid (ATRA) and AEG-1 knockdown synergistically inhibited growth of human HCC xenografts. The present study establishes AEG-1 as a novel homeostatic regulator of RXR and RXR/RAR that might contribute to hepatocarcinogenesis. Targeting AEG-1 could sensitize patients with HCC and AML to retinoid- and rexinoid-based therapeutics.


Cancer Research | 2014

Genetic deletion of AEG-1 prevents hepatocarcinogenesis.

Chadia L. Robertson; Jyoti Srivastava; Ayesha Siddiq; Rachel Gredler; Luni Emdad; Devaraja Rajasekaran; Maaged Akiel; Xue-Ning Shen; Chunqing Guo; Shah Giashuddin; Xiang-Yang Wang; Shobha Ghosh; Mark A. Subler; Jolene J. Windle; Paul B. Fisher; Devanand Sarkar

Activation of the oncogene AEG-1 (MTDH, LYRIC) has been implicated recently in the development of hepatocellular carcinoma (HCC). In mice, HCC can be initiated by exposure to the carcinogen DEN, which has been shown to rely upon activation of NF-κB in liver macrophages. Because AEG-1 is an essential component of NF-κB activation, we interrogated the susceptibility of mice lacking the AEG-1 gene to DEN-induced hepatocarcinogenesis. AEG-1-deficient mice displayed resistance to DEN-induced HCC and lung metastasis. No difference was observed in the response to growth factor signaling or activation of AKT, ERK, and β-catenin, compared with wild-type control animals. However, AEG-1-deficient hepatocytes and macrophages exhibited a relative defect in NF-κB activation. Mechanistic investigations showed that IL6 production and STAT3 activation, two key mediators of HCC development, were also deficient along with other biologic and epigenetics findings in the tumor microenvironment, confirming that AEG-1 supports an NF-κB-mediated inflammatory state that drives HCC development. Overall, our findings offer in vivo proofs that AEG-1 is essential for NF-κB activation and hepatocarcinogenesis, and they reveal new roles for AEG-1 in shaping the tumor microenvironment for HCC development.


Journal of Biological Chemistry | 2012

Late SV40 factor (LSF) enhances angiogenesis by transcriptionally up-regulating matrix metalloproteinase-9 (MMP-9).

Prasanna K. Santhekadur; Rachel Gredler; Dong Chen; Ayesha Siddiq; Xue-Ning Shen; Swadesh K. Das; Luni Emdad; Paul B. Fisher; Devanand Sarkar

Background: The transcription factor Late SV40 Factor (LSF) is overexpressed in human hepatocellular carcinoma (HCC). Results: LSF augments tumor angiogenesis by transcriptionally up-regulating matrix metalloproteinase-9 (MMP-9). Conclusion: A novel target of LSF is identified contributing to its oncogenic properties. Significance: LSF regulates a network of proteins, including osteopontin, MMP-9, and c-Met, thereby establishing the rationale for LSF inhibition as a potential therapeutic strategy for HCC. The transcription factor late SV40 factor (LSF) is overexpressed in human hepatocellular carcinoma (HCC) fostering a highly aggressive and metastatic phenotype. Angiogenesis is an essential component of cancer aggression and metastasis and HCC is a highly aggressive and angiogenic cancer. In the present studies, we analyzed the molecular mechanism of LSF-induced angiogenesis in HCC. Employing human umbilical vein endothelial cells (HUVEC) differentiation assay and chicken chorioallantoic membrane (CAM) assay we document that stable LSF overexpression augments and stable dominant negative inhibition of LSF (LSFdn) abrogates angiogenesis by human HCC cells. A quest for LSF-regulated factors contributing to angiogenesis, by chromatin immunoprecipitation-on-chip (ChIP-on-chip) assay, identified matrix metalloproteinase-9 (MMP-9) as a direct target of LSF. MMP-9 expression and enzymatic activity were higher in LSF-overexpressing cells and lower in LSFdn-expressing cells. Deletion mutation analysis identified the LSF-responsive regions in the MMP-9 promoter and ChIP assay confirmed LSF binding to the MMP-9 promoter. Inhibition of MMP-9 significantly abrogated LSF-induced angiogenesis as well as in vivo tumorigenesis, thus reinforcing the role of MMP-9 in facilitating LSF function. The present findings identify a novel target of LSF contributing to its oncogenic properties.


Hepatology | 2015

Astrocyte elevated gene-1 and c-Myc cooperate to promote hepatocarcinogenesis in mice

Jyoti Srivastava; Ayesha Siddiq; Rachel Gredler; Xue Ning Shen; Devaraja Rajasekaran; Chadia L. Robertson; Mark A. Subler; Jolene J. Windle; Catherine I. Dumur; Nitai D. Mukhopadhyay; Dawn Garcia; Zhao Lai; Yidong Chen; Uthra Balaji; Paul B. Fisher; Devanand Sarkar

Astrocyte elevated gene‐1 (AEG‐1) and c‐Myc are overexpressed in human hepatocellular carcinoma (HCC) functioning as oncogenes. AEG‐1 is transcriptionally regulated by c‐Myc, and AEG‐1 itself induces c‐Myc by activating the Wnt/β‐catenin–signaling pathway. We now document the cooperation of AEG‐1 and c‐Myc in promoting hepatocarcinogenesis by analyzing hepatocyte‐specific transgenic mice expressing either AEG‐1 (albumin [Alb]/AEG‐1), c‐Myc (Alb/c‐Myc), or both (Alb/AEG‐1/c‐Myc). Wild‐type and Alb/AEG‐1 mice did not develop spontaneous HCC. Alb/c‐Myc mice developed spontaneous HCC without distant metastasis, whereas Alb/AEG‐1/c‐Myc mice developed highly aggressive HCC with frank metastasis to the lungs. Induction of carcinogenesis by N‐nitrosodiethylamine significantly accelerated the kinetics of tumor formation in all groups. However, in Alb/AEG‐1/c‐Myc, the effect was markedly pronounced with lung metastasis. In vitro analysis showed that Alb/AEG‐1/c‐Myc hepatocytes acquired increased proliferation and transformative potential with sustained activation of prosurvival and epithelial‐mesenchymal transition–signaling pathways. RNA‐sequencing analysis identified a unique gene signature in livers of Alb/AEG‐1/c‐Myc mice that was not observed when either AEG‐1 or c‐Myc was overexpressed. Specifically, Alb/AEG‐1/c‐Myc mice overexpressed maternally imprinted noncoding RNAs (ncRNAs), such as Rian, Meg‐3, and Mirg, which are implicated in hepatocarcinogenesis. Knocking down these ncRNAs significantly inhibited proliferation and invasion by Alb/AEG‐1/c‐Myc hepatocytes. Conclusion: Our studies reveal a novel cooperative oncogenic effect of AEG‐1 and c‐Myc that might explain the mechanism of aggressive HCC. Alb/AEG‐1/c‐Myc mice provide a useful model to understand the molecular mechanism of cooperation between these two oncogenes and other molecules involved in hepatocarcinogenesis. This model might also be of use for evaluating novel therapeutic strategies targeting HCC. (Hepatology 2015;61:915–929)


Proceedings of the National Academy of Sciences of the United States of America | 2012

Antiproliferative small-molecule inhibitors of transcription factor LSF reveal oncogene addiction to LSF in hepatocellular carcinoma

Trevor Grant; Joshua A. Bishop; Lisa M. Christadore; Girish Barot; Hang Gyeong Chin; Sarah Woodson; John Kavouris; Ayesha Siddiq; Rachel Gredler; Xue-Ning Shen; Jennifer Sherman; Tracy L. Meehan; Kevin Fitzgerald; Sriharsa Pradhan; Laura Briggs; William H. Andrews; Devanand Sarkar; Scott E. Schaus; Ulla Hansen

Hepatocellular carcinoma (HCC) is the fifth most common cancer worldwide. Despite the prevalence of HCC, there is no effective, systemic treatment. The transcription factor LSF is a promising protein target for chemotherapy; it is highly expressed in HCC patient samples and cell lines, and promotes oncogenesis in rodent xenograft models of HCC. Here, we identify small molecules that effectively inhibit LSF cellular activity. The lead compound, factor quinolinone inhibitor 1 (FQI1), inhibits LSF DNA-binding activity both in vitro, as determined by electrophoretic mobility shift assays, and in cells, as determined by ChIP. Consistent with such inhibition, FQI1 eliminates transcriptional stimulation of LSF-dependent reporter constructs. FQI1 also exhibits antiproliferative activity in multiple cell lines. In LSF-overexpressing cells, including HCC cells, cell death is rapidly induced; however, primary or immortalized hepatocytes are unaffected by treatment with FQI1. The highly concordant structure–activity relationship of a panel of 23 quinolinones strongly suggests that the growth inhibitory activity is due to a single biological target or family. Coupled with the striking agreement between the concentrations required for antiproliferative activity (GI50s) and for inhibition of LSF transactivation (IC50s), we conclude that LSF is the specific biological target of FQIs. Based on these in vitro results, we tested the efficacy of FQI1 in inhibiting HCC tumor growth in a mouse xenograft model. As a single agent, tumor growth was dramatically inhibited with no observable general tissue cytotoxicity. These findings support the further development of LSF inhibitors for cancer chemotherapy.


Bioconjugate Chemistry | 2015

Combination of Nanoparticle-Delivered siRNA for Astrocyte Elevated Gene-1 (AEG-1) and All-trans Retinoic Acid (ATRA): An Effective Therapeutic Strategy for Hepatocellular Carcinoma (HCC)

Devaraja Rajasekaran; Jyoti Srivastava; Kareem Ebeid; Rachel Gredler; Maaged Akiel; Nidhi Jariwala; Chadia L. Robertson; Xue-Ning Shen; Ayesha Siddiq; Paul B. Fisher; Aliasger K. Salem; Devanand Sarkar

Hepatocellular carcinoma (HCC) is a fatal cancer with no effective therapy. Astrocyte elevated gene-1 (AEG-1) plays a pivotal role in hepatocarcinogenesis and inhibits retinoic acid-induced gene expression and cell death. The combination of a lentivirus expressing AEG-1 shRNA and all-trans retinoic acid (ATRA) profoundly and synergistically inhibited subcutaneous human HCC xenografts in nude mice. We have now developed liver-targeted nanoplexes by conjugating poly(amidoamine) (PAMAM) dendrimers with polyethylene glycol (PEG) and lactobionic acid (Gal) (PAMAM-PEG-Gal) which were complexed with AEG-1 siRNA (PAMAM-AEG-1si). The polymer conjugate was characterized by (1)H-NMR, MALDI, and mass spectrometry; and optimal nanoplex formulations were characterized for surface charge, size, and morphology. Orthotopic xenografts of human HCC cell QGY-7703 expressing luciferase (QGY-luc) were established in the livers of athymic nude mice and tumor development was monitored by bioluminescence imaging (BLI). Tumor-bearing mice were treated with PAMAM-siCon, PAMAM-siCon+ATRA, PAMAM-AEG-1si, and PAMAM-AEG-1si+ATRA. In the control group the tumor developed aggressively. ATRA showed little effect due to high AEG-1 levels in QGY-luc cells. PAMAM-AEG-1si showed significant reduction in tumor growth, and the combination of PAMAM-AEG-1si+ATRA showed profound and synergistic inhibition so that the tumors were almost undetectable by BLI. A marked decrease in AEG-1 level was observed in tumor samples treated with PAMAM-AEG-1si. The group treated with PAMAM-AEG-1si+ATRA nanoplexes showed increased necrosis, inhibition of proliferation, and increased apoptosis when compared to other groups. Liver is an ideal organ for RNAi therapy and ATRA is an approved anticancer agent. Our exciting observations suggest that the combinatorial approach might be an effective way to combat HCC.


Journal of Biological Chemistry | 2015

Astrocyte elevated gene-1 (AEG-1) regulates lipid homeostasis

Chadia L. Robertson; Jyoti Srivastava; Ayesha Siddiq; Rachel Gredler; Luni Emdad; Devaraja Rajasekaran; Maaged Akiel; Xue-Ning Shen; Frank Corwin; Gobalakrishnan Sundaresan; Jamal Zweit; Colleen M. Croniger; Xiaoli Gao; Shobha Ghosh; Philip B. Hylemon; Mark A. Subler; Jolene J. Windle; Paul B. Fisher; Devanand Sarkar

