1
|
Xiang X, Bhowmick K, Shetty K, Ohshiro K, Yang X, Wong LL, Yu H, Latham PS, Satapathy SK, Brennan C, Dima RJ, Chambwe N, Sharifova G, Cacaj F, John S, Crawford JM, Huang H, Dasarathy S, Krainer AR, He AR, Amdur RL, Mishra L. Mechanistically based blood proteomic markers in the TGF-β pathway stratify risk of hepatocellular cancer in patients with cirrhosis. Genes Cancer 2024; 15:1-14. [PMID: 38323119 PMCID: PMC10843195 DOI: 10.18632/genesandcancer.234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 12/05/2023] [Indexed: 02/08/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of death from cancer worldwide but is often diagnosed at an advanced incurable stage. Yet, despite the urgent need for blood-based biomarkers for early detection, few studies capture ongoing biology to identify risk-stratifying biomarkers. We address this gap using the TGF-β pathway because of its biological role in liver disease and cancer, established through rigorous animal models and human studies. Using machine learning methods with blood levels of 108 proteomic markers in the TGF-β family, we found a pattern that differentiates HCC from non-HCC in a cohort of 216 patients with cirrhosis, which we refer to as TGF-β based Protein Markers for Early Detection of HCC (TPEARLE) comprising 31 markers. Notably, 20 of the patients with cirrhosis alone presented an HCC-like pattern, suggesting that they may be a group with as yet undetected HCC or at high risk for developing HCC. In addition, we found two other biologically relevant markers, Myostatin and Pyruvate Kinase M2 (PKM2), which were significantly associated with HCC. We tested these for risk stratification of HCC in multivariable models adjusted for demographic and clinical variables, as well as batch and site. These markers reflect ongoing biology in the liver. They potentially indicate the presence of HCC early in its evolution and before it is manifest as a detectable lesion, thereby providing a set of markers that may be able to stratify risk for HCC.
Collapse
Affiliation(s)
- Xiyan Xiang
- The Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research and Cold Spring Harbor Laboratory, Division of Gastroenterology and Hepatology, Northwell Health, Manhasset, NY 11030, USA
- These authors contributed equally to this work
| | - Krishanu Bhowmick
- The Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research and Cold Spring Harbor Laboratory, Division of Gastroenterology and Hepatology, Northwell Health, Manhasset, NY 11030, USA
- These authors contributed equally to this work
| | - Kirti Shetty
- Division of Gastroenterology and Hepatology, University of Maryland, Baltimore, MD 21201, USA
| | - Kazufumi Ohshiro
- The Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research and Cold Spring Harbor Laboratory, Division of Gastroenterology and Hepatology, Northwell Health, Manhasset, NY 11030, USA
| | - Xiaochun Yang
- The Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research and Cold Spring Harbor Laboratory, Division of Gastroenterology and Hepatology, Northwell Health, Manhasset, NY 11030, USA
| | - Linda L. Wong
- Department of Surgery, University of Hawaii, Honolulu, HI 96813, USA
| | - Herbert Yu
- Department of Epidemiology, University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Patricia S. Latham
- Department of Pathology, The George Washington University, Washington, DC 20037, USA
| | - Sanjaya K. Satapathy
- Department of Medicine, Sandra Atlas Bass Center for Liver Diseases and Transplantation, North Shore University Hospital/Northwell Health, Manhasset, NY 11030, USA
| | - Christina Brennan
- Office of Clinical Research, Northwell Health, Lake Success, NY 11042, USA
- The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Richard J. Dima
- Office of Clinical Research, Northwell Health, Lake Success, NY 11042, USA
| | - Nyasha Chambwe
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Gulru Sharifova
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Fellanza Cacaj
- The Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research and Cold Spring Harbor Laboratory, Division of Gastroenterology and Hepatology, Northwell Health, Manhasset, NY 11030, USA
| | - Sahara John
- The Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research and Cold Spring Harbor Laboratory, Division of Gastroenterology and Hepatology, Northwell Health, Manhasset, NY 11030, USA
| | | | - Hai Huang
- The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Srinivasan Dasarathy
- Division of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, OH 44106, USA
| | | | - Aiwu R. He
- Georgetown Lombardi Comprehensive Cancer Center, Washington, DC 20007, USA
| | - Richard L. Amdur
- The Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research and Cold Spring Harbor Laboratory, Division of Gastroenterology and Hepatology, Northwell Health, Manhasset, NY 11030, USA
- Quantitative Intelligence, The Institutes for Health Systems Science, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Lopa Mishra
- The Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research and Cold Spring Harbor Laboratory, Division of Gastroenterology and Hepatology, Northwell Health, Manhasset, NY 11030, USA
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Department of Surgery, The George Washington University, Washington, DC 20037, USA
| |
Collapse
|
2
|
Xie Y, Chen X, Wang X, Liu S, Chen S, Yu Z, Wang W. Transforming growth factor-β1 protects against white matter injury and reactive astrogliosis via the p38 MAPK pathway in rodent demyelinating model. J Neurochem 2024; 168:83-99. [PMID: 38183677 DOI: 10.1111/jnc.16037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/25/2023] [Accepted: 11/28/2023] [Indexed: 01/08/2024]
Abstract
In central nervous system (CNS), demyelination is a pathological process featured with a loss of myelin sheaths around axons, which is responsible for the diseases of multiple sclerosis, neuromyelitis optica, and so on. Transforming growth factor-beta1 (TGF-β1) is a multifunctional cytokine participating in abundant physiological and pathological processes in CNS. However, the effects of TGF-β1 on CNS demyelinating disease and its underlying mechanisms are controversial and not well understood. Herein, we evaluated the protective potential of TGF-β1 in a rodent demyelinating model established by lysophosphatidylcholine (LPC) injection. It was identified that supplement of TGF-β1 evidently rescued the cognitive deficit and motor dysfunction in LPC modeling mice assessed by novel object recognition and balance beam behavioral tests. Besides, quantified by luxol fast blue staining, immunofluorescence, and western blot, administration of TGF-β1 was found to significantly ameliorate the demyelinating lesion and reactive astrogliosis by suppressing p38 MAPK pathway. Mechanistically, the results of in vitro experiments indicated that treatment of TGF-β1 could directly promote the differentiation and migration of cultured oligodendrocytes. Our study revealed that modulating TGF-β1 activity might serve as a promising and innovative therapeutic strategy in CNS demyelinating diseases.
Collapse
Affiliation(s)
- Yi Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Xuejiao Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyue Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Shuai Liu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Wuhan, China
| | - Simiao Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, China
| | - Zhiyuan Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Fu X, Zhang Y, Luo Q, Ju Y, Song G. Targeting the mechano-microenvironment and liver cancer stem cells: a promising therapeutic strategy for liver cancer. Cancer Biol Med 2023; 20:j.issn.2095-3941.2023.0229. [PMID: 38009775 PMCID: PMC10690881 DOI: 10.20892/j.issn.2095-3941.2023.0229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 10/30/2023] [Indexed: 11/29/2023] Open
Abstract
Over the past 2 decades, cancer stem cells (CSCs) have been identified as the root cause of cancer occurrence, progression, chemoradioresistance, recurrence, and metastasis. Targeting CSCs is a novel therapeutic strategy for cancer management and treatment. Liver cancer (LC) is a malignant disease that can endanger human health. Studies are increasingly suggesting that changes in the liver mechanical microenvironment are a primary driver triggering the occurrence and development of liver cancer. In this review, we summarize current understanding of the roles of the liver mechano-microenvironment and liver cancer stem cells (LCSCs) in liver cancer progression. We also discuss the relationship between the mechanical heterogeneity of liver cancer tissues and LCSC recruitment and metastasis. Finally, we highlight potential mechanosensitive molecules in LCSCs and mechanotherapy in liver cancer. Understanding the roles and regulatory mechanisms of the mechano-microenvironment and LCSCs may provide fundamental insights into liver cancer progression and aid in further development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Xiaorong Fu
- School of Biology and Engineering, Guizhou Medical University, Guiyang 550000, China
- College of Bioengineering, Chongqing University, Chongqing 400030, China
- Department of Mechanical Science and Engineering, Nagoya University, Nagoya 4648603, Japan
| | - Yi Zhang
- Department of Mechanical Science and Engineering, Nagoya University, Nagoya 4648603, Japan
| | - Qing Luo
- College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Yang Ju
- Department of Mechanical Science and Engineering, Nagoya University, Nagoya 4648603, Japan
| | - Guanbin Song
- College of Bioengineering, Chongqing University, Chongqing 400030, China
| |
Collapse
|
4
|
Lominadze Z, Shaik MR, Choi D, Zaffar D, Mishra L, Shetty K. Hepatocellular Carcinoma Genetic Classification. Cancer J 2023; 29:249-258. [PMID: 37796642 PMCID: PMC10686192 DOI: 10.1097/ppo.0000000000000682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
ABSTRACT Hepatocellular carcinoma (HCC) represents a significant global burden, with management complicated by its heterogeneity, varying presentation, and relative resistance to therapy. Recent advances in the understanding of the genetic, molecular, and immunological underpinnings of HCC have allowed a detailed classification of these tumors, with resultant implications for diagnosis, prognostication, and selection of appropriate treatments. Through the correlation of genomic features with histopathology and clinical outcomes, we are moving toward a comprehensive and unifying framework to guide our diagnostic and therapeutic approach to HCC.
Collapse
Affiliation(s)
- Zurabi Lominadze
- Division of Gastroenterology and Hepatology, University of Maryland School of Medicine
| | | | - Dabin Choi
- Department of Medicine, University of Maryland Medical Center
| | - Duha Zaffar
- Department of Medicine, University of Maryland Midtown Medical Center
| | - Lopa Mishra
- Feinstein Institutes for Medical Research and Cold Spring Harbor Laboratory; Divisions of Gastroenterology and Hepatology, Northwell Health
| | - Kirti Shetty
- Division of Gastroenterology and Hepatology, University of Maryland School of Medicine
| |
Collapse
|
5
|
Kern AE, Ortmayr G, Assinger A, Starlinger P. The role of microRNAs in the different phases of liver regeneration. Expert Rev Gastroenterol Hepatol 2023; 17:959-973. [PMID: 37811642 DOI: 10.1080/17474124.2023.2267422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023]
Abstract
INTRODUCTION Since the first discovery of microRNAs (miRs) extensive evidence reveals their indispensable role in different patho-physiological processes. They are recognized as critical regulators of hepatic regeneration, as they modulate multiple complex signaling pathways affecting liver regeneration. MiR-related translational suppression and degradation of target mRNAs and proteins are not limited to one specific gene, but act on multiple targets. AREAS COVERED In this review, we are going to explore the role of miRs in the context of liver regeneration and discuss the regulatory effects attributed to specific miRs. Moreover, specific pathways crucial for liver regeneration will be discussed, with a particular emphasis on the involvement of miRs within the respective signaling cascades. EXPERT OPINION The considerable amount of studies exploring miR functions in a variety of diseases paved the way for the development of miR-directed therapeutics. Clinical implementation has already shown promising results, but additional research is warranted to assure safe and efficient delivery. Nevertheless, given the broad functional properties of miRs and their critical involvement during hepatic regeneration, they represent an attractive treatment target to promote liver recovery after hepatic resection.
Collapse
Affiliation(s)
- Anna Emilia Kern
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Gregor Ortmayr
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Alice Assinger
- Department of Vascular Biology and Thrombosis Research, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Patrick Starlinger
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, Mayo Clinic, Rochester, MN, USA
- Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
6
|
Li X, Yao Y, Wei L. Indirubin alleviates CCl 4-induced liver fibrosis by regulation of TGF-β-mediated signaling pathways. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:1047-1052. [PMID: 37605732 PMCID: PMC10440143 DOI: 10.22038/ijbms.2023.70476.15319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/10/2023] [Indexed: 08/23/2023]
Abstract
Objectives Liver fibrosis is a common liver disease caused by chronic liver damage. However, there are currently no approved drugs available to treat it. Therefore, the therapeutic effect of indirubin on liver fibrosis was evaluated. This study investigated the protective effect and related molecular mechanism of indirubin against CCl4-induced liver fibrosis in mice. Materials and Methods We first detected the effect of indirubin on liver fibrosis in mice (n=8 per group, 32 mice total) by ELISA, HE, and Masson staining. Subsequently, the proliferation of activated HSCs was detected by MTT and EdU. Finally, the changes of related proteins and signaling pathways in mice treated with indirubin were investigated by qRT-PCR and Western blot. One-way ANOVA or two-tailed student's t-test was used for comparison between groups. Results Firstly, we found that indirubin (25 mg/kg) therapy could attenuate liver injury and significantly down-regulate α-SMA (P=0.0038) and collagen 1 (P=0.0057) in the liver using CCl4-induced liver fibrosis in mice. Secondly, we showed that indirubin (25 μM) could significantly inhibit hepatic stellate cell (HSC) trans-differentiation into myofibroblasts and proliferation (P=0.0063) in HSC-T6 cells treated by TGF-β. Finally, we showed that indirubin could greatly reduce the protein levels of p-Smad2/3, p38, p-ERK, and p-JNK in vivo and in vitro. Conclusion Our results suggested that indirubin alleviated liver fibrosis and HSC activation mainly through TGF-β-mediated signaling pathways in vivo and in vitro. In conclusion, our data showed that indirubin could be a promising clinical therapeutic drug for the prevention and treatment of liver fibrosis.
Collapse
Affiliation(s)
- Xiaoying Li
- College of Biology and Food Engineering, Huaihua University. Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, Hunan, 418000, China
| | - Yuanzhi Yao
- College of Biology and Food Engineering, Huaihua University. Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, Hunan, 418000, China
| | - Lin Wei
- College of Biology and Food Engineering, Huaihua University. Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, Hunan, 418000, China
- College of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
- Daosheng Biology (Shenzhen) Co., Ltd, Shenzhen, 518107, China
| |
Collapse
|
7
|
Xu Z, Jiang N, Xiao Y, Yuan K, Wang Z. The role of gut microbiota in liver regeneration. Front Immunol 2022; 13:1003376. [PMID: 36389782 PMCID: PMC9647006 DOI: 10.3389/fimmu.2022.1003376] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/12/2022] [Indexed: 12/02/2022] Open
Abstract
The liver has unique regeneration potential, which ensures the continuous dependence of the human body on hepatic functions. As the composition and function of gut microbiota has been gradually elucidated, the vital role of gut microbiota in liver regeneration through gut-liver axis has recently been accepted. In the process of liver regeneration, gut microbiota composition is changed. Moreover, gut microbiota can contribute to the regulation of the liver immune microenvironment, thereby modulating the release of inflammatory factors including IL-6, TNF-α, HGF, IFN-γ and TGF-β, which involve in different phases of liver regeneration. And previous research have demonstrated that through enterohepatic circulation, bile acids (BAs), lipopolysaccharide, short-chain fatty acids and other metabolites of gut microbiota associate with liver and may promote liver regeneration through various pathways. In this perspective, by summarizing gut microbiota-derived signaling pathways that promote liver regeneration, we unveil the role of gut microbiota in liver regeneration and provide feasible strategies to promote liver regeneration by altering gut microbiota composition.
Collapse
Affiliation(s)
- Zhe Xu
- Department of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Nan Jiang
- Department of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Yuanyuan Xiao
- Department of Obstetrics and Gynecology, West China Second Hospital of Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- *Correspondence: Zhen Wang, ; Kefei Yuan, ; Yuanyuan Xiao,
| | - Kefei Yuan
- Department of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
- *Correspondence: Zhen Wang, ; Kefei Yuan, ; Yuanyuan Xiao,
| | - Zhen Wang
- Department of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
- Laboratory of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
- *Correspondence: Zhen Wang, ; Kefei Yuan, ; Yuanyuan Xiao,
| |
Collapse
|
8
|
Xie Y, Chen X, Li Y, Chen S, Liu S, Yu Z, Wang W. Transforming growth factor-β1 protects against LPC-induced cognitive deficit by attenuating pyroptosis of microglia via NF-κB/ERK1/2 pathways. J Neuroinflammation 2022; 19:194. [PMID: 35902863 PMCID: PMC9336072 DOI: 10.1186/s12974-022-02557-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 07/06/2022] [Indexed: 11/30/2022] Open
Abstract
Background Demyelinating diseases in central nervous system (CNS) are a group of diseases characterized by myelin damage or myelin loss. Transforming growth factor beta1 (TGF-β1) is widely recognized as an anti-inflammatory cytokine, which can be produced by both glial and neuronal cells in CNS. However, the effects of TGF-β1 on demyelinating diseases and its underlying mechanisms have not been well investigated. Methods A demyelinating mouse model using two-point injection of lysophosphatidylcholine (LPC) to the corpus callosum in vivo was established. Exogenous TGF-β1 was delivered to the lesion via brain stereotactic injection. LFB staining, immunofluorescence, and Western blot were applied to examine the severity of demyelination and pyroptosis process in microglia. Morris water maze test was used to assess the cognitive abilities of experimental mice. Furthermore, lipopolysaccharide (LPS) was applied to induce pyroptosis in primary cultured microglia in vitro, to explore potential molecular mechanism. Results The degree of demyelination in LPC-modeling mice was found improved with supplement of TGF-β1. Besides, TGF-β1 treatment evidently ameliorated the activated proinflammatory pyroptosis of microglia, with downregulated levels of the key pyroptosis effector Gasdermin D (GSDMD), inflammasomes, and cleaved-IL-1β, which effectively attenuated neuroinflammation in vivo. Evaluated by behavioral tests, the cognitive deficit in LPC-modeling mice was found mitigated with application of TGF-β1. Mechanistically, TGF-β1 could reverse pyroptosis-like morphology in LPS-stimulated primary cultured microglia observed by scanning electron microscopy, as well as decrease the protein levels of cleaved-GSDMD, inflammasomes, and cleaved-IL-1β. Activation of ERK1/2 and NF-κB pathways largely abolished the protective effects of TGF-β1, which indicated that TGF-β1 alleviated the pyroptosis possibly via regulating NF-κB/ERK1/2 signal pathways. Conclusions Our studies demonstrated TGF-β1 notably relieved the demyelinating injury and cognitive disorder in LPC-modeling mice, by attenuating the inflammatory pyroptosis of microglia via ERK1/2 and NF-κB pathways. Targeting TGF-β1 activity might serve as a promising therapeutic strategy in demyelinating diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02557-0.
Collapse
Affiliation(s)
- Yi Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xuejiao Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ying Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Simiao Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, 310003, China
| | - Shuai Liu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhiyuan Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
9
|
Rojas Á, Gil-Gómez A, de la Cruz-Ojeda P, Muñoz-Hernández R, Sánchez-Torrijos Y, Gallego-Durán R, Millán R, Rico MC, Montero-Vallejo R, Gato-Zambrano S, Maya-Miles D, Ferrer MT, Muntané J, Robles-Frías MJ, Ampuero J, Padillo FJ, Romero-Gómez M. Long non-coding RNA H19 as a biomarker for hepatocellular carcinoma. Liver Int 2022; 42:1410-1422. [PMID: 35243752 DOI: 10.1111/liv.15230] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 12/06/2021] [Accepted: 12/21/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND AIMS Liver cancer stem cells (CSCs) could be involved in the carcinogenesis, recurrence, metastasis and chemoresistance of hepatocellular carcinoma (HCC). The aim of this study was to explore the role of lncRNA-H19 as a biomarker for liver cancer. METHODS LncRNA-H19 expression levels and the functional assays were conducted in EpCAM+ CD133+ CSCs and C57BL/6J mice fed with a high-fat high-cholesterol carbohydrate (HFHCC) or standard diet for 52 weeks. Liver tissue and plasma samples from patients with cirrhosis, with or without HCC, were used for the analyses of gene expression and circulating lncRNA-H19 levels in an estimation and validation cohort. RESULTS EpCAM+ CD133+ cells showed a stem cell-like phenotype, self-renewal capacity, upregulation of pluripotent gene expression and overexpressed lncRNA-H19 (p < .001). Suppression of lncRNA-H19 by antisense oligonucleotide treatment significantly reduced the self-renewal capacity (p < .001). EpCAM, CD133 and lncRNA-h19 expression increased accordingly with disease progression in HFHCC-fed mice (p < .05) and also in liver tissue from HCC patients (p = .0082). Circulating lncRNA-H19 levels were significantly increased in HCC patients in both cohorts (p = .013; p < .0001). In addition, lncRNA-H19 levels increased accordingly with BCLC staging (p < .0001) and decreased after a partial and complete therapeutic response (p < .05). In addition, patients with cirrhosis who developed HCC during follow-up showed higher lncRNA-H19 levels (p = .0025). CONCLUSION LncRNA-H19 expression was increased in CSCs, in liver tissue and plasma of patients with HCC and decreased after partial/complete therapeutic response. Those patients who developed HCC during the follow-up showed higher levels of lncRNA-H19. LncRNA-H19 could constitute a new biomarker of HCC.
Collapse
Affiliation(s)
- Ángela Rojas
- Seliver Group, Institute of Biomedicine of Seville/ /Hospital, Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain.,Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain
| | - Antonio Gil-Gómez
- Seliver Group, Institute of Biomedicine of Seville/ /Hospital, Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain.,Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain
| | - Patricia de la Cruz-Ojeda
- Institute of Biomedicine of Seville/ /Hospital, Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Rocío Muñoz-Hernández
- Seliver Group, Institute of Biomedicine of Seville/ /Hospital, Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain.,Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain
| | - Yolanda Sánchez-Torrijos
- Seliver Group, Institute of Biomedicine of Seville/ /Hospital, Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain.,Digestive Diseases Unit, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Rocío Gallego-Durán
- Seliver Group, Institute of Biomedicine of Seville/ /Hospital, Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain.,Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain
| | - Raquel Millán
- Seliver Group, Institute of Biomedicine of Seville/ /Hospital, Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - María Carmen Rico
- Seliver Group, Institute of Biomedicine of Seville/ /Hospital, Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Rocío Montero-Vallejo
- Seliver Group, Institute of Biomedicine of Seville/ /Hospital, Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Sheila Gato-Zambrano
- Seliver Group, Institute of Biomedicine of Seville/ /Hospital, Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Douglas Maya-Miles
- Seliver Group, Institute of Biomedicine of Seville/ /Hospital, Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - M Teresa Ferrer
- Digestive Diseases Unit, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Jordi Muntané
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain.,Institute of Biomedicine of Seville/ /Hospital, Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain.,Department of Medical Physiology and Biophysics, University of Seville, Seville, Spain
| | | | - Javier Ampuero
- Seliver Group, Institute of Biomedicine of Seville/ /Hospital, Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain.,Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain.,Digestive Diseases Unit, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Francisco J Padillo
- Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain.,Institute of Biomedicine of Seville/ /Hospital, Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain.,General Surgery Department, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Manuel Romero-Gómez
- Seliver Group, Institute of Biomedicine of Seville/ /Hospital, Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain.,Hepatic and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Madrid, Spain.,Digestive Diseases Unit, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| |
Collapse
|
10
|
Baba AB, Rah B, Bhat GR, Mushtaq I, Parveen S, Hassan R, Hameed Zargar M, Afroze D. Transforming Growth Factor-Beta (TGF-β) Signaling in Cancer-A Betrayal Within. Front Pharmacol 2022; 13:791272. [PMID: 35295334 PMCID: PMC8918694 DOI: 10.3389/fphar.2022.791272] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 02/09/2022] [Indexed: 12/11/2022] Open
Abstract
A ubiquitously expressed cytokine, transforming growth factor-beta (TGF-β) plays a significant role in various ongoing cellular mechanisms. The gain or loss-of-function of TGF-β and its downstream mediators could lead to a plethora of diseases includes tumorigenesis. Specifically, at the early onset of malignancy TGF-β act as tumour suppressor and plays a key role in clearing malignant cells by reducing the cellular proliferation and differentiation thus triggers the process of apoptosis. Subsequently, TGF-β at an advanced stage of malignancy promotes tumorigenesis by augmenting cellular transformation, epithelial-mesenchymal-transition invasion, and metastasis. Besides playing the dual roles, depending upon the stage of malignancy, TGF-β also regulates cell fate through immune and stroma components. This oscillatory role of TGF-β to fight against cancer or act as a traitor to collaborate and crosstalk with other tumorigenic signaling pathways and its betrayal within the cell depends upon the cellular context. Therefore, the current review highlights and understands the dual role of TGF-β under different cellular conditions and its crosstalk with other signaling pathways in modulating cell fate.
Collapse
|
11
|
Chen Z, Xiang L, Li L, Ou H, Fang Y, Xu Y, Liu Q, Hu Z, Huang Y, Li X, Yang D. TGF-β1 induced deficiency of linc00261 promotes epithelial–mesenchymal-transition and stemness of hepatocellular carcinoma via modulating SMAD3. J Transl Med 2022; 20:75. [PMID: 35123494 PMCID: PMC8818189 DOI: 10.1186/s12967-022-03276-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 01/24/2022] [Indexed: 12/28/2022] Open
Abstract
Emerging evidence suggests that long non-coding RNAs (lncRNAs) play important roles in the metastasis and recurrence of hepatocellular carcinoma (HCC). A kinds of lncRNAs were found to be involved in regulating epithelial–mesenchymal transition (EMT) or stem-like traits in human cancers, however, the molecular mechanism and signaling pathways targeting EMT and stemness remains largely unknown. Previously, we found that linc00261 was down-regulated in HCC and associated with multiple worse clinical pathological parameters and poor prognosis. Here, we show that linc00261 was down-regulated in TGF-β1 stimulated cells, and forced expression of linc00261 attenuated EMT and stem-like traits in HCC. Linc00261 also inhibited the tumor sphere forming in vitro and decreased the tumorigenicity in vivo. Furthermore, we revealed that linc00261 suppressed the expression and phosphorylation of SMAD3 (p-SMAD3), which could be core transcriptional modulator in TGF-β1 signaling mediated EMT and the acquisition of stemness traits. A negative correlation between linc00261 and p-SMAD3 was determined in HCC samples. Conclusion: Our study revealed that linc00261 suppressed EMT and stem-like traits in HCC cells by inhibiting TGF-β1/SMAD3 signaling.
Collapse
|
12
|
Zaidi S, Gough NR, Mishra L. Mechanisms and clinical significance of TGF-β in hepatocellular cancer progression. Adv Cancer Res 2022; 156:227-248. [DOI: 10.1016/bs.acr.2022.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
13
|
Wang Z, Hopson LM, Singleton SS, Yang X, Jogunoori W, Mazumder R, Obias V, Lin P, Nguyen BN, Yao M, Miller L, White J, Rao S, Mishra L. Mice with dysfunctional TGF-β signaling develop altered intestinal microbiome and colorectal cancer resistant to 5FU. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166179. [PMID: 34082069 DOI: 10.1016/j.bbadis.2021.166179] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 05/13/2021] [Accepted: 05/17/2021] [Indexed: 12/20/2022]
Abstract
Emerging data show a rise in colorectal cancer (CRC) incidence in young men and women that is often chemoresistant. One potential risk factor is an alteration in the microbiome. Here, we investigated the role of TGF-β signaling on the intestinal microbiome and the efficacy of chemotherapy for CRC induced by azoxymethane and dextran sodium sulfate in mice. We used two genotypes of TGF-β-signaling-deficient mice (Smad4+/- and Smad4+/-Sptbn1+/-), which developed CRC with similar phenotypes and had similar alterations in the intestinal microbiome. Using these mice, we evaluated the intestinal microbiome and determined the effect of dysfunctional TGF-β signaling on the response to the chemotherapeutic agent 5-Fluoro-uracil (5FU) after induction of CRC. Using shotgun metagenomic sequencing, we determined gut microbiota composition in mice with CRC and found reduced amounts of beneficial species of Bacteroides and Parabacteroides in the mutants compared to the wild-type (WT) mice. Furthermore, the mutant mice with CRC were resistant to 5FU. Whereas the abundances of E. boltae, B.dorei, Lachnoclostridium sp., and Mordavella sp. were significantly reduced in mice with CRC, these species only recovered to basal amounts after 5FU treatment in WT mice, suggesting that the alterations in the intestinal microbiome resulting from compromised TGF-β signaling impaired the response to 5FU. These findings could have implications for inhibiting the TGF-β pathway in the treatment of CRC or other cancers.
Collapse
Affiliation(s)
- Zhuanhuai Wang
- Center for Translational Medicine, Department of Surgery, The George Washington University, Washington, DC, USA; Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lindsay M Hopson
- Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, DC, USA
| | - Stephanie S Singleton
- Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, DC, USA
| | - Xiaochun Yang
- Center for Translational Medicine, Department of Surgery, The George Washington University, Washington, DC, USA; The Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research & Cold Spring Harbor Laboratory, Department of Medicine, Division of Gastroenterology and Hepatology, Northwell Health, NY, USA
| | - Wilma Jogunoori
- Research and Development, Veterans Affairs Medical Center, Washington, DC, USA
| | - Raja Mazumder
- Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, DC, USA
| | - Vincent Obias
- Department of Surgery, The George Washington University, Washington, DC, USA
| | - Paul Lin
- Department of Surgery, The George Washington University, Washington, DC, USA
| | - Bao-Ngoc Nguyen
- Center for Translational Medicine, Department of Surgery, The George Washington University, Washington, DC, USA
| | - Michael Yao
- Department of Gastroenterology, Veterans Affairs Medical Center, Washington, DC, USA
| | - Larry Miller
- Department of Medicine, Division of Gastroenterology, Zucker School of Medicine at Hofstra/Northwell Health System, New Hyde Park, NY, USA
| | - Jon White
- Department of Surgery, The George Washington University, Washington, DC, USA
| | - Shuyun Rao
- Center for Translational Medicine, Department of Surgery, The George Washington University, Washington, DC, USA; The Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research & Cold Spring Harbor Laboratory, Department of Medicine, Division of Gastroenterology and Hepatology, Northwell Health, NY, USA.
| | - Lopa Mishra
- Center for Translational Medicine, Department of Surgery, The George Washington University, Washington, DC, USA; The Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research & Cold Spring Harbor Laboratory, Department of Medicine, Division of Gastroenterology and Hepatology, Northwell Health, NY, USA.
| |
Collapse
|
14
|
Li F, Xu J, Liu S. Cancer Stem Cells and Neovascularization. Cells 2021; 10:cells10051070. [PMID: 33946480 PMCID: PMC8147173 DOI: 10.3390/cells10051070] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/19/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cells (CSCs) refer to a subpopulation of cancer cells responsible for tumorigenesis, metastasis, and drug resistance. Increasing evidence suggests that CSC-associated tumor neovascularization partially contributes to the failure of cancer treatment. In this review, we discuss the roles of CSCs on tumor-associated angiogenesis via trans-differentiation or forming the capillary-like vasculogenic mimicry, as well as the roles of CSCs on facilitating endothelial cell-involved angiogenesis to support tumor progression and metastasis. Furthermore, we discuss the underlying regulation mechanisms, including the intrinsic signals of CSCs and the extrinsic signals such as cytokines from the tumor microenvironment. Further research is required to identify and verify some novel targets to develop efficient therapeutic approaches for more efficient cancer treatment through interfering CSC-mediated neovascularization.
Collapse
Affiliation(s)
- Fengkai Li
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Cancer Institutes, Fudan University, Shanghai 200032, China; (F.L.); (J.X.)
- Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
- The International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jiahui Xu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Cancer Institutes, Fudan University, Shanghai 200032, China; (F.L.); (J.X.)
- Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
- The International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Suling Liu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Cancer Institutes, Fudan University, Shanghai 200032, China; (F.L.); (J.X.)
- Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
- The International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Correspondence: ; Tel.: +86-21-34771023
| |
Collapse
|
15
|
Paraiso IL, Tran TQ, Magana AA, Kundu P, Choi J, Maier CS, Bobe G, Raber J, Kioussi C, Stevens JF. Xanthohumol ameliorates Diet-Induced Liver Dysfunction via Farnesoid X Receptor-Dependent and Independent Signaling. Front Pharmacol 2021; 12:643857. [PMID: 33959012 PMCID: PMC8093804 DOI: 10.3389/fphar.2021.643857] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/22/2021] [Indexed: 12/20/2022] Open
Abstract
The farnesoid X receptor (FXR) plays a critical role in the regulation of lipid and bile acid (BA) homeostasis. Hepatic FXR loss results in lipid and BA accumulation, and progression from hepatic steatosis to nonalcoholic steatohepatitis (NASH). This study aimed to evaluate the effects of xanthohumol (XN), a hop-derived compound mitigating metabolic syndrome, on liver damage induced by diet and FXR deficiency in mice. Wild-type (WT) and liver-specific FXR-null mice (FXRLiver−/−) were fed a high-fat diet (HFD) containing XN or the vehicle formation followed by histological characterization, lipid, BA and gene profiling. HFD supplemented with XN resulted in amelioration of hepatic steatosis and decreased BA concentrations in FXRLiver−/− mice, the effect being stronger in male mice. XN induced the constitutive androstane receptor (CAR), pregnane X receptor (PXR) and glucocorticoid receptor (GR) gene expression in the liver of FXRLiver−/− mice. These findings suggest that activation of BA detoxification pathways represents the predominant mechanism for controlling hydrophobic BA concentrations in FXRLiver−/− mice. Collectively, these data indicated sex-dependent relationship between FXR, lipids and BAs, and suggest that XN ameliorates HFD-induced liver dysfunction via FXR-dependent and independent signaling.
Collapse
Affiliation(s)
- Ines L Paraiso
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States.,Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States
| | - Thai Q Tran
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States
| | - Armando Alcazar Magana
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States.,Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States.,Department of Chemistry, Oregon State University, Corvallis, OR, United States
| | - Payel Kundu
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States
| | - Jaewoo Choi
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Claudia S Maier
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
| | - Gerd Bobe
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States.,Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR, United States
| | - Jacob Raber
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States.,Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States.,Department of Neurology, Psychiatry and Radiation Medicine, Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Portland, OR, United States
| | - Chrissa Kioussi
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States
| | - Jan F Stevens
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States.,Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States
| |
Collapse
|
16
|
Zhu C, Dong B, Sun L, Wang Y, Chen S. Cell Sources and Influencing Factors of Liver Regeneration: A Review. Med Sci Monit 2020; 26:e929129. [PMID: 33311428 PMCID: PMC7747472 DOI: 10.12659/msm.929129] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Liver regeneration (LR) is a set of complicated mechanisms between cells and molecules in which the processes of initiation, maintenance, and termination of liver repair are regulated. Although LR has been studied extensively, there are still numerous challenges in gaining its full understanding. Cells for LR have a wide range of sources and the feature of plasticity, and regeneration patterns are not the same under different conditions. Many patients undergoing partial hepatectomy develop cirrhosis or steatosis. The changes of LR in these cases are not clear. Many types of cells participate in LR. Hepatocytes, biliary epithelial cells, hepatic progenitor cells, and human liver stem cells can serve as the cell sources for LR. However, different types and degrees of damage trigger the response from the most suitable cells. Exploring the cell sources of LR is of great significance for accelerating recovery of liver function under different pathological patterns and developing a cell therapy strategy to cope with the shortage of donors for liver transplantation. In clinical practice, the background of the liver influences regeneration. Fibrosis and steatosis create different LR microenvironments and signal molecule interaction patterns. In addition, factors such as partial hepatectomy, aging, platelets, nerves, hormones, bile acids, and gut microbiota are widely involved in this process. Understanding the influencing factors of LR has practical value for individualized treatment of patients with liver diseases. In this review, we have summarized recent studies and proposed our views. We discuss cell sources and the influential factors on LR to help in solving clinical problems.
Collapse
Affiliation(s)
- Chengzhan Zhu
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland).,Shandong Key Laboratory of Digital Medicine and Computer Assisted Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Bingzi Dong
- Shandong Key Laboratory of Digital Medicine and Computer Assisted Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Leqi Sun
- Department of Oncological Medical Services, Institute of Health Sciences, Tokushima University of Graduate School, Tokushima City, Tokushima, Japan
| | - Yixiu Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Shuhai Chen
- Department of Surgery, Institute of Biomedical Sciences, Tokushima University of Graduate School, Tokushima City, Tokushima, Japan
| |
Collapse
|
17
|
Liao Z, Chen L, Zhang X, Zhang H, Tan X, Dong K, Lu X, Zhu H, Liu Q, Zhang Z, Ding Z, Dong W, Zhu P, Chu L, Liang H, Datta PK, Zhang B, Chen X. PTPRε Acts as a Metastatic Promoter in Hepatocellular Carcinoma by Facilitating Recruitment of SMAD3 to TGF-β Receptor 1. Hepatology 2020; 72:997-1012. [PMID: 31903610 DOI: 10.1002/hep.31104] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 12/17/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND AIMS Transforming growth factor beta (TGF-β) suppresses early stages of tumorigenesis, but contributes to the migration and metastasis of cancer cells. However, the role of TGF-β signaling in invasive prometastatic hepatocellular carcinoma (HCC) is poorly understood. In this study, we investigated the roles of canonical TGF-β/mothers against decapentaplegic homolog 3 (SMAD3) signaling and identified downstream effectors on HCC migration and metastasis. APPROACH AND RESULTS By using in vitro trans-well migration and invasion assays and in vivo metastasis models, we demonstrated that SMAD3 and protein tyrosine phosphatase receptor epsilon (PTPRε) promote migration, invasion, and metastasis of HCC cells in vitro and in vivo. Further mechanistic studies revealed that, following TGF-β stimulation, SMAD3 binds directly to PTPRε promoters to activate its expression. PTPRε interacts with TGFBR1/SMAD3 and facilitates recruitment of SMAD3 to TGFBR1, resulting in a sustained SMAD3 activation status. The tyrosine phosphatase activity of PTPRε is important for binding with TGFBR1, recruitment and activation of SMAD3, and its prometastatic role in vitro. A positive correlation between pSMAD3/SMAD3 and PTPRε expression was determined in HCC samples, and high expression of SMAD3 or PTPRε was associated with poor prognosis of patients with HCC. CONCLUSIONS PTPRε positive feedback regulates TGF-β/SMAD3 signaling to promote HCC metastasis.
Collapse
Affiliation(s)
- Zhibin Liao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Lin Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Xuewu Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Hongwei Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Xiaolong Tan
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Keshuai Dong
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Xun Lu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - He Zhu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Qiumeng Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Zhanguo Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Zeyang Ding
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Wei Dong
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Peng Zhu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Liang Chu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Huifang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Pran K Datta
- Division of Hematology and Oncology, Department of Medicine, UAB Comprehensive Cancer Center, Birmingham, AL.,Birmingham Veterans Affairs Medical Center, Birmingham, AL
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.,Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China.,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.,Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China.,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
18
|
Hyslip J, Martins PN. Liver Repair and Regeneration in Transplant: State of the Art. CURRENT TRANSPLANTATION REPORTS 2020. [DOI: 10.1007/s40472-020-00269-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
19
|
Guo H, Li P, Su L, Wu K, Huang K, Lai R, Xu J, Sun D, Li S, Deng Z, Wang Y, Guo H, Chen Z, Wang S. Low expression of IL-37 protein is correlated with high Oct4 protein expression in hepatocellular carcinoma. Gene 2020; 737:144445. [PMID: 32035244 DOI: 10.1016/j.gene.2020.144445] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 01/31/2020] [Accepted: 02/03/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The function of IL-37 in cancer remains largely unclear. The present research was to probe the protein expression of IL-37 and Oct4 in hepatocellular carcinoma (HCC), para-cancerous tissues (PT) and cancer cell lines, and discuss their relationship. METHODS Forty-nine HCC specimens and forty-nine PT samples were collected for immunohistochemical staining of IL-37 and Oct4 protein. Then, the correlations among IL-37, Oct4 and the clinical indicators were analyzed. In further in vitro studies, IL-37 was over expressed in HepG2 and MHCC97H cancer cell lines by gene transfection using a lipo3000 kit. Finally, the protein expression of IL-37 and Oct4 was detected by immunofluorescence and western blot to verify the in vivo correlation between IL and 37 and Oct4. RESULTS In HCC, IL-37 protein expression was weakly positive with a positive rate of 12.2% while Oct4 expression was strongly positive with a positive rate of 91.8%. In PT, strong positive IL-37 (83.7%) and weakly positive Oct4 (91.8%) were shown. The increased IL-37 and decreased Oct4 induced by IL-37 gene transfection were observed through IF in cells. In terms of clinical significance, the difference of IL-37 expression between HCC and PT was statistically significant (χ2 = 51.815, P = 3.2796 × 10-11). IL-37 in tumor tissues was associated with serum AFP (χ2 = 5.515, P = 0.048) and cirrhosis (χ2 = 7.451, P = 0.014). IL-37 expression of PT was link to gender (χ2 = 10.376, P = 0.013) and tumor size (χ2 = 8.118, P = 0.04). The expression of Oct4 in HCC was related to the patient's gender and cirrhosis. Importantly, there was a negative correlation between IL and 37 and Oct4 in tumor tissues (r = -0.299, P = 0.047) but not in PT (P > 0.05). Oct4 protein expression was down-regulated by IL-37 by 63.35% in HepG2 cells (P < 0.05) and 95.20% in MHCC97H cells (P < 0.05). CONCLUSION IL-37 expression in tumor tissues and PT was related to serum AFP and liver cirrhosis, tumor size, respectively. IL-37 protein expression was correlated with Oct4 in cancer cell lines and tumor tissues but not PT. The present study indicated that IL-37 might play a role in the development of HCC.
Collapse
Affiliation(s)
- Hongsheng Guo
- Department of Histology and Embryology of the Basic Medical College, Guangdong Medical University, Dongguan, Guangdong Province 523808, China
| | - Peng Li
- Department of Histology and Embryology of the Basic Medical College, Guangdong Medical University, Dongguan, Guangdong Province 523808, China
| | - Liudan Su
- Institute of Laboratory Medicine, Guangdong Medical University, Dongguan, Guangdong Province 523808, China
| | - Kun Wu
- Institute of Laboratory Medicine, Guangdong Medical University, Dongguan, Guangdong Province 523808, China
| | - Kai Huang
- Second Clinical Medical College, Guangdong Medical University, Dongguan, Guangdong Province 523808, China
| | - Ruizhi Lai
- Second Clinical Medical College, Guangdong Medical University, Dongguan, Guangdong Province 523808, China
| | - Jing Xu
- Second Clinical Medical College, Guangdong Medical University, Dongguan, Guangdong Province 523808, China
| | - Dingbao Sun
- Second Clinical Medical College, Guangdong Medical University, Dongguan, Guangdong Province 523808, China
| | - Shuxian Li
- Department of Histology and Embryology of the Basic Medical College, Guangdong Medical University, Dongguan, Guangdong Province 523808, China
| | - Ziliang Deng
- Department of Histology and Embryology of the Basic Medical College, Guangdong Medical University, Dongguan, Guangdong Province 523808, China
| | - Yan Wang
- Department of Histology and Embryology of the Basic Medical College, Guangdong Medical University, Dongguan, Guangdong Province 523808, China
| | - Haina Guo
- Dongguan Maternal and Child Healthcare Hospital, Dongguan, Guangdong Province 520300, China
| | - Zhangquan Chen
- Institute of Laboratory Medicine, Guangdong Medical University, Dongguan, Guangdong Province 523808, China
| | - Sen Wang
- Department of Histology and Embryology of the Basic Medical College, Guangdong Medical University, Dongguan, Guangdong Province 523808, China.
| |
Collapse
|
20
|
El-Houseini ME, Ismail A, Abdelaal AA, El-Habashy AH, Abdallah ZF, Mohamed MZ, El-Hadidi M, Cho WCS, Ahmed H, Al-Shafie TA. Role of TGF-β1 and C-Kit Mutations in the Development of Hepatocellular Carcinoma in Hepatitis C Virus-Infected Patients: in vitro Study. BIOCHEMISTRY (MOSCOW) 2019; 84:941-953. [PMID: 31522676 DOI: 10.1134/s0006297919080108] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Transforming growth factor beta (TGF-β) acts as a tumor-suppressing cytokine in healthy tissues and non-malignant tumors. Yet, in malignancy, TGF-β can exert the opposite effects that can promote proliferation of cancer cells. C-Kit plays a prominent role in stem cell activation and liver regeneration after injury. However, little is known about the cross-talk between TGF-β and C-Kit and its role in the progression of hepatocellular carcinoma (HCC). Here, we studied the effect of increasing doses of TGF-β1 on CD44+CD90+ liver stem cells (LSCs) and C-Kit gene expression in malignant and adjacent non-malignant liver tissues excised from 32 HCC patients. The percentage of LSCs in malignant tumors was two times higher compared to their counterparts from the non-malignant tissues. When treated with increasing doses of TGF-β1, proliferation of both malignant and non-malignant LSCs was progressively suppressed, but low TGF-β1 dose failed to suppress the growth of malignant LSCs. Moreover, C-Kit exons 9 and 11 were expressed in malignant LSCs, but not in their non-malignant counterparts. Analysis of C-Kit detected mutations in exon 9 (but not in exon 11) in some malignant liver cells resulting in the changes in the amino acid sequence and dysregulation of protein structure and function. Interestingly, in malignant liver cells, mutations in exon 9 were associated with high-viremia hepatitis C virus (HCV), and expression of this exon was not suppressed by the TGF-β1 treatment at all doses. To our knowledge, this is the first report that mutations in the C-Kit gene in HCC patients are associated with high- viremia HCV. Our study emphasizes the need for investigation of the TGF-β1 level and C-Kit mutations in patients with chronic HCV for HCC prevention and better therapy management.
Collapse
Affiliation(s)
- M E El-Houseini
- Cairo University, National Cancer Institute, Department of Cancer Biology, Cairo, 11796, Egypt
| | - A Ismail
- Ain Shams University, Faculty of Medicine, Department of Surgery, Cairo, 11566, Egypt
| | - A A Abdelaal
- Ain Shams University, Faculty of Medicine, Department of Surgery, Cairo, 11566, Egypt
| | - A H El-Habashy
- Cairo University, National Cancer Institute, Department of Pathology, Cairo, 11796, Egypt
| | - Z F Abdallah
- Cairo University, National Cancer Institute, Department of Cancer Biology, Cairo, 11796, Egypt
| | - M Z Mohamed
- Medical Center of Egyptian Railways, Department of Medical Laboratory, Cairo, 11669, Egypt
| | - M El-Hadidi
- Nile University, Center of Informatics Science, Giza, 12525, Egypt
| | - W C S Cho
- Queen Elizabeth Hospital, Department of Clinical Oncology, Kowloon, Hong Kong, China
| | - H Ahmed
- GlycoMantra, Inc., Baltimore, MD 21227, USA
| | - T A Al-Shafie
- Cairo University, National Cancer Institute, Department of Cancer Biology, Cairo, 11796, Egypt. .,Pharos University in Alexandria, Faculty of Pharmacy and Drug Manufacturing, Department of Pharmacology and Therapeutics, Alexandria, 21311, Egypt
| |
Collapse
|
21
|
Valizadeh A, Majidinia M, Samadi-Kafil H, Yousefi M, Yousefi B. The roles of signaling pathways in liver repair and regeneration. J Cell Physiol 2019; 234:14966-14974. [PMID: 30770551 DOI: 10.1002/jcp.28336] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 12/23/2018] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
The liver has remarkable regeneration potency that restores liver mass and sustains body hemostasis. Liver regeneration through signaling pathways following resection or moderate damages are well studied. Various cell signaling, growth factors, cytokines, receptors, and cell types implicated in liver regeneration undergo controlled hypertrophy and proliferation. Some aspects of liver regeneration have been discovered and many investigations have been carried out to identify its mechanisms. However, for optimizing liver regeneration more should be understood about mechanisms that control the growth of hepatocytes and other liver cell types in adults. The current paper deals with the possible applicability of liver regeneration signaling pathways as a target for therapeutic approaches and preventing various liver damages. Furthermore, the latest findings of spectrum-specific signaling pathway mechanisms that underlie liver regeneration are briefly described.
Collapse
Affiliation(s)
- Amir Valizadeh
- Stem Cells Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Hossein Samadi-Kafil
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
22
|
Qiu Y, Shan W, Yang Y, Jin M, Dai Y, Yang H, Jiao R, Xia Y, Liu Q, Ju L, Huang G, Zhang J, Yang L, Li L, Li Y. Reversal of sorafenib resistance in hepatocellular carcinoma: epigenetically regulated disruption of 14-3-3η/hypoxia-inducible factor-1α. Cell Death Discov 2019; 5:120. [PMID: 31341646 PMCID: PMC6642098 DOI: 10.1038/s41420-019-0200-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 06/15/2019] [Accepted: 06/23/2019] [Indexed: 02/07/2023] Open
Abstract
Sorafenib resistance is one of the main obstacles to the treatment of advanced/recurrent hepatocellular carcinoma (HCC). Here, sorafenib-resistant HCC cells and xenografts in nude mice were used as experimental models. A cohort of patients with advanced recurrent HCC who were receiving sorafenib therapy was used to assess the clinical significance of this therapy. Our data showed that 14-3-3η maintained sorafenib resistance in HCC. An analysis of the underlying molecular mechanisms revealed that 14-3-3η stabilizes hypoxia-inducible factor 1α (HIF-1α) through the inhibition of ubiquitin-dependent proteasome protein degradation, which leads to the maintenance of cancer stem cell (CSC) properties. We further found that microRNA-16 (miR-16) is a competent miRNA that reverses sorafenib resistance by targeting the 3'-UTR of 14-3-3η and thereby inhibits 14-3-3η/HIF-1α/CSC properties. In HCC patients, significant negative correlations were found between the expression of miR-16 and 14-3-3η, HIF-1α, or CSC properties. Further analysis showed that low miR-16 expression but high 14-3-3η expression can prognosticate sorafenib resistance and poor survival. Collectively, our present study indicated that miR-16/14-3-3η is involved in sorafenib resistance in HCC and that these two factors could be potential therapeutic targets and biomarkers for predicting the response to sorafenib treatment.
Collapse
Affiliation(s)
- Yongxin Qiu
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166 China
- Department of Medical Center for Digestive Diseases, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011 China
| | - Wenqi Shan
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166 China
- Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166 China
| | - Ye Yang
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166 China
- Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166 China
| | - Ming Jin
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166 China
- Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166 China
| | - Yi Dai
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166 China
- Department of Medical Center for Digestive Diseases, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011 China
| | - Hanyu Yang
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166 China
- Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166 China
| | - Ruonan Jiao
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166 China
- Department of Medical Center for Digestive Diseases, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011 China
| | - Yunwei Xia
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166 China
- Department of Medical Center for Digestive Diseases, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011 China
| | - Qinqiang Liu
- Department of Medical Center for Digestive Diseases, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011 China
| | - Liang Ju
- Department of Medical Center for Digestive Diseases, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011 China
| | - Guangming Huang
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166 China
- Department of Medical Center for Digestive Diseases, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011 China
| | - Jianping Zhang
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166 China
- Department of Medical Center for Digestive Diseases, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011 China
| | - Lihua Yang
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166 China
- Department of Medical Center for Digestive Diseases, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011 China
| | - Lei Li
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166 China
- Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166 China
| | - Yuan Li
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166 China
- Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166 China
| |
Collapse
|
23
|
Chen S, Li J, Zhou P, Zhi X. SPTBN1 and cancer, which links? J Cell Physiol 2019; 235:17-25. [PMID: 31206681 DOI: 10.1002/jcp.28975] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 05/28/2019] [Indexed: 12/16/2022]
Abstract
SPTBN1 is a dynamic intracellular nonpleckstrin homology-domain protein, functioning as a transforming growth factor-β signal transducing adapter protein which is necessary to form Smad3/Smad4 complex. Recently SPTBN1 is considered to be associated with many kinds of cancers. SPTBN1 expression and function differ between different tumor states or types. This review summarizes the recent advances in the expression patterns of SPTBN1 in cancers, and in understanding the mechanisms by which SPTBN1 affects the occurrence, progression, and metastasis of cancer. Identifying SPTBN1 expression and function in cancers will contribute to the clinical diagnosis and treatment of cancer and the investigation of anticancer drugs.
Collapse
Affiliation(s)
- Shuyi Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jiajia Li
- Department of Gynecology, Affiliated Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Ping Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiuling Zhi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
24
|
Ogunwobi OO, Harricharran T, Huaman J, Galuza A, Odumuwagun O, Tan Y, Ma GX, Nguyen MT. Mechanisms of hepatocellular carcinoma progression. World J Gastroenterol 2019; 25:2279-2293. [PMID: 31148900 PMCID: PMC6529884 DOI: 10.3748/wjg.v25.i19.2279] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 03/27/2019] [Accepted: 04/10/2019] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary malignancy of the liver. It is the second leading cause of cancer-related deaths worldwide, with a very poor prognosis. In the United States, there has been only minimal improvement in the prognosis for HCC patients over the past 15 years. Details of the molecular mechanisms and other mechanisms of HCC progression remain unclear. Consequently, there is an urgent need for better understanding of these mechanisms. HCC is often diagnosed at advanced stages, and most patients will therefore need systemic therapy, with sorafenib being the most common at the present time. However, sorafenib therapy only minimally enhances patient survival. This review provides a summary of some of the known mechanisms that either cause HCC or contribute to its progression. Included in this review are the roles of viral hepatitis, non-viral hepatitis, chronic alcohol intake, genetic predisposition and congenital abnormalities, toxic exposures, and autoimmune diseases of the liver. Well-established molecular mechanisms of HCC progression such as epithelial-mesenchymal transition, tumor-stromal interactions and the tumor microenvironment, cancer stem cells, and senescence bypass are also discussed. Additionally, we discuss the roles of circulating tumor cells, immunomodulation, and neural regulation as potential new mechanisms of HCC progression. A better understanding of these mechanisms could have implications for the development of novel and more effective therapeutic and prognostic strategies, which are critically needed.
Collapse
Affiliation(s)
- Olorunseun O Ogunwobi
- Department of Biological Sciences, Hunter College of The City University of New York, New York, NY 10065, United States
- The Graduate Center Departments of Biology and Biochemistry, The City University of New York, New York, NY 10016, United States
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10065, United States
- Hunter College Center for Cancer Health Disparities Research (CCHDR), New York, NY 10065, United States
| | - Trisheena Harricharran
- Department of Biological Sciences, Hunter College of The City University of New York, New York, NY 10065, United States
- The Graduate Center Departments of Biology and Biochemistry, The City University of New York, New York, NY 10016, United States
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10065, United States
- Hunter College Center for Cancer Health Disparities Research (CCHDR), New York, NY 10065, United States
| | - Jeannette Huaman
- Department of Biological Sciences, Hunter College of The City University of New York, New York, NY 10065, United States
- The Graduate Center Departments of Biology and Biochemistry, The City University of New York, New York, NY 10016, United States
- Hunter College Center for Cancer Health Disparities Research (CCHDR), New York, NY 10065, United States
| | - Anna Galuza
- Department of Biological Sciences, Hunter College of The City University of New York, New York, NY 10065, United States
- Hunter College Center for Cancer Health Disparities Research (CCHDR), New York, NY 10065, United States
| | - Oluwatoyin Odumuwagun
- Department of Biological Sciences, Hunter College of The City University of New York, New York, NY 10065, United States
- Hunter College Center for Cancer Health Disparities Research (CCHDR), New York, NY 10065, United States
| | - Yin Tan
- Center for Asian Health, School of Medicine, Temple University, Philadelphia, PA 19140, United States
| | - Grace X Ma
- Center for Asian Health, School of Medicine, Temple University, Philadelphia, PA 19140, United States
| | - Minhhuyen T Nguyen
- Department of Medicine, Fox Chase Cancer Center, Philadelphia, PA 19111, United States
| |
Collapse
|
25
|
Subcutaneous Inoculation of Echinococcus multilocularis Induces Delayed Regeneration after Partial Hepatectomy. Sci Rep 2019; 9:462. [PMID: 30679666 PMCID: PMC6345980 DOI: 10.1038/s41598-018-37293-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 12/04/2018] [Indexed: 02/07/2023] Open
Abstract
Alveolar echinococcosis (AE) is caused by the larval stage of echinococcus multilocularis (E. multilocularis), and hepatectomy is the main modality in hepatic AE patients. Liver regeneration after partial hepatectomy (PHx) in such patients is challenging, and further investigation is needed. Thus far, knowledge regarding the possible impact of E. multilocularis on liver regeneration after PHx is limited. Herein, a subcutaneous infection model of E. multilocularis was developed in C57 BL/6 mice, and after 3 months, PHx was performed. Plasma and liver samples were harvested under inhalational isofluorane (2%) anaesthesia at designated post-PHx time points (0, 24, 48, 96 and 168 h). The parameters included the future remnant liver/body weight ratio (FLR/BW), liver function tests (AST and ALT) and related cytokines (TNF-α, IL-6, Factor V, HMGB1, TGF-β, TSP-1, and TLR4) and proteins (MyD88 and STAT3). To assess the proliferation intensity of hepatocytes, BrdU, Ki67 and PAS staining were carried out in regenerated liver tissue. The FLR/BW in the infected group from 48 h after surgery was lower than that in the control group. The BrdU positive hepatocyte proportions reached their peak at 48 h in the control group and 96 h in the infected group and then gradually decreased. During the first 48 h after surgery, both the AST and ALT levels in the infected group were lower; however, these levels were altered from 96 h after surgery. In the infected group, the concentrations and mRNA expression levels of the pre-inflammatory cytokines TNF-α and IL-6 demonstrated a delayed peak. Moreover, post-operatively, the TGF-β and TSP-1 levels showed high levels in the infected group at each different time-point compared to those in the control group; however, high levels of TGF-β were observed at 96 h in the control group. The MyD88 and STAT3 protein expression levels in the infected group were markedly higher than those in the control group 96 h after surgery. Delayed liver regeneration after PHx was observed in the C57 BL/6 mice with the subcutaneous infection of E. multilocularis in the current study. This phenomenon could be partially explained by the alteration in the pro-inflammatory cytokines in the immunotolerant milieu induced by chronic E. multilocularis infection.
Collapse
|
26
|
Wang N, Wang S, Li MY, Hu BG, Liu LP, Yang SL, Yang S, Gong Z, Lai PBS, Chen GG. Cancer stem cells in hepatocellular carcinoma: an overview and promising therapeutic strategies. Ther Adv Med Oncol 2018; 10:1758835918816287. [PMID: 30622654 PMCID: PMC6304707 DOI: 10.1177/1758835918816287] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 11/06/2018] [Indexed: 12/12/2022] Open
Abstract
The poor clinical outcome of hepatocellular carcinoma (HCC) patients is ascribed to the resistance of HCC cells to traditional treatments and tumor recurrence after curative therapies. Cancer stem cells (CSCs) have been identified as a small subset of cancer cells which have high capacity for self-renewal, differentiation and tumorigenesis. Recent advances in the field of liver CSCs (LCSCs) have enabled the identification of CSC surface markers and the isolation of CSC subpopulations from HCC cells. Given their central role in cancer initiation, metastasis, recurrence and therapeutic resistance, LCSCs constitute a therapeutic opportunity to achieve cure and prevent relapse of HCC. Thus, it is necessary to develop therapeutic strategies to selectively and efficiently target LCSCs. Small molecular inhibitors targeting the core stemness signaling pathways have been actively pursued and evaluated in preclinical and clinical studies. Other alternative therapeutic strategies include targeting LCSC surface markers, interrupting the CSC microenvironment, and altering the epigenetic state. In this review, we summarize the properties of CSCs in HCC and discuss novel therapeutic strategies that can be used to target LCSCs.
Collapse
Affiliation(s)
- Nuozhou Wang
- Department of Surgery, The Chinese University of
Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong SAR,
China
| | - Shanshan Wang
- Department of Otorhinolaryngology, Head and Neck
Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Prince of
Wales Hospital, Hong Kong, China
| | - Ming-Yue Li
- Department of Surgery, Faculty of Medicine, The
Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong,
China
- Shenzhen Research Institute, The Chinese
University of Hong Kong, Shenzhen, Guangdong, China
| | - Bao-guang Hu
- Department of Gastrointestinal Surgery, The
Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong,
China
| | - Li-ping Liu
- Department of Hepatobiliary and Pancreas
Surgery, The Second Clinical Medical College of Jinan University (Shenzhen
People’s Hospital), Shenzhen, Guangdong Province, China
| | - Sheng-li Yang
- Cancer Center, Union Hospital, Tongji Medical
College, Huazhong University of Science and Technology, Wuhan, China
| | - Shucai Yang
- Department of Clinical Laboratory, Pingshan
District People’s Hospital of Shenzhen, Shenzhen, Guangdong Province,
China
| | - Zhongqin Gong
- Department of Surgery, The Chinese University of
Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong SAR,
China
| | - Paul B. S. Lai
- Department of Surgery, The Chinese University
of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong
SAR, China
| | - George G. Chen
- Department of Surgery, The Chinese University
of Hong Kong, Prince of Wales Hospital, Shatin, New Territories, Hong Kong
SAR, China
- Shenzhen Research Institute, The Chinese
University of Hong Kong, Shenzhen, Guangdong, China
| |
Collapse
|
27
|
Yang L, Zhang S, Wang G. Keratin 17 in disease pathogenesis: from cancer to dermatoses. J Pathol 2018; 247:158-165. [PMID: 30306595 DOI: 10.1002/path.5178] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 09/17/2018] [Accepted: 10/02/2018] [Indexed: 12/16/2022]
Abstract
Keratin 17 (K17) is a type I intermediate filament mainly expressed in the basal cells of epithelia. As a multifaceted cytoskeletal protein, K17 regulates a myriad of biological processes, including cell proliferation and growth, skin inflammation and hair follicle cycling. Aberrant overexpression of K17 is found in various diseases ranging from psoriasis to malignancies such as breast, cervical, oral squamous and gastric carcinomas. Moreover, genetic mutation in KRT17 is related to tissue-specific diseases, represented by steatocystoma multiplex and pachyonychia congenita. In this review, we summarize our findings concerning the regulatory mechanisms of K17 overexpression in psoriasis and compare them to the literature relating to other diseases. We discuss data that proinflammatory cytokines, including interleukin-17 (IL-17), IL-22, interferon-gamma (IFN-γ), transforming growth factor-beta (TGF-β) and transcription factors glioma-associated oncogene homolog 1/2 (Gli1/2), Nrf2 and p53 can regulate K17 by transcriptional and translational control. Moreover, post-translational modification, including phosphorylation and ubiquitination, is involved in the regulation of K17 stability and biological functions. We therefore review the current understanding of the K17 regulatory mechanism and its pathogenic role in diseases from dermatoses to cancer. Prospects for anti-K17 therapy in diagnosis, prognosis and disease treatment are also discussed. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Luting Yang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR, China
| | - Shaolong Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR, China
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR, China
| |
Collapse
|
28
|
Newsted D, Banerjee S, Watt K, Nersesian S, Truesdell P, Blazer LL, Cardarelli L, Adams JJ, Sidhu SS, Craig AW. Blockade of TGF-β signaling with novel synthetic antibodies limits immune exclusion and improves chemotherapy response in metastatic ovarian cancer models. Oncoimmunology 2018; 8:e1539613. [PMID: 30713798 DOI: 10.1080/2162402x.2018.1539613] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 09/26/2018] [Accepted: 10/04/2018] [Indexed: 12/21/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is a leading cause of cancer-related death in women. EOC is often diagnosed at late stages, with peritoneal metastases and ascites production. Current surgery and platinum-based chemotherapy regimes fail to prevent recurrence in most patients. High levels of Transforming growth factor-β (TGF-β) within ascites has been linked to poor prognosis. TGF-β signaling promotes epithelial-mesenchymal transition (EMT) in EOC tumor cells, and immune suppression within the tumor microenvironment, with both contributing to chemotherapy resistance and metastasis. The goal of this study was to develop specific synthetic inhibitory antibodies to the Type II TGF-β receptor (TGFBR2), and test these antibodies in EOC cell and tumor models. Following screening of a phage-displayed synthetic antigen-binding fragment (Fab) library with the extracellular domain of TGFBR2, we identified a lead inhibitory Fab that suppressed TGF-β signaling in mouse and human EOC cell lines. Affinity maturation of the lead inhibitory Fab resulted in several derivative Fabs with increased affinity for TGFBR2 and efficacy as suppressors of TGF-β signaling, EMT and EOC cell invasion. In EOC xenograft and syngeneic tumor models, blockade of TGFBR2 with our lead antibodies led to improved chemotherapy response. This correlated with reversal of EMT and immune exclusion in these tumor models with TGFBR2 blockade. Together, these results describe new inhibitors of the TGF-β pathway that improve antitumor immunity, and response to chemotherapy in preclinical EOC models.
Collapse
Affiliation(s)
- Daniel Newsted
- Department of Biomedical and Molecular Sciences, Queen's University; Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada
| | | | - Kathleen Watt
- Department of Biomedical and Molecular Sciences, Queen's University; Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada
| | - Sarah Nersesian
- Department of Biomedical and Molecular Sciences, Queen's University; Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada
| | - Peter Truesdell
- Department of Biomedical and Molecular Sciences, Queen's University; Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada
| | - Levi L Blazer
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Lia Cardarelli
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Jarrett J Adams
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Sachdev S Sidhu
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Andrew W Craig
- Department of Biomedical and Molecular Sciences, Queen's University; Cancer Biology & Genetics division, Queen's Cancer Research Institute, Kingston, ON, Canada
| |
Collapse
|
29
|
Wang X, Chen X, Sun L, Bi X, He H, Chen L, Pang J. The function of MMP-28/TGF-β induced cell apoptosis in human glioma cells. Exp Ther Med 2018; 16:2867-2874. [PMID: 30214508 PMCID: PMC6125874 DOI: 10.3892/etm.2018.6566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 07/02/2018] [Indexed: 12/22/2022] Open
Abstract
The aim of the present study was to assess the expression status of matrix metalloproteinase (MMP)-28 and to investigate its molecular mechanisms in glioma cells. MicroRNA (miRNA) reverse transcription-quantitative polymerase chain reaction was used to analyze the expression of MMP-28 and transforming growth factor (TGF)-β expression in glioma patients and healthy volunteers. MTT and Transwell assays were conducted to determine cell growth and metastasis, respectively. Annexin V/propidium iodide staining was also employed to measure cell apoptosis. MMP-28 and TGF-β protein expression were measured using western Blot analysis. The results indicated that MMP-28 and TGF-β expression was downregulated in glioma patients, when compared with the normal group. Overall survival and disease-free survival of patients with a low expression of MMP-28 were lower than those with high MMP-28 expression. Overexpression of MMP-28 induced TGF-β protein expression, while downregulation of MMP-28 suppressed TGF-β protein expression in glioma cell. The downregulation of MMP-28 reduced the cell growth and apoptosis of glioma cell via the suppression of TGF-β. By contrast, upregulation of MMP-28 induced cell growth and reduced the apoptosis of glioma cells by activating TGF-β. In addition, the TGF-β inhibitor attenuated the effects of MMP-28 in glioma cells. Collectively, the results indicated that MMP-28 was able to induce TGF-β in human glioma cells.
Collapse
Affiliation(s)
- Xuepeng Wang
- Department of Neurosurgery, Affiliated Hospital of Beihua University, Jilin 132000, P.R. China
| | - Xi Chen
- Department of Neurology, Affiliated Hospital of Beihua University, Jilin 132000, P.R. China
| | - Lin Sun
- Department of Production, Affiliated Hospital of Beihua University, Jilin 132000, P.R. China
| | - Xiaoli Bi
- Department of CT, Affiliated Hospital of Beihua University, Jilin 132000, P.R. China
| | - Haitao He
- Department of Cycle of Internal, Affiliated Hospital of Beihua University, Jilin 132000, P.R. China
| | - Lei Chen
- Department of Neurosurgery, Affiliated Hospital of Beihua University, Jilin 132000, P.R. China
| | - Jinfeng Pang
- Department of Neurosurgery, Affiliated Hospital of Beihua University, Jilin 132000, P.R. China
| |
Collapse
|
30
|
Fabregat I, Caballero-Díaz D. Transforming Growth Factor-β-Induced Cell Plasticity in Liver Fibrosis and Hepatocarcinogenesis. Front Oncol 2018; 8:357. [PMID: 30250825 PMCID: PMC6139328 DOI: 10.3389/fonc.2018.00357] [Citation(s) in RCA: 232] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/13/2018] [Indexed: 12/11/2022] Open
Abstract
The Transforming Growth Factor-beta (TGF-β) family plays relevant roles in the regulation of different cellular processes that are essential for tissue and organ homeostasis. In the case of the liver, TGF-β signaling participates in different stages of disease progression, from initial liver injury toward fibrosis, cirrhosis and cancer. When a chronic injury takes place, mobilization of lymphocytes and other inflammatory cells occur, thus setting the stage for persistence of an inflammatory response. Macrophages produce profibrotic mediators, among them, TGF-β, which is responsible for activation -transdifferentiation- of quiescent hepatic stellate cells (HSC) to a myofibroblast (MFB) phenotype. MFBs are the principal source of extracellular matrix protein (ECM) accumulation and prominent mediators of fibrogenesis. TGF-β also mediates an epithelial-mesenchymal transition (EMT) process in hepatocytes that may contribute, directly or indirectly, to increase the MFB population. In hepatocarcinogenesis, TGF-β plays a dual role, behaving as a suppressor factor at early stages, but contributing to later tumor progression, once cells escape from its cytostatic effects. As part of its potential pro-tumorigenic actions, TGF-β induces EMT in liver tumor cells, which increases its pro-migratory and invasive potential. In parallel, TGF-β also induces changes in tumor cell plasticity, conferring properties of a migratory tumor initiating cell (TIC). The main aim of this review is to shed light about the pleiotropic actions of TGF-β that explain its effects on the different liver cell populations. The cross-talk with other signaling pathways that contribute to TGF-β effects, in particular the Epidermal Growth Factor Receptor (EGFR), will be presented. Finally, we will discuss the rationale for targeting the TGF-β pathway in liver pathologies.
Collapse
Affiliation(s)
- Isabel Fabregat
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute, Barcelona, Spain.,Department of Physiological Sciences, School of Medicine, University of Barcelona, Barcelona, Spain.,Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Barcelona, Spain
| | - Daniel Caballero-Díaz
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute, Barcelona, Spain.,Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Barcelona, Spain
| |
Collapse
|
31
|
Abstract
Resistance to chemotherapy and cancer relapse are major clinical challenges attributed to a sub population of cancer stem cells (CSCs). The concept of CSCs has been the subject of intense research by the oncology community since evidence for their existence was first published over twenty years ago. Emerging data indicates that they are also able to evade novel therapies such as targeted agents, immunotherapies and anti-angiogenics. The inability to appropriately identify and isolate CSCs is a major hindrance to the field and novel technologies are now being utilized. Agents that target CSC-associated cell surface receptors and signaling pathways have generated promising pre-clinical results and are now entering clinical trial. Here we discuss and evaluate current therapeutic strategies to target CSCs.
Collapse
Affiliation(s)
- Stephanie Annett
- Molecular and Cellular Therapeutics, Royal College of Surgeons Ireland, Ireland
| | - Tracy Robson
- Molecular and Cellular Therapeutics, Royal College of Surgeons Ireland, Ireland.
| |
Collapse
|
32
|
Yao J, Dai Q, Liu Z, Zhou L, Xu J. Circular RNAs in Organ Fibrosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1087:259-273. [DOI: 10.1007/978-981-13-1426-1_21] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|