401
|
Lv M, Xu Y, Tang R, Ren J, Shen S, Chen Y, Liu B, Hou Y, Wang T. miR141–CXCL1–CXCR2 Signaling–Induced Treg Recruitment Regulates Metastases and Survival of Non–Small Cell Lung Cancer. Mol Cancer Ther 2014; 13:3152-62. [DOI: 10.1158/1535-7163.mct-14-0448] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
402
|
Lin L, Yin X, Hu X, Wang Q, Zheng L. The impact of hepatitis B virus x protein and microRNAs in hepatocellular carcinoma: a comprehensive analysis. Tumour Biol 2014; 35:11695-700. [PMID: 25286757 DOI: 10.1007/s13277-014-2658-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 09/18/2014] [Indexed: 02/06/2023] Open
Abstract
microRNAs (miRNAs) are evolutionarily conserved small non-coding RNAs, approximately 22 nucleotides (nts) in length, widely found in animals, plants, and viruses. Mature miRNAs control gene expression at a post-transcriptional level through blocking protein translation or inducing mRNA degradation. Many recent studies have shown that hepatitis B virus x protein (HBx), a viral protein with a crucial role in hepatogenesis, is associated with the regulation of miRNAs. This interaction impacts fundamental tumor processes, such as cell proliferation, differentiation, and apoptosis. In this review, we summarized the recent literature on the roles of HBx-regulated miRNAs in the pathogenesis of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Li Lin
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Road, Baiyun District, Guangzhou, 510515, People's Republic of China
| | | | | | | | | |
Collapse
|
403
|
Wang F, Yuan JH, Wang SB, Yang F, Yuan SX, Ye C, Yang N, Zhou WP, Li WL, Li W, Sun SH. Oncofetal long noncoding RNA PVT1 promotes proliferation and stem cell-like property of hepatocellular carcinoma cells by stabilizing NOP2. Hepatology 2014; 60:1278-90. [PMID: 25043274 DOI: 10.1002/hep.27239] [Citation(s) in RCA: 359] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Accepted: 05/21/2014] [Indexed: 12/14/2022]
Abstract
UNLABELLED Many protein-coding oncofetal genes are highly expressed in murine and human fetal liver and silenced in adult liver. The protein products of these hepatic oncofetal genes have been used as clinical markers for the recurrence of hepatocellular carcinoma (HCC) and as therapeutic targets for HCC. Herein we examined the expression profiles of long noncoding RNAs (lncRNAs) found in fetal and adult liver in mice. Many fetal hepatic lncRNAs were identified; one of these, lncRNA-mPvt1, is an oncofetal RNA that was found to promote cell proliferation, cell cycling, and the expression of stem cell-like properties of murine cells. Interestingly, we found that human lncRNA-hPVT1 was up-regulated in HCC tissues and that patients with higher lncRNA-hPVT1 expression had a poor clinical prognosis. The protumorigenic effects of lncRNA-hPVT1 on cell proliferation, cell cycling, and stem cell-like properties of HCC cells were confirmed both in vitro and in vivo by gain-of-function and loss-of-function experiments. Moreover, mRNA expression profile data showed that lncRNA-hPVT1 up-regulated a series of cell cycle genes in SMMC-7721 cells. By RNA pulldown and mass spectrum experiments, we identified NOP2 as an RNA-binding protein that binds to lncRNA-hPVT1. We confirmed that lncRNA-hPVT1 up-regulated NOP2 by enhancing the stability of NOP2 proteins and that lncRNA-hPVT1 function depends on the presence of NOP2. CONCLUSION Our study demonstrates that the expression of many lncRNAs is up-regulated in early liver development and that the fetal liver can be used to search for new diagnostic markers for HCC. LncRNA-hPVT1 promotes cell proliferation, cell cycling, and the acquisition of stem cell-like properties in HCC cells by stabilizing NOP2 protein. Regulation of the lncRNA-hPVT1/NOP2 pathway may have beneficial effects on the treatment of HCC.
Collapse
Affiliation(s)
- Fang Wang
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China; Center of Reproductive Medicine, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
404
|
Misso G, Di Martino MT, De Rosa G, Farooqi AA, Lombardi A, Campani V, Zarone MR, Gullà A, Tagliaferri P, Tassone P, Caraglia M. Mir-34: a new weapon against cancer? MOLECULAR THERAPY-NUCLEIC ACIDS 2014; 3:e194. [PMID: 25247240 PMCID: PMC4222652 DOI: 10.1038/mtna.2014.47] [Citation(s) in RCA: 390] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Accepted: 08/11/2014] [Indexed: 02/07/2023]
Abstract
The microRNA(miRNA)-34a is a key regulator of tumor suppression. It controls the
expression of a plethora of target proteins involved in cell cycle, differentiation
and apoptosis, and antagonizes processes that are necessary for basic cancer cell
viability as well as cancer stemness, metastasis, and chemoresistance. In this
review, we focus on the molecular mechanisms of miR-34a-mediated tumor suppression,
giving emphasis on the main miR-34a targets, as well as on the principal regulators
involved in the modulation of this miRNA. Moreover, we shed light on the miR-34a role
in modulating responsiveness to chemotherapy and on the phytonutrients-mediated
regulation of miR-34a expression and activity in cancer cells. Given the broad
anti-oncogenic activity of miR-34a, we also discuss the substantial benefits of a new
therapeutic concept based on nanotechnology delivery of miRNA mimics. In fact, the
replacement of oncosuppressor miRNAs provides an effective strategy against tumor
heterogeneity and the selective RNA-based delivery systems seems to be an excellent
platform for a safe and effective targeting of the tumor.
Collapse
Affiliation(s)
- Gabriella Misso
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Magna Graecia University and Medical OncologyUnit, T. Campanella Cancer Center, Salvatore Venuta University Campus, Catanzaro, Italy
| | - Giuseppe De Rosa
- Department of Pharmacy, University "Federico II" of Naples, Naples, Italy
| | - Ammad Ahmad Farooqi
- Laboratory for Translational Oncology and Personalized Medicine, Rashid Latif Medical College, Lahore, Pakistan
| | - Angela Lombardi
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Virginia Campani
- Department of Pharmacy, University "Federico II" of Naples, Naples, Italy
| | - Mayra Rachele Zarone
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Annamaria Gullà
- Department of Experimental and Clinical Medicine, Magna Graecia University and Medical OncologyUnit, T. Campanella Cancer Center, Salvatore Venuta University Campus, Catanzaro, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University and Medical OncologyUnit, T. Campanella Cancer Center, Salvatore Venuta University Campus, Catanzaro, Italy
| | - Pierfrancesco Tassone
- 1] Department of Experimental and Clinical Medicine, Magna Graecia University and Medical OncologyUnit, T. Campanella Cancer Center, Salvatore Venuta University Campus, Catanzaro, Italy [2] Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, USA
| | - Michele Caraglia
- 1] Department of Experimental and Clinical Medicine, Magna Graecia University and Medical OncologyUnit, T. Campanella Cancer Center, Salvatore Venuta University Campus, Catanzaro, Italy [2] Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
405
|
Xie KL, Zhang YG, Liu J, Zeng Y, Wu H. MicroRNAs associated with HBV infection and HBV-related HCC. Theranostics 2014; 4:1176-92. [PMID: 25285167 PMCID: PMC4183996 DOI: 10.7150/thno.8715] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 08/10/2014] [Indexed: 02/05/2023] Open
Abstract
Hepatitis B virus (HBV) infection is a global problem and a major risk factor for hepatocellular carcinoma (HCC). microRNAs (miRNAs) comprise a group of small noncoding RNAs regulating gene expression at the posttranslational level, thereby participating in fundamental biological processes, including cell proliferation, differentiation, and apoptosis. In this review, we summarize the roles of miRNAs in HBV infection, the recently identified mechanism underlying dysregulation of miRNAs in HBV-associated HCC, and their association with hepatocarcinogenesis. Moreover, we discuss the recent advances in the use of circulating miRNAs in the early diagnosis of HCC as well as therapies based on these aberrantly expressed miRNAs.
Collapse
|
406
|
Yang P, Markowitz GJ, Wang XF. The hepatitis B virus-associated tumor microenvironment in hepatocellular carcinoma. Natl Sci Rev 2014; 1:396-412. [PMID: 25741453 DOI: 10.1093/nsr/nwu038] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In contrast to a majority of cancer types, the initiation of hepatocellular carcinoma (HCC) is intimately associated with a chronically diseased liver tissue, with one of the most prevalent etiological factors being hepatitis B virus (HBV). Transformation of the liver in HBV-associated HCC often follows from or accompanies long-term symptoms of chronic hepatitis, inflammation and cirrhosis, and viral load is a strong predictor for both incidence and progression of HCC. Besides aiding in transformation, HBV plays a crucial role in modulating the accumulation and activation of both cellular components of the microenvironment, such as immune cells and fibroblasts, and non-cellular components of the microenvironment, such as cytokines and growth factors, markedly influencing disease progression and prognosis. This review will explore some of these components and mechanisms to demonstrate both underlying themes and the inherent complexity of these interacting systems in the initiation, progression, and metastasis of HBV-positive HCC.
Collapse
Affiliation(s)
- Pengyuan Yang
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA ; CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Geoffrey J Markowitz
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Xiao-Fan Wang
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
407
|
Xiao Z, Li CH, Chan SL, Xu F, Feng L, Wang Y, Jiang JD, Sung JJY, Cheng CHK, Chen Y. A small-molecule modulator of the tumor-suppressor miR34a inhibits the growth of hepatocellular carcinoma. Cancer Res 2014; 74:6236-47. [PMID: 25217526 DOI: 10.1158/0008-5472.can-14-0855] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Small molecules that restore the expression of growth-inhibitory microRNAs (miRNA) downregulated in tumors may have potential as anticancer agents. miR34a functions as a tumor suppressor and is downregulated or silenced commonly in a variety of human cancers, including hepatocellular carcinoma (HCC). In this study, we used an HCC cell-based miR34a luciferase reporter system to screen for miR34a modulators that could exert anticancer activity. One compound identified as a lead candidate, termed Rubone, was identified through its ability to specifically upregulate miR34a in HCC cells. Rubone activated miR34a expression in HCC cells with wild-type or mutated p53 but not in cells with p53 deletions. Notably, Rubone lacked growth-inhibitory effects on nontumorigenic human hepatocytes. In a mouse xenograft model of HCC, Rubone dramatically inhibited tumor growth, exhibiting stronger anti-HCC activity than sorafenib both in vitro and in vivo. Mechanistic investigations showed that Rubone decreased expression of cyclin D1, Bcl-2, and other miR34a target genes and that it enhanced the occupancy of p53 on the miR34a promoter. Taken together, our results offer a preclinical proof of concept for Rubone as a lead candidate for further investigation as a new class of HCC therapeutic based on restoration of miR34a tumor-suppressor function.
Collapse
Affiliation(s)
- Zhangang Xiao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Chi Han Li
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Stephen L Chan
- Department of Clinical Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Feiyue Xu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Lu Feng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Yan Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian-Dong Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Joseph J Y Sung
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Christopher H K Cheng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong.
| | - Yangchao Chen
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong. State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, NT, Hong Kong. Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.
| |
Collapse
|
408
|
Marra F, Tacke F. Roles for chemokines in liver disease. Gastroenterology 2014; 147:577-594.e1. [PMID: 25066692 DOI: 10.1053/j.gastro.2014.06.043] [Citation(s) in RCA: 613] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 06/05/2014] [Accepted: 06/26/2014] [Indexed: 02/08/2023]
Abstract
Sustained hepatic inflammation is an important factor in progression of chronic liver diseases, including hepatitis C or non-alcoholic steatohepatitis. Liver inflammation is regulated by chemokines, which regulate the migration and activities of hepatocytes, Kupffer cells, hepatic stellate cells, endothelial cells, and circulating immune cells. However, the effects of the different chemokines and their receptors vary during pathogenesis of different liver diseases. During development of chronic viral hepatitis, CCL5 and CXCL10 regulate the cytopathic versus antiviral immune responses of T cells and natural killer cells. During development of nonalcoholic steatohepatitis, CCL2 and its receptor are up-regulated in the liver, where they promote macrophage accumulation, inflammation, fibrosis, and steatosis, as well as in adipose tissue. CCL2 signaling thereby links hepatic and systemic inflammation related to metabolic disorders and insulin resistance. Several chemokine signaling pathways also promote hepatic fibrosis. Recent studies have shown that other chemokines and immune cells have anti-inflammatory and antifibrotic activities. Chemokines and their receptors can also contribute to the pathogenesis of hepatocellular carcinoma, promoting proliferation of cancer cells, the inflammatory microenvironment of the tumor, evasion of the immune response, and angiogenesis. We review the roles of different chemokines in the pathogenesis of liver diseases and their potential use as biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Fabio Marra
- Dipartimento di Medicina Sperimentale e Clinica, University of Florence, Florence, Italy.
| | - Frank Tacke
- Department of Medicine III, RWTH University Hospital Aachen, Aachen, Germany.
| |
Collapse
|
409
|
Cheng BB, Qu MJ, Wu LL, Shen Y, Yan ZQ, Zhang P, Qi YX, Jiang ZL. MicroRNA-34a targets Forkhead box j2 to modulate differentiation of endothelial progenitor cells in response to shear stress. J Mol Cell Cardiol 2014; 74:4-12. [PMID: 24792364 DOI: 10.1016/j.yjmcc.2014.04.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 04/14/2014] [Accepted: 04/21/2014] [Indexed: 10/25/2022]
Abstract
Flow shear stress plays important roles in modulating differentiation of endothelial progenitor cells (EPCs). MicroRNAs are crucial for diverse cellular processes, but the expressions and functions of microRNAs in EPCs responding to mechanical stimuli remain unclear. We sought to determine the effects of microRNA-34a (miR-34a) and a novel target Forkhead box j2 (Foxj2) on shear stress-induced EPC differentiation. Human umbilical cord blood-derived EPCs were exposed to laminar shear stress of 15dyn/cm(2) with parallel plate flow chamber system. Real time RT-PCR showed that shear stress significantly increased miR-34a expression, which was accompanied by the endothelial differentiation of EPCs. Whereas Foxj2, a putative target of miR-34a predicted by multiple algorithms, was suppressed in this process. Dual luciferase reporter assays, as well as miR-34a mimics and inhibitor treatment were used to confirm the interplay between miR-34a and Foxj2. Our results revealed an inverse correlation of miR-34a and Foxj2 expressions implicated in the endothelial differentiation of EPCs. MiR-34a contributed to this process by up-regulating the expressions of endothelial cell markers, and down-regulating smooth muscular cell markers. In addition, Foxj2 overexpression attenuated endothelial differentiation of EPCs, while Foxj2 siRNA had the opposite effect. These data suggested a unique mechanism that shear stress induces the expression of miR-34a, which targets to Foxj2 and promotes endothelial differentiation of EPCs. The results provide new insights into miR-34a/Foxj2 on shear stress-induced EPC differentiation.
Collapse
Affiliation(s)
- Bin-Bin Cheng
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ming-Juan Qu
- Department of Cell Biology & Genetics, School of Life Sciences, Ludong University, Yantai, China
| | - Lei-Lei Wu
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Shen
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi-Qiang Yan
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Zhang
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ying-Xin Qi
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| | - Zong-Lai Jiang
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
410
|
Caja F, Vannucci L. TGFβ: A player on multiple fronts in the tumor microenvironment. J Immunotoxicol 2014; 12:300-7. [DOI: 10.3109/1547691x.2014.945667] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
411
|
The role of miRNA-34a as a prognostic biomarker for cirrhotic patients with portal hypertension receiving TIPS. PLoS One 2014; 9:e103779. [PMID: 25068403 PMCID: PMC4113430 DOI: 10.1371/journal.pone.0103779] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 07/02/2014] [Indexed: 12/12/2022] Open
Abstract
Background Circulating miRNA-34a is increased in blood of patients with different liver diseases when compared to healthy controls. However, the origin of miRNA-34a and its possible relationship with hemodynamics and outcome in cirrhotic patients with portal hypertension is unknown. We analyzed the levels of miRNA-34a in cirrhotic patients with severe portal hypertension. Methods We included 60 cirrhotic patients receiving TIPS for prevention of rebleeding and/or therapy-refractory ascites. miRNA-34a levels were measured using qPCR and normalized by SV-40 in the portal and hepatic venous blood of these patients taken at TIPS procedure. Hemodynamic and clinical parameters were assessed before TIPS and during follow-up. Results Levels of miRNA-34a were higher in the hepatic vein than in the portal vein. Circulating miRNA-34a in the hepatic vein correlated with ALT, CHE and sodium excretion after TIPS. miRNA-34a showed no correlation with portal pressure, but its levels in the portal vein correlated inversely with the congestion index. Interestingly, the levels of miRNA-34a in the portal and hepatic vein showed inverse correlation with arterial pressure. Furthermore, levels of miRNA-34a in the hepatic vein had a predictive value for survival, but MELD, creatinine at short-time follow-up 14 days after TIPS-insertion and portal pressure after TIPS performed better. Conclusion This study demonstrates for the first time, that miRNA-34a may originate to a large extent from the liver. Even though higher levels of miRNA-34a are possibly associated with better survival at long-term follow-up in cirrhotic patients with severe portal hypertension receiving TIPS, classical prognostic parameters predict the survival better.
Collapse
|
412
|
Xu HZ, Liu YP, Guleng B, Ren JL. Hepatitis B Virus-Related Hepatocellular Carcinoma: Pathogenic Mechanisms and Novel Therapeutic Interventions. Gastrointest Tumors 2014; 1:135-45. [PMID: 26676160 DOI: 10.1159/000365307] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Infection with the hepatitis B virus (HBV) is one of most important risk factors for hepatocellular carcinoma (HCC). Indeed, HBV is considered a group 1 human carcinogen and is a highly oncogenic agent. HBV cannot be effectively controlled or completely eliminated, so chronic HBV infection is a public health challenge worldwide. SUMMARY It is now believed that HBV-induced HCC involves a complex interaction between multiple viral and host factors. Many factors contribute to HBV-associated HCC, including products of HBV, viral integration and mutation, and host susceptibility. This review outlines the main pathogenic mechanisms with a focus on those that suggest novel targets for the prevention and treatment of HCC. KEY MESSAGE HBV infection is an important risk factor for HCC. Understanding the interaction between viral and host factors in HBV-induced HCC will reveal potential targets for future therapies. PRACTICAL IMPLICATIONS The two main therapeutic strategies consist of antiviral agents and immunotherapy-based approaches. Dendritic cell-based immunotherapy is promising for restoring the T cell-mediated antiviral immune response. Another approach is the specific expansion of the host's pool of HBV-specific T cells. Stimulation of the Toll-like receptors (TLRs), particularly TLR9, provides another means of boosting the antiviral response. Combination therapy with cytokines (interferon gamma and tumor necrosis factor alpha) plus lamivudine is more effective than these agents used alone. Therapeutic vaccines are being developed as an alternative to long-term antiviral treatment or as an adjunct.
Collapse
Affiliation(s)
- Hong-Zhi Xu
- Department of Gastroenterology, Zhongshan Hospital affiliated with Xiamen University, Xiamen, China
| | - Yun-Peng Liu
- Department of Gastroenterology, Zhongshan Hospital affiliated with Xiamen University, Xiamen, China
| | - Bayasi Guleng
- Department of Gastroenterology, Zhongshan Hospital affiliated with Xiamen University, Xiamen, China ; Medical College of Xiamen University, Xiamen, China
| | - Jian-Lin Ren
- Department of Gastroenterology, Zhongshan Hospital affiliated with Xiamen University, Xiamen, China
| |
Collapse
|
413
|
Complex metastatic niches: already a target for therapy? Curr Opin Cell Biol 2014; 31:29-38. [PMID: 25036901 DOI: 10.1016/j.ceb.2014.06.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 06/24/2014] [Accepted: 06/26/2014] [Indexed: 01/06/2023]
Abstract
Metastatic spread is an inefficient process which requires generation of supportive microenvironments in which cancer cells can survive, proliferate and escape from immune attack. These niches are induced by systemic and locally produced factors and establish a tumor-supportive and immune suppressive environment which is molecularly and functionally different from the niche at the primary site. Tumor dormancy may result if the niche is not sufficiently supportive/protective. Co-evolution of cancer cells and the surrounding microenvironment creates a large number of such dynamic niches, and we are just beginning to elucidate the complexity of these interactions and their tissue-specific differences. We will discuss exciting possibilities but also challenges which are immanent when trying to target these stromal responses for diagnosis and therapy.
Collapse
|
414
|
Siemens H, Jackstadt R, Kaller M, Hermeking H. Repression of c-Kit by p53 is mediated by miR-34 and is associated with reduced chemoresistance, migration and stemness. Oncotarget 2014; 4:1399-415. [PMID: 24009080 PMCID: PMC3824539 DOI: 10.18632/oncotarget.1202] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The c-Kit receptor tyrosine kinase is commonly over-expressed in different types of cancer. p53 activation is known to result in the down-regulation of c-Kit. However, the underlying mechanism has remained unknown. Here, we show that the p53-induced miR-34 microRNA family mediates repression of c-Kit by p53 via a conserved seed-matching sequence in the c-Kit 3'-UTR. Ectopic miR-34a resulted in a decrease in Erk signaling and transformation, which was dependent on the down-regulation of c-Kit expression. Furthermore, ectopic expression of c-Kit conferred resistance of colorectal cancer (CRC) cells to treatment with 5-fluorouracil (5-FU), whereas ectopic miR-34a sensitized the cells to 5-FU. After stimulation with c-Kit ligand/stem cell factor (SCF) Colo320 CRC cells displayed increased migration/invasion, whereas ectopic miR-34a inhibited SCF-induced migration/invasion. Activation of a conditional c-Kit allele induced several stemness markers in DLD-1 CRC cells. In primary CRC samples elevated c-Kit expression also showed a positive correlation with markers of stemness, such as Lgr5, CD44, OLFM4, BMI-1 and β-catenin. On the contrary, activation of a conditional miR-34a allele in DLD-1 cells diminished the expression of c-Kit and several stemness markers (CD44, Lgr5 and BMI-1) and suppressed sphere formation. MiR-34a also suppressed enhanced sphere-formation after exposure to SCF. Taken together, our data establish c-Kit as a new direct target of miR-34 and demonstrate that this regulation interferes with several c-Kit-mediated effects on cancer cells. Therefore, this regulation may be potentially relevant for future diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Helge Siemens
- Experimental and Molecular Pathology, Institute of Pathology, Ludwig-Maximilians-University München, D-80337 Munich, Germany
| | | | | | | |
Collapse
|
415
|
Chen L, Ma H, Hu H, Gao L, Wang X, Ma J, Gao Q, Liu B, Zhou G, Liang C. Special role of Foxp3 for the specifically altered microRNAs in Regulatory T cells of HCC patients. BMC Cancer 2014; 14:489. [PMID: 25000974 PMCID: PMC4099493 DOI: 10.1186/1471-2407-14-489] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 07/04/2014] [Indexed: 12/17/2022] Open
Abstract
Background Regulatory T cells (Tregs) exhibit functional abnormalities in the context of hepatocellular carcinoma (HCC). The microRNAs (miRNAs) are identified as the key modulators in Tregs. This study was to explore whether the expression profiles of miRNAs of Tregs were different in HCC-activated Tregs and whether Foxp3 had special effects on them. Methods We isolated HCC-activated Tregs from mice bearing HCC and compared the expression profiles of miRNAs between HCC-activated Tregs and control Tregs by microarray. RNA interference against Foxp3 was also performed through transfection of synthetic siRNAs to Tregs for analyzing the effect of Foxp3 on the expression of miRNAs. Tregs isolated from HCC patients (n = 12) and healthy controls (n = 7) were used for validation of the differentially expressed miRNAs. Finally, bioinformatic analysis was applied to infer their possible roles. Results We found nine specifically altered miRNAs in HCC-activated Tregs from the murine model. After transfection with siRNAs against Foxp3, control Tregs showed obvious reduction of Foxp3 and five miRNAs were significantly changed; HCC-activated Tregs exhibited a slight reduction of Foxp3 with three miRNAs significantly changed. Tregs from HCC patients and healthy controls finally confirmed the up-regulation of four miRNAs (hsa-miR-182-5p, hsa-miR-214-3p, hsa-miR-129-5p and hsa-miR-30b-5p). Following bioinformatic analysis suggested these altered miRNAs would target eight important signaling pathways that could affect the functions of Tregs. Conclusions Our studies provided the first evidence that Tregs in HCC had the specifically altered expression of miRNAs, which was affected by Foxp3. These results are useful both in finding new biomarkers and in further exploring the functions of Tregs in HCC patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Chunmin Liang
- Lab of Tumor Immunology, Department of Anatomy and Histology & Embryology, Shanghai Medical College of Fudan University, 138 Yixueyuan Road, 200032 Shanghai, PR China.
| |
Collapse
|
416
|
De Antonellis P, Carotenuto M, Vandenbussche J, De Vita G, Ferrucci V, Medaglia C, Boffa I, Galiero A, Di Somma S, Magliulo D, Aiese N, Alonzi A, Spano D, Liguori L, Chiarolla C, Verrico A, Schulte JH, Mestdagh P, Vandesompele J, Gevaert K, Zollo M. Early targets of miR-34a in neuroblastoma. Mol Cell Proteomics 2014; 13:2114-31. [PMID: 24912852 DOI: 10.1074/mcp.m113.035808] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Several genes encoding for proteins involved in proliferation, invasion, and apoptosis are known to be direct miR-34a targets. Here, we used proteomics to screen for targets of miR-34a in neuroblastoma (NBL), a childhood cancer that originates from precursor cells of the sympathetic nervous system. We examined the effect of miR-34a overexpression using a tetracycline inducible system in two NBL cell lines (SHEP and SH-SY5Y) at early time points of expression (6, 12, and 24 h). Proteome analysis using post-metabolic labeling led to the identification of 2,082 proteins, and among these 186 were regulated (112 proteins down-regulated and 74 up-regulated). Prediction of miR-34a targets via bioinformatics showed that 32 transcripts held miR-34a seed sequences in their 3'-UTR. By combining the proteomics data with Kaplan Meier gene-expression studies, we identified seven new gene products (ALG13, TIMM13, TGM2, ABCF2, CTCF, Ki67, and LYAR) that were correlated with worse clinical outcomes. These were further validated in vitro by 3'-UTR seed sequence regulation. In addition, Michigan Molecular Interactions searches indicated that together these proteins affect signaling pathways that regulate cell cycle and proliferation, focal adhesions, and other cellular properties that overall enhance tumor progression (including signaling pathways such as TGF-β, WNT, MAPK, and FAK). In conclusion, proteome analysis has here identified early targets of miR-34a with relevance to NBL tumorigenesis. Along with the results of previous studies, our data strongly suggest miR-34a as a useful tool for improving the chance of therapeutic success with NBL.
Collapse
Affiliation(s)
- Pasqualino De Antonellis
- From the ‡Centro di Ingegneria Genetica e Biotecnologie Avanzate (CEINGE), 80145 Naples, Italy; §Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, 80131 Naples, Italy
| | - Marianeve Carotenuto
- From the ‡Centro di Ingegneria Genetica e Biotecnologie Avanzate (CEINGE), 80145 Naples, Italy; §Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, 80131 Naples, Italy
| | - Jonathan Vandenbussche
- ‖Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium; **Department of Biochemistry, Ghent University, B-9000 Ghent, Belgium
| | - Gennaro De Vita
- From the ‡Centro di Ingegneria Genetica e Biotecnologie Avanzate (CEINGE), 80145 Naples, Italy; §Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, 80131 Naples, Italy
| | - Veronica Ferrucci
- From the ‡Centro di Ingegneria Genetica e Biotecnologie Avanzate (CEINGE), 80145 Naples, Italy; §Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, 80131 Naples, Italy
| | - Chiara Medaglia
- From the ‡Centro di Ingegneria Genetica e Biotecnologie Avanzate (CEINGE), 80145 Naples, Italy; §Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, 80131 Naples, Italy
| | - Iolanda Boffa
- From the ‡Centro di Ingegneria Genetica e Biotecnologie Avanzate (CEINGE), 80145 Naples, Italy; §Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, 80131 Naples, Italy
| | - Alessandra Galiero
- From the ‡Centro di Ingegneria Genetica e Biotecnologie Avanzate (CEINGE), 80145 Naples, Italy; §Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, 80131 Naples, Italy
| | - Sarah Di Somma
- From the ‡Centro di Ingegneria Genetica e Biotecnologie Avanzate (CEINGE), 80145 Naples, Italy; §Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, 80131 Naples, Italy
| | - Daniela Magliulo
- From the ‡Centro di Ingegneria Genetica e Biotecnologie Avanzate (CEINGE), 80145 Naples, Italy; §Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, 80131 Naples, Italy
| | - Nadia Aiese
- From the ‡Centro di Ingegneria Genetica e Biotecnologie Avanzate (CEINGE), 80145 Naples, Italy; §Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, 80131 Naples, Italy
| | - Alessandro Alonzi
- From the ‡Centro di Ingegneria Genetica e Biotecnologie Avanzate (CEINGE), 80145 Naples, Italy; §Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, 80131 Naples, Italy
| | - Daniela Spano
- From the ‡Centro di Ingegneria Genetica e Biotecnologie Avanzate (CEINGE), 80145 Naples, Italy; §Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, 80131 Naples, Italy
| | - Lucia Liguori
- From the ‡Centro di Ingegneria Genetica e Biotecnologie Avanzate (CEINGE), 80145 Naples, Italy; §Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, 80131 Naples, Italy
| | - Cristina Chiarolla
- From the ‡Centro di Ingegneria Genetica e Biotecnologie Avanzate (CEINGE), 80145 Naples, Italy; §Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, 80131 Naples, Italy
| | - Antonio Verrico
- From the ‡Centro di Ingegneria Genetica e Biotecnologie Avanzate (CEINGE), 80145 Naples, Italy; §Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, 80131 Naples, Italy; ‡‡Department of Translational Medical Science, Section of Pediatrics, University of Naples Federico II, 80131 Naples, Italy
| | | | - Pieter Mestdagh
- ¶¶Center for Medical Genetics, Ghent University Hospital, B-9000 Ghent, Belgium
| | - Jo Vandesompele
- ¶¶Center for Medical Genetics, Ghent University Hospital, B-9000 Ghent, Belgium
| | - Kris Gevaert
- ‖Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium; **Department of Biochemistry, Ghent University, B-9000 Ghent, Belgium
| | - Massimo Zollo
- From the ‡Centro di Ingegneria Genetica e Biotecnologie Avanzate (CEINGE), 80145 Naples, Italy; §Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Federico II, 80131 Naples, Italy; ‖‖Centro di Medicina Trasfusionale, Azienda Ospedaliera Federico II, 80131 Naples, Italy
| |
Collapse
|
417
|
Yuan JH, Yang F, Wang F, Ma JZ, Guo YJ, Tao QF, Liu F, Pan W, Wang TT, Zhou CC, Wang SB, Wang YZ, Yang Y, Yang N, Zhou WP, Yang GS, Sun SH. A long noncoding RNA activated by TGF-β promotes the invasion-metastasis cascade in hepatocellular carcinoma. Cancer Cell 2014; 25:666-81. [PMID: 24768205 DOI: 10.1016/j.ccr.2014.03.010] [Citation(s) in RCA: 1251] [Impact Index Per Article: 113.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Revised: 01/07/2014] [Accepted: 03/11/2014] [Indexed: 02/06/2023]
Abstract
The role of TGF-β-induced epithelial-mesenchymal transition (EMT) in cancer cell dissemination is well established, but the involvement of lncRNAs in TGF-β signaling is still unknown. In this study, we observed that the lncRNA-activated by TGF-β (lncRNA-ATB) was upregulated in hepatocellular carcinoma (HCC) metastases and associated with poor prognosis. lncRNA-ATB upregulated ZEB1 and ZEB2 by competitively binding the miR-200 family and then induced EMT and invasion. In addition, lncRNA-ATB promoted organ colonization of disseminated tumor cells by binding IL-11 mRNA, autocrine induction of IL-11, and triggering STAT3 signaling. Globally, lncRNA-ATB promotes the invasion-metastasis cascade. Thus, these findings suggest that lncRNA-ATB, a mediator of TGF-β signaling, could predispose HCC patients to metastases and may serve as a potential target for antimetastatic therapies.
Collapse
Affiliation(s)
- Ji-hang Yuan
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Fu Yang
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Fang Wang
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Jin-zhao Ma
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Ying-jun Guo
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Qi-fei Tao
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Feng Liu
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Wei Pan
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Tian-tian Wang
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Chuan-chuan Zhou
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Shao-bing Wang
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Yu-zhao Wang
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China
| | - Yuan Yang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Ning Yang
- The Fifth Department of Hepatic Surgery, Eastern Hepatobiliary Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Wei-ping Zhou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Guang-shun Yang
- The Fifth Department of Hepatic Surgery, Eastern Hepatobiliary Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Shu-han Sun
- Department of Medical Genetics, Second Military Medical University, Shanghai, 200433, China.
| |
Collapse
|
418
|
Rokavec M, Li H, Jiang L, Hermeking H. The p53/miR-34 axis in development and disease. J Mol Cell Biol 2014; 6:214-30. [DOI: 10.1093/jmcb/mju003] [Citation(s) in RCA: 239] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
419
|
Zhang D, Zhou J, Dong M. Dysregulation of microRNA-34a expression in colorectal cancer inhibits the phosphorylation of FAK via VEGF. Dig Dis Sci 2014; 59:958-67. [PMID: 24370784 DOI: 10.1007/s10620-013-2983-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Accepted: 12/04/2013] [Indexed: 01/07/2023]
Abstract
BACKGROUND MicroRNAs (miRs) are small non-coding RNAs that play important roles in cancer development where they can act as oncogenes or as tumor-suppressors. MiR-34a is a tumor suppressor that is frequently downregulated in a number of tumor types. However, little is known about the role of miR-34a in colorectal cancer (CRC). AIM This study aims to show the dysregulation of miR-34a in CRC and to characterize the function and mechanism of miR-34a in CRC cell lines. METHODS The expression of miR-34a was detected using real-time PCR on CRC tissues and adjacent non-tumorous tissues. The ELISA was used to assess vascular endothelial growth factor (VEGF). The focal adhesion kinase (FAK) and phosphorylated FAK Y397 (pY397FAK) were measured by Western blot. The functions of miR-34a in vivo were measured by migration, invasion, CCK-8 assay and flow cytometry. RESULTS MiR-34a was significantly downregulated and pY397FAK was upregulated in CRC cancer tissues. It plays an important role in inhibiting migration and invasion and in increasing apoptosis of CRC cells. Bioinformatic analysis suggested that VEGF may be a target of miR-34a, and this hypothesis was proved by ELISA and RT-PCR. The level of pY397FAK that could be activated by VEGF was downregulated in miR-34a overexpression CRC cell lines. The phosphorylation of FAK at 397 sites in miR-34a-stable cell lines was completely rescued by extra VEGF treatment. CONCLUSION MiR-34a is frequently downregulated in CRC and modulates the phosphorylation of FAK by negatively regulating VEGF.
Collapse
Affiliation(s)
- Danhua Zhang
- Department of General Surgery, Gastrointestinal Surgery, The First Hospital, China Medical University, Shenyang, 110001, China,
| | | | | |
Collapse
|
420
|
Xu C, Zhou W, Wang Y, Qiao L. Hepatitis B virus-induced hepatocellular carcinoma. Cancer Lett 2014; 345:216-222. [PMID: 23981576 DOI: 10.1016/j.canlet.2013.08.035] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 08/10/2013] [Accepted: 08/18/2013] [Indexed: 12/12/2022]
Abstract
Many factors are considered to contribute to hepatitis B virus (HBV) associated hepatocellular carcinoma (HCC), including products of HBV, HBV integration and mutation, and host susceptibility. HBV X protein (HBx) can interfere with several signal pathways that associated with cell proliferation and apoptosis, and the impact of HBx C-terminal truncation in the development of HCC has been implicated. Recent studies by advanced sequencing technologies have revealed recurrent HBV DNA integration sites in hepatoma cells and susceptible genes/SNPs play an important role in the pathogenesis of liver cancer. Epigenetic changes, immune and inflammatory factors are also important contributing factors for liver cancer. This mini-review provides an overview on the recent development of HBV induced HCC.
Collapse
Affiliation(s)
- Cheng Xu
- Institute for Infectious Diseases, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Wence Zhou
- The Department of General Surgery II, The First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Yuming Wang
- Institute for Infectious Diseases, Southwest Hospital, Third Military Medical University, Chongqing 400038, China.
| | - Liang Qiao
- Storr Liver Unit, University of Sydney, Westmead Millennium Institute, Westmead Hospital, Westmead, NSW 2145, Australia.
| |
Collapse
|
421
|
Toomer KH, Chen Z. Autoimmunity as a double agent in tumor killing and cancer promotion. Front Immunol 2014; 5:116. [PMID: 24672527 PMCID: PMC3957029 DOI: 10.3389/fimmu.2014.00116] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Accepted: 03/05/2014] [Indexed: 12/19/2022] Open
Abstract
Cancer immunotherapy through manipulation of the immune system holds great potential for the treatment of human cancers. However, recent trials targeting the negative immune regulators cytotoxic T-lymphocyte antigen 4, programed death 1 (PD-1), and PD-1 receptor ligand (PD-L1) demonstrated that clinically significant antitumor responses were often associated with the induction of autoimmune toxicity. This finding suggests that the same immune mechanisms that elicit autoimmunity may also contribute to the destruction of tumors. Given the fact that the immunological identity of tumors might be largely an immunoprivileged self, autoimmunity may not represent a wholly undesirable outcome in the context of cancer immunotherapy. Rather, targeted killing of cancer cells and autoimmune damage to healthy tissues may be intricately linked through molecular mechanisms, in particular inflammatory cytokine signaling. On the other hand, since chronic inflammation is a well-recognized condition that promotes tumor development, it appears that autoimmunity can be a "double agent" in mediating either pro-tumor or antitumor effects. This review surveys the tumor-promoting and tumoricidal activities of several prominent cytokines: IFN-γ, TNF-α, TGF-β, IL-17, IL-23, IL-4, and IL-13, produced by three major subsets of T helper cells that interact with innate immune cells. Many of these cytokines exert divergent and seemingly contradictory effects on cancer development in different human and animal models, suggesting a high degree of context dependence in their functions. We hypothesize that these inflammatory cytokines could mediate a feedback loop of autoimmunity, antitumor immunity, and tumorigenesis. Understanding the diverse and paradoxical roles of cytokines from autoimmune responses in the setting of cancer will advance the long-term goal of improving cancer immunotherapy, while minimizing the hazards of immune-mediated tissue damage and the possibility of de novo tumorigenesis, through proper monitoring and preventive measures.
Collapse
Affiliation(s)
- Kevin H Toomer
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine , Miami, FL , USA
| | - Zhibin Chen
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine , Miami, FL , USA ; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine , Miami, FL , USA
| |
Collapse
|
422
|
Giannelli G, Villa E, Lahn M. Transforming growth factor-β as a therapeutic target in hepatocellular carcinoma. Cancer Res 2014; 74:1890-4. [PMID: 24638984 DOI: 10.1158/0008-5472.can-14-0243] [Citation(s) in RCA: 215] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Hepatocellular carcinoma arises in patients as a consequence of long-standing preexisting liver illnesses, including viral hepatitis, alcohol abuse, or metabolic disease. In such preexisting liver diseases, TGF-β plays an important role in orchestrating a favorable microenvironment for tumor cell growth and promoting epithelial-mesenchymal transition (EMT). TGF-β signaling promotes hepatocellular carcinoma progression by two mechanisms: first, via an intrinsic activity as an autocrine or paracrine growth factor and, second, via an extrinsic activity by inducing microenvironment changes, including cancer-associated fibroblasts, T regulatory cells, and inflammatory mediators. Although there is an increasing understanding on how TGF-β signaling is associated with tumor progression in hepatocellular carcinoma, it is not clear whether TGF-β signaling is limited to a certain subgroup of patients with hepatocellular carcinoma or is a key driver of hepatocellular carcinoma during the entire tumorigenesis of hepatocellular carcinoma. Inhibitors of the TGF-β signaling have been shown to block hepatocellular carcinoma growth and progression by modulating EMT in different experimental models, leading to the clinical investigation of the TGF-β inhibitor LY2157299 monohydrate in hepatocellular carcinoma. Preliminary results from a phase II clinical trial have shown improved clinical outcome and also changes consistent with a reduction of EMT.
Collapse
Affiliation(s)
- Gianluigi Giannelli
- Authors' Affiliations: Department of Biomedical Science and Human Oncology, University of Bari Medical School, Bari; Department of Internal Medicine, University of Modena and Reggio, Modena, Italy; and Early Phase Clinical Investigation, Eli Lilly and Company, Indianapolis, Indiana
| | | | | |
Collapse
|
423
|
Turato C, Simonato D, Quarta S, Gatta A, Pontisso P. MicroRNAs and SerpinB3 in hepatocellular carcinoma. Life Sci 2014; 100:9-17. [PMID: 24496037 DOI: 10.1016/j.lfs.2014.01.073] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 12/30/2013] [Accepted: 01/22/2014] [Indexed: 12/27/2022]
Abstract
miRNAs are small non-coding RNAs which target complementary mRNA sequences, usually resulting in gene silencing. They can exhibit oncogenic or tumor suppressor properties, modulating cell homeostasis. Several data have documented that miRNAs are typically deregulated in different types of cancers, including hepatocellular carcinoma (HCC). Some of the miRNAs such as miR-122, miR-221, miR-1 and miR-21 have been found to repress post-transcriptionally the expression of genes involved in cell cycle regulation, cell proliferation, apoptosis, cell migration and invasion. In HCC serum levels of miR-122, miR-221 and miR-16 have been described deregulated, suggesting that they may be used as molecular targets for early detection, prognosis and treatment. The ov-serpin SerpinB3 was found previously increased in liver tumor cancers and associated with apoptosis resistance, increased cell proliferation and invasiveness. Recent data indicate that this serpin may enhance its oncogenic potential through inhibition of several tumor suppressive miRNAs, typically described in HCC.
Collapse
Affiliation(s)
- Cristian Turato
- Department of Medicine, University of Padua, Via Giustiniani, 2, 35128 Padua, Italy
| | - Davide Simonato
- Department of Medicine, University of Padua, Via Giustiniani, 2, 35128 Padua, Italy
| | - Santina Quarta
- Department of Medicine, University of Padua, Via Giustiniani, 2, 35128 Padua, Italy
| | - Angelo Gatta
- Department of Medicine, University of Padua, Via Giustiniani, 2, 35128 Padua, Italy
| | - Patrizia Pontisso
- Department of Medicine, University of Padua, Via Giustiniani, 2, 35128 Padua, Italy.
| |
Collapse
|
424
|
Otsuka M, Kishikawa T, Yoshikawa T, Ohno M, Takata A, Shibata C, Koike K. The role of microRNAs in hepatocarcinogenesis: current knowledge and future prospects. J Gastroenterol 2014; 49:173-184. [PMID: 24258409 DOI: 10.1007/s00535-013-0909-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 11/04/2013] [Indexed: 02/04/2023]
Abstract
MicroRNAs (miRNAs) are small, noncoding RNA molecules that regulate gene expression post-transcriptionally through complementary base pairing with thousands of messenger RNAs. Although the precise biological functions of individual miRNAs are still unknown, miRNAs are speculated to play important roles in diverse biological processes through fine regulation of their target gene expression. A growing body of data indicates the deregulation of miRNAs during hepatocarcinogenesis. In this review, we summarize recent findings regarding deregulated miRNA expression and their possible target genes in hepatocarcinogenesis, with emphasis on inflammation-related hepatocarcinogenesis. Because miRNA-based strategies are being applied to clinical therapeutics, precise knowledge of miRNA functions is crucial both scientifically and clinically. We discuss the current open questions from these points of view, which must be clarified in the near future.
Collapse
Affiliation(s)
- Motoyuki Otsuka
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 5-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan,
| | | | | | | | | | | | | |
Collapse
|
425
|
Xu WP, Yi M, Li QQ, Zhou WP, Cong WM, Yang Y, Ning BF, Yin C, Huang ZW, Wang J, Qian H, Jiang CF, Chen YX, Xia CY, Wang HY, Zhang X, Xie WF. Perturbation of MicroRNA-370/Lin-28 homolog A/nuclear factor kappa B regulatory circuit contributes to the development of hepatocellular carcinoma. Hepatology 2013; 58:1977-91. [PMID: 23728999 DOI: 10.1002/hep.26541] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 04/17/2003] [Accepted: 05/16/2013] [Indexed: 01/09/2023]
Abstract
UNLABELLED MicroRNA 370 (miR-370) is located within the DLK1/DIO3 imprinting region on human chromosome 14, which has been identified as a cancer-associated genomic region. However, the role of miR-370 in malignances remains controversial. Here, we report that miR-370 was repressed in human hepatocellular carcinoma (HCC) tissues and hepatoma cell lines. Using gain-of-function and loss-of-function experiments, we demonstrated that miR-370 inhibited the malignant phenotype of HCC cells in vitro. Overexpression of miR-370 inhibited growth and metastasis of HCC cells in vivo. Moreover, the RNA-binding protein, LIN28A, was identified as a direct functional target of miR-370, which, in turn, blocked the biogenesis of miR-370 by binding to its precursor. LIN28A also mediated the suppressive effects of miR-370 on migration and invasion of HCC cells by post-transcriptionally regulating RelA/p65, which is an important effector of the canonical nuclear factor kappa B (NF-κB) pathway. Interleukin-6 (IL-6), a well-known NF-κB downstream inflammatory molecule, reduced miR-370 but increased LIN28A levels in HCC. Furthermore, miR-370 levels were inversely correlated with LIN28A and IL-6 messenger RNA (mRNA) levels, whereas LIN28A mRNA expression was positively correlated with IL-6 expression in human HCC samples. Interestingly, reduction of miR-370 expression was associated with the development of HCC in rats, as well as with aggressive tumor behavior and short survival in HCC patients. CONCLUSIONS These data demonstrate the involvement of a novel regulatory circuit consisting of miR-370, LIN28A, RelA/p65 and IL-6 in HCC progression. Manipulating this feedback loop may have beneficial effect in HCC treatment.
Collapse
Affiliation(s)
- Wen-Ping Xu
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
426
|
Fan Q, Gu D, Liu H, Yang L, Zhang X, Yoder MC, Kaplan MH, Xie J. Defective TGF-β signaling in bone marrow-derived cells prevents hedgehog-induced skin tumors. Cancer Res 2013; 74:471-483. [PMID: 24282281 DOI: 10.1158/0008-5472.can-13-2134-t] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Hedgehog signaling in cancer cells drives changes in the tumor microenvironment that are incompletely understood. Here, we report that hedgehog-driven tumors exhibit an increase in myeloid-derived suppressor cells (MDSC) and a decrease in T cells, indicative of an immunosuppressive tumor microenvironment. This change was associated with activated TGF-β signaling in several cell types in basal cell carcinomas. We determined that TGF-β signaling in bone marrow-derived cells, not keratinocytes, regulates MDSC and promotes tumor development. Tgfbr2 deficiency in the bone marrow-derived cells also reduced the size of previously developed tumors in mice. We identified CCL2 as the major chemokine attracting MDSCs to tumor, whose expression was Tgfbr2-dependent, whereas its receptor CCR2 was highly expressed in MDSC population. CCL2 alone was sufficient to induce migration of MDSCs. Moreover, the CCR2 inhibitors prevented MDSC migration toward skin cells in vitro, and reduced MDSC accumulation and hedgehog signaling-driven tumor development in mice. Our results reveal a signaling network critical for hedgehog signaling in cancer cells to establish an effective immunosuppressive microenvironment during tumor development.
Collapse
Affiliation(s)
- Qipeng Fan
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Dongsheng Gu
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Hailan Liu
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Ling Yang
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Xiaoli Zhang
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Mervin C Yoder
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Mark H Kaplan
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Jingwu Xie
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
427
|
Cheng Q, Yi B, Wang A, Jiang X. Exploring and exploiting the fundamental role of microRNAs in tumor pathogenesis. Onco Targets Ther 2013; 6:1675-84. [PMID: 24273410 PMCID: PMC3836659 DOI: 10.2147/ott.s52730] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
miRNAs (miRs) are short RNA molecules that are involved in the posttranscriptional regulation of mRNA. The roles of miRs in tumor pathogenesis have only recently become a focus of research. It is becoming increasingly clear that miRs are important regulators of apoptosis, proliferation, invasion, and metastasis in cancer cells during cancer genesis and progression, furthering our understanding of cancer. In the present review, we summarize and evaluate the recent advances in our understanding of the characteristics of miRs as well as their regulated functions in cancer stem cells (CSCs), the epithelial-mesenchymal transition (EMT), and the tumor microenvironment (TM), describing their roles in tumor pathogenesis and their possible use as new therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Qingbao Cheng
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | | | | | | |
Collapse
|
428
|
Hasmim M, Noman MZ, Messai Y, Bordereaux D, Gros G, Baud V, Chouaib S. Cutting Edge: Hypoxia-Induced Nanog Favors the Intratumoral Infiltration of Regulatory T Cells and Macrophages via Direct Regulation of TGF-β1. THE JOURNAL OF IMMUNOLOGY 2013; 191:5802-6. [DOI: 10.4049/jimmunol.1302140] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
429
|
Abstract
The influence of the microenvironment on tumour progression is becoming clearer. In this Review we address the role of an essential signalling pathway, that of transforming growth factor-β, in the regulation of components of the tumour microenvironment and how this contributes to tumour progression.
Collapse
Affiliation(s)
- Michael Pickup
- Vanderbilt University Medical Center, Vanderbilt-Ingram Comprehensive Cancer Center, Medicine and Pathology, Cancer Biology, 2220 Pierce Avenue, 691 Preston Research Building, Nashville, Tennessee 37232, USA
| | | | | |
Collapse
|
430
|
Regulatory T-cell directed therapies in liver diseases. J Hepatol 2013; 59:1127-34. [PMID: 23727305 DOI: 10.1016/j.jhep.2013.05.034] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Revised: 05/17/2013] [Accepted: 05/22/2013] [Indexed: 01/07/2023]
|
431
|
Zhang Y, Yang P, Wang XF. Microenvironmental regulation of cancer metastasis by miRNAs. Trends Cell Biol 2013; 24:153-60. [PMID: 24125906 DOI: 10.1016/j.tcb.2013.09.007] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 09/12/2013] [Accepted: 09/13/2013] [Indexed: 12/19/2022]
Abstract
miRNAs are a class of small, non-coding RNAs that regulate cancer progression, especially the processes of invasion and metastasis. Although earlier studies in metastasis primarily focused on the impact that miRNAs have on the intrinsic properties of cancer cells, recent reports reveal that miRNAs also shape interactions between cancer cells and their associated stroma. In this review, we discuss current known mechanisms by which miRNAs execute their microenvironmental regulation of cancer metastasis, including regulating expression of cell membrane-bound and secreted proteins or directly transmitting mature miRNAs between different cell types. The significance of miRNA-mediated tumor-stroma interactions in regulating metastasis suggests that miRNAs may be a potential therapeutic target.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Pengyuan Yang
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA; Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiao-Fan Wang
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
432
|
Li YQ, Liu FF, Zhang XM, Guo XJ, Ren MJ, Fu L. Tumor secretion of CCL22 activates intratumoral Treg infiltration and is independent prognostic predictor of breast cancer. PLoS One 2013; 8:e76379. [PMID: 24124553 PMCID: PMC3790712 DOI: 10.1371/journal.pone.0076379] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 08/26/2013] [Indexed: 12/27/2022] Open
Abstract
It has been reported that dense intratumoral infiltration of Foxp3 +Tregs (Tregs) was an independent factor for poor prognosis of breast cancer (BC) patients. However, the cytokines activating the Treg infiltration are not known. This study was undertaken to evaluate the role of CCL22 and TGF-β1 in this cascade and their prognostic significance for BC patients. 417 cases of invasive breast cancer were selected from the prior study cohort and the expressions of CCL22 and TGF-β1 were assessed by immunohistochemistry. It was identified that tumor secretion of CCL22 was positively correlated with the intratumoral Treg infiltration (P<0.0001), but its association with lymphoid aggregates surrounding the tumor was not proven to be significant (P=0.056). Moreover, CCL22 expression was found to be associated with the tumor histological features known to be related with unfavorable prognosis of patients, including high histological grade (P<0.0001), negative ER (P<0.0001), negative PR (P=0.001), and HER2 amplification (P=0.028). Similar to intratumoral Treg infiltrates, CCL22 tumor secretion correlated with the prognosis of the molecular subtypes of breast carcinoma (P<0.0001). Univariate analysis revealed CCL22 to be an independent prognostic factor for overall survival (OS, P<0.0001) and progression-free survival (PFS, P<0.0001) of BC patients that were confirmed by multivariate analysis (P=0.011 and P=0.010 respectively). In contrast, although TGF-β1 expression was positively correlated with both Tregs infiltrates into the tumor bed and lymphoid aggregates surrounding the tumor (P=0.023; P=0.046, respectively), its expression was not significantly associated with the molecular subtypes of breast carcinoma and the prognosis of the patients. Our study indicates that both CCL22 and TGF-β1 are candidate chemoattractants for intratumoral Foxp3 +Tregs infiltration; however, unlike the later, CCL22 is an independent prognostic predictor of BC patients, and it therefore may have the potential to serve as a target for immunotherapeutic strategy of BC.
Collapse
Affiliation(s)
- Ya-Qing Li
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Fang-Fang Liu
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xin-Min Zhang
- Department of Pathology and Laboratory Medicine, Temple University Hospital, Philadelphia, Pennsylvania, United States of America
| | - Xiao-Jing Guo
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Mei-Jing Ren
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Li Fu
- Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin, China
- * E-mail:
| |
Collapse
|
433
|
Zhang YH, Dong XQ, Zou Y, Wu JL, Bai LP, Liu JH, Wang M. Role of TGF-β in metastasis of hepatocellular carcinoma. Shijie Huaren Xiaohua Zazhi 2013; 21:2508-2514. [DOI: 10.11569/wcjd.v21.i25.2508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the role of transforming growth factor-b (TGF-β) derived from regulatory T cells in metastasis of hepatocellular carcinoma.
METHODS: Forty-eight primary liver cancer patients without metastasis, 32 liver cancer patients with distant metastasis (17 cases of lung metastasis and 15 cases of brain metastasis), and 54 healthy subjects were included in this study. Serum concentrations of interleukin-10 (IL-10), IL-17A, tumor necrosis factor-α (TNF-α) and TGF-β were measured by ELISA. Levels of TGF-β in peripheral blood mononuclear cells (PBMCs) and regulatory T cells were determined by flow cytometry and RT-PCR, respectively.
RESULTS: Serum levels of TNF-α and TGF-β were significantly increased in liver cancer patients with metastasis compared with normal controls. Serum level of TGF-β was significantly higher in liver cancer patients with metastasis than in liver cancer without metastasis and normal controls (both P < 0.05). Furthermore, we confirmed that TGF-β in liver cancer patients with metastasis was derived from regulatory T cells by quantitative real-time PCR and flow cytometry.
CONCLUSION: TGF-β has a dual role in tumorigenesis; it acts as a tumor suppressor in early stage but promotes tumor cell invasion and metastasis in advanced stage. High level of TGF-β was found only in liver cancer patients with metastasis.
Collapse
|
434
|
Karimi-Googheri M, Daneshvar H, Nosratabadi R, Zare-Bidaki M, Hassanshahi G, Ebrahim M, Arababadi MK, Kennedy D. Important roles played by TGF-β in hepatitis B infection. J Med Virol 2013; 86:102-8. [PMID: 24009084 DOI: 10.1002/jmv.23727] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2013] [Indexed: 12/14/2022]
Abstract
Hepatitis B virus (HBV) which includes, fulminant, acute, chronic, asymptomatic, and occult HBV infection is the most prevalent virus that leads to human liver diseases. Chronic, asymptomatic, and occult infection can induce further sever diseases such as hepatocellular carcinoma (HCC) and cirrhosis of the liver. The underlying mechanisms that allow progression of the prolonged forms of the infection and subsequent HCC or cirrhosis of the liver are yet to be clarified. However, many researchers have suggested that immunological and genetic parameters may play important roles in the etiology of hepatitis B. Transforming growth factor beta (TGF-β) is an important cytokine with dual regulatory functions in the immune system and in the responses against viral infections. However, the pathways and mechanisms controlling these are not fully understood. The crucial roles of TGF-β in the development of Th17 and T regulatory lymphocytes, the main cell types involved in autoimmunity and destructive immune related diseases, have been documented and this provides insights into TGF-β function during hepatitis infection and subsequent HCC and cirrhosis of the liver. Recent findings also confirm that TGF-β directly alters hepatocyte function during hepatitis B, hence, the aim of this review is to address the current data regarding the association and status of TGF-β with hepatitis B infection and its related disorders including HCC and cirrhosis of the liver.
Collapse
|
435
|
Hanks BA, Holtzhausen A, Evans KS, Jamieson R, Gimpel P, Campbell OM, Hector-Greene M, Sun L, Tewari A, George A, Starr M, Nixon A, Augustine C, Beasley G, Tyler DS, Osada T, Morse MA, Ling L, Lyerly HK, Blobe GC. Type III TGF-β receptor downregulation generates an immunotolerant tumor microenvironment. J Clin Invest 2013; 123:3925-40. [PMID: 23925295 PMCID: PMC3754240 DOI: 10.1172/jci65745] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 06/13/2013] [Indexed: 01/02/2023] Open
Abstract
Cancers subvert the host immune system to facilitate disease progression. These evolved immunosuppressive mechanisms are also implicated in circumventing immunotherapeutic strategies. Emerging data indicate that local tumor-associated DC populations exhibit tolerogenic features by promoting Treg development; however, the mechanisms by which tumors manipulate DC and Treg function in the tumor microenvironment remain unclear. Type III TGF-β receptor (TGFBR3) and its shed extracellular domain (sTGFBR3) regulate TGF-β signaling and maintain epithelial homeostasis, with loss of TGFBR3 expression promoting progression early in breast cancer development. Using murine models of breast cancer and melanoma, we elucidated a tumor immunoevasion mechanism whereby loss of tumor-expressed TGFBR3/sTGFBR3 enhanced TGF-β signaling within locoregional DC populations and upregulated both the immunoregulatory enzyme indoleamine 2,3-dioxygenase (IDO) in plasmacytoid DCs and the CCL22 chemokine in myeloid DCs. Alterations in these DC populations mediated Treg infiltration and the suppression of antitumor immunity. Our findings provide mechanistic support for using TGF-β inhibitors to enhance the efficacy of tumor immunotherapy, indicate that sTGFBR3 levels could serve as a predictive immunotherapy biomarker, and expand the mechanisms by which TGFBR3 suppresses cancer progression to include effects on the tumor immune microenvironment.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Chemokine CCL22/metabolism
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Down-Regulation
- Female
- Humans
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasm Transplantation
- Proteoglycans/genetics
- Proteoglycans/metabolism
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/metabolism
- Transforming Growth Factor beta/metabolism
- Tumor Escape
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Brent A. Hanks
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Alisha Holtzhausen
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Katherine S. Evans
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Rebekah Jamieson
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Petra Gimpel
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Olivia M. Campbell
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Melissa Hector-Greene
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Lihong Sun
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Alok Tewari
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Amanda George
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Mark Starr
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Andrew Nixon
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Christi Augustine
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Georgia Beasley
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Douglas S. Tyler
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Takayu Osada
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Michael A. Morse
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Leona Ling
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - H. Kim Lyerly
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Gerard C. Blobe
- Department of Medicine and
Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA.
Freie Universität Berlin, Institut für Chemie und Biochemie, Berlin, Germany.
Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, USA.
Biogen Idec Inc., Cambridge, Massachusetts, USA.
Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
436
|
Ma Y, Bao-Han W, Lv X, Su Y, Zhao X, Yin Y, Zhang X, Zhou Z, MacNaughton WK, Wang H. MicroRNA-34a mediates the autocrine signaling of PAR2-activating proteinase and its role in colonic cancer cell proliferation. PLoS One 2013; 8:e72383. [PMID: 23991105 PMCID: PMC3753253 DOI: 10.1371/journal.pone.0072383] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 07/09/2013] [Indexed: 12/15/2022] Open
Abstract
The tumor microenvironment is replete with proteinases. As a sensor of proteinases, proteinase activated receptor 2 (PAR2) plays critical roles in tumorigenesis. We showed that PAR2 and its activating proteinase were coexpressed in different colon cancer cell lines, including HT29. Inactivating proteinase or knockdown of PAR2 significantly not only reduced cell proliferation in vitro but also inhibited tumorigenicity of HT29 in vivo. In addition, activation of PAR2 promoted DNA synthesis and upregulated Cyclin D1 activity at both transcriptional and post-transcriptional levels. Further studies showed that miRNA-34a mediated PAR2-induced Cyclin D1 upregulation. Inhibition of miR-34a partially abolished the suppression of Cyclin D1 induced by PAR2 deficiency. In addition, we showed that TGF-β contributed to the regulation of miR-34a by PAR2. Finally, in colorectal carcinoma samples, upregulation of PAR2 and downregulation of miR-34a were significantly correlated with grade and lymphomatic metastasis. Our findings provide the first evidence that miRNA mediates autocrine proteinase signaling-mediated cancer cell proliferation.
Collapse
Affiliation(s)
- Yiming Ma
- State Key Laboratory of Molecular Oncology, Cancer Institute/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Wuyun Bao-Han
- State Key Laboratory of Molecular Oncology, Cancer Institute/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xue Lv
- State Key Laboratory of Molecular Oncology, Cancer Institute/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yuntao Su
- First affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Xinhua Zhao
- State Key Laboratory of Molecular Oncology, Cancer Institute/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yongmei Yin
- First affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Xingmao Zhang
- Department of gastrointestinal cancer surgery, Cancer Institute/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zhixiang Zhou
- Department of gastrointestinal cancer surgery, Cancer Institute/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Wallace K. MacNaughton
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Biophysics, University of Calgary, Calgary, Alberta, Canada
| | - Hongying Wang
- State Key Laboratory of Molecular Oncology, Cancer Institute/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
437
|
A miR-34a-SIRT6 axis in the squamous cell differentiation network. EMBO J 2013; 32:2248-63. [PMID: 23860128 DOI: 10.1038/emboj.2013.156] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 06/17/2013] [Indexed: 01/15/2023] Open
Abstract
Squamous cell carcinomas (SCCs) are highly heterogeneous tumours, resulting from deranged expression of genes involved in squamous cell differentiation. Here we report that microRNA-34a (miR-34a) functions as a novel node in the squamous cell differentiation network, with SIRT6 as a critical target. miR-34a expression increases with keratinocyte differentiation, while it is suppressed in skin and oral SCCs, SCC cell lines, and aberrantly differentiating primary human keratinocytes (HKCs). Expression of this miRNA is restored in SCC cells, in parallel with differentiation, by reversion of genomic DNA methylation or wild-type p53 expression. In normal HKCs, the pro-differentiation effects of increased p53 activity or UVB exposure are miR-34a-dependent, and increased miR-34a levels are sufficient to induce differentiation of these cells both in vitro and in vivo. SIRT6, a sirtuin family member not previously connected with miR-34a function, is a direct target of this miRNA in HKCs, and SIRT6 down-modulation is sufficient to reproduce the miR-34a pro-differentiation effects. The findings are of likely biological significance, as SIRT6 is oppositely expressed to miR-34a in normal keratinocytes and keratinocyte-derived tumours.
Collapse
|
438
|
MicroRNA-19a-3p inhibits breast cancer progression and metastasis by inducing macrophage polarization through downregulated expression of Fra-1 proto-oncogene. Oncogene 2013; 33:3014-23. [PMID: 23831570 DOI: 10.1038/onc.2013.258] [Citation(s) in RCA: 144] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 03/22/2013] [Accepted: 04/29/2013] [Indexed: 12/11/2022]
Abstract
One of the hallmarks of malignancy is the polarization of tumor-associated macrophages (TAMs) from a pro-immune (M1-like) phenotype to an immune-suppressive (M2-like) phenotype. However, the molecular basis of the process is still unclear. MicroRNA (miRNA) comprises a group of small, non-coding RNAs that are broadly expressed by a variety of organisms and are involved in cell behaviors such as suppression or promotion of tumorigenesis. Here, we demonstrate that miR-19a-3p, broadly conserved among vertebrates, was downregulated in RAW264.7 macrophage cells of the M2 phenotype in conditoned medium of 4T1 mouse breast tumor cells. This downregulation correlated with an increased expression of the Fra-1 gene, which was reported to act as a pro-oncogene by supporting the invasion and progression of breast tumors. We found significant upregulation of miR-19a-3p in RAW264.7 macrophages after transfection with a miR-19a-3p mimic that resulted in a significant suppression of the expression of this gene. In addition, we could measure the activity of binding between miR-19a-3p and Fra-1 with a psiCHECK luciferase reporter system. Further, transfection of RAW264.7 macrophage cells with the miR-19a-3p mimic decreased the expression of the Fra-1 downstream genes VEGF, STAT3 and pSTAT3. Most importantly, the capacity of 4T1 breast tumor cells to migrate and invade was impaired in vivo by the intratumoral injection of miR-19a-3p. Taken together, these findings indicate that miR-19a-3p is capable of downregulating the M2 phenotype in M2 macrophages and that the low expression of this miRNA has an important role in the upregulation of Fra-1 expression and induction of M2 macrophage polarization.
Collapse
|
439
|
Insulin receptor tyrosine kinase substrate activates EGFR/ERK signalling pathway and promotes cell proliferation of hepatocellular carcinoma. Cancer Lett 2013; 337:96-106. [PMID: 23693078 DOI: 10.1016/j.canlet.2013.05.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2013] [Revised: 05/07/2013] [Accepted: 05/14/2013] [Indexed: 11/21/2022]
Abstract
Insulin receptor tyrosine kinase substrate (IRTKS) is closely associated with actin remodelling and membrane protrusion, but its role in the pathogenesis of malignant tumours, including hepatocellular carcinoma (HCC), is still unknown. In this study, we showed that IRTKS was frequently upregulated in HCC samples, and its expression level was significantly associated with tumour size. Enforced expression of IRTKS in human HCC cell lines significantly promoted their proliferation and colony formation in vitro, and their capacity to develop tumour xenografts in vivo, whereas knockdown of IRTKS resulted in the opposite effects. Furthermore, the bromodeoxyuridine (BrdU) incorporation analyses and propidium iodide staining indicated that IRTKS can promote the entry into S phase of cell cycle progression. Significantly, IRTKS can interact with epidermal growth factor receptor (EGFR), results in the phosphorylation of extracellular signal-regulated kinase (ERK). By contrast, inhibition of ERK activation can attenuate the effects of IRTKS overexpression on cellular proliferation. Taken together, these data demonstrate that IRTKS promotes the proliferation of HCC cells by enhancing EGFR-ERK signalling pathway.
Collapse
|
440
|
Huang CS, Ho CT, Tu SH, Pan MH, Chuang CH, Chang HW, Chang CH, Wu CH, Ho YS. Long-term ethanol exposure-induced hepatocellular carcinoma cell migration and invasion through lysyl oxidase activation are attenuated by combined treatment with pterostilbene and curcumin analogues. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2013; 61:4326-4335. [PMID: 23560895 DOI: 10.1021/jf4004175] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Ethanol consumption induces hepatocellular carcinoma (HCC) cell metastasis by changing the extracellular matrix (ECM). Lysyl oxidase (LOX) catalyzes the cross-linkage of collagen or elastin in the ECM. LOX protein and mRNA overexpression (>21-fold compared with controls, n = 6) was detected in cirrhotic HCC patients with a history of alcoholism. LOX protein expression was induced in HCC cells after long-term treatment with ethanol (10 mM) for 20-40 passages (denoted E20-E40 cells). Pterostilbene (PSB, 1 μM) displayed significant potency to reduce LOX-mediated activity in E40 cells when combined with curcumin and its analogues. The ability of E40 cells to form colonies in soft agar was reduced by both genetic depletion of LOX and by chemical inhibitors of LOX expression. This study suggests that targeting LOX expression with food components such as PSB and curcumin may be a novel strategy to overcome ethanol-induced HCC cell metastasis in liver cancer patients.
Collapse
Affiliation(s)
- Ching-Shui Huang
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
441
|
Yamashita T, Wang XW. Cancer stem cells in the development of liver cancer. J Clin Invest 2013; 123:1911-8. [PMID: 23635789 DOI: 10.1172/jci66024] [Citation(s) in RCA: 413] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Liver cancer is an aggressive disease with a poor outcome. Several hepatic stem/progenitor markers are useful for isolating a subset of liver cells with stem cell features, known as cancer stem cells (CSCs). These cells are responsible for tumor relapse, metastasis, and chemoresistance. Liver CSCs dictate a hierarchical organization that is shared in both organogenesis and tumorigenesis. An increased understanding of the molecular signaling events that regulate cellular hierarchy and stemness, and success in defining key CSC-specific genes, have opened up new avenues to accelerate the development of novel diagnostic and treatment strategies. This Review highlights recent advances in understanding the pathogenesis of liver CSCs and discusses unanswered questions about the concept of liver CSCs.
Collapse
Affiliation(s)
- Taro Yamashita
- Department of General Medicine, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan.
| | | |
Collapse
|
442
|
Gurtan AM, Sharp PA. The role of miRNAs in regulating gene expression networks. J Mol Biol 2013; 425:3582-600. [PMID: 23500488 DOI: 10.1016/j.jmb.2013.03.007] [Citation(s) in RCA: 318] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 02/28/2013] [Accepted: 03/04/2013] [Indexed: 01/03/2023]
Abstract
MicroRNAs (miRNAs) are key regulators of gene expression. They are conserved across species, expressed across cell types, and active against a large proportion of the transcriptome. The sequence-complementary mechanism of miRNA activity exploits combinatorial diversity, a property conducive to network-wide regulation of gene expression, and functional evidence supporting this hypothesized systems-level role has steadily begun to accumulate. The emerging models are exciting and will yield deep insight into the regulatory architecture of biology. However, because of the technical challenges facing the network-based study of miRNAs, many gaps remain. Here, we review mammalian miRNAs by describing recent advances in understanding their molecular activity and network-wide function.
Collapse
Affiliation(s)
- Allan M Gurtan
- David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, USA.
| | | |
Collapse
|
443
|
miR-126 and miR-126* repress recruitment of mesenchymal stem cells and inflammatory monocytes to inhibit breast cancer metastasis. Nat Cell Biol 2013; 15:284-94. [PMID: 23396050 DOI: 10.1038/ncb2690] [Citation(s) in RCA: 290] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 01/11/2013] [Indexed: 02/07/2023]
Abstract
The tumour stroma is an active participant during cancer progression. Stromal cells promote tumour progression and metastasis through multiple mechanisms including enhancing tumour invasiveness and angiogenesis, and suppressing immune surveillance. We report here that miR-126/miR-126(*), a microRNA pair derived from a single precursor, independently suppress the sequential recruitment of mesenchymal stem cells and inflammatory monocytes into the tumour stroma to inhibit lung metastasis by breast tumour cells in a mouse xenograft model. miR-126/miR-126(*) directly inhibit stromal cell-derived factor-1 alpha (SDF-1α) expression, and indirectly suppress the expression of chemokine (C-C motif) ligand 2 (Ccl2) by cancer cells in an SDF-1α-dependent manner. miR-126/miR-126(*) expression is downregulated in cancer cells by promoter methylation of their host gene Egfl7. These findings determine how this microRNA pair alters the composition of the primary tumour microenvironment to favour breast cancer metastasis, and demonstrate a correlation between miR-126/126(*) downregulation and poor metastasis-free survival of breast cancer patients.
Collapse
|
444
|
MicroRNAs in hepatocellular carcinoma: regulation, function, and clinical implications. ScientificWorldJournal 2013; 2013:924206. [PMID: 23431261 PMCID: PMC3575633 DOI: 10.1155/2013/924206] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 01/13/2013] [Indexed: 12/21/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide and the third cause of cancer-related death. Poor understanding of the mechanisms underlying the pathogenesis of HCC makes it difficult to be diagnosed and treated at early stage. MicroRNAs (miRNAs), a class of noncoding single-stranded RNAs of ~22 nucleotides in length, posttranscriptionally regulate gene expression by base pairing with the 3' untranslated regions (3'UTRs) of target messenger RNAs (mRNAs). Aberrant expression of miRNAs is found in many if not all cancers, and many deregulated miRNAs have been proved to play crucial roles in the initiation and progression of cancers by regulating the expression of various oncogenes or tumor suppressor genes. In this Paper, we will summarize the regulations and functions of miRNAs aberrantly expressed in HCC and discuss the potential application of miRNAs as diagnostic and prognostic biomarkers of HCC and their potential roles in the intervention of HCC.
Collapse
|
445
|
Abstract
Creating a permissive microenvironment is a strategy employed by tumor cells to disseminate. In this issue of Cancer Cell, Yang et al. identify the molecular signaling events that connect hepatitis infection with TGFβ activity and T regulatory cell recruitment to establish a favorable microenvironment for tumor metastasis.
Collapse
Affiliation(s)
- Anuradha Budhu
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, 37 Convent Drive, Bethesda, MD 20892, USA
| | | |
Collapse
|
446
|
LIN BIYUN, XIAO CHUANXING, ZHAO WENXIU, XIAO LI, CHEN XU, LI PING, WANG XIAOMIN. Enoyl-coenzyme A hydratase short chain 1 silencing attenuates the proliferation of hepatocellular carcinoma by inhibiting epidermal growth factor signaling in vitro and in vivo. Mol Med Rep 2012; 12:1421-8. [DOI: 10.3892/mmr.2015.3453] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 02/17/2015] [Indexed: 11/06/2022] Open
|