Background: The physiological function of the oncogene astrocyte elevated gene-1 (AEG-1) was analyzed using a knock-out mouse (AEG-1KO). Results: The AEG-1KO mouse shows a lean phenotype, which may be due to decreased intestinal fat absorption because of hyperactivation of LXR and PPARα. Conclusion: A novel role of AEG-1 is identified, regulating lipid metabolism. Significance: AEG-1 may play a role in regulating obesity and its associated disorders. Astrocyte elevated gene-1 (AEG-1), also known as MTDH (metadherin) or LYRIC, is an established oncogene. However, the physiological function of AEG-1 is not known. To address this question, we generated an AEG-1 knock-out mouse (AEG-1KO) and characterized it. Although AEG-1KO mice were viable and fertile, they were significantly leaner with prominently less body fat and lived significantly longer compared with wild type (WT). When fed a high fat and cholesterol diet (HFD), WT mice rapidly gained weight, whereas AEG-1KO mice did not gain weight at all. This phenotype of AEG-1KO mice is due to decreased fat absorption from the intestines, not because of decreased fat synthesis or increased fat consumption. AEG-1 interacts with retinoid X receptor (RXR) and inhibits RXR function. In enterocytes of AEG-1KO mice, we observed increased activity of RXR heterodimer partners, liver X receptor and peroxisome proliferator-activated receptor-α, key inhibitors of intestinal fat absorption. Inhibition of fat absorption in AEG-1KO mice was further augmented when fed an HFD providing ligands to liver X receptor and peroxisome proliferator-activated receptor-α. Our studies reveal a novel role of AEG-1 in regulating nuclear receptors controlling lipid metabolism. AEG-1 may significantly modulate the effects of HFD and thereby function as a unique determinant of obesity.


Oncotarget | 2015

Small molecule inhibitors of Late SV40 Factor (LSF) abrogate hepatocellular carcinoma (HCC): Evaluation using an endogenous HCC model.

Devaraja Rajasekaran; Ayesha Siddiq; Jennifer L.S. Willoughby; Jessica M. Biagi; Lisa M. Christadore; Sarah A. Yunes; Rachel Gredler; Nidhi Jariwala; Chadia L. Robertson; Maaged Akiel; Xue-Ning Shen; Mark A. Subler; Jolene J. Windle; Scott E. Schaus; Paul B. Fisher; Ulla Hansen; Devanand Sarkar

Hepatocellular carcinoma (HCC) is a lethal malignancy with high mortality and poor prognosis. Oncogenic transcription factor Late SV40 Factor (LSF) plays an important role in promoting HCC. A small molecule inhibitor of LSF, Factor Quinolinone Inhibitor 1 (FQI1), significantly inhibited human HCC xenografts in nude mice without harming normal cells. Here we evaluated the efficacy of FQI1 and another inhibitor, FQI2, in inhibiting endogenous hepatocarcinogenesis. HCC was induced in a transgenic mouse with hepatocyte-specific overexpression of c-myc (Alb/c-myc) by injecting N-nitrosodiethylamine (DEN) followed by FQI1 or FQI2 treatment after tumor development. LSF inhibitors markedly decreased tumor burden in Alb/c-myc mice with a corresponding decrease in proliferation and angiogenesis. Interestingly, in vitro treatment of human HCC cells with LSF inhibitors resulted in mitotic arrest with an accompanying increase in CyclinB1. Inhibition of CyclinB1 induction by Cycloheximide or CDK1 activity by Roscovitine significantly prevented FQI-induced mitotic arrest. A significant induction of apoptosis was also observed upon treatment with FQI. These effects of LSF inhibition, mitotic arrest and induction of apoptosis by FQI1s provide multiple avenues by which these inhibitors eliminate HCC cells. LSF inhibitors might be highly potent and effective therapeutics for HCC either alone or in combination with currently existing therapies.

Collaboration


Dive into the Ayesha Siddiq's collaboration.

Top Co-Authors

Avatar

Devanand Sarkar

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Paul B. Fisher

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Rachel Gredler

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Chadia L. Robertson

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Devaraja Rajasekaran

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Jyoti Srivastava

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Maaged Akiel

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Xue-Ning Shen

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Jolene J. Windle

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Mark A. Subler

Virginia Commonwealth University

View shared research outputs
Researchain Logo
Decentralizing Knowledge