Network


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

Hotspot


Dive into the research topics where Shu-hao Hsu is active.

Publication


Featured researches published by Shu-hao Hsu.


Journal of Clinical Investigation | 2012

Essential metabolic, anti-inflammatory, and anti-tumorigenic functions of miR-122 in liver

Shu-hao Hsu; Bo Wang; Janaiah Kota; Jianhua Yu; Stefan Costinean; Huban Kutay; Lianbo Yu; Shoumei Bai; Krista La Perle; Raghu R. Chivukula; Hsiaoyin Mao; Min Wei; K. Reed Clark; Michael A. Caligiuri; Samson T. Jacob; Joshua T. Mendell; Kalpana Ghoshal

miR-122, an abundant liver-specific microRNA (miRNA), regulates cholesterol metabolism and promotes hepatitis C virus (HCV) replication. Reduced miR-122 expression in hepatocellular carcinoma (HCC) correlates with metastasis and poor prognosis. Nevertheless, the consequences of sustained loss of function of miR-122 in vivo have not been determined. Here, we demonstrate that deletion of mouse Mir122 resulted in hepatosteatosis, hepatitis, and the development of tumors resembling HCC. These pathologic manifestations were associated with hyperactivity of oncogenic pathways and hepatic infiltration of inflammatory cells that produce pro-tumorigenic cytokines, including IL-6 and TNF. Moreover, delivery of miR-122 to a MYC-driven mouse model of HCC strongly inhibited tumorigenesis, further supporting the tumor suppressor activity of this miRNA. These findings reveal critical functions for miR-122 in the maintenance of liver homeostasis and have important therapeutic implications, including the potential utility of miR-122 delivery for selected patients with HCC and the need for careful monitoring of patients receiving miR-122 inhibition therapy for HCV.


Journal of Biological Chemistry | 2009

MicroRNA-122 inhibits tumorigenic properties of hepatocellular carcinoma cells and sensitizes these cells to Sorafenib

Shoumei Bai; Mohd W. Nasser; Bo Wang; Shu-hao Hsu; Jharna Datta; Huban Kutay; Arti Yadav; Gerard J. Nuovo; Pawan Kumar; Kalpana Ghoshal

MicroRNAs are negative regulators of protein coding genes. The liver-specific microRNA-122 (miR-122) is frequently suppressed in primary hepatocellular carcinomas (HCCs). In situ hybridization demonstrated that miR-122 is abundantly expressed in hepatocytes but barely detectable in primary human HCCs. Ectopic expression of miR-122 in nonexpressing HepG2, Hep3B, and SK-Hep-1 cells reversed their tumorigenic properties such as growth, replication potential, clonogenic survival, anchorage-independent growth, migration, invasion, and tumor formation in nude mice. Further, miR-122-expressing HCC cells retained an epithelial phenotype that correlated with reduced Vimentin expression. ADAM10 (a distintegrin and metalloprotease family 10), serum response factor (SRF), and insulin-like growth factor 1 receptor (Igf1R) that promote tumorigenesis were validated as targets of miR-122 and were repressed by the microRNA. Conversely, depletion of the endogenous miR-122 in Huh-7 cells facilitated their tumorigenic properties with concomitant up-regulation of these targets. Expression of SRF or Igf1R partially reversed tumor suppressor function of miR-122. Further, miR-122 impeded angiogenic properties of endothelial cells in vitro. Notably, ADAM10, SRF, and Igf1R were up-regulated in primary human HCCs compared with the matching liver tissue. Co-labeling studies demonstrated exclusive localization of miR-122 in the benign livers, whereas SRF predominantly expressed in HCC. More importantly, growth and clonogenic survival of miR-122-expressing HCC cells were significantly reduced upon treatment with sorafenib, a multi-kinase inhibitor clinically effective against HCC. Collectively, these results suggest that the loss of multifunctional miR-122 contributes to the malignant phenotype of HCC cells, and miR-122 mimetic alone or in combination with anticancer drugs can be a promising therapeutic regimen against liver cancer.


Oncogene | 2010

TGFβ-mediated upregulation of hepatic miR-181b promotes hepatocarcinogenesis by targeting TIMP3

Bo Wang; Shu-hao Hsu; Sarmila Majumder; Huban Kutay; Wei Huang; Samson T. Jacob; Kalpana Ghoshal

To identify microRNAs (miRNAs) that may have a causal role in hepatocarcinogenesis, we used an animal model in which C57BL/6 mice fed choline-deficient and amino acid defined (CDAA) diet develop preneoplastic lesions at 65 weeks and hepatocellular carcinomas after 84 weeks. miRNA expression profiling showed significant upregulation of miR-181b and miR-181d in the livers of mice as early as 32 weeks that persisted at preneoplastic stage. The expression of tissue inhibitor of metalloprotease 3 (TIMP3), a tumor suppressor and a validated miR-181 target, was markedly suppressed in the livers of mice fed CDAA diet. Upregulation of hepatic transforming growth factor (TGF)β and its downstream mediators Smad 2, 3 and 4 and increase in phospho-Smad2 in the liver nuclear extract correlated with elevated miR-181b/d in mice fed CDAA diet. The levels of the precursor and mature miR-181b were augmented on exposure of hepatic cells to TGFβ and were significantly reduced by small interference RNA-mediated depletion of Smad4, showing the involvement of TGFβ signaling pathway in miR-181b expression. Ectopic expression and depletion of miR-181b showed that miR-181b enhanced matrix metallopeptidases (MMP)2 and MMP9 activity and promoted growth, clonogenic survival, migration and invasion of hepatocellular carcinoma (HCC) cells that could be reversed by modulating TIMP3 level. Further, depletion of miR-181b inhibited tumor growth of HCC cells in nude mice. miR-181b also enhanced resistance of HCC cells to the anticancer drug doxorubicin. On the basis of these results, we conclude that upregulation of miR-181b at early stages of feeding CDAA diet promotes hepatocarcinogenesis.


Molecular Cell | 2014

mRNA destabilization is the dominant effect of mammalian microRNAs by the time substantial repression ensues.

Stephen W. Eichhorn; Huili Guo; Sean Edward McGeary; Ricard A. Rodriguez-Mias; Chanseok Shin; Daehyun Baek; Shu-hao Hsu; Kalpana Ghoshal; Judit Villén; David P. Bartel

MicroRNAs (miRNAs) regulate target mRNAs through a combination of translational repression and mRNA destabilization, with mRNA destabilization dominating at steady state in the few contexts examined globally. Here, we extend the global steady-state measurements to additional mammalian contexts and find that regardless of the miRNA, cell type, growth condition, or translational state, mRNA destabilization explains most (66%->90%) miRNA-mediated repression. We also determine the relative dynamics of translational repression and mRNA destabilization for endogenous mRNAs as a miRNA is induced. Although translational repression occurs rapidly, its effect is relatively weak, such that by the time consequential repression ensues, the effect of mRNA destabilization dominates. These results imply that consequential miRNA-mediated repression is largely irreversible and provide other insights into the nature of miRNA-mediated regulation. They also simplify future studies, dramatically extending the known contexts and time points for which monitoring mRNA changes captures most of the direct miRNA effects.


Hepatology | 2012

Stat3-mediated activation of microRNA-23a suppresses gluconeogenesis in hepatocellular carcinoma by down-regulating Glucose-6-phosphatase and peroxisome proliferator-activated receptor gamma, coactivator 1 alpha†

Bo Wang; Shu-hao Hsu; Wendy L. Frankel; Kalpana Ghoshal; Samson T. Jacob

Considerable effort has been made in elucidating the mechanism and functional significance of high levels of aerobic glycolysis in cancer cells, commonly referred to as the Warburg effect. Here we investigated whether the gluconeogenic pathway is significantly modulated in hepatocarcinogenesis, resulting in altered levels of glucose homeostasis. To test this possibility, we used a mouse model (mice fed a choline‐deficient diet) that develops nonalcoholic steatohepatitis (NASH), preneoplastic nodules, and hepatocellular carcinoma (HCC), along with human primary HCCs and HCC cells. This study demonstrated marked reduction in the expressions of G6pc, Pepck, and Fbp1 encoding the key gluconeogenic enzymes glucose‐6‐phosphatase, phosphoenolpyruvate carboxykinase, fructose‐1,6‐phosphatase, respectively, and the transcription factor Pgc‐1α in HCCs developed in the mouse model that correlated with reduction in serum glucose in tumor‐bearing mice. The messenger RNA (mRNA) levels of these genes were also reduced by ≈80% in the majority of primary human HCCs compared with matching peritumoral livers. The expression of microRNA (miR)‐23a, a candidate miR targeting PGC‐1α and G6PC, was up‐regulated in the mouse liver tumors as well as in primary human HCC. We confirmed PGC‐1α and G6PC as direct targets of miR‐23a and their expressions negatively correlated with miR‐23a expression in human HCCs. G6PC expression also correlated with tumor grade in human primary HCCs. Finally, this study showed that the activation of interleukin (IL)‐6‐Stat3 signaling caused the up‐regulation of miR‐23a expression in HCC. Conclusion: Based on these data, we conclude that gluconeogenesis is severely compromised in HCC by IL6‐Stat3‐mediated activation of miR‐23a, which directly targets PGC‐1α and G6PC, leading to decreased glucose production. (HEPATOLOGY 2012;56:186–197)


Nanomedicine: Nanotechnology, Biology and Medicine | 2013

Cationic Lipid Nanoparticles for Therapeutic Delivery of siRNA and miRNA to Murine Liver Tumor

Shu-hao Hsu; Bo Yu; Xinmei Wang; Yuanzhi Lu; Carl Schmidt; Robert J. Lee; L. James Lee; Samson T. Jacob; Kalpana Ghoshal

UNLABELLED miR-122, a liver-specific tumor suppressor microRNA, is frequently down-regulated in hepatocellular carcinoma (HCC). LNP-DP1, a cationic lipid nanoparticle formulation, was developed as a vehicle to restore deregulated gene expression in HCC cells by miR-122 delivery. LNP-DP1 consists of 2-dioleyloxy-N,N-dimethyl-3-aminopropane (DODMA), egg phosphatidylcholine, cholesterol and cholesterol-polyethylene glycol. In vitro, LNP-DP1-mediated transfection of a miR-122 mimic to HCC cells down-regulated miR-122 target genes by >95%. In vivo, siRNAs/miRNAs encapsulated in LNP-DP1 were preferentially taken up by hepatocytes and tumor cells in a mouse HCC model. The miR-122 mimic in LNP-DP1 was functional in HCC cells without causing systemic toxicity. To demonstrate its therapeutic potential, LNP-DP1 encapsulating miR-122 mimic was intratumorally injected and resulted in ~50% growth suppression of HCC xenografts within 30 days, which correlated well with suppression of target genes and impairment of angiogenesis. These data demonstrate the potential of LNP-DP1-mediated microRNA delivery as a novel strategy for HCC therapy. FROM THE CLINICAL EDITOR In this study, LNP-DP1 -a cationic lipid nanoparticle formulation -is reported as a vehicle to restore deregulated gene expression in hepatic carcinoma cells by siRNA and miRNA delivery using a mouse model. Further expansions to this study may enable transition to clinical trials of this system.


Hepatology | 2014

Reciprocal regulation of microRNA‐122 and c‐Myc in hepatocellular cancer: Role of E2F1 and transcription factor dimerization partner 2

Bo Wang; Shu-hao Hsu; Xinmei Wang; Huban Kutay; Hemant Kumar Bid; Jianhua Yu; Ramesh K. Ganju; Samson T. Jacob; Mariia Yuneva; Kalpana Ghoshal

c‐Myc is a well‐known oncogene frequently up‐regulated in different malignancies, whereas liver‐specific microRNA (miR)‐122, a bona fide tumor suppressor, is down‐regulated in hepatocellular cancer (HCC). Here we explored the underlying mechanism of reciprocal regulation of these two genes. Real‐time reverse‐transcription polymerase chain reaction (RT‐PCR) and northern blot analysis demonstrated reduced expression of the primary, precursor, and mature miR‐122 in c‐MYC‐induced HCCs compared to the benign livers, indicating transcriptional suppression of miR‐122 upon MYC overexpression. Indeed, chromatin immunoprecipitation (ChIP) assay showed significantly reduced association of RNA polymerase II and histone H3K9Ac, markers of active chromatin, with the miR‐122 promoter in tumors relative to the c‐MYC‐uninduced livers, indicating transcriptional repression of miR‐122 in c‐MYC‐overexpressing tumors. The ChIP assay also demonstrated a significant increase in c‐Myc association with the miR‐122 promoter region that harbors a conserved noncanonical c‐Myc binding site in tumors compared to the livers. Ectopic expression and knockdown studies showed that c‐Myc indeed suppresses expression of primary and mature miR‐122 in hepatic cells. Additionally, Hnf‐3β, a liver enriched transcription factor that activates miR‐122 gene, was suppressed in c‐MYC‐induced tumors. Notably, miR‐122 also repressed c‐Myc transcription by targeting transcriptional activator E2f1 and coactivator Tfdp2, as evident from ectopic expression and knockdown studies and luciferase reporter assays in mouse and human hepatic cells. Conclusion: c‐Myc represses miR‐122 gene expression by associating with its promoter and by down‐regulating Hnf‐3β expression, whereas miR‐122 indirectly inhibits c‐Myc transcription by targeting Tfdp2 and E2f1. In essence, these results suggest a double‐negative feedback loop between a tumor suppressor (miR‐122) and an oncogene (c‐Myc). (Hepatology 2014;59:555–566)


Hepatology | 2016

MicroRNA‐122 Regulates Polyploidization in the Murine Liver

Shu-hao Hsu; Evan Delgado; P. Anthony Otero; Kun-yu Teng; Huban Kutay; Kolin Meehan; Justin B. Moroney; Jappmann K. Monga; Nicholas J. Hand; Joshua R. Friedman; Kalpana Ghoshal; Andrew W. Duncan

A defining feature of the mammalian liver is polyploidy, a numerical change in the entire complement of chromosomes. The first step of polyploidization involves cell division with failed cytokinesis. Although polyploidy is common, affecting ∼90% of hepatocytes in mice and 50% in humans, the specialized role played by polyploid cells in liver homeostasis and disease remains poorly understood. The goal of this study was to identify novel signals that regulate polyploidization, and we focused on microRNAs (miRNAs). First, to test whether miRNAs could regulate hepatic polyploidy, we examined livers from Dicer1 liver‐specific knockout mice, which are devoid of mature miRNAs. Loss of miRNAs resulted in a 3‐fold reduction in binucleate hepatocytes, indicating that miRNAs regulate polyploidization. Second, we surveyed age‐dependent expression of miRNAs in wild‐type mice and identified a subset of miRNAs, including miR‐122, that is differentially expressed at 2‐3 weeks, a period when extensive polyploidization occurs. Next, we examined Mir122 knockout mice and observed profound, lifelong depletion of polyploid hepatocytes, proving that miR‐122 is required for complete hepatic polyploidization. Moreover, the polyploidy defect in Mir122 knockout mice was ameliorated by adenovirus‐mediated overexpression of miR‐122, underscoring the critical role miR‐122 plays in polyploidization. Finally, we identified direct targets of miR‐122 (Cux1, Rhoa, Iqgap1, Mapre1, Nedd4l, and Slc25a34) that regulate cytokinesis. Inhibition of each target induced cytokinesis failure and promoted hepatic binucleation. Conclusion: Among the different signals that have been associated with hepatic polyploidy, miR‐122 is the first liver‐specific signal identified; our data demonstrate that miR‐122 is both necessary and sufficient in liver polyploidization, and these studies will serve as the foundation for future work investigating miR‐122 in liver maturation, homeostasis, and disease. (Hepatology 2016;64:599‐615)


Biochimica et Biophysica Acta | 2015

Indole-3-carbinol inhibits tumorigenicity of hepatocellular carcinoma cells via suppression of microRNA-21 and upregulation of phosphatase and tensin homolog

Xinmei Wang; Hongyan He; Yuanzhi Lu; Wei Ren; Kun-yu Teng; Chi-Ling Chiang; Zhaogang Yang; Bo Yu; Shu-hao Hsu; Samson T. Jacob; Kalpana Ghoshal; L. James Lee

A major obstacle to successful treatment of hepatocellular carcinoma (HCC) is its high resistance to cytotoxic chemotherapy due to overexpression of multidrug resistance genes. Activation of the AKT pathway is known to be involved in chemoresistance in HCC; however, the underlying mechanisms modulating the AKT pathway by chemopreventive agents remain unclear. In the present study, we found that indole-3-carbinol (I3C) treatment for tumor cells repressed the AKT pathway by increasing the expression of phosphatase and tensin homolog (PTEN) in HCC xenograft tumor and HCC cell lines. qRT-PCR data showed that the expression of miR-21 and miR-221&222 was significantly reduced by I3C in HCC cells in vitro and in vivo. Reactivation of the AKT pathway via restoration of miR-21 was reversed by I3C. Ectopic expression of miR-21 mediated-accelerated wound healing was abrogated by I3C. Moreover, reducing the expression of miR-21 by anti-miR decreased the resistance of HCC cells to I3C. These results provide experimental evidences that I3C could function as a miR-21 regulator, leading to repression of the PTEN/AKT pathway and opening a new avenue for eradication of drug-resistant cells, thus potentially helping to improve the therapeutic outcome in patients diagnosed with HCC.


PLOS ONE | 2011

Male germ cell-specific RNA binding protein RBMY: a new oncogene explaining male predominance in liver cancer.

Daw-Jen Tsuei; Po-Huang Lee; Hsiao-Yu Peng; Shau-Lin Lu; De‐Shiuan Su; Yung-Ming Jeng; Hey-Chi Hsu; Shu-hao Hsu; Jia-Feng Wu; Yen-Hsuan Ni; Mei-Hwei Chang

Male gender is a risk factor for the development of hepatocellular carcinoma (HCC) but the mechanisms are not fully understood. The RNA binding motif gene on the Y chromosome (RBMY), encoding a male germ cell-specific RNA splicing regulator during spermatogenesis, is aberrantly activated in human male liver cancers. This study investigated the in vitro oncogenic effect and the possible mechanism of RBMY in human hepatoma cell line HepG2 and its in vivo effect with regards to the livers of human and transgenic mice. RBMY expression in HepG2 cells was knocked down by RNA interference and the cancer cell phenotype was characterized by soft-agar colony formation and sensitivity to hydrogen-peroxide-induced apoptosis. The results revealed that RBMY knockdown reduced the transformation and anti-apoptotic efficiency of HepG2 cells. The expression of RBMY, androgen receptor (AR) and its inhibitory variant AR45, AR-targeted genes insulin-like growth factor 1 (IGF-1) and insulin-like growth factor binding protein 3 (IGFBP-3) was analyzed by quantitative RT-PCR. Up-regulation of AR45 variant and reduction of IGF-1 and IGFBP-3 expression was only detected in RBMY knockdown cells. Moreover, RBMY positive human male HCC expressed lower level of AR45 as compared to RBMY negative HCC tissues. The oncogenic properties of RBMY were further assessed in a transgenic mouse model. Liver-specific RBMY transgenic mice developed hepatic pre-cancerous lesions, adenoma, and HCC. RBMY also accelerated chemical carcinogen-induced hepatocarcinogenesis in transgenic mice. Collectively, these findings suggest that Y chromosome-specific RBMY is likely involved in the regulation of androgen receptor activity and contributes to male predominance of HCC.

Collaboration


Dive into the Shu-hao Hsu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bo Wang

Ohio State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bo Yu

Ohio State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lianbo Yu

Ohio State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge