Network


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

Hotspot


Dive into the research topics where Chadia L. Robertson is active.

Publication


Featured researches published by Chadia L. Robertson.


Journal of Biological Chemistry | 2012

Multifunction protein staphylococcal nuclease domain containing 1 (SND1) promotes tumor angiogenesis in human hepatocellular carcinoma through novel pathway that involves nuclear factor κB and miR-221.

Prasanna K. Santhekadur; Swadesh K. Das; Rachel Gredler; Dong Chen; Jyoti Srivastava; Chadia L. Robertson; Albert S. Baldwin; Paul B. Fisher; Devanand Sarkar

Background: Staphylococcal nuclease domain-containing 1 (SND1) is overexpressed in human hepatocellular carcinoma (HCC). Results: SND1 augments tumor angiogenesis by activating NF-κB, resulting in the induction of miR-221, which subsequently induces angiogenin and CXCL16. Conclusion: A novel pathway activated by SND1 is identified as contributing to tumor angiogenesis. Significance: SND1 promotes hepatocarcinogenesis by multiple ways indicating that small molecule inhibitors of SND1 might be effective therapies for HCC. Staphylococcal nuclease domain-containing 1 (SND1) is a multifunctional protein that is overexpressed in multiple cancers, including hepatocellular carcinoma (HCC). Stable overexpression of SND1 in Hep3B cells expressing a low level of SND1 augments, whereas stable knockdown of SND1 in QGY-7703 cells expressing a high level of SND1 inhibits establishment of xenografts in nude mice, indicating that SND1 promotes an aggressive tumorigenic phenotype. In this study we analyzed the role of SND1 in regulating tumor angiogenesis, a hallmark of cancer. Conditioned medium from Hep3B-SND1 cells stably overexpressing SND1 augmented, whereas that from QGY-SND1si cells stably overexpressing SND1 siRNA significantly inhibited angiogenesis, as analyzed by a chicken chorioallantoic membrane assay and a human umbilical vein endothelial cell differentiation assay. We unraveled a linear pathway in which SND1-induced activation of NF-κB resulted in induction of miR-221 and subsequent induction of angiogenic factors Angiogenin and CXCL16. Inhibition of either of these components resulted in significant inhibition of SND1-induced angiogenesis, thus highlighting the importance of this molecular cascade in regulating SND1 function. Because SND1 regulates NF-κB and miR-221, two important determinants of HCC controlling the aggressive phenotype, SND1 inhibition might be an effective strategy to counteract this fatal malady.


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.


International Journal of Oncology | 2015

Role of the staphylococcal nuclease and tudor domain containing 1 in oncogenesis (Review)

Nidhi Jariwala; Devaraja Rajasekaran; Jyoti Srivastava; Rachel Gredler; Maaged Akiel; Chadia L. Robertson; Luni Emdad; Paul B. Fisher; Devanand Sarkar

The staphylococcal nuclease and tudor domain containing 1 (SND1) is a multifunctional protein overexpressed in breast, prostate, colorectal and hepatocellular carcinomas and malignant glioma. Molecular studies have revealed the multifaceted activities of SND1 involved in regulating gene expression at transcriptional as well as post-transcriptional levels. Early studies identified SND1 as a transcriptional co-activator. SND1 is also a component of RNA-induced silencing complex (RISC) thus mediating RNAi function, a regulator of mRNA splicing, editing and stability, and plays a role in maintenance of cell viability. Such diverse actions allow the SND1 to modulate a complex array of molecular networks, thereby promoting carcinogenesis. Here, we describe the crucial role of SND1 in cancer development and progression, and highlight SND1 as a potential target for therapeutic intervention.


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)


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.


FEBS Open Bio | 2014

Staphylococcal nuclease domain containing-1 (SND1) promotes migration and invasion via angiotensin II type 1 receptor (AT1R) and TGFβ signaling

Prasanna K. Santhekadur; Maaged Akiel; Luni Emdad; Rachel Gredler; Jyoti Srivastava; Devaraja Rajasekaran; Chadia L. Robertson; Nitai D. Mukhopadhyay; Paul B. Fisher; Devanand Sarkar

Staphylococcal nuclease domain containing‐1 (SND1) is overexpressed in human hepatocellular carcinoma (HCC) patients and promotes tumorigenesis by human HCC cells. We now document that SND1 increases angiotensin II type 1 receptor (AT1R) levels by increasing AT1R mRNA stability. This results in activation of ERK, Smad2 and subsequently the TGFβ signaling pathway, promoting epithelial–mesenchymal transition (EMT) and migration and invasion by human HCC cells. A positive correlation was observed between SND1 and AT1R expression levels in human HCC patients. Small molecule inhibitors of SND1, alone or in combination with AT1R blockers, might be an effective therapeutic strategy for late‐stage aggressive HCC.


Journal of Biological Chemistry | 2016

Staphylococcal Nuclease and Tudor Domain Containing 1 (SND1 Protein) Promotes Hepatocarcinogenesis by Inhibiting Monoglyceride Lipase (MGLL).

Devaraja Rajasekaran; Nidhi Jariwala; Rachel G. Mendoza; Chadia L. Robertson; Maaged Akiel; Mikhail G. Dozmorov; Paul B. Fisher; Devanand Sarkar

Staphylococcal nuclease and tudor domain containing 1 (SND1) is overexpressed in multiple cancers, including hepatocellular carcinoma (HCC), and functions as an oncogene. This study was carried out to identify novel SND1-interacting proteins to better understand its molecular mechanism of action. SND1-interacting proteins were identified by a modified yeast two-hybrid assay. Protein-protein interaction was confirmed by co-immunoprecipitation analysis. Monoglyceride lipase (MGLL) expression was analyzed by quantitative RT-PCR, Western blot, and immunohistochemistry. MGLL-overexpressing clones were analyzed for cell proliferation and cell cycle analysis and in vivo tumorigenesis in nude mice. MGLL was identified as an SND1-interacting protein. Interaction of SND1 with MGLL resulted in ubiquitination and proteosomal degradation of MGLL. MGLL expression was detected in normal human hepatocytes and mouse liver, although it was undetected in human HCC cell lines. An inverse correlation between SND1 and MGLL levels was identified in a human HCC tissue microarray as well as in the TCGA database. Forced overexpression of MGLL in human HCC cells resulted in marked inhibition in cell proliferation with a significant delay in cell cycle progression and a marked decrease in tumor growth in nude mouse xenograft assays. MGLL overexpression inhibited Akt activation that is independent of enzymatic activity of MGLL and overexpression of a constitutively active Akt rescued cells from inhibition of proliferation and restored normal cell cycle progression. This study unravels a novel mechanism of SND1 function and identifies MGLL as a unique tumor suppressor for HCC. MGLL might function as a homeostatic regulator of Akt restraining its activation.

Collaboration


Dive into the Chadia L. Robertson'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

Devaraja Rajasekaran

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Rachel Gredler

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Jyoti Srivastava

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Ayesha Siddiq

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Maaged Akiel

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
Top Co-Authors

Avatar

Nidhi Jariwala

Virginia Commonwealth University

View shared research outputs
Researchain Logo
Decentralizing Knowledge