Network


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

Hotspot


Dive into the research topics where Bo Wang is active.

Publication


Featured researches published by Bo Wang.


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.


Journal of Biological Chemistry | 2008

Down-regulation of Micro-RNA-1 (miR-1) in Lung Cancer SUPPRESSION OF TUMORIGENIC PROPERTY OF LUNG CANCER CELLS AND THEIR SENSITIZATION TO DOXORUBICIN-INDUCED APOPTOSIS BY miR-1

Mohd W. Nasser; Jharna Datta; Gerard J. Nuovo; Huban Kutay; Tasneem Motiwala; Sarmila Majumder; Bo Wang; Saul Suster; Samson T. Jacob; Kalpana Ghoshal

Micro-RNAs are ∼21–25-nucleotide-long noncoding RNAs that regulate gene expression primarily at the post-transcriptional level in animals. Here, we report that micro-RNA-1 (miR-1), abundant in the cardiac and smooth muscles, is expressed in the lung and is down-regulated in human primary lung cancer tissues and cell lines. In situ hybridization demonstrated localization of miR-1 in bronchial epithelial cells. The tumor suppressor C/EBPα, frequently suppressed in lung cancer, reactivated miR-1 expression in the lung cancer cells. Repressed miR-1 was also activated in lung cancer cells upon treatment with a histone deacetylase inhibitor. These observations led us to examine the antitumorigenic potential of miR-1 in lung cancer cells. Expression of miR-1 in nonexpressing A549 and H1299 cells reversed their tumorigenic properties, such as growth, replication potential, motility/migration, clonogenic survival, and tumor formation in nude mice. Exogenous miR-1 significantly reduced expression of oncogenic targets, such as MET, a receptor tyrosine kinase, and Pim-1, a Ser/Thr kinase, frequently up-regulated in lung cancer. Similarly, the levels of two additional targets, FoxP1, a transcription factor with oncogeneic property, and HDAC4 that represses differentiation-promoting genes, were reduced in miR-1-expressing cells. Conversely, depletion of miR-1 facilitated N417 cell growth with concomitant elevation of these targets. Further, ectopic miR-1 induced apoptosis in A549 cells in response to the potent anticancer drug doxorubicin. Enhanced activation of caspases 3 and 7, cleavage of their substrate PARP-1, and depletion of anti-apoptotic Mcl-1 contributed to the sensitivity of miR-1-expressing cells to doxorubicin. Thus, miR-1 has potential therapeutic application against lung cancers.


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.


Hepatology | 2009

Role of microRNA‐155 at early stages of hepatocarcinogenesis induced by choline‐deficient and amino acid–defined diet in C57BL/6 mice

Bo Wang; Sarmila Majumder; Gerard J. Nuovo; Huban Kutay; Stefano Volinia; Tushar Patel; Thomas D. Schmittgen; Carlo M. Croce; Kalpana Ghoshal; Samson T. Jacob

MicroRNAs (miRs) are conserved, small (20‐25 nucleotide) noncoding RNAs that negatively regulate expression of messenger RNAs (mRNAs) at the posttranscriptional level. Aberrant expression of certain microRNAs plays a causal role in tumorigenesis. Here, we report identification of hepatic microRNAs that are dysregulated at early stages of feeding C57BL/6 mice choline‐deficient and amino acid–defined (CDAA) diet that is known to promote nonalcoholic steatohepatitis (NASH)‐induced hepatocarcinogenesis after 84 weeks. Microarray analysis identified 30 hepatic microRNAs that are significantly (P ≤ 0.01) altered in mice fed CDAA diet for 6, 18, 32, and 65 weeks compared with those fed choline‐sufficient and amino acid–defined (CSAA) diet. Real‐time reverse transcription polymerase chain reaction (RT‐PCR) analysis demonstrated up‐regulation of oncogenic miR‐155, miR‐221/222, and miR‐21 and down‐regulation of the most abundant liver‐specific miR‐122 at early stages of hepatocarcinogenesis. Western blot analysis showed reduced expression of hepatic phosphatase and tensin homolog (PTEN) and CCAAT/enhancer binding protein beta (C/EBPβ), respective targets of miR‐21 and miR‐155, in these mice at early stages. DNA binding activity of nuclear factor kappa B (NF‐κB) that transactivates miR‐155 gene was significantly (P = 0.002) elevated in the liver nuclear extract of mice fed CDAA diet. Furthermore, the expression of miR‐155, as measured by in situ hybridization and real‐time RT‐PCR, correlated with diet‐induced histopathological changes in the liver. Ectopic expression of miR‐155 promoted growth of hepatocellular carcinoma (HCC) cells, whereas its depletion inhibited cell growth. Notably, miR‐155 was significantly (P = 0.0004) up‐regulated in primary human HCCs with a concomitant decrease (P = 0.02) in C/EBPβ level compared with matching liver tissues. Conclusion: Temporal changes in microRNA profile occur at early stages of CDAA diet‐induced hepatocarcinogenesis. Reciprocal regulation of specific oncomirs and their tumor suppressor targets implicate their role in NASH‐induced hepatocarcinogenesis and suggest their use in the diagnosis, prognosis, and therapy of liver cancer. (HEPATOLOGY 2009.)


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)


Journal of Controlled Release | 2013

Lactosylated Gramicidin-based lipid nanoparticles (Lac-GLN) for targeted delivery of anti-miR-155 to hepatocellular carcinoma

Mengzi Zhang; Xiaoju Zhou; Bo Wang; Bryant C. Yung; Ly James Lee; Kalpana Ghoshal; Robert J. Lee

Lactosylated gramicidin-containing lipid nanoparticles (Lac-GLN) were developed for delivery of anti-microRNA-155 (anti-miR-155) to hepatocellular carcinoma (HCC) cells. MiR-155 is an oncomiR frequently elevated in HCC. The Lac-GLN formulation contained N-lactobionyl-dioleoyl phosphatidylethanolamine (Lac-DOPE), a ligand for the asialoglycoprotein receptor (ASGR), and an antibiotic peptide gramicidin A. The nanoparticles exhibited a mean particle diameter of 73 nm, zeta potential of +3.5mV, anti-miR encapsulation efficiency of 88%, and excellent colloidal stability at 4°C. Lac-GLN effectively delivered anti-miR-155 to HCC cells with a 16.1- and 4.1-fold up-regulation of miR-155 targets C/EBPβ and FOXP3 genes, respectively, and exhibited significant greater efficiency over Lipofectamine 2000. In mice, intravenous injection of Lac-GLN containing Cy3-anti-miR-155 led to preferential accumulation of the anti-miR-155 in hepatocytes. Intravenous administration of 1.5 mg/kg anti-miR-155 loaded Lac-GLN resulted in up-regulation of C/EBPβ and FOXP3 by 6.9- and 2.2-fold, respectively. These results suggest potential application of Lac-GLN as a liver-specific delivery vehicle for anti-miR therapy.


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)


Cancer Research | 2010

Loss of metallothionein predisposes mice to diethylnitrosamine-induced hepatocarcinogenesis by activating NF-kappaB target genes.

Sarmila Majumder; Satavisha Roy; Thomas Kaffenberger; Bo Wang; Stefan Costinean; Wendy L. Frankel; Anna Bratasz; Periannan Kuppusamy; Tsonwin Hai; Kalpana Ghoshal; Samson T. Jacob

Metallothioneins (MT) are potent scavengers of free radicals that are silenced in primary hepatocellular carcinomas (HCC) of human and rodent origin. To examine whether loss of MT promotes hepatocarcinogenesis, male Mt-1 and Mt-2 double knockout (MTKO) and wild-type (WT) mice were exposed to diethylnitrosamine (DEN) and induction of HCC was monitored at 23 and 33 weeks. The size and number of liver tumors, the ratio between liver and body weight, and liver damage were markedly elevated in the MTKO mice at both time points compared with the WT mice. At 23 weeks, MTKO mice developed HCC whereas WT mice developed only preneoplastic nodules suggesting that loss of MT accelerates hepatocarcinogenesis. MTKO tumors also exhibited higher superoxide anion levels. Although NF-κB activity increased in the liver nuclear extracts of both genotypes after DEN exposure, the complex formed in MTKO mice was predominantly p50/65 heterodimer (transcriptional activator) as opposed to p50 homodimer (transcriptional repressor) in WT mice. Phosphorylation of p65 at Ser276 causing its activation was also significantly augmented in DEN-exposed MTKO livers. NF-κB targets that include early growth response genes and proinflammatory cytokines were significantly upregulated in MTKO mice. Concurrently, there was a remarkable increase (∼100-fold) in Pai-1 expression; significant increase in c-Jun, c-Fos, c-Myc, Ets2, and ATF3 expressions; and growth factor signaling that probably contributed to the increased tumor growth in MTKO mice. Taken together, these results demonstrate that MTs protect mice from hepatocarcinogen-induced liver damage and carcinogenesis, underscoring their potential therapeutic application against hepatocellular cancer.


Genome Medicine | 2011

Role of cancer stem cells in hepatocarcinogenesis

Bo Wang; Samson T. Jacob

There has been considerable interest in cancer stem cells (CSCs) among cancer biologists and clinicians, most likely because of their role in the heterogeneity of cancer and their potential application in cancer therapeutics. Recent studies suggest that CSCs play a key role in liver carcinogenesis. A small subpopulation of cancer cells with CSC properties has been identified and characterized from hepatocellular carcinoma (HCC) cell lines, animal models and human primary HCCs. Considering the high mortality and ineffectiveness of current therapies for HCC, understanding the characteristics and function of CSCs is likely to lead to development of new therapies resulting in improvement of patient survival. This review summarizes recent progress in liver cancer stem cell research with regard to the identification, cell origin, regulation of self-renewal capacity, and therapeutic implications of liver CSCs.

Collaboration


Dive into the Bo Wang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wendy L. Frankel

The Ohio State University Wexner Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge