1
|
Chen H, Nisar MA, Mulla J, Li X, Cao K, Lu S, Nagaoka K, Wu S, Ting PS, Tseng TS, Lin HY, Yin XM, Feng W, Wu Z, Cheng Z, Mueller W, Bay A, Schechner L, Bai X, Huang CK. Liver TET1 promotes metabolic dysfunction-associated steatotic liver disease. EMBO Mol Med 2025; 17:1101-1117. [PMID: 40164757 DOI: 10.1038/s44321-025-00224-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 03/07/2025] [Accepted: 03/12/2025] [Indexed: 04/02/2025] Open
Abstract
Global hepatic DNA methylation change has been linked to human patients with metabolic dysfunction-associated steatotic liver disease (MASLD). DNA demethylation is regulated by the TET family proteins, whose enzymatic activities require 2-oxoglutarate (2-OG) and iron that both are elevated in human MASLD patients. We aimed to investigate liver TET1 in MASLD progression. Depleting TET1 using two different strategies substantially alleviated MASLD progression. Knockout (KO) of TET1 slightly improved diet induced obesity and glucose homeostasis. Intriguingly, hepatic cholesterols, triglycerides, and CD36 were significantly decreased upon TET1 depletion. Consistently, liver specific TET1 KO led to improvement of MASLD progression. Mechanistically, TET1 promoted CD36 expression through transcriptional upregulation via DNA demethylation control. Overexpression of CD36 reversed the impacts of TET1 downregulation on fatty acid uptake in hepatocytes. More importantly, targeting TET1 with a small molecule inhibitor significantly suppressed MASLD progression. Conclusively, liver TET1 plays a deleterious role in MASLD, suggesting the potential of targeting TET1 in hepatocytes to suppress MASLD.
Collapse
Affiliation(s)
- Hongze Chen
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Pancreatic and Biliary Surgery, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Muhammad Azhar Nisar
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Joud Mulla
- Liver Research Center, Division of Gastroenterology & Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI, USA
| | - Xinjian Li
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Pancreatic and Biliary Surgery, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Kevin Cao
- Liver Research Center, Division of Gastroenterology & Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI, USA
| | - Shaolei Lu
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, USA
| | - Katsuya Nagaoka
- Liver Research Center, Division of Gastroenterology & Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI, USA
| | - Shang Wu
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Peng-Sheng Ting
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Tung-Sung Tseng
- School of Public Health, Louisiana State University Health Sciences Center, New Orleans, USA
| | - Hui-Yi Lin
- School of Public Health, Louisiana State University Health Sciences Center, New Orleans, USA
| | - Xiao-Ming Yin
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Wenke Feng
- Department Structural Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Zhijin Wu
- Department of Biostatistics, School of Public Health, Brown University, Providence, RI, USA
| | - Zhixiang Cheng
- Liver Research Center, Division of Gastroenterology & Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI, USA
| | - William Mueller
- Liver Research Center, Division of Gastroenterology & Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI, USA
| | - Amalia Bay
- Liver Research Center, Division of Gastroenterology & Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI, USA
| | - Layla Schechner
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Xuewei Bai
- Department of Pancreatic and Biliary Surgery, First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang Province, China
- Liver Research Center, Division of Gastroenterology & Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI, USA
| | - Chiung-Kuei Huang
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
2
|
Da FF, Meng YT, Chen YF, Yuan ZW, Liu Y, Dai ZH. Tetracera asiatica flavonoids attenuate alcohol-induced liver injury by suppressing oxidative stress and inflammation mediated by the Keap-1/Nrf2/HO-1, NF-κB/MAPK and PERK/Nrf2 signaling pathways in alcoholic liver injury rats. Tissue Cell 2025; 96:102913. [PMID: 40334394 DOI: 10.1016/j.tice.2025.102913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/26/2025] [Accepted: 04/08/2025] [Indexed: 05/09/2025]
Abstract
Alcoholic liver disease is regarded as a leading reason for liver cirrhosis. This study aimed to investigate the protective effect of tetracera asiatica flavonoids (TAF) on alcoholic liver injury (ALI) and explore the associated mechanisms. An ALI rat model was established and then divided into four groups, including ALI group, low-dose TAF (l-TAF) group, medium-dose TAF (m-TAF) group, and high-dose TAF (h-TAF) group. Levels of ALT, AST, ALB, SOD, MDA, NO, CAT, TG, TNF-α, IL-1β, Nrf2, Keap1, HO-1, NQO-1, and GSH-Px were measured in ALI rats in different groups. Pathological changes and inflammatory infiltration were examined using HE staining. Western blot was used to detect expressions of Nrf2, MAPK p38, PERK, NF-κB, ERK1/2 and anti-JNK1/2/3. The results showed that TAF protected against alcoholic liver injury in ALI rats by decreasing ALT and AST levels and inhibiting inflammatory response. TAF significantly reversed alcohol-induced increase in NO (P < 0.05), and remarkably decreased levels of TNF-α (P < 0.001) and IL-1β (P < 0.01), compared with the ALI group. TAF significantly increased the transcription of Nrf2, Keap1, HO-1, NQO-1 and GSH-Px gene (all P < 0.05) and inhibited the alcohol-induced upregulation of MAPK p38 expression (P < 0.001), p-NF-κB/NF-κB ratio (P < 0.001), p-ERK/1/2/ERK1/2 ratio (P < 0.05), and p-JNK1/2/3/JNK1/2/2 ratio (P < 0.05), compared with the ALI group (all P < 0.001). TAF obviously reversed effects of ALI modeling, and remarkably downregulated the expression of PERK and upregulated Nrf2 (all P < 0.001) compared with the ALI rats. In conclusion, TAF attenuates alcohol-induced livery injury through suppressing Keap-1/Nrf2/HO-1, NF-κB/MAPK and PERK/Nrf2 signaling pathways mediated oxidative stress and inflammation in ALI rats.
Collapse
Affiliation(s)
- Fang-Fang Da
- School of Chinese Ethnic Medicine, Guizhou Minzu University, Guiyang 550025, China; Key Laboratory of Guizhou Ethnic Medicine Resource Development and Utilization in Guizhou Minzu University, State Ethnic Affairs Commission, Guiyang 550025, China.
| | - Yao-Ting Meng
- School of Chinese Ethnic Medicine, Guizhou Minzu University, Guiyang 550025, China; Key Laboratory of Guizhou Ethnic Medicine Resource Development and Utilization in Guizhou Minzu University, State Ethnic Affairs Commission, Guiyang 550025, China
| | - Yu-Feng Chen
- School of Chinese Ethnic Medicine, Guizhou Minzu University, Guiyang 550025, China; Key Laboratory of Guizhou Ethnic Medicine Resource Development and Utilization in Guizhou Minzu University, State Ethnic Affairs Commission, Guiyang 550025, China
| | - Zi-Wan Yuan
- School of Chinese Ethnic Medicine, Guizhou Minzu University, Guiyang 550025, China
| | - Ying Liu
- School of Chinese Ethnic Medicine, Guizhou Minzu University, Guiyang 550025, China
| | - Zhong-Hua Dai
- School of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530200, China.
| |
Collapse
|
3
|
Nagaoka K, Bai X, Liu D, Cao K, Mulla J, Ji C, Chen H, Nisar MA, Bay A, Mueller W, Hildebrand G, Gao JS, Lu S, Setoyama H, Tanaka Y, Wands JR, Huang CK. Elevated 2-oxoglutarate antagonizes DNA damage responses in cholangiocarcinoma chemotherapy through regulating aspartate beta-hydroxylase. Cancer Lett 2024; 580:216493. [PMID: 37977350 DOI: 10.1016/j.canlet.2023.216493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/22/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023]
Abstract
Cholangiocarcinoma (CCA) is resistant to systemic chemotherapies that kill malignant cells mainly through DNA damage responses (DDRs). Recent studies suggest that the involvement of 2-oxoglutarate (2-OG) dependent dioxygenases in DDRs may be associated with chemoresistance in malignancy, but how 2-OG impacts DDRs in CCA chemotherapy remains elusive. We examined serum 2-OG levels in CCA patients before receiving chemotherapy. CCA patients are classified as progressive disease (PD), partial response (PR), and stable disease (SD) after receiving chemotherapy. CCA patients classified as PD showed significantly higher serum 2-OG levels than those defined as SD and PR. Treating CCA cells with 2-OG reduced DDRs. Overexpression of full-length aspartate beta-hydroxylase (ASPH) could mimic the effects of 2-OG on DDRs, suggesting the important role of ASPH in chemoresistance. Indeed, the knockdown of ASPH improved chemotherapy in CCA cells. Targeting ASPH with a specific small molecule inhibitor also enhanced the effects of chemotherapy. Mechanistically, ASPH modulates DDRs by affecting ATM and ATR, two of the major regulators finely controlling DDRs. More importantly, targeting ASPH improved the therapeutic potential of chemotherapy in two preclinical CCA models. Our data suggested the impacts of elevated 2-OG and ASPH on chemoresistance through antagonizing DDRs. Targeting ASPH may enhance DDRs, improving chemotherapy in CCA patients.
Collapse
Affiliation(s)
- Katsuya Nagaoka
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA; Department of Gastroenterology & Hepatology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Xuewei Bai
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Dan Liu
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Kevin Cao
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Joud Mulla
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Chengcheng Ji
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Hongze Chen
- Department of Pathology and Laboratory Medicine, Tulane University, New Orleans, LA, USA
| | - Muhammad Azhar Nisar
- Department of Pathology and Laboratory Medicine, Tulane University, New Orleans, LA, USA
| | - Amalia Bay
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - William Mueller
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Grace Hildebrand
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Jin-Song Gao
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Shaolei Lu
- Department of Pathology and Laboratory Medicine, Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Hiroko Setoyama
- Department of Gastroenterology & Hepatology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yasuhito Tanaka
- Department of Gastroenterology & Hepatology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Jack R Wands
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Chiung-Kuei Huang
- Liver Research Center, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA; Department of Pathology and Laboratory Medicine, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
4
|
Yu S, Yu S, Liu H, Liao N, Liu X. Enhancing mesenchymal stem cell survival and homing capability to improve cell engraftment efficacy for liver diseases. Stem Cell Res Ther 2023; 14:235. [PMID: 37667383 PMCID: PMC10478247 DOI: 10.1186/s13287-023-03476-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/25/2023] [Indexed: 09/06/2023] Open
Abstract
Although mesenchymal stem cell (MSC) transplantation provides an alternative strategy for end-stage liver disease (ESLD), further widespread application of MSC therapy is limited owing to low cell engraftment efficiency. Improving cell engraftment efficiency plays a critical role in enhancing MSC therapy for liver diseases. In this review, we summarize the current status and challenges of MSC transplantation for ESLD. We also outline the complicated cell-homing process and highlight how low cell engraftment efficiency is closely related to huge differences in extracellular conditions involved in MSC homing journeys ranging from constant, controlled conditions in vitro to variable and challenging conditions in vivo. Improving cell survival and homing capabilities enhances MSC engraftment efficacy. Therefore, we summarize the current strategies, including hypoxic priming, drug pretreatment, gene modification, and cytokine pretreatment, as well as splenectomy and local irradiation, used to improve MSC survival and homing capability, and enhance cell engraftment and therapeutic efficiency of MSC therapy. We hope that this review will provide new insights into enhancing the efficiency of MSC engraftment in liver diseases.
Collapse
Affiliation(s)
- Shaoxiong Yu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, People's Republic of China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, People's Republic of China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, People's Republic of China
| | - Saihua Yu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, People's Republic of China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, People's Republic of China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, People's Republic of China
| | - Haiyan Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, People's Republic of China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, People's Republic of China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, People's Republic of China
| | - Naishun Liao
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, People's Republic of China.
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, People's Republic of China.
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, People's Republic of China.
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, People's Republic of China.
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, People's Republic of China.
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, People's Republic of China.
| |
Collapse
|
5
|
Liu D, Shi Y, Chen H, Nisar MA, Jabara N, Langwinski N, Mattson S, Nagaoka K, Bai X, Lu S, Huang CK. Molecular profiling reveals potential targets in cholangiocarcinoma. World J Gastroenterol 2023; 29:4053-4071. [PMID: 37476584 PMCID: PMC10354586 DOI: 10.3748/wjg.v29.i25.4053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/16/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND Cholangiocarcinoma (CCA) is a devastating malignancy and has a very poor prognosis if tumors spread outside the liver. Understanding the molecular mechanisms underlying the CCA progression will likely yield therapeutic approaches toward treating this deadly disease. AIM To determine the molecular pathogenesis in CCA progression. METHODS In silico analysis, in vitro cell culture, CCA transgenic animals, histological, and molecular assays were adopted to determine the molecular pathogenesis. RESULTS The transcriptomic data of human CCA samples were retrieved from The Cancer Genome Atlas (TGCA, CHOL), European Bioinformatics Institute (EBI, GAD00001001076), and Gene Expression Omnibus (GEO, GSE107943) databases. Using Gene set enrichment analysis, the cell cycle and Notch related pathways were demonstrated to be significantly activated in CCA in TCGA and GEO datasets. We, through differentially expressed genes, found several cell cycle and notch associated genes were significantly up-regulated in cancer tissues when compared with the non-cancerous control samples. The associated genes, via quantitative real-time PCR and western blotting assays, were further examined in normal human cholangiocytes, CCA cell lines, mouse normal bile ducts, and mouse CCA tumors established by specifically depleting P53 and expressing KrasG12D mutation in the liver. Consistently, we validated that the cell cycle and Notch pathways are up-regulated in CCA cell lines and mouse CCA tumors. Interestingly, targeting cell cycle and notch pathways using small molecules also exhibited significant beneficial effects in controlling tumor malignancy. More importantly, we demonstrated that several cell cycle and Notch associated genes are significantly associated with poor overall survival and disease-free survival using the Log-Rank test. CONCLUSION In summary, our study comprehensively analyzed the gene expression pattern of CCA samples using publicly available datasets and identified the cell cycle and Notch pathways are potential therapeutic targets in this deadly disease.
Collapse
Affiliation(s)
- Dan Liu
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Yang Shi
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Hongze Chen
- Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Muhammad Azhar Nisar
- Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Nicholas Jabara
- Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Noah Langwinski
- Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Sophia Mattson
- Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Katsuya Nagaoka
- Department of Medicine, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI 02903, United States
| | - Xuewei Bai
- Department of Medicine, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI 02903, United States
| | - Shaolei Lu
- Department of Pathology, Alpert Medical School of Brown University, Providence, RI 02903, United States
| | - Chiung-Kuei Huang
- Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA 70112, United States
| |
Collapse
|
6
|
Abdel-Hamid MS, Mansour AM, Hassan MH, Abdelhady R, Elsadek BEM, El-Sayed ESM, Salama SA. Estrogen Attenuates Diethylnitrosamine-Induced Hepatocellular Carcinoma in Female Rats via Modulation of Estrogen Receptor/FASN/CD36/IL-6 Axis. Biol Pharm Bull 2023; 46:1558-1568. [PMID: 37914358 DOI: 10.1248/bpb.b23-00342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
This study was designed to evaluate the potential protective impact of estrogen and estrogen receptor against diethylnitrosamine (DEN)-induced hepatocellular carcinoma (HCC) in rats. The levels of liver injury serum biomarkers, liver content of interleukin-6 (IL-6), relative liver weight and distortion of liver histological pictures were significantly increased in ovariectomized (OVX) rats and SHAM rats that received DEN alone and were further exaggerated when DEN was combined with fulvestrant (F) compared to non-DEN treated rats. The OVX rats showed higher insults than SHAM rats. The tapering impact on these parameters was clear in OVX rats that received estradiol benzoate (EB), silymarin (S) or orlistat (ORS). The immunohistochemistry and/or Western blot analysis of liver tissues showed a prominent increase in fatty acid synthase (FASN) and cluster of differentiation 36 (CD36) expressions in OVX and SHAM rats who received DEN and/ or F compared to SHAM rats. In contrast to S, treatment of OVX rats with EB mitigated DEN-induced expression of FASN and CD36 in liver tissue, while ORS improved DEN-induced expression of FASN. In conclusion, the protective effect against HCC was mediated via estrogen receptor alpha (ER-α) which abrogates its downstream genes involved in lipid metabolism namely FASN and CD36 depriving the tumor from survival vital energy source. In addition, ORS induced similar mitigating effect against DEN-induced HCC which could be attributed to FASN inhibition and anti-inflammatory effect. Furthermore, S alleviated DEN-induced HCC, independent of its estrogenic effect.
Collapse
Affiliation(s)
| | - Ahmed M Mansour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University
| | - Memy H Hassan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University
| | - Rasha Abdelhady
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Fayoum University
| | - Bakheet E M Elsadek
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Al-Azhar University
| | - El-Sayed M El-Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University
| | - Salama A Salama
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University
| |
Collapse
|
7
|
Estrogen as a key regulator of energy homeostasis and metabolic health. Biomed Pharmacother 2022; 156:113808. [DOI: 10.1016/j.biopha.2022.113808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/02/2022] [Accepted: 10/03/2022] [Indexed: 11/23/2022] Open
|
8
|
Wang N, Li X, Zhong Z, Qiu Y, Liu S, Wu H, Tang X, Chen C, Fu Y, Chen Q, Guo T, Li J, Zhang S, Zern MA, Ma K, Wang B, Ou Y, Gu W, Cao J, Chen H, Duan Y. 3D hESC exosomes enriched with miR-6766-3p ameliorates liver fibrosis by attenuating activated stellate cells through targeting the TGFβRII-SMADS pathway. J Nanobiotechnology 2021; 19:437. [PMID: 34930304 PMCID: PMC8686281 DOI: 10.1186/s12951-021-01138-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 11/13/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Exosomes secreted from stem cells exerted salutary effects on the fibrotic liver. Herein, the roles of exosomes derived from human embryonic stem cell (hESC) in anti-fibrosis were extensively investigated. Compared with two-dimensional (2D) culture, the clinical and biological relevance of three-dimensional (3D) cell spheroids were greater because of their higher regeneration potential since they behave more like cells in vivo. In our study, exosomes derived from 3D human embryonic stem cells (hESC) spheroids and the monolayer (2D) hESCs were collected and compared the therapeutic potential for fibrotic liver in vitro and in vivo. RESULTS In vitro, PKH26 labeled-hESC-Exosomes were shown to be internalized and integrated into TGFβ-activated-LX2 cells, and reduced the expression of profibrogenic markers, thereby regulating cellular phenotypes. TPEF imaging indicated that PKH26-labeled-3D-hESC-Exsomes possessed an enhanced capacity to accumulate in the livers and exhibited more dramatic therapeutic potential in the injured livers of fibrosis mouse model. 3D-hESC-Exosomes decreased profibrogenic markers and liver injury markers, and improved the level of liver functioning proteins, eventually restoring liver function of fibrosis mice. miRNA array revealed a significant enrichment of miR-6766-3p in 3D-hESC-Exosomes, moreover, bioinformatics and dual luciferase reporter assay identified and confirmed the TGFβRII gene as the target of miR-6766-3p. Furthermore, the delivery of miR-6766-3p into activated-LX2 cells decreased cell proliferation, chemotaxis and profibrotic effects, and further investigation demonstrated that the expression of target gene TGFβRII and its downstream SMADs proteins, especially phosphorylated protein p-SMAD2/3 was also notably down-regulated by miR-6766-3p. These findings unveiled that miR-6766-3p in 3D-hESC-Exosomes inactivated SMADs signaling by inhibiting TGFβRII expression, consequently attenuating stellate cell activation and suppressing liver fibrosis. CONCLUSIONS Our results showed that miR-6766-3p in the 3D-hESC-Exosomes inactivates smads signaling by restraining TGFβRII expression, attenuated LX2 cell activation and suppressed liver fibrosis, suggesting that 3D-hESC-Exosome enriched-miR-6766-3p is a novel anti-fibrotic therapeutics for treating chronic liver disease. These results also proposed a significant strategy that 3D-Exo could be used as natural nanoparticles to rescue liver injury via delivering antifibrotic miR-6766-3p.
Collapse
Affiliation(s)
- Ning Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, People's Republic of China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, No.382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China
| | - Xiajing Li
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, No.382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China
- School of Medicine, South China University of Technology, Guangzhou, 510180, People's Republic of China
| | - Zhiyong Zhong
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, People's Republic of China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, No.382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China
| | - Yaqi Qiu
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, No.382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China
| | - Shoupei Liu
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, No.382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China
| | - Haibin Wu
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, No.382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China
| | - Xianglian Tang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, People's Republic of China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, No.382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China
| | - Chuxin Chen
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, People's Republic of China
| | - Yingjie Fu
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, No.382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China
| | - Qicong Chen
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, People's Republic of China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, No.382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China
| | - Tingting Guo
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, No.382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China
| | - Jinsong Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Shuai Zhang
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, No.1 Panfu Road, Guangzhou, 510180, People's Republic of China
| | - Mark A Zern
- Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA, 95817, USA
| | - Keqiang Ma
- Department of Hepatobiliary Pancreatic Surgery, Huadu District People's Hospital of Guangzhou, Guangzhou, 510800, People's Republic of China
| | - Bailin Wang
- Department of General Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, People's Republic of China
| | - Yimeng Ou
- Department of General Surgery, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, People's Republic of China
| | - Weili Gu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, No.1 Panfu Road, Guangzhou, 510180, People's Republic of China.
| | - Jie Cao
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, No.1 Panfu Road, Guangzhou, 510180, People's Republic of China.
| | - Honglin Chen
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, No.382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China.
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, People's Republic of China.
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, People's Republic of China.
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, People's Republic of China.
- Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, People's Republic of China.
| | - Yuyou Duan
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences and School of Medicine, South China University of Technology, No.382 Waihuan East Road, Suite 406, Higher Education Mega Center, Guangzhou, 510006, People's Republic of China.
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, People's Republic of China.
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, People's Republic of China.
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, People's Republic of China.
- Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, People's Republic of China.
| |
Collapse
|
9
|
Gorczyca G, Wartalski K, Wiater J, Samiec M, Tabarowski Z, Duda M. Anabolic Steroids-Driven Regulation of Porcine Ovarian Putative Stem Cells Favors the Onset of Their Neoplastic Transformation. Int J Mol Sci 2021; 22:ijms222111800. [PMID: 34769230 PMCID: PMC8583785 DOI: 10.3390/ijms222111800] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/27/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022] Open
Abstract
Nandrolone (Ndn) and boldenone (Bdn), the synthetic testosterone analogues with strong anabolic effects, despite being recognized as potentially carcinogenic compounds, are commonly abused by athletes and bodybuilders, which includes women, worldwide. This study tested the hypothesis that different doses of Ndn and Bdn can initiate neoplastic transformation of porcine ovarian putative stem cells (poPSCs). Immunomagnetically isolated poPSCs were expanded ex vivo in the presence of Ndn or Bdn, for 7 and 14 days. Results show that pharmacological doses of both Ndn and Bdn, already after 7 days of poPSCs culture, caused a significant increase of selected, stemness-related markers of cancer cells: CD44 and CD133. Notably, Ndn also negatively affected poPSCs growth not only by suppressing their proliferation and mitochondrial respiration but also by inducing apoptosis. This observation shows, for the first time, that chronic exposure to Ndn or Bdn represents a precondition that might enhance risk of poPSCs neoplastic transformation. These studies carried out to accomplish detailed molecular characterization of the ex vivo expanded poPSCs and their potentially cancerous derivatives (PCDs) might be helpful to determine their suitability as nuclear donor cells (NDCs) for further investigations focused on cloning by somatic cell nuclear transfer (SCNT). Such investigations might also be indispensable to estimate the capabilities of nuclear genomes inherited from poPSCs and their PCDs to be epigenetically reprogrammed (dedifferentiated) in cloned pig embryos generated by SCNT. This might open up new possibilities for biomedical research aimed at more comprehensively recognizing genetic and epigenetic mechanisms underlying not only tumorigenesis but also reversal/retardation of pro-tumorigenic intracellular events.
Collapse
Affiliation(s)
- Gabriela Gorczyca
- Department of Endocrinology, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa 9 Street, 30-387 Krakow, Poland;
| | - Kamil Wartalski
- Department of Histology, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 7 Street, 31-034 Krakow, Poland; (K.W.); (J.W.)
| | - Jerzy Wiater
- Department of Histology, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 7 Street, 31-034 Krakow, Poland; (K.W.); (J.W.)
| | - Marcin Samiec
- Department of Reproductive Biotechnology and Cryoconservation, National Research Institute of Animal Production, Krakowska 1 Street, 32-083 Balice near Kraków, Poland
- Correspondence: (M.S.); (M.D.)
| | - Zbigniew Tabarowski
- Department of Experimental Hematology, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa 9 Street, 30-387 Krakow, Poland;
| | - Małgorzata Duda
- Department of Endocrinology, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa 9 Street, 30-387 Krakow, Poland;
- Correspondence: (M.S.); (M.D.)
| |
Collapse
|
10
|
Deng Y, Xia B, Chen Z, Wang F, Lv Y, Chen G. Stem Cell-based Therapy Strategy for Hepatic Fibrosis by Targeting Intrahepatic Cells. Stem Cell Rev Rep 2021; 18:77-93. [PMID: 34668120 DOI: 10.1007/s12015-021-10286-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2021] [Indexed: 12/11/2022]
Abstract
The whole liver transplantation is the most effective treatment for end-stage fibrosis. However, the lack of available donors, immune rejection and total cost of surgery remain as the key challenges in advancing liver fibrosis therapeutics. Due to the multi-differentiation and low immunogenicity of stem cells, treatment of liver fibrosis with stem cells has been considered as a valuable new therapeutic modality. The pathological progression of liver fibrosis is closely related to the changes in the activities of intrahepatic cells. Damaged hepatocytes, activated Kupffer cells and other inflammatory cells lead to hepatic stellate cells (HSCs) activation, further promoting apoptosis of damaged hepatocytes, while stem cells can work on fibrosis-related intrahepatic cells through relevant transduction pathways. Herein, this article elucidates the phenomena and the mechanisms of the crosstalk between various types of stem cells and intrahepatic cells including HSCs and hepatocytes in the treatment of liver fibrosis. Then, the important influences of chemical compositions, mechanical properties and blood flow on liver fibrosis models with stem cell treatment are emphasized. Clinical trials on stem cell-based therapy for liver fibrosis are also briefly summarized. Finally, continuing challenges and future directions of stem cell-based therapy for hepatic fibrosis are discussed. In short, stem cells play an important advantage and have a great potential in treating liver fibrosis by interacting with intrahepatic cells. Clarifying how stem cells interact with intrahepatic cells to change the progression of liver fibrosis is of great significance for a deeper understanding of liver fibrosis mechanisms and targeted therapy.
Collapse
Affiliation(s)
- Yaxin Deng
- School of Pharmacy and Bioengineering, Chongqing University of Technology, No. 69 Hongguang Avenue, Banan District, Chongqing, 400054, People's Republic of China.,Chongqing Key Laboratory of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing, 400054, People's Republic of China
| | - Bin Xia
- Engineering Research Center for Waste Oil Recovery Technology and Equipment, Ministry of Education, Chongqing Technology and Business University, Chongqing, 400067, People's Republic of China
| | - Zhongmin Chen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, No. 69 Hongguang Avenue, Banan District, Chongqing, 400054, People's Republic of China.,Chongqing Key Laboratory of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing, 400054, People's Republic of China
| | - Fuping Wang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, No. 69 Hongguang Avenue, Banan District, Chongqing, 400054, People's Republic of China.,Chongqing Key Laboratory of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing, 400054, People's Republic of China
| | - Yonggang Lv
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, 400044, People's Republic of China.,State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan, 430200, People's Republic of China
| | - Guobao Chen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, No. 69 Hongguang Avenue, Banan District, Chongqing, 400054, People's Republic of China. .,Chongqing Key Laboratory of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing, 400054, People's Republic of China.
| |
Collapse
|
11
|
Bai X, Zhang H, Zhou Y, Nagaoka K, Meng J, Ji C, Liu D, Dong X, Cao K, Mulla J, Cheng Z, Mueller W, Bay A, Hildebrand G, Lu S, Wallace J, Wands JR, Sun B, Huang CK. Ten-Eleven Translocation 1 Promotes Malignant Progression of Cholangiocarcinoma With Wild-Type Isocitrate Dehydrogenase 1. Hepatology 2021; 73:1747-1763. [PMID: 32740973 PMCID: PMC7855500 DOI: 10.1002/hep.31486] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Cholangiocarcinoma (CCA) is a highly lethal disease without effective therapeutic approaches. The whole-genome sequencing data indicate that about 20% of patients with CCA have isocitrate dehydrogenase 1 (IDH1) mutations, which have been suggested to target 2-oxoglutarate (OG)-dependent dioxygenases in promoting CCA carcinogenesis. However, the clinical study indicates that patients with CCA and mutant IDH1 have better prognosis than those with wild-type IDH1, further complicating the roles of 2-OG-dependent enzymes. APPROACH AND RESULTS This study aimed to clarify if ten-eleven translocation 1 (TET1), which is one of the 2-OG-dependent enzymes functioning in regulating 5-hydroxymethylcytosine (5hmC) formation, is involved in CCA progression. By analyzing The Cancer Genome Atlas (TCGA) data set, TET1 mRNA was found to be substantially up-regulated in patients with CCA when compared with noncancerous bile ducts. Additionally, TET1 protein expression was significantly elevated in human CCA tumors. CCA cells were challenged with α-ketoglutarate (α-KG) and dimethyl-α-KG (DM-α-KG), which are cosubstrates for TET1 dioxygenase. The treatments with α-KG and DM-α-KG promoted 5hmC formation and malignancy of CCA cells. Molecular and pharmacological approaches were used to inhibit TET1 activity, and these treatments substantially suppressed 5hmC and CCA carcinogenesis. Mechanistically, it was found that knockdown of TET1 may suppress CCA progression by targeting cell growth and apoptosis through epigenetic regulation. Consistently, targeting TET1 significantly inhibited CCA malignant progression in a liver orthotopic xenograft model by targeting cell growth and apoptosis. CONCLUSIONS Our data suggest that expression of TET1 is highly associated with CCA carcinogenesis. It will be important to evaluate TET1 expression in CCA tumors before application of the IDH1 mutation inhibitor because the inhibitor suppresses 2-hydroxyglutarate expression, which may result in activation of TET, potentially leading to CCA malignancy.
Collapse
Affiliation(s)
- Xuewei Bai
- Department of Pancreatic and Biliary Surgery, First
Affiliated Hospital of Harbin Medical University, Harbin 150001, China; Liver
Research Center, Division of Gastroenterology & Liver Research Center, Warren
Alpert Medical School of Brown University and Rhode Island Providence, RI, USA,Liver Research Center, Division of Gastroenterology &
Liver Research Center, Warren Alpert Medical School of Brown University and Rhode
Island Hospital, Providence, RI, USA
| | - Hongyu Zhang
- Liver Research Center, Division of Gastroenterology &
Liver Research Center, Warren Alpert Medical School of Brown University and Rhode
Island Hospital, Providence, RI, USA
| | - Yamei Zhou
- Department of Pancreatic and Biliary Surgery, First
Affiliated Hospital of Harbin Medical University, Harbin 150001, China; Liver
Research Center, Division of Gastroenterology & Liver Research Center, Warren
Alpert Medical School of Brown University and Rhode Island Providence, RI, USA,Liver Research Center, Division of Gastroenterology &
Liver Research Center, Warren Alpert Medical School of Brown University and Rhode
Island Hospital, Providence, RI, USA
| | - Katsuya Nagaoka
- Liver Research Center, Division of Gastroenterology &
Liver Research Center, Warren Alpert Medical School of Brown University and Rhode
Island Hospital, Providence, RI, USA
| | - Jialin Meng
- Department of Urology, The First Affiliated Hospital of
Anhui Medical University; Institute of Urology & Anhui Province Key Laboratory
of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, China
| | - Chengcheng Ji
- Liver Research Center, Division of Gastroenterology &
Liver Research Center, Warren Alpert Medical School of Brown University and Rhode
Island Hospital, Providence, RI, USA
| | - Dan Liu
- Liver Research Center, Division of Gastroenterology &
Liver Research Center, Warren Alpert Medical School of Brown University and Rhode
Island Hospital, Providence, RI, USA
| | - Xianghui Dong
- Department of Pathology, The First Affiliated Hospital of
Harbin Medical University, Harbin 150001, Heilongjiang Province, P.R. China
| | - Kevin Cao
- Liver Research Center, Division of Gastroenterology &
Liver Research Center, Warren Alpert Medical School of Brown University and Rhode
Island Hospital, Providence, RI, USA
| | - Joud Mulla
- Liver Research Center, Division of Gastroenterology &
Liver Research Center, Warren Alpert Medical School of Brown University and Rhode
Island Hospital, Providence, RI, USA
| | - Zhixiang Cheng
- Liver Research Center, Division of Gastroenterology &
Liver Research Center, Warren Alpert Medical School of Brown University and Rhode
Island Hospital, Providence, RI, USA
| | - William Mueller
- Liver Research Center, Division of Gastroenterology &
Liver Research Center, Warren Alpert Medical School of Brown University and Rhode
Island Hospital, Providence, RI, USA
| | - Amalia Bay
- Liver Research Center, Division of Gastroenterology &
Liver Research Center, Warren Alpert Medical School of Brown University and Rhode
Island Hospital, Providence, RI, USA
| | - Grace Hildebrand
- Liver Research Center, Division of Gastroenterology &
Liver Research Center, Warren Alpert Medical School of Brown University and Rhode
Island Hospital, Providence, RI, USA
| | - Shaolei Lu
- Department of Pathology and Laboratory Medicine, Warren
Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI,
USA
| | - Joselynn Wallace
- Center for Computational Biology of Human Disease and
Center for Computation and Visualization, Brown University, Providence, RI,
USA
| | - Jack R. Wands
- Liver Research Center, Division of Gastroenterology &
Liver Research Center, Warren Alpert Medical School of Brown University and Rhode
Island Hospital, Providence, RI, USA
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, First
Affiliated Hospital of Harbin Medical University, Harbin 150001, China; Liver
Research Center, Division of Gastroenterology & Liver Research Center, Warren
Alpert Medical School of Brown University and Rhode Island Providence, RI, USA,Correspondence to: Chiung-Kuei Huang,
Ph.D., Liver Research Center, Rhode Island Hospital, Brown Alpert Medical
School, Brown University, 55 Claverick Street, Providence, RI 02903,
; Bei Sun, M.D., Ph.D. Department
of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin
Medical University. Key Laboratory of Hepatosplenic Surgery, Ministry of
Education, The First Affiliated Hospital of Harbin Medical University, No. 23
Youzheng Road, Nangang District, Harbin 150081, Heilongjiang Province, P.R.
China. Tel: 86-451-85555721; Fax: 86-451-53643849;
| | - Chiung-Kuei Huang
- Liver Research Center, Division of Gastroenterology &
Liver Research Center, Warren Alpert Medical School of Brown University and Rhode
Island Hospital, Providence, RI, USA,Correspondence to: Chiung-Kuei Huang,
Ph.D., Liver Research Center, Rhode Island Hospital, Brown Alpert Medical
School, Brown University, 55 Claverick Street, Providence, RI 02903,
; Bei Sun, M.D., Ph.D. Department
of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin
Medical University. Key Laboratory of Hepatosplenic Surgery, Ministry of
Education, The First Affiliated Hospital of Harbin Medical University, No. 23
Youzheng Road, Nangang District, Harbin 150081, Heilongjiang Province, P.R.
China. Tel: 86-451-85555721; Fax: 86-451-53643849;
| |
Collapse
|
12
|
Nagaoka K, Ogawa K, Ji C, Cao KY, Bai X, Mulla J, Cheng Z, Wands JR, Huang CK. Targeting Aspartate Beta-Hydroxylase with the Small Molecule Inhibitor MO-I-1182 Suppresses Cholangiocarcinoma Metastasis. Dig Dis Sci 2021; 66:1080-1089. [PMID: 32445050 DOI: 10.1007/s10620-020-06330-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 05/08/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Cholangiocarcinoma is a devastating disease with a 2% 5-year survival if the disease has spread outside the liver. The enzyme aspartate beta-hydroxylase (ASPH) has been demonstrated to be highly expressed in cholangiocarcinoma but not in normal bile ducts and found to stimulate tumor cell migration. In addition, it was found that targeting ASPH inhibits cholangiocarcinoma malignant progression. However, it is not clear whether targeting ASPH with the small molecule inhibitor MO-I-1182 suppresses cholangiocarcinoma metastasis. The current study aims to study the efficacy of MO-I-1182 in suppressing cholangiocarcinoma metastasis. METHODS The analysis was performed in vitro and in vivo with a preclinical animal model by using molecular and biochemical strategies to regulate ASPH expression and function. RESULTS Knockdown of ASPH substantially inhibited cell migration and invasion in two human cholangiocarcinoma cell lines. Targeting ASPH with a small molecule inhibitor suppressed cholangiocarcinoma progression. Molecular mechanism studies demonstrated that knockdown of ASPH subsequently suppressed protein levels of the matrix metalloproteinases. The ASPH knockdown experiments suggest that this enzyme may modulate cholangiocarcinoma metastasis by regulating matrix metalloproteinases expression. Furthermore, using an ASPH inhibitor in a rat cholangiocarcinoma intrahepatic model established with BED-Neu-CL#24 cholangiocarcinoma cells, it was found that targeting ASPH inhibited intrahepatic cholangiocarcinoma metastasis and downstream expression of the matrix metalloproteinases. CONCLUSION ASPH may modulate cholangiocarcinoma metastasis via matrix metalloproteinases expression. Taken together, targeting ASPH function may inhibit intrahepatic cholangiocarcinoma metastasis and improve survival.
Collapse
Affiliation(s)
- Katsuya Nagaoka
- Liver Research Center, Division of Gastroenterology and Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, 55 Claverick St, Providence, RI, 02903, USA
| | - Kousuke Ogawa
- Liver Research Center, Division of Gastroenterology and Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, 55 Claverick St, Providence, RI, 02903, USA
| | - Chengcheng Ji
- Liver Research Center, Division of Gastroenterology and Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, 55 Claverick St, Providence, RI, 02903, USA
| | - Kevin Y Cao
- Liver Research Center, Division of Gastroenterology and Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, 55 Claverick St, Providence, RI, 02903, USA
| | - Xuewei Bai
- Liver Research Center, Division of Gastroenterology and Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, 55 Claverick St, Providence, RI, 02903, USA
| | - Joud Mulla
- Liver Research Center, Division of Gastroenterology and Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, 55 Claverick St, Providence, RI, 02903, USA
| | - Zhixiang Cheng
- Liver Research Center, Division of Gastroenterology and Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, 55 Claverick St, Providence, RI, 02903, USA
| | - Jack R Wands
- Liver Research Center, Division of Gastroenterology and Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, 55 Claverick St, Providence, RI, 02903, USA
| | - Chiung-Kuei Huang
- Liver Research Center, Division of Gastroenterology and Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, 55 Claverick St, Providence, RI, 02903, USA.
| |
Collapse
|
13
|
Hu H, Zhou H, Xu D. A review of the effects and molecular mechanisms of dimethylcurcumin (ASC-J9) on androgen receptor-related diseases. Chem Biol Drug Des 2021; 97:821-835. [PMID: 33277796 DOI: 10.1111/cbdd.13811] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 11/19/2020] [Indexed: 12/17/2022]
Abstract
Dimethylcurcumin (ASC-J9) is a curcumin analogue capable of inhibiting prostate cancer cell proliferation. The mechanism is associated with the unique role of ASC-J9 in enhancing androgen receptor (AR) degradation. So far, ASC-J9 has been investigated in typical AR-associated diseases such as prostate cancer, benign prostatic hypertrophy, bladder cancer, renal diseases, liver diseases, cardiovascular diseases, cutaneous wound, spinal and bulbar muscular atrophy, ovarian cancer and melanoma, exhibiting great potentials in disease control. In this review, the effects and molecular mechanisms of ASC-J9 on various AR-associated diseases are summarized. Importantly, the effects of ASC-J9 and AR antagonists enzalutamide/bicalutamide on prostate cancer are compared in detail and crucial differences are highlighted. At last, the pharmacological effects of ASC-J9 are summarized and the future applications of ASC-J9 in AR-associated disease control are discussed.
Collapse
Affiliation(s)
- Hang Hu
- National & Local Joint Engineering Research Center for High-efficiency Refining and High-quality Utilization of Biomass, School of Pharmaceutical Engineering and Life Sciences, Changzhou University, Changzhou, China
| | - Huan Zhou
- Center for Health Science and Engineering, School of Materials Science and Engineering, Hebei University of Technology, Tianjin, China
| | - Defeng Xu
- National & Local Joint Engineering Research Center for High-efficiency Refining and High-quality Utilization of Biomass, School of Pharmaceutical Engineering and Life Sciences, Changzhou University, Changzhou, China
| |
Collapse
|
14
|
Zheng W, Yang Y, Sequeira RC, Bishop CE, Atala A, Gu Z, Zhao W. Effects of Extracellular Vesicles Derived from Mesenchymal Stem/Stromal Cells on Liver Diseases. Curr Stem Cell Res Ther 2019; 14:442-452. [PMID: 30854976 DOI: 10.2174/1574888x14666190308123714] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/17/2018] [Accepted: 02/13/2019] [Indexed: 12/18/2022]
Abstract
Therapeutic effects of Mesenchymal Stem/Stromal Cells (MSCs) transplantation have been observed in various disease models. However, it is thought that MSCs-mediated effects largely depend on the paracrine manner of secreting cytokines, growth factors, and Extracellular Vesicles (EVs). Similarly, MSCs-derived EVs also showed therapeutic benefits in various liver diseases through alleviating fibrosis, improving regeneration of hepatocytes, and regulating immune activity. This review provides an overview of the MSCs, their EVs, and their therapeutic potential in treating various liver diseases including liver fibrosis, acute and chronic liver injury, and Hepatocellular Carcinoma (HCC). More specifically, the mechanisms by which MSC-EVs induce therapeutic benefits in liver diseases will be covered. In addition, comparisons between MSCs and their EVs were also evaluated as regenerative medicine against liver diseases. While the mechanisms of action and clinical efficacy must continue to be evaluated and verified, MSCs-derived EVs currently show tremendous potential and promise as a regenerative medicine treatment for liver disease in the future.
Collapse
Affiliation(s)
- Wenjie Zheng
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China.,Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Medical Center Blvd, Winston-Salem, NC 27157, United States
| | - Yumin Yang
- Co-Innovation Center of Neuro-regeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Russel Clive Sequeira
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Medical Center Blvd, Winston-Salem, NC 27157, United States
| | - Colin E Bishop
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Medical Center Blvd, Winston-Salem, NC 27157, United States
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Medical Center Blvd, Winston-Salem, NC 27157, United States
| | - Zhifeng Gu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Weixin Zhao
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Medical Center Blvd, Winston-Salem, NC 27157, United States
| |
Collapse
|
15
|
Chen JF, Lin PW, Tsai YR, Yang YC, Kang HY. Androgens and Androgen Receptor Actions on Bone Health and Disease: From Androgen Deficiency to Androgen Therapy. Cells 2019; 8:cells8111318. [PMID: 31731497 PMCID: PMC6912771 DOI: 10.3390/cells8111318] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 12/12/2022] Open
Abstract
Androgens are not only essential for bone development but for the maintenance of bone mass. Therefore, conditions with androgen deficiency, such as male hypogonadism, androgen-insensitive syndromes, and prostate cancer with androgen deprivation therapy are strongly associated with bone loss and increased fracture risk. Here we summarize the skeletal effects of androgens—androgen receptors (AR) actions based on in vitro and in vivo studies from animals and humans, and discuss bone loss due to androgens/AR deficiency to clarify the molecular basis for the anabolic action of androgens and AR in bone homeostasis and unravel the functions of androgen/AR signaling in healthy and disease states. Moreover, we provide evidence for the skeletal benefits of androgen therapy and elucidate why androgens are more beneficial than male sexual hormones, highlighting their therapeutic potential as osteoanabolic steroids in improving bone fracture repair. Finally, the application of selective androgen receptor modulators may provide new approaches for the treatment of osteoporosis and fractures as well as building stronger bones in diseases dependent on androgens/AR status.
Collapse
Affiliation(s)
- Jia-Feng Chen
- Division of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Kaohsiung Chang-Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung 833, Taiwan;
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Kaohsiung 833, Taiwan; (P.-W.L.); (Y.-R.T.); (Y.-C.Y.)
| | - Pei-Wen Lin
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Kaohsiung 833, Taiwan; (P.-W.L.); (Y.-R.T.); (Y.-C.Y.)
- Center for Menopause and Reproductive Medicine Research, Department of Obstetrics and Gynecology, Kaohsiung Chang-Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung 833, Taiwan
| | - Yi-Ru Tsai
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Kaohsiung 833, Taiwan; (P.-W.L.); (Y.-R.T.); (Y.-C.Y.)
- Center for Menopause and Reproductive Medicine Research, Department of Obstetrics and Gynecology, Kaohsiung Chang-Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung 833, Taiwan
- An-Ten Obstetrics and Gynecology Clinic, Kaohsiung 802, Taiwan
| | - Yi-Chien Yang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Kaohsiung 833, Taiwan; (P.-W.L.); (Y.-R.T.); (Y.-C.Y.)
- Department of Dermatology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Hong-Yo Kang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Kaohsiung 833, Taiwan; (P.-W.L.); (Y.-R.T.); (Y.-C.Y.)
- Center for Menopause and Reproductive Medicine Research, Department of Obstetrics and Gynecology, Kaohsiung Chang-Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung 833, Taiwan
- Correspondence: ; Tel.: +886-7-731-7123 (ext. 8898)
| |
Collapse
|
16
|
Grossmann M, Wierman ME, Angus P, Handelsman DJ. Reproductive Endocrinology of Nonalcoholic Fatty Liver Disease. Endocr Rev 2019; 40:417-446. [PMID: 30500887 DOI: 10.1210/er.2018-00158] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 11/19/2018] [Indexed: 02/07/2023]
Abstract
The liver and the reproductive system interact in a multifaceted bidirectional fashion. Sex steroid signaling influences hepatic endobiotic and xenobiotic metabolism and contributes to the pathogenesis of functional and structural disorders of the liver. In turn, liver function affects the reproductive axis via modulating sex steroid metabolism and transport to tissues via sex hormone-binding globulin (SHBG). The liver senses the body's metabolic status and adapts its energy homeostasis in a sex-dependent fashion, a dimorphism signaled by the sex steroid milieu and possibly related to the metabolic costs of reproduction. Sex steroids impact the pathogenesis of nonalcoholic fatty liver disease, including development of hepatic steatosis, fibrosis, and carcinogenesis. Preclinical studies in male rodents demonstrate that androgens protect against hepatic steatosis and insulin resistance both via androgen receptor signaling and, following aromatization to estradiol, estrogen receptor signaling, through regulating genes involved in hepatic lipogenesis and glucose metabolism. In female rodents in contrast to males, androgens promote hepatic steatosis and dysglycemia, whereas estradiol is similarly protective against liver disease. In men, hepatic steatosis is associated with modest reductions in circulating testosterone, in part consequent to a reduction in circulating SHBG. Testosterone treatment has not been demonstrated to improve hepatic steatosis in randomized controlled clinical trials. Consistent with sex-dimorphic preclinical findings, androgens promote hepatic steatosis and dysglycemia in women, whereas endogenous estradiol appears protective in both men and women. In both sexes, androgens promote hepatic fibrosis and the development of hepatocellular carcinoma, whereas estradiol is protective.
Collapse
Affiliation(s)
- Mathis Grossmann
- Department of Medicine Austin Health, University of Melbourne, Heidelberg, Victoria, Australia.,Department of Endocrinology, Austin Health, Heidelberg, Victoria, Australia
| | - Margaret E Wierman
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,Research Service, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado
| | - Peter Angus
- Department of Medicine Austin Health, University of Melbourne, Heidelberg, Victoria, Australia.,Departments of Gastroenterology and Hepatology, Heidelberg, Victoria, Australia
| | - David J Handelsman
- ANZAC Research Institute, University of Sydney, Concord Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
17
|
Hu C, Zhao L, Duan J, Li L. Strategies to improve the efficiency of mesenchymal stem cell transplantation for reversal of liver fibrosis. J Cell Mol Med 2019; 23:1657-1670. [PMID: 30635966 PMCID: PMC6378173 DOI: 10.1111/jcmm.14115] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 12/06/2018] [Accepted: 12/06/2018] [Indexed: 12/12/2022] Open
Abstract
End‐stage liver fibrosis frequently progresses to portal vein thrombosis, formation of oesophageal varices, hepatic encephalopathy, ascites, hepatocellular carcinoma and liver failure. Mesenchymal stem cells (MSCs), when transplanted in vivo, migrate into fibrogenic livers and then differentiate into hepatocyte‐like cells or fuse with hepatocytes to protect liver function. Moreover, they can produce various growth factors and cytokines with anti‐inflammatory effects to reverse the fibrotic state of the liver. In addition, only a small number of MSCs migrate to the injured tissue after cell transplantation; consequently, multiple studies have investigated effective strategies to improve the survival rate and activity of MSCs for the treatment of liver fibrosis. In this review, we intend to arrange and analyse the current evidence related to MSC transplantation in liver fibrosis, to summarize the detailed mechanisms of MSC transplantation for the reversal of liver fibrosis and to discuss new strategies for this treatment. Finally, and most importantly, we will identify the current problems with MSC‐based therapies to repair liver fibrosis that must be addressed in order to develop safer and more effective routes for MSC transplantation. In this way, it will soon be possible to significantly improve the therapeutic effects of MSC transplantation for liver regeneration, as well as enhance the quality of life and prolong the survival time of patients with liver fibrosis.
Collapse
Affiliation(s)
- Chenxia Hu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Lingfei Zhao
- Kidney Disease Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, PR China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Jinfeng Duan
- The Key Laboratory of Mental Disorder Management of Zhejiang Province, Department of Psychiatry, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Lanjuan Li
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, PR China
| |
Collapse
|
18
|
Hajighasemlou S, Pakzad S, Ai J, Muhammadnejad S, Mirmoghtadaei M, Hosseinzadeh F, Gharibzadeh S, Kamali A, Ahmadi A, Verdi J. Characterization and Validation of Hepatocellular Carcinoma (HCC) Xenograft tumor as a Suitable Liver Cancer Model for
Preclinical Mesenchymal Stem Cell Studies. Asian Pac J Cancer Prev 2018; 19:1627-1631. [PMID: 29936790 PMCID: PMC6103601 DOI: 10.22034/apjcp.2018.19.6.1627] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is the fifth most diagnosed cancer and the third leading cause of cancer-related death. sorafenib is used as a standard therapy to treat HCC. mesenchymal stromal cells (MSCs) have also been used to suppress HCC. Here we investigate the development of a xenograft model of liver cancer to study the homing of hpMSC-GFP cells, tumor kinetics and molecular characterizations of HCC. Methods: To create xenograft models of HCC, HepG2 cell lines were inoculated into the flanks of 9 nude mice bilaterally. Animals were then divided into three groups: the first group received hpMSC-GFP systemically, the second received intra-tumoral hpMSC-GFP and the third received PBS. The first two groups were sacrificed after 72 hours of MSCs injection but the third group was followed up for forty days. One tumor from each animal was then transferred to formalin buffer for H&E staining and immunohistochemistry analysis (KI67 and CD34), and the other tumor was used for ex-vivo imaging. Blood samples were taken from all subjects before sacrificing them. Results: Histopathological fidelity of heterotopic HePG2 xenograft models to human HCC tumors was demonstrated. Biochemical evaluation suggested the health of the animal’s liver and kidneys. Ex-vivo imaging illustrated homing of more hpMSC-GFP cells in tumor tissues derived from the group receiving intra-tumoral hpMSC-GFP. Conclusion: A standard method was used to inoculate tumor cells and the intervention was shown to be safe to liver and kidneys. Local injection of MSCs can be used as cell therapy to fight neoplasms.
Collapse
Affiliation(s)
- Saieh Hajighasemlou
- Tissue Engineering and Applied Cell Sciences, Tehran University of Medical Science, Tehran, Iran.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Iwagami Y, Zou J, Zhang H, Cao K, Ji C, Kim M, Huang CK. Alcohol-mediated miR-34a modulates hepatocyte growth and apoptosis. J Cell Mol Med 2018; 22:3987-3995. [PMID: 29873178 PMCID: PMC6050481 DOI: 10.1111/jcmm.13681] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 04/11/2018] [Indexed: 01/02/2023] Open
Abstract
MicroRNAs (miRs) have been recently shown to be heavily involved in the development of alcoholic liver disease (ALD) and suggested as a potential therapeutic target in ALD. The miR‐34a was consistently reported to be significantly elevated in several ALD rodent models, but it remains unclear how miR‐34a modulates the cellular behaviours of hepatocytes in ALD development and progression. This study aims to characterize alcohol‐induced miR‐34a impact on hepatocytes growth and apoptosis. The miRNA array was performed to assess changes in miRNA after chronic alcohol feeding. Liver and blood samples were used to examine ALD progression. The miR‐34a was overexpressed in human hepatocytes to evaluate its impact on cell growth and apoptosis. Real‐time quantitative PCR and Western blot were used to determine the growth and apoptosis molecular signalling pathways associated with miR‐34a. Alcohol feeding significantly promoted fatty liver progression, serum ALT levels, apoptosis and miR‐34a expression in rat liver. Overexpression of miR‐34a in human hepatocytes suppressed cell growth signallings, including c‐Met, cyclin D1 and cyclin‐dependent kinase 6 (CDK6). The miR‐34a might also inhibit the expression of sirtuin 1 (Sirt1) and its target, B‐cell lymphoma 2. Interestingly, the expression of miR‐34a reverses the suppressive effects of ethanol on cell growth. But, miR‐34a promotes hepatocyte senescence and apoptosis. Although the miR‐34a‐mediated down‐regulation of cell growth‐associated genes may contribute to cell growth retardation, other miR‐34a targets, such as Sirt1, may reverse this phenotype. Future studies will be needed to clarify the role of miR‐34a in ALD progression.
Collapse
Affiliation(s)
- Yoshifumi Iwagami
- Department of Medicine, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Jing Zou
- Department of Medicine, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Hongyu Zhang
- Department of Medicine, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Kevin Cao
- Department of Medicine, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Chengcheng Ji
- Department of Medicine, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Miran Kim
- Department of Medicine, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Chiung-Kuei Huang
- Department of Medicine, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| |
Collapse
|
20
|
Omrani MR, Yaqubi M, Mohammadnia A. Transcription Factors in Regulatory and Protein Subnetworks during Generation of Neural Stem Cells and Neurons from Direct Reprogramming of Non-fibroblastic Cell Sources. Neuroscience 2018; 380:63-77. [PMID: 29653196 DOI: 10.1016/j.neuroscience.2018.03.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 03/19/2018] [Accepted: 03/20/2018] [Indexed: 12/31/2022]
Abstract
Direct reprogramming of non-fibroblastic cells to the neuronal cell types including induced neurons (iNs) and induced neural stem cells (iNSCs) has provided an alternative approach for the direct reprogramming of fibroblasts to those cells. However, to increase the efficiency of the reprogramming process the underlying mechanisms should be clarified. In the current study, we analyzed the gene expression profiles of five different cellular conversions to understand the most significant molecular mechanisms and transcription factors (TFs) underlying each conversion. For each conversion, we found the list of differentially expressed genes (DEGs) and the list of differentially expressed TFs (DE-TFs) which regulate expression of DEGs. Moreover, we constructed gene regulatory networks based on the TF-binding sites' data and found the most central regulators and the most active part of the networks. Furthermore, protein complexes were identified from constructed protein-protein interaction networks for DE-TFs. Finally, we proposed a list of main regulators for each conversion; for example, in the direct conversion of epithelial-like cells (ECs) to iNSCs, combination of centrality with active modules or protein complex analyses highlighted the role of POU3F2, BACH1, AR, PBX1, SOX2 and NANOG genes in this conversion. To the best of our knowledge, this study is the first one that analyzed the direct conversion of non-fibroblastic cells toward iNs and iNSCs and we believe that the expression manipulation of identified genes may increase efficiency of these processes.
Collapse
Affiliation(s)
- Mohammad Reza Omrani
- National Institute of Genetics Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Moein Yaqubi
- Department of Psychiatry, Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada.
| | | |
Collapse
|
21
|
Lee CW, Chen YF, Wu HH, Lee OK. Historical Perspectives and Advances in Mesenchymal Stem Cell Research for the Treatment of Liver Diseases. Gastroenterology 2018; 154:46-56. [PMID: 29107021 DOI: 10.1053/j.gastro.2017.09.049] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 09/23/2017] [Accepted: 09/27/2017] [Indexed: 12/14/2022]
Abstract
Liver transplantation is the only effective therapy for patients with decompensated cirrhosis and fulminant liver failure. However, due to a shortage of donor livers and complications associated with immune suppression, there is an urgent need for new therapeutic strategies for patients with end-stage liver diseases. Given their unique function in self-renewal and differentiation potential, stem cells might be used to regenerate damaged liver tissue. Recent studies have shown that stem cell-based therapies can improve liver function in a mouse model of hepatic failure. Moreover, acellular liver scaffolds seeded with hepatocytes produced functional bioengineered livers for organ transplantation in preclinical studies. The therapeutic potential of stem cells or their differentiated progenies will depend on their capacity to differentiate into mature and functional cell types after transplantation. It will also be important to devise methods to overcome their genomic instability, immune reactivity, and tumorigenic potential. We review directions and advances in the use of mesenchymal stem cells and their derived hepatocytes for liver regeneration. We also discuss the potential applications of hepatocytes derived from human pluripotent stem cells and challenges to using these cells in treating end-stage liver disease.
Collapse
Affiliation(s)
- Chien-Wei Lee
- Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan; Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Fan Chen
- Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan; Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hao-Hsiang Wu
- Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan; Institute of Biophotonics, National Yang-Ming University, Taipei, Taiwan
| | - Oscar K Lee
- Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan; Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; Taipei City Hospital, Taipei, Taiwan.
| |
Collapse
|
22
|
Alhomrani M, Correia J, Zavou M, Leaw B, Kuk N, Xu R, Saad MI, Hodge A, Greening DW, Lim R, Sievert W. The Human Amnion Epithelial Cell Secretome Decreases Hepatic Fibrosis in Mice with Chronic Liver Fibrosis. Front Pharmacol 2017; 8:748. [PMID: 29114223 PMCID: PMC5660722 DOI: 10.3389/fphar.2017.00748] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 10/03/2017] [Indexed: 12/22/2022] Open
Abstract
Background: Hepatic stellate cells (HSCs) are the primary collagen-secreting cells in the liver. While HSCs are the major cell type involved in the pathogenesis of liver fibrosis, hepatic macrophages also play an important role in mediating fibrogenesis and fibrosis resolution. Previously, we observed a reduction in HSC activation, proliferation, and collagen synthesis following exposure to human amnion epithelial cells (hAEC) and hAEC-conditioned media (hAEC-CM). This suggested that specific factors secreted by hAEC might be effective in ameliorating liver fibrosis. hAEC-derived extracellular vesicles (hAEC-EVs), which are nanosized (40–100 nm) membrane bound vesicles, may act as novel cell–cell communicators. Accordingly, we evaluated the efficacy of hAEC-EV in modulating liver fibrosis in a mouse model of chronic liver fibrosis and in human HSC. Methods: The hAEC-EVs were isolated and characterized. C57BL/6 mice with CCl4-induced liver fibrosis were administered hAEC-EV, hAEC-CM, or hAEC-EV depleted medium (hAEC-EVDM). LX2 cells, a human HSC line, and bone marrow-derived mouse macrophages were exposed to hAEC-EV, hAEC-CM, and hAEC-EVDM. Mass spectrometry was used to examine the proteome profile of each preparation. Results: The extent of liver fibrosis and number of activated HSCs were reduced significantly in CCl4-treated mice given hAEC-EVs, hAEC-CM, and hAEC EVDM compared to untreated controls. Hepatic macrophages were significantly decreased in all treatment groups, where a predominant M2 phenotype was observed. Human HSCs cultured with hAEC-EV and hAEC-CM displayed a significant reduction in collagen synthesis and hAEC-EV, hAEC-CM, and hAEC-EVDM altered macrophage polarization in bone marrow-derived mouse macrophages. Proteome analysis showed that 164 proteins were unique to hAEC-EV in comparison to hAEC-CM and hAEC-EVDM, and 51 proteins were co-identified components with the hAEC-EV fraction. Conclusion: This study provides novel data showing that hAEC-derived EVs significantly reduced liver fibrosis and macrophage infiltration to an extent similar to hAEC-EVDM and hAEC-CM. hAEC-EV-based therapy may be a potential therapeutic option for liver fibrosis.
Collapse
Affiliation(s)
- Majid Alhomrani
- Centre for Inflammatory Diseases, Monash University, Clayton, VIC, Australia.,Hudson Institute of Medical Research, Clayton, VIC, Australia.,Medical College, Taif University, Taif, Saudi Arabia
| | - Jeanne Correia
- Centre for Inflammatory Diseases, Monash University, Clayton, VIC, Australia.,Gastroenterology and Hepatology Unit, Monash Health, Clayton, VIC, Australia
| | - Marcus Zavou
- Centre for Inflammatory Diseases, Monash University, Clayton, VIC, Australia.,Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Bryan Leaw
- Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Nathan Kuk
- Centre for Inflammatory Diseases, Monash University, Clayton, VIC, Australia.,Gastroenterology and Hepatology Unit, Monash Health, Clayton, VIC, Australia
| | - Rong Xu
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Mohamed I Saad
- Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Alexander Hodge
- Centre for Inflammatory Diseases, Monash University, Clayton, VIC, Australia.,Gastroenterology and Hepatology Unit, Monash Health, Clayton, VIC, Australia
| | - David W Greening
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Rebecca Lim
- Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - William Sievert
- Centre for Inflammatory Diseases, Monash University, Clayton, VIC, Australia.,Gastroenterology and Hepatology Unit, Monash Health, Clayton, VIC, Australia
| |
Collapse
|
23
|
Pchelintseva E, Djamgoz MBA. Mesenchymal stem cell differentiation: Control by calcium-activated potassium channels. J Cell Physiol 2017; 233:3755-3768. [PMID: 28776687 DOI: 10.1002/jcp.26120] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 08/01/2017] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are widely used in modern medicine for which understanding the mechanisms controlling their differentiation is fundamental. Ion channels offer novel insights to this process because of their role in modulating membrane potential and intracellular milieu. Here, we evaluate the contribution of calcium-activated potassium (KCa ) channels to the three main components of MSC differentiation: initiation, proliferation, and migration. First, we demonstrate the importance of the membrane potential (Vm ) and the apparent association of hyperpolarization with differentiation. Of KCa subtypes, most evidence points to activity of big-conductance channels in inducing initiation. On the other hand, intermediate-conductance currents have been shown to promote progression through the cell cycle. While there is no information on the role of KCa channels in migration of MSCs, work from other stem cells and cancer cells suggest that intermediate-conductance and to a lesser extent big-conductance channels drive migration. In all cases, these effects depend on species, tissue origin and lineage. Finally, we present a conceptual model that demonstrates how KCa activity could influence differentiation by regulating Vm and intracellular Ca2+ oscillations. We conclude that KCa channels have significant involvement in MSC differentiation and could potentially enable novel tissue engineering approaches and therapies.
Collapse
Affiliation(s)
- Ekaterina Pchelintseva
- Department of Life Sciences, Imperial College London, South Kensington Campus, Neuroscience Solution to Cancer Research Group, London, UK.,Department of Bioengineering, Imperial College London, South Kensington Campus, London, UK
| | - Mustafa B A Djamgoz
- Department of Life Sciences, Imperial College London, South Kensington Campus, Neuroscience Solution to Cancer Research Group, London, UK
| |
Collapse
|
24
|
Lou G, Yang Y, Liu F, Ye B, Chen Z, Zheng M, Liu Y. MiR-122 modification enhances the therapeutic efficacy of adipose tissue-derived mesenchymal stem cells against liver fibrosis. J Cell Mol Med 2017; 21:2963-2973. [PMID: 28544786 PMCID: PMC5661245 DOI: 10.1111/jcmm.13208] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 03/11/2017] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cell (MSC) transplantation alone may be insufficient for treatment of liver fibrosis because of complicated histopathological changes in the liver. Given that miR‐122 plays an essential role in liver fibrosis by negatively regulating the proliferation and transactivation of hepatic stellate cells (HSCs), this study investigated whether miR‐122 modification can improve the therapeutic efficacy of adipose tissue‐derived MSCs in treating liver fibrosis. MiR‐122‐modified AMSCs (AMSC‐122) were constructed through lentivirus‐mediated transfer of pre‐miR‐122. MiR‐122‐modified AMSCs expressed high level of miR‐122, while they retained their phenotype and differentiation potential as naïve AMSCs. AMSC‐122 more effectively suppressed the proliferation of and collagen maturation in HSCs than scramble miRNA‐modified AMSCs. In addition, AMSC‐derived exosomes mediated the miR‐122 communication between AMSCs and HSCs, further affecting the expression levels of miR‐122 target genes, such as insulin‐like growth factor receptor 1 (IGF1R), Cyclin G(1) (CCNG1) and prolyl‐4‐hydroxylase α1 (P4HA1), which are involved in proliferation of and collagen maturation in HSCs. Moreover, miR‐122 modification enhanced the therapeutic efficacy of AMSCs in the treatment of carbon tetrachloride (CCl4)‐induced liver fibrosis by suppressing the activation of HSCs and alleviating collagen deposition. Results demonstrate that miR‐122 modification improves the therapeutic efficacy of AMSCs through exosome‐mediated miR‐122 communication; thus, miR‐122 modification is a new potential strategy for treatment of liver fibrosis.
Collapse
Affiliation(s)
- Guohua Lou
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ying Yang
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Feifei Liu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bingjue Ye
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhi Chen
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Min Zheng
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yanning Liu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
25
|
Shiota G, Itaba N. Progress in stem cell-based therapy for liver disease. Hepatol Res 2017; 47:127-141. [PMID: 27188253 DOI: 10.1111/hepr.12747] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 05/03/2016] [Accepted: 05/12/2016] [Indexed: 12/16/2022]
Abstract
Liver transplantation has been accepted as a useful therapeutic approach for patients with end-stage liver disease. However, the mismatch between the great demand for liver transplants and the number of available donor organs underscores the urgent need for alternative therapeutic strategies for patients with acute and chronic liver failure. The rapidly growing knowledge on stem cell biology has opened new avenues toward stem cell-based therapy for liver disease. As stem cells have capacity for high proliferation and multipotent differentiation, the characteristics of stem cells fit the cell therapy. Several types of cells have been investigated as possible sources of liver regeneration: mesenchymal stem cells, hematopoietic stem cells, liver progenitor cells, induced pluripotent stem cells, and bone marrow mononuclear cells. In vitro and in vivo experiments revealed that these cells have great potential as candidates of stem cell therapy. We reviewed the reports on clinical trials of cell therapy for liver disease that have been recently undertaken using mesenchymal stem cells, hematopoietic stem cells, bone marrow mononuclear cells, and liver progenitor cells. These reports have heterogeneity of description of trial design, types of infused cells, patient population, and efficacy of therapies. We addressed these reports from these viewpoints and clarified their significance. We hope that this review article will provide a perspective on the available approaches based on stem cell-based therapy for liver disease.
Collapse
Affiliation(s)
- Goshi Shiota
- Division of Molecular and Genetic Medicine, Graduate School of Medicine, Tottori University, Yonago, Japan
| | - Noriko Itaba
- Departments of Genetic Medicine and Regenerative Therapeutics, Graduate School of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
26
|
Chen L, Zhang C, Chen L, Wang X, Xiang B, Wu X, Guo Y, Mou X, Yuan L, Chen B, Wang J, Xiang C. Human Menstrual Blood-Derived Stem Cells Ameliorate Liver Fibrosis in Mice by Targeting Hepatic Stellate Cells via Paracrine Mediators. Stem Cells Transl Med 2016; 6:272-284. [PMID: 28170193 PMCID: PMC5442725 DOI: 10.5966/sctm.2015-0265] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 06/16/2016] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) may have potential applications in regenerative medicine for the treatment of chronic liver diseases (CLDs). Human menstrual blood is a novel source of MSCs, termed menstrual blood-derived stem cells (MenSCs). Compared with bone marrow MSCs, MenSCs exhibit a higher proliferation rate and they can be obtained through a simple, safe, painless procedure without ethical concerns. Although the therapeutic efficacy of MenSCs has been explored in some diseases, their effects on liver fibrosis are still unclear. In the present study, we investigated the therapeutic effects of MenSC transplantation in a carbon tetrachloride-induced mouse model of liver fibrosis. These results revealed that MenSCs markedly improved liver function, attenuated collagen deposition, and inhibited activated hepatic stellate cells up to 2 weeks after transplantation. Moreover, tracking of green fluorescent protein-expressing MenSCs demonstrated that transplanted cells migrated to the sites of injury, but few differentiated into functional hepatocyte-like cells. Transwell coculturing experiments also showed that MenSCs suppressed proliferation of LX-2 cells (an immortalized hepatic stellate cell line) through secretion of monocyte chemoattractant protein-1, interleukin-6, hepatocyte growth factor, growth-related oncogene, interleukin-8, and osteoprotegerin. Collectively, our results provided preliminary evidence for the antifibrotic capacity of MenSCs in liver fibrosis and suggested that these cells may be an alternative therapeutic approach for the treatment of CLDs. Stem Cells Translational Medicine 2017;6:272-284.
Collapse
Affiliation(s)
- Lijun Chen
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, People's Republic of China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases and Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Molecular Diagnosis Division, Zhejiang‐California International Nanosystem Institute, Zhejiang University, Hangzhou, People's Republic of China
| | - Chunfeng Zhang
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, People's Republic of China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases and Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Molecular Diagnosis Division, Zhejiang‐California International Nanosystem Institute, Zhejiang University, Hangzhou, People's Republic of China
| | - Lu Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases and Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Xiaojun Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases and Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Bingyu Xiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases and Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Xiaoxing Wu
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Yang Guo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases and Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Xiaozhou Mou
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou, People's Republic of China
| | - Li Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases and Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Bo Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases and Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Jinfu Wang
- Institute of Cell and Development, College of Life Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Charlie Xiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases and Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
- Molecular Diagnosis Division, Zhejiang‐California International Nanosystem Institute, Zhejiang University, Hangzhou, People's Republic of China
- Institute for Cell‐Based Drug Development of Zhejiang Province, Hangzhou, People's Republic of China
| |
Collapse
|
27
|
Yuan D, Su G, Liu Y, Chi X, Feng J, Zhu Q, Cai J, Luo G, Hei Z. Propofol attenuated liver transplantation-induced acute lung injury via connexin43 gap junction inhibition. J Transl Med 2016; 14:194. [PMID: 27364362 PMCID: PMC4929774 DOI: 10.1186/s12967-016-0954-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 06/21/2016] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Postoperative acute lung injury (ALI) is a severe complication after liver transplantation, which influences patient survival rate obviously. However, its mechanisms are unclear and effective therapies are still lacking. The current study focused on effects of propofol on liver transplantation-induced ALI and whether its underlying mechanism was relative with connexin43 (Cx43) alternation. The authors postulated that endotoxin induced enhancement of Cx43 gap junction (GJ) plays a critical role in mediating post liver transplantation ALI and that pretreatment with the anesthetic propofol, known to inhibit gap junction, can confer effective protection. METHODS Male Sprague-Dawley rats underwent autologous orthotopic liver transplantation (AOLT) in the absence or presence of treatments with the selective Cx43 inhibitor, enanthol (0.1 mg/kg) and propofol (50 mg/kg), a commonly used anesthetic in clinical anesthesia. In vitro study, BEAS-2B cells, a kind of lung epithelial cell line expressing Cx43, exposed to lipopolysaccharide (LPS), which mainly contributed to ALI. Function of Cx43 GJ was regulated by Cx43 specific inhibitors, gap26 (300 μM) or enhancer, retinoic acid (10 μM) and two specific siRNAs. RESULTS Compared with the sham group, AOLT results in ALI obviously with plasma endotoxin increase. Cx43 inhibition decreased ALI through inflammatory reaction reduction. In vitro studies, LPS-induced BEAS-2B cells damage was attenuated by Cx43 function inhibition, but amplified by enhancement. Another important finding was propofol reduced Cx43 function and protected against LPS-mediated BEAS-2B cells damage or AOLT-induced ALI, mechanisms of which were also associated with inflammatory reaction decrease. CONCLUSION Cx43 plays a vital role in liver transplantation-induced ALI. Propofol decreased Cx43 function and protected against ALI in vivo and in vitro. This finding provide a new basis for targeted intervention of organ protection in liver transplantation, even in other kinds of operations.
Collapse
Affiliation(s)
- Dongdong Yuan
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Tianhe Road, Guangzhou, People’s Republic of China
| | - Guangjie Su
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Tianhe Road, Guangzhou, People’s Republic of China
| | - Yue Liu
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Tianhe Road, Guangzhou, People’s Republic of China
| | - Xinjin Chi
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Tianhe Road, Guangzhou, People’s Republic of China
| | - Jiayu Feng
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Tianhe Road, Guangzhou, People’s Republic of China
| | - Qianqian Zhu
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Tianhe Road, Guangzhou, People’s Republic of China
| | - Jun Cai
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Tianhe Road, Guangzhou, People’s Republic of China
| | - Gangjian Luo
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Tianhe Road, Guangzhou, People’s Republic of China
| | - Ziqing Hei
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Tianhe Road, Guangzhou, People’s Republic of China
| |
Collapse
|
28
|
Huang CK, Aihara A, Iwagami Y, Yu T, Carlson R, Koga H, Kim M, Zou J, Casulli S, Wands JR. Expression of transforming growth factor β1 promotes cholangiocarcinoma development and progression. Cancer Lett 2016; 380:153-62. [PMID: 27364974 DOI: 10.1016/j.canlet.2016.05.038] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 04/26/2016] [Accepted: 05/05/2016] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND AIMS The role of transforming growth factor beta 1 (TGFβ1) in cholangiocarcinoma (CCA) initiation and growth requires further definition. METHODS We employed pharmacological and genetic approaches to inhibit or enhance TGFβ1 signaling, respectively, and determine the cellular mechanisms involved. RESULTS It was observed that inhibiting TGFβ1 activity with short hairpin RNA (shRNA) or pharmaceutical agents suppressed CCA development and growth, whereas overexpression of TGFβ1 enhanced CCA tumor size and promoted intrahepatic metastasis in a rat model. Suppression of TGFβ1 activity inhibits downstream target gene expression mediated by miR-34a that includes cyclin D1, CDK6, and c-Met. In addition, "knockdown" of TGFβ1 expression revealed a miR-34a positive feedback mechanism for enhanced p21 expression in CCAs. A miR-34a inhibitor reversed the effects of "knocking down" TGFβ1 on cell growth, migration, cyclin D1, CDK6 and c-Met expression, suggesting that TGFβ1 mediated effects occur, in part, through this miR-34a signaling pathway. Overexpression of TGFβ1 was associated with CCA tumor progression. CONCLUSIONS This study suggests that TGFβ1 is involved in CCA tumor progression and participates through miR-34a mediated downstream cascades, and is a target to inhibit CCA development and growth.
Collapse
Affiliation(s)
- Chiung-Kuei Huang
- Division of Gastroenterology & Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI 02903, USA
| | - Arihiro Aihara
- Division of Gastroenterology & Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI 02903, USA
| | - Yoshifumi Iwagami
- Division of Gastroenterology & Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI 02903, USA
| | - Tunan Yu
- Division of Gastroenterology & Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI 02903, USA
| | - Rolf Carlson
- Division of Gastroenterology & Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI 02903, USA
| | - Hironori Koga
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan
| | - Miran Kim
- Division of Gastroenterology & Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI 02903, USA
| | - Jing Zou
- Division of Gastroenterology & Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI 02903, USA
| | - Sarah Casulli
- Division of Gastroenterology & Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI 02903, USA
| | - Jack R Wands
- Division of Gastroenterology & Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI 02903, USA.
| |
Collapse
|
29
|
ASC-J9(®) suppresses castration resistant prostate cancer progression via degrading the enzalutamide-induced androgen receptor mutant AR-F876L. Cancer Lett 2016; 379:154-60. [PMID: 27233475 DOI: 10.1016/j.canlet.2016.05.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 04/27/2016] [Accepted: 05/18/2016] [Indexed: 12/17/2022]
Abstract
Androgen deprivation therapy (ADT) with the newly developed powerful anti-androgen enzalutamide (Enz, also known as MDV3100) has promising therapeutic effects to suppress castration resistant prostate cancer (CRPC) and extending patients' lives an extra 4.8 months. However, most Enz therapy eventually fails with the development of Enz resistance. The detailed mechanisms how CRPC develops Enz resistance remain unclear and may involve multiple mechanisms. Among them, the induction of the androgen receptor (AR) mutant AR-F876L in some CRPC patients may represent one driving force that confers Enz resistance. Here, we demonstrate that the AR degradation enhancer, ASC-J9(®), not only degrades wild-type AR, but also has the ability to target AR-F876L. The consequence of suppressing AR-F876L may then abrogate AR-F876L mediated CRPC cell proliferation and metastasis. Thus, developing ASC-J9(®) as a new therapeutic approach may represent a novel therapy to better suppress CRPC that has already developed Enz resistance.
Collapse
|
30
|
Huang CK, Iwagami Y, Aihara A, Chung W, de la Monte S, Thomas JM, Olsen M, Carlson R, Yu T, Dong X, Wands J. Anti-Tumor Effects of Second Generation β-Hydroxylase Inhibitors on Cholangiocarcinoma Development and Progression. PLoS One 2016; 11:e0150336. [PMID: 26954680 PMCID: PMC4783022 DOI: 10.1371/journal.pone.0150336] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 02/11/2016] [Indexed: 12/19/2022] Open
Abstract
Cholangiocarcinoma (CCA) has a poor prognosis due to widespread intrahepatic spread. Aspartate β-hydroxylase (ASPH) is a transmembrane protein and catalyzes the hydroxylation of aspartyl and asparaginyl residues in calcium binding epidermal growth factor (cbEGF)-like domains of various proteins, including Notch receptors and ligands. ASPH is highly overexpressed (>95%) in human CCA tumors. We explored the molecular mechanisms by which ASPH mediated the CCA malignant phenotype and evaluated the potential of ASPH as a therapeutic target for CCA. The importance of expression and enzymatic activity of ASPH for CCA growth and progression was examined using shRNA "knockdown" and a mutant construct that reduced its catalytic activity. Second generation small molecule inhibitors (SMIs) of β-hydroxylase activity were developed and used to target ASPH in vitro and in vivo. Subcutaneous and intrahepatic xenograft rodent models were employed to determine anti-tumor effects on CCA growth and development. It was found that the enzymatic activity of ASPH was critical for mediating CCA progression, as well as inhibiting apoptosis. Mechanistically, ASPH overexpression promoted Notch activation and modulated CCA progression through a Notch1-dependent cyclin D1 pathway. Targeting ASPH with shRNAs or a SMI significantly suppressed CCA growth in vivo.
Collapse
Affiliation(s)
- Chiung-Kuei Huang
- Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island, United States of America
| | - Yoshifumi Iwagami
- Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island, United States of America
| | - Arihiro Aihara
- Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island, United States of America
| | - Waihong Chung
- Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island, United States of America
| | - Suzanne de la Monte
- Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island, United States of America
| | - John-Michael Thomas
- Department of Medical Chemistry, College of Pharmacy Glendale, Midwestern University, Glendale, Arizona, United States of America
| | - Mark Olsen
- Department of Medical Chemistry, College of Pharmacy Glendale, Midwestern University, Glendale, Arizona, United States of America
| | - Rolf Carlson
- Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island, United States of America
| | - Tunan Yu
- Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island, United States of America
| | - Xiaoqun Dong
- Department of Biomedical and Pharmaceutical Science, College of Pharmacy, The University of Rhode Island, Pharmacy Building, 7 Greenhouse Road, Kingston, Rhode Island, United States of America
| | - Jack Wands
- Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island, United States of America
- * E-mail:
| |
Collapse
|
31
|
The Androgen Receptor Bridges Stem Cell-Associated Signaling Nodes in Prostate Stem Cells. Stem Cells Int 2016; 2016:4829602. [PMID: 26880966 PMCID: PMC4737002 DOI: 10.1155/2016/4829602] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 12/04/2015] [Accepted: 12/13/2015] [Indexed: 11/18/2022] Open
Abstract
The therapeutic potential of stem cells relies on dissecting the complex signaling networks that are thought to regulate their pluripotency and self-renewal. Until recently, attention has focused almost exclusively on a small set of "core" transcription factors for maintaining the stem cell state. It is now clear that stem cell regulatory networks are far more complex. In this review, we examine the role of the androgen receptor (AR) in coordinating interactions between signaling nodes that govern the balance of cell fate decisions in prostate stem cells.
Collapse
|
32
|
Chen L, Lv D, Wang D, Chen X, Zhu Z, Cao Y, Chai Y. A novel strategy of profiling the mechanism of herbal medicines by combining network pharmacology with plasma concentration determination and affinity constant measurement. MOLECULAR BIOSYSTEMS 2016; 12:3347-3356. [DOI: 10.1039/c6mb00500d] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Herbal medicines have long been widely used in the treatment of various complex diseases in China.
Collapse
Affiliation(s)
- Langdong Chen
- Department of Pharmaceutical Analysis
- School of Pharmacy
- Second Military Medical University
- Shanghai 200433
- China
| | - Diya Lv
- Center of Analysis and Testing
- School of Pharmacy
- Second Military Medical University
- Shanghai 200433
- China
| | - Dongyao Wang
- Department of Pharmaceutical Analysis
- School of Pharmacy
- Second Military Medical University
- Shanghai 200433
- China
| | - Xiaofei Chen
- Department of Pharmaceutical Analysis
- School of Pharmacy
- Second Military Medical University
- Shanghai 200433
- China
| | - Zhenyu Zhu
- Center of Analysis and Testing
- School of Pharmacy
- Second Military Medical University
- Shanghai 200433
- China
| | - Yan Cao
- Department of Biochemical Pharmacy
- School of Pharmacy
- Second Military Medical University
- Shanghai 200433
- China
| | - Yifeng Chai
- Department of Pharmaceutical Analysis
- School of Pharmacy
- Second Military Medical University
- Shanghai 200433
- China
| |
Collapse
|
33
|
Huang CK, Luo J, Lee SO, Chang C. Concise review: androgen receptor differential roles in stem/progenitor cells including prostate, embryonic, stromal, and hematopoietic lineages. Stem Cells 2015; 32:2299-308. [PMID: 24740898 DOI: 10.1002/stem.1722] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 03/07/2014] [Accepted: 03/12/2014] [Indexed: 01/07/2023]
Abstract
Stem/progenitor (S/P) cells are special types of cells that have the ability to generate tissues throughout their entire lifetime and play key roles in the developmental process. Androgen and the androgen receptor (AR) signals are the critical determinants in male gender development, suggesting that androgen and AR signals might modulate the behavior of S/P cells. In this review, we summarize the AR effects on the behavior of S/P cells, including self-renewal, proliferation, apoptosis, and differentiation in normal S/P cells, as well as proliferation, invasion, and self-renewal in prostate cancer S/P cells. AR plays a protective role in the oxidative stress-induced apoptosis in embryonic stem cells. AR inhibits the self-renewal of embryonic stem cells, bone marrow stromal cells, and prostate S/P cells, but promotes their differentiation except for adipogenesis. However, AR promotes the proliferation of hematopoietic S/P cells and stimulates hematopoietic lineage differentiation. In prostate cancer S/P cells, AR suppresses their self-renewal, metastasis, and invasion. Together, AR differentially influences the characteristics of normal S/P cells and prostate cancer S/P cells, and targeting AR might improve S/P cell transplantation therapy, especially in embryonic stem cells and bone marrow stromal cells.
Collapse
Affiliation(s)
- Chiung-Kuei Huang
- Departments of Pathology, Urology, Radiation Oncology, the George Whipple Lab for Cancer Research, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester, New York, USA
| | | | | | | |
Collapse
|
34
|
Huang CK, Luo J, Lai KP, Wang R, Pang H, Chang E, Yan C, Sparks J, Lee SO, Cho J, Chang C. Androgen receptor promotes abdominal aortic aneurysm development via modulating inflammatory interleukin-1α and transforming growth factor-β1 expression. Hypertension 2015; 66:881-91. [PMID: 26324502 DOI: 10.1161/hypertensionaha.115.05654] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Sex difference is a risk factor for abdominal aortic aneurysm (AAA) formation yet the reason for male predominance remains unclear. Androgen and the androgen receptor (AR) influence the male sex difference, indicating that AR signaling may affect AAA development. Using angiotensin II–induced AAA in apolipoprotein E null mouse models (82.4% AAA incidence), we found that mice lacking AR failed to develop AAA and aorta had dramatically reduced macrophages infiltration and intact elastic fibers. These findings suggested that AR expression in endothelial cells, macrophages, or smooth muscle cells might play a role in AAA development. Selective knockout of AR in each of these cell types further demonstrated that mice lacking AR in macrophages (20% AAA incidence) or smooth muscle cells (12.5% AAA incidence) but not in endothelial cells (71.4% AAA incidence) had suppressed AAA development. Mechanism dissection showed that AR functioned through modulation of interleukin-1α (IL-1α) and transforming growth factor-β1 signals and by targeting AR with the AR degradation enhancer ASC-J9 led to significant suppression of AAA development. These results demonstrate the underlying mechanism by which AR influences AAA development is through IL-1α and transforming growth factor-β1, and provides a potential new therapy to suppress/prevent AAA by targeting AR with ASC-J9.
Collapse
Affiliation(s)
- Chiung-Kuei Huang
- George Whipple Laboratory for Cancer Research, Departments of Pathology, Urology, Radiation Oncology, and The Wilmot Cancer Center, University of Rochester Medical Center, Rochester
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Christ B, Brückner S, Winkler S. The Therapeutic Promise of Mesenchymal Stem Cells for Liver Restoration. Trends Mol Med 2015; 21:673-686. [PMID: 26476857 DOI: 10.1016/j.molmed.2015.09.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 09/04/2015] [Accepted: 09/11/2015] [Indexed: 12/17/2022]
Abstract
Hepatocyte transplantation aims to provide a functional substitution of liver tissue lost due to trauma or toxins. Chronic liver diseases are associated with inflammation, deterioration of tissue homeostasis, and deprivation of metabolic capacity. Recent advances in liver biology have focused on the pro-regenerative features of mesenchymal stem cells (MSCs). We argue that MSCs represent an attractive therapeutic option to treat liver disease. Indeed, their pleiotropic actions include the modulation of immune reactions, the stimulation of cell proliferation, and the attenuation of cell death responses. These characteristics are highly warranted add-ons to their capacity for hepatocyte differentiation. Undoubtedly, the elucidation of the regenerative mechanisms of MSCs in different liver diseases will promote their versatile and disease-specific therapeutic use.
Collapse
Affiliation(s)
- Bruno Christ
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, University Hospital Leipzig, University of Leipzig, Leipzig, Germany.
| | - Sandra Brückner
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, University Hospital Leipzig, University of Leipzig, Leipzig, Germany
| | - Sandra Winkler
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, University Hospital Leipzig, University of Leipzig, Leipzig, Germany
| |
Collapse
|
36
|
Sun C, Hu JJ, Pan Q, Cao Y, Fan JG, Li GM. Hepatic differentiation of rat induced pluripotent stem cells in vitro. World J Gastroenterol 2015; 21:11118-11126. [PMID: 26494966 PMCID: PMC4607909 DOI: 10.3748/wjg.v21.i39.11118] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 04/23/2015] [Accepted: 08/25/2015] [Indexed: 02/06/2023] Open
Abstract
AIM To show the efficient generation of hepatocyte-like cells (HLCs) differentiated from the induced pluripotent stem cells (iPSCs) of rats. METHODS Hepatic differentiation was achieved using a three-step protocol with several growth factors. First, rat iPSCs were differentiated into definitive endoderm cells using Activin A and Wnt3a treatment. Then fibroblast growth factor 4 and bone morphogenetic protein 2 were added to the culture medium and used to induce hepatic differentiation. Finally, hepatocyte growth factor, Oncostatin M and dexamethasone were used for hepatic maturation. The liver-related markers and functions of HLCs were assessed at the gene and protein levels. RESULTS After endodermal induction, the differentiated cells expressed endodermal markers forkhead box protein A2 and SRY-box containing gene 17 at the mRNA and protein levels. After 20 d of culture, the iPSCs were differentiated into HLCs. These differentiated cells expressed hepatic markers including α-fetoprotein, albumin CK8, CK18, CK19, and transcription factor HNF-4α. In addition, the cells expressed functional proteins such as α1-antitrypsin, cytochrome P450 1A2 and CYP 3A4. They acted like healthy hepatic cells, storing glycogen and taking up indocyanine green and low-density lipoproteins. Also, the rates of urea synthesis (20 d 1.202 ± 0.080 mg/dL vs 0 d 0.317 ± 0.021 mg/dL, P < 0.01) and albumin secretion (20 d 1.601 ± 0.102 mg/dL vs 0 d 0.313 ± 0.015 mg/dL, P < 0.01) increased significantly as differentiation progressed. CONCLUSION Rat iPSCs can differentiate into HLCs rapidly and efficiently. These differentiated cells may be an attractive resource for treatment of end-stage liver disease.
Collapse
|
37
|
Baligar P, Mukherjee S, Kochat V, Rastogi A, Mukhopadhyay A. Molecular and Cellular Functions Distinguish Superior Therapeutic Efficiency of Bone Marrow CD45 Cells Over Mesenchymal Stem Cells in Liver Cirrhosis. Stem Cells 2015; 34:135-47. [PMID: 26389810 DOI: 10.1002/stem.2210] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 08/24/2015] [Indexed: 12/12/2022]
Abstract
Liver fibrosis is strongly associated with chronic inflammation. As an alternative to conventional treatments for fibrosis, mesenchymal stem cells (MSCs) therapy is found to be attractive due to its immunomodulatory functions. However, low survival rate and profibrogenic properties of MSCs remain the major concerns, leading to skepticism in many investigators. Here, we have asked the question whether bone marrow (BM)-derived CD45 cells is the better candidate than MSCs to treat fibrosis, if so, what are the molecular mechanisms that make such distinction. Using CCl4 -induced liver fibrosis mouse model of a Metavir fibrosis score 3, we showed that BM-CD45 cells have better antifibrotic effect than adipose-derived (AD)-MSCs. In fact, our study revealed that antifibrotic potential of CD45 cells are compromised by the presence of MSCs. This difference was apparently due to significantly high level expressions of matrix metalloproteinases-9 and 13, and the suppression of hepatic stellate cells' (HpSCs) activation in the CD45 cells transplantation group. Mechanism dissection studied in vitro supported the above opposing results and revealed that CD45 cell-secreted FasL induced apoptotic death of activated HpSCs. Further analyses suggest that MSC-secreted transforming growth factor β and insulin-like growth factor-1 promoted myofibroblastic differentiation of HpSCs and their proliferation. Additionally, the transplantation of CD45 cells led to functional improvement of the liver through repair and regeneration. Thus, BM-derived CD45 cells appear as a superior candidate for the treatment of liver fibrosis due to structural and functional improvement of CCl4 -induced fibrotic liver, which were much lower in case of AD-MSC therapy.
Collapse
Affiliation(s)
- Prakash Baligar
- Stem Cell Biology Laboratory, National Institute of Immunology, New Delhi, India
| | - Snehasish Mukherjee
- Stem Cell Biology Laboratory, National Institute of Immunology, New Delhi, India
| | - Veena Kochat
- Stem Cell Biology Laboratory, National Institute of Immunology, New Delhi, India
| | - Archana Rastogi
- Department of Pathology, Institute of Liver & Biliary Sciences, Vasant Kunj, New Delhi, India
| | - Asok Mukhopadhyay
- Stem Cell Biology Laboratory, National Institute of Immunology, New Delhi, India
| |
Collapse
|
38
|
In Vitro and In Vivo Hepatic Differentiation of Adult Somatic Stem Cells and Extraembryonic Stem Cells for Treating End Stage Liver Diseases. Stem Cells Int 2015; 2015:871972. [PMID: 26347063 PMCID: PMC4541019 DOI: 10.1155/2015/871972] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 06/17/2015] [Accepted: 07/13/2015] [Indexed: 02/06/2023] Open
Abstract
The shortage of liver donors is a major handicap that prevents most patients from receiving liver transplantation and places them on a waiting list for donated liver tissue. Then, primary hepatocyte transplantation and bioartificial livers have emerged as two alternative treatments for these often fatal diseases. However, another problem has emerged. Functional hepatocytes for liver regeneration are in short supply, and they will dedifferentiate immediately in vitro after they are isolated from liver tissue. Alternative stem-cell-based therapeutic strategies, including hepatic stem cells (HSCs), embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and mesenchymal stem cells (MSCs), are more promising, and more attention has been devoted to these approaches because of the high potency and proliferation ability of the cells. This review will focus on the general characteristics and the progress in hepatic differentiation of adult somatic stem cells and extraembryonic stem cells
in vitro and in vivo for the treatment of end stage liver diseases. The hepatic differentiation of stem cells would offer an ideal and promising source for cell therapy and tissue engineering for treating liver diseases.
Collapse
|
39
|
Huang CK, Yu T, de la Monte SM, Wands JR, Derdak Z, Kim M. Restoration of Wnt/β-catenin signaling attenuates alcoholic liver disease progression in a rat model. J Hepatol 2015; 63:191-8. [PMID: 25724365 PMCID: PMC4475483 DOI: 10.1016/j.jhep.2015.02.030] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 01/14/2015] [Accepted: 02/17/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS Alcoholic liver disease (ALD) is characterized by the development of fatty liver, alcoholic hepatitis, fibrosis and cirrhosis. However, the underlying mechanism(s) associated with progression remains elusive. Pro-inflammatory cytokines have been implicated in ALD progression due to pro-apoptotic effects on hepatocytes. Wnt/β-catenin signaling recently has been shown to promote inflammation and apoptosis, suggesting that activation of this signaling pathway may modulate ALD progression. The current study was designed to test whether pharmacological activation of Wnt/β-catenin signaling altered ALD development and progression in a rat model. METHODS Adult male Long Evans rats were fed with isocaloric liquid diets containing 0% or 37% ethanol for 8 weeks, and also treated with Wnt agonist during the last 3 weeks of the feeding regimen. Liver and blood samples were subjected to histology, TUNEL assay, immunoblot analysis, real-time quantitative PCR, and alanine transaminase (ALT) assay. RESULTS Wnt/β-catenin signaling was negatively correlated with Foxo3A expression and reduced steatosis, cellular injury and apoptosis in ALD rats. Mutation experiments demonstrated that Foxo3A was critical for modulating these effects. Activation of Wnt/β-catenin signaling suppressed Foxo3A-induced apoptosis through upregulation of serum/glucocorticoid regulated kinase 1 (SGK1). Moreover, pharmacological restoration of Wnt/β-catenin signaling reduced ALD progression in vivo. CONCLUSIONS Wnt/β-catenin signaling plays a protective role in ALD progression via antagonizing Foxo3A-induced apoptosis, and activation of the Wnt/β-catenin signaling cascade attenuates ALD progression.
Collapse
Affiliation(s)
- Chiung-Kuei Huang
- Liver Research Center, Division of Gastroenterology, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Tunan Yu
- Liver Research Center, Division of Gastroenterology, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Suzanne M de la Monte
- Departments of Medicine and Pathology, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Jack R Wands
- Liver Research Center, Division of Gastroenterology, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Zoltan Derdak
- Liver Research Center, Division of Gastroenterology, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Miran Kim
- Liver Research Center, Division of Gastroenterology, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA.
| |
Collapse
|
40
|
Kanda T, Yokosuka O. The androgen receptor as an emerging target in hepatocellular carcinoma. J Hepatocell Carcinoma 2015; 2:91-9. [PMID: 27508198 PMCID: PMC4918288 DOI: 10.2147/jhc.s48956] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the male-dominant liver diseases with poor prognosis, although treatments for HCC have been progressing in the past decades. Androgen receptor (AR) is a member of the nuclear receptor superfamily. Previous studies reported that AR was expressed in human HCC and non-HCC tissues. AR is activated both ligand-dependently and ligand-independently. The latter is associated with a mitogen-activated protein kinase–, v-akt murine thymoma viral oncogene homolog 1–, or signal-transducer and activator of transcription–signaling pathway, which has been implicated in the development of HCC. It has been reported that more than 200 RNA expression levels are altered by androgen treatment. In the liver, androgen-responsive genes are cytochrome P450s, transforming growth factor β, vascular endothelial growth factor, and glucose-regulated protein 78 kDa, which are also associated with human hepatocarcinogenesis. Recent studies also revealed that AR plays a role in cell migration and metastasis. It is possible that cross-talk among AR-signaling, endoplasmic reticulum stress, and innate immune response is important for human hepatocarcinogenesis and HCC development. This review shows that AR could play a potential role in human HCC and represent one of the important target molecules for the treatment of HCC.
Collapse
Affiliation(s)
- Tatsuo Kanda
- Department of Gastroenterology and Nephrology, Chiba University, Graduate School of Medicine, Chiba, Japan
| | - Osamu Yokosuka
- Department of Gastroenterology and Nephrology, Chiba University, Graduate School of Medicine, Chiba, Japan
| |
Collapse
|
41
|
Lin TH, Sato T, Barcay KR, Waters H, Loi F, Zhang R, Pajarinen J, Egashira K, Yao Z, Goodman SB. NF-κB decoy oligodeoxynucleotide enhanced osteogenesis in mesenchymal stem cells exposed to polyethylene particle. Tissue Eng Part A 2015; 21:875-83. [PMID: 25518013 DOI: 10.1089/ten.tea.2014.0144] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Excessive generation of wear particles after total joint replacement may lead to local inflammation and periprosthetic osteolysis. Modulation of the key transcription factor NF-κB in immune cells could potentially mitigate the osteolytic process. We previously showed that local delivery of ultrahigh-molecular-weight polyethylene (UHMWPE) particles recruited osteoprogenitor cells and reduced osteolysis. However, the biological effects of modulating the NF-κB signaling pathway on osteoprogenitor/mesenchymal stem cells (MSCs) remain unclear. Here we showed that decoy oligodeoxynucleotide (ODN) increased cell viability when primary murine MSCs were exposed to UHMWPE particles, but had no effects on cellular apoptosis. Decoy ODN increased transforming growth factor-beta 1 (TGF-β1) and osteoprotegerin (OPG) in MSCs exposed to UHMWPE particles. Mechanistic studies showed that decoy ODN upregulated OPG expression through a TGF-β1-dependent pathway. By measuring the alkaline phosphatase activity, osteocalcin levels, Runx2 and osteopontin expression, and performing a bone mineralization assay, we found that decoy ODN increased MSC osteogenic ability when the cells were exposed to UHMWPE particles. Furthermore, the cellular response to decoy ODN and UHMWPE particles with regard to cell phenotype, cell viability, and osteogenic ability was confirmed using primary human MSCs. Our results suggest that modulation of wear particle-induced inflammation by NF-κB decoy ODN had no adverse effects on MSCs and may potentially further mitigate periprosthetic osteolysis by protecting MSC viability and osteogenic ability.
Collapse
Affiliation(s)
- Tzu-Hua Lin
- 1 Department of Orthopaedic Surgery, Stanford University , Stanford, California
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Myocardin related transcription factor A programs epigenetic activation of hepatic stellate cells. J Hepatol 2015; 62:165-74. [PMID: 25109772 DOI: 10.1016/j.jhep.2014.07.029] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 07/17/2014] [Accepted: 07/23/2014] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Activation of hepatic stellate cells (HSCs) represents a key process in liver injury and, in the absence of intervention, leads to irreversible cirrhosis contributing significantly to the mortality of patients with liver disease. A missing link in the current understanding of HSC activation is the involvement of the epigenetic machinery. We investigated the role of the myocardin related transcription factor A (MRTF-A) in HSC activation. METHODS Liver fibrosis was induced in wild type (WT) and MRTF-A deficient (KO) mice by CCl4 injection. Expression of mRNA and protein was measured by real-time PCR, Western blotting, and immunohistochemistry. Protein binding to DNA was assayed by chromatin immunoprecipitation (ChIP). Knockdown of endogenous proteins was mediated by either small interfering RNA (siRNA) or short hairpin RNA (shRNA), carried by lentiviral particles. RESULTS KO mice exhibited resistance to CCl4-induced liver fibrosis compared to WT littermates. The expression of activated HSC signature genes was suppressed in the absence of MRTF-A. ChIP assays revealed that MRTF-A deficiency led to the erasure of key histone modifications, associated with transcriptional activation, such as H3K4 di- and tri-methylation, on the promoter regions of fibrogenic genes. Mechanistically, MRTF-A recruited a histone methyltransferase complex (COMPASS) to the promoters of fibrogenic genes to activate transcription. Silencing of individual COMPASS components dampened transactivation of fibrogenic genes in vitro and blocked liver fibrosis in mice. Oestradiol suppressed HSC activation by dampening the expression and binding activity of COMPASS. CONCLUSIONS Our data illustrate a novel mechanism that connects MRTF-A dependent histone H3K4 methylation to HSC activation.
Collapse
|
43
|
Lin J, Wu JF, Zhang Q, Zhang HW, Cao GW. Virus-related liver cirrhosis: molecular basis and therapeutic options. World J Gastroenterol 2014; 20:6457-69. [PMID: 24914367 PMCID: PMC4047331 DOI: 10.3748/wjg.v20.i21.6457] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 01/10/2014] [Accepted: 03/08/2014] [Indexed: 02/07/2023] Open
Abstract
Chronic infections with hepatitis B virus (HBV) and/or hepatitis C virus (HCV) are the major causes of cirrhosis globally. It takes 10-20 years to progress from viral hepatitis to cirrhosis. Intermediately active hepatic inflammation caused by the infections contributes to the inflammation-necrosis-regeneration process, ultimately cirrhosis. CD8(+) T cells and NK cells cause liver damage via targeting the infected hepatocytes directly and releasing pro-inflammatory cytokine/chemokines. Hepatic stellate cells play an active role in fibrogenesis via secreting fibrosis-related factors. Under the inflammatory microenvironment, the viruses experience mutation-selection-adaptation to evade immune clearance. However, immune selection of some HBV mutations in the evolution towards cirrhosis seems different from that towards hepatocellular carcinoma. As viral replication is an important driving force of cirrhosis pathogenesis, antiviral treatment with nucleos(t)ide analogs is generally effective in halting the progression of cirrhosis, improving liver function and reducing the morbidity of decompensated cirrhosis caused by chronic HBV infection. Interferon-α plus ribavirin and/or the direct acting antivirals such as Vaniprevir are effective for compensated cirrhosis caused by chronic HCV infection. The standard of care for the treatment of HCV-related cirrhosis with interferon-α plus ribavirin should consider the genotypes of IL-28B. Understanding the mechanism of fibrogenesis and hepatocyte regeneration will facilitate the development of novel therapies for decompensated cirrhosis.
Collapse
|
44
|
Wu J, Hadoke PWF, Mair I, Lim WG, Miller E, Denvir MA, Smith LB. Modulation of neointimal lesion formation by endogenous androgens is independent of vascular androgen receptor. Cardiovasc Res 2014; 103:281-90. [PMID: 24903497 PMCID: PMC4094672 DOI: 10.1093/cvr/cvu142] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aims Low androgen levels have been linked with an increased risk of cardiovascular disease in men. Previous studies have suggested that androgens directly inhibit atherosclerotic lesion formation although the underlying mechanisms for this remain unclear. This study addressed the hypothesis that endogenous androgens inhibit arterial remodelling by a direct action on the androgen receptor (AR) in the vascular wall. Methods and results We studied a series of novel mouse lines with cell-specific deletion of the AR in either the endothelium or in smooth muscle cells or both cell types. Findings were compared with a model of global androgen deficiency in wild-type mice (castrated). We characterized the cardiovascular phenotype, vascular pharmacology and histology, and assessed neointimal lesion formation following vascular injury to the femoral artery. Cell-specific AR deletion did not alter body weight, circulating testosterone levels or seminal vesicle weight, but caused limited alterations in arterial contractility and blood pressure. Neointimal lesion formation was unaltered by selective deletion of AR from the vascular endothelium, smooth muscle, or both cell types. Castration in wild-type mice increased neointimal lesion volume (Sham vs. Castration: 2.4 × 107 ± 4.5 × 106 vs. 3.9 × 107 ± 4.9 × 106 µm3, P = 0.04, n = 9–10). Conclusion Vascular cell-specific AR deletion had no effect on neointimal lesion formation, while low systemic androgen levels adversely affect neointimal lesion size. These findings suggest that the cardio-protective effects of androgens are mediated either by AR outside the vasculature or by AR-independent mechanisms.
Collapse
Affiliation(s)
- Junxi Wu
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Patrick W F Hadoke
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Iris Mair
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Win Gel Lim
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Eileen Miller
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Martin A Denvir
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Lee B Smith
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
45
|
Ma WL, Lai HC, Yeh S, Cai X, Chang C. Androgen receptor roles in hepatocellular carcinoma, fatty liver, cirrhosis and hepatitis. Endocr Relat Cancer 2014; 21:R165-82. [PMID: 24424503 PMCID: PMC4165608 DOI: 10.1530/erc-13-0283] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Androgen/androgen receptor (AR) signaling plays important roles in normal liver function and in progression of liver diseases. In studies of noncancerous liver diseases, AR knockout mouse models of liver disease have revealed that androgen/AR signaling suppresses the development of steatosis, virus-related hepatitis, and cirrhosis. In addition, studies have shown that targeting AR in bone marrow-derived mesenchymal stem cells (BM-MSCs) improves their self-renewal and migration potentials, thereby increasing the efficacy of BM-MSC transplantation as a way to control the progression of cirrhosis. Androgen/AR signaling is known to be involved in the initiation of carcinogen- or hepatitis B virus-related hepatocellular carcinoma (HCC). However, studies have demonstrated that AR, rather than androgen, plays the dominant role in cancer initiation. Therefore, targeting AR might be an appropriate therapy for patients with early-stage HCC. In contrast, androgen/AR signaling has been shown to suppress metastasis of HCC in patients with late-stage disease. In addition, there is evidence that therapy comprising Sorafenib and agents that enhance the functional expression of AR may suppress the progression of late-stage HCC.
Collapse
Affiliation(s)
- Wen-Lung Ma
- Sex Hormone Research Center, Department of
Gastroenterology, and Graduate Institute of Clinical Medical Science, China Medical
University/Hospital, Taichung 404, Taiwan
- George Whipple Lab for Cancer Research, Departments of
Pathology and Urology and The Wilmot Cancer Center, University of Rochester Medical
Center, Rochester, NY 14642, USA
| | - Hsueh-Chou Lai
- Sex Hormone Research Center, Department of
Gastroenterology, and Graduate Institute of Clinical Medical Science, China Medical
University/Hospital, Taichung 404, Taiwan
| | - Shuyuan Yeh
- George Whipple Lab for Cancer Research, Departments of
Pathology and Urology and The Wilmot Cancer Center, University of Rochester Medical
Center, Rochester, NY 14642, USA
| | - Xiujun Cai
- Department of General Surgery, Chawnshang Chang Liver
Cancer Center, Sir Run-run Shaw Hospital, Zhejiang University, Hangzhou, China
- Corresponding author: Chawnshang
Chang () and Xiujun Cai
()
| | - Chawnshang Chang
- Sex Hormone Research Center, Department of
Gastroenterology, and Graduate Institute of Clinical Medical Science, China Medical
University/Hospital, Taichung 404, Taiwan
- George Whipple Lab for Cancer Research, Departments of
Pathology and Urology and The Wilmot Cancer Center, University of Rochester Medical
Center, Rochester, NY 14642, USA
- Corresponding author: Chawnshang
Chang () and Xiujun Cai
()
| |
Collapse
|
46
|
Peng SY, Chou CJ, Cheng PJ, Ko IC, Kao YJ, Chen YH, Cheng WTK, Shaw SWS, Wu SC. Therapeutic potential of amniotic-fluid-derived stem cells on liver fibrosis model in mice. Taiwan J Obstet Gynecol 2014; 53:151-157. [PMID: 25017258 DOI: 10.1016/j.tjog.2014.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 02/10/2014] [Indexed: 01/18/2023] Open
Abstract
OBJECTIVE Liver fibrosis results from the wound healing response to chronic liver damage. Advanced liver fibrosis results in cirrhosis and liver failure, and liver transplantation is often the only option for effective therapy; however, the shortage of available donor livers limits this treatment. Thus, new therapies for advanced liver fibrosis are essential. MATERIALS AND METHODS Amniotic fluid contains an abundance of stem cells, which are derived from all three germ layers of the developing fetus. These cells do not induce teratomas in vivo and do not pose any ethical concerns. To generate liver fibrosis models, male ICR mice were treated with CCl4 via oral gavage for 4 weeks, and the serum levels of glutamate oxaloacetate transaminase, glutamate pyruvate transaminase, and albumin were higher than in the control group following chemical induction. To assess the potential of amniotic-fluid-derived stem cells (mAFSCs) to ameliorate liver fibrosis in vivo, mAFSCs were isolated from amniotic fluid of 13.5-day-old transgenic mice, which globally express the fluorescent protein, enhanced green fluorescent protein (EGFP), for tracing purposes (EGFP-mAFSCs). Single cells were injected via the mesentery (1 × 10(6) cells/mouse) of transplanted mice with liver fibrosis. RESULTS Four weeks after EGFP-mAFSC transplantation, the serum glutamate oxaloacetate transaminase, glutamate pyruvate transaminase, and albumin levels of recipient mice in the EGFP-mAFSC-injected group were significantly decreased when compared with mice in the saline-injected group. Additionally, fibrotic tissues were evaluated using Masson's trichrome staining 4 weeks after cell transplantation. Shrinkage of the fibrotic area was observed in the EGFP-mAFSC-injected group. The tissue-repair effects were also confirmed by hydroxyproline content analysis. CONCLUSION The possible repair mechanism from our data revealed that EGFP-mAFSCs may fuse with the recipient liver cells. Overall, EGFP-mAFSCs can ameliorate liver fibrosis in mice, thus providing insight into the future development of regenerative medicine.
Collapse
Affiliation(s)
- Shao-Yu Peng
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Chih-Jen Chou
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Po-Jen Cheng
- Department of Obstetrics and Gynaecology, Chang Gung Memorial Hospital at Linkou and Chang Gung University, College of Medicine, Taoyuan, Taiwan
| | - I-Chen Ko
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Yi-Jung Kao
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Yu-Hsu Chen
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan; Department of Surgery, Hualien Armed Forces General Hospital, Hualien, Taiwan
| | - Winston Teng-Kui Cheng
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan; Department of Animal Science and Biotechnology, Tunghai University, Taichung, Taiwan
| | - S W Steven Shaw
- Department of Obstetrics and Gynaecology, Chang Gung Memorial Hospital at Linkou and Chang Gung University, College of Medicine, Taoyuan, Taiwan; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan; Prenatal Cell and Gene Therapy Group, Institute for Women's Health, University College London, London, UK.
| | - Shinn-Chih Wu
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan; Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
47
|
Stock P, Brückner S, Winkler S, Dollinger MM, Christ B. Human bone marrow mesenchymal stem cell-derived hepatocytes improve the mouse liver after acute acetaminophen intoxication by preventing progress of injury. Int J Mol Sci 2014; 15:7004-28. [PMID: 24758938 PMCID: PMC4013675 DOI: 10.3390/ijms15047004] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 04/02/2014] [Accepted: 04/09/2014] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells from human bone marrow (hMSC) have the potential to differentiate into hepatocyte-like cells in vitro and continue to maintain important hepatocyte functions in vivo after transplantation into host mouse livers. Here, hMSC were differentiated into hepatocyte-like cells in vitro (hMSC-HC) and transplanted into livers of immunodeficient Pfp/Rag2⁻/⁻ mice treated with a sublethal dose of acetaminophen (APAP) to induce acute liver injury. APAP induced a time- and dose-dependent damage of perivenous areas of the liver lobule. Serum levels of aspartate aminotransferase (AST) increased to similar levels irrespective of hMSC-HC transplantation. Yet, hMSC-HC resided in the damaged perivenous areas of the liver lobules short-term preventing apoptosis and thus progress of organ destruction. Disturbance of metabolic protein expression was lower in the livers receiving hMSC-HC. Seven weeks after APAP treatment, hepatic injury had completely recovered in groups both with and without hMSC-HC. Clusters of transplanted cells appeared predominantly in the periportal portion of the liver lobule and secreted human albumin featuring a prominent quality of differentiated hepatocytes. Thus, hMSC-HC attenuated the inflammatory response and supported liver regeneration after acute injury induced by acetaminophen. They hence may serve as a novel source of hepatocyte-like cells suitable for cell therapy of acute liver diseases.
Collapse
Affiliation(s)
- Peggy Stock
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Applied Molecular Hepatology Laboratory, University Hospital Leipzig, Liebigstraße 21, D-04103 Leipzig, Germany.
| | - Sandra Brückner
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Applied Molecular Hepatology Laboratory, University Hospital Leipzig, Liebigstraße 21, D-04103 Leipzig, Germany.
| | - Sandra Winkler
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Applied Molecular Hepatology Laboratory, University Hospital Leipzig, Liebigstraße 21, D-04103 Leipzig, Germany.
| | - Matthias M Dollinger
- Clinics for Internal Medicine I, University Hospital Ulm, Albert-Einstein-Allee 23, D-89081 Ulm, Germany.
| | - Bruno Christ
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Applied Molecular Hepatology Laboratory, University Hospital Leipzig, Liebigstraße 21, D-04103 Leipzig, Germany.
| |
Collapse
|
48
|
Liu Y, El-Serag HB, Jiao L, Lee J, Moore D, Franco LM, Tavakoli-Tabasi S, Tsavachidis S, Kuzniarek J, Ramsey DJ, White DL. WNT signaling pathway gene polymorphisms and risk of hepatic fibrosis and inflammation in HCV-infected patients. PLoS One 2013; 8:e84407. [PMID: 24386373 PMCID: PMC3875538 DOI: 10.1371/journal.pone.0084407] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 11/14/2013] [Indexed: 12/17/2022] Open
Abstract
Background Chronic hepatitis C infection is the leading cause of hepatocellular carcinoma (HCC), a highly lethal malignancy with rapidly increasing prevalence in the United States. Little is known about genetic variations and HCC risk. This study aimed to determine if genetic variation in Wnt signaling pathway genes are associated with advanced hepatic fibrosis and inflammation risk in a hepatitis C virus (HCV) infected population. Methods We performed a genetic association cross-sectional study evaluating single nucleotide polymorphisms (SNPs) in 58 candidate genes and risk of FibroSURE-Acti Test determined advanced fibrosis (F3/F4-F4 advanced cases vs. F0-F3 mild controls) and inflammation (A2/A3-A3 advanced cases vs. A0-A2 mild controls). We calculated odds ratios (ORs) and 95% confidence intervals (CIs) employing multivariate logistic regression. Haplotypes were inferred by the HAPLO.STAT program, interactions were evaluated using multifactor dimensionality reduction (MDR) analysis. Results Among 425 chronically HCV-infected male veterans, 155 (37%) had advanced fibrosis and 180 (42%) had advanced inflammation. Of 3016 SNPs evaluated, eight were significantly associated with fibrosis risk (e.g., SFRP2 rs11937424: OR = 2.19, 95% CI 1.48-3.23, P = 0.00004), and seven were significantly associated with inflammation risk (e.g., SFRP1 rs16890282: OR = 2.15, 95% CI 1.39-3.16, P = 0.0004). MDR analysis identified overweight/obese, SOST rs1405952, SFRP2 rs11937424, and FZD4 rs11234870 as the best interaction model for predicting risk of fibrosis; whereas race/ethnicity, FZD1 rs1346665, and TBX3 rs1520177 as the best interaction model for predicting risk of inflammation. Conclusions Polymorphisms in several genes involved in the Wnt signaling pathway were associated with hepatic fibrosis or inflammation risk in HCV-infected males. Additional studies in other multi-ethnic HCV cohorts are needed to validate our findings in males and to assess if similar associations exist in chronically HCV-infected females.
Collapse
Affiliation(s)
- Yanhong Liu
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
- Dan L. Duncan Cancer Center at Baylor College of Medicine, Houston, Texas, United States of America
| | - Hashem B. El-Serag
- Section of Gastroenterology and Hepatology, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, United States of America
- Clinical Epidemiology and Comparative Effectiveness Program, Section of Health Services Research (IQuEST), Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, United States of America
- Dan L. Duncan Cancer Center at Baylor College of Medicine, Houston, Texas, United States of America
- Texas Medical Center Digestive Disease Center, Houston, Texas, United States of America
| | - Li Jiao
- Section of Gastroenterology and Hepatology, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, United States of America
- Clinical Epidemiology and Comparative Effectiveness Program, Section of Health Services Research (IQuEST), Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, United States of America
- Dan L. Duncan Cancer Center at Baylor College of Medicine, Houston, Texas, United States of America
- Texas Medical Center Digestive Disease Center, Houston, Texas, United States of America
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, United States of America
| | - JuSeog Lee
- Department of Systems Biology, MD Anderson Cancer Center, Houston, Texas, United States of America
| | - David Moore
- Department Molecular and Cell Biology and Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Luis M. Franco
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Shahriar Tavakoli-Tabasi
- Section of Infectious Diseases, Michael E. DeBakey Veterans Affairs Medical Center and Baylor College of Medicine, Houston, Texas, United States of America
| | - Spiridon Tsavachidis
- Dan L. Duncan Cancer Center at Baylor College of Medicine, Houston, Texas, United States of America
| | - Jill Kuzniarek
- Clinical Epidemiology and Comparative Effectiveness Program, Section of Health Services Research (IQuEST), Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, United States of America
| | - David J. Ramsey
- Clinical Epidemiology and Comparative Effectiveness Program, Section of Health Services Research (IQuEST), Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, United States of America
| | - Donna L. White
- Section of Gastroenterology and Hepatology, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, United States of America
- Clinical Epidemiology and Comparative Effectiveness Program, Section of Health Services Research (IQuEST), Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, United States of America
- Dan L. Duncan Cancer Center at Baylor College of Medicine, Houston, Texas, United States of America
- Texas Medical Center Digestive Disease Center, Houston, Texas, United States of America
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
49
|
Tanimoto H, Terai S, Taro T, Murata Y, Fujisawa K, Yamamoto N, Sakaida I. Improvement of liver fibrosis by infusion of cultured cells derived from human bone marrow. Cell Tissue Res 2013; 354:717-28. [PMID: 24104560 DOI: 10.1007/s00441-013-1727-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 08/29/2013] [Indexed: 01/12/2023]
Abstract
We develop "autologous bone marrow cell infusion (ABMi) therapy" for the treatment of human decompensated liver cirrhosis and confirm the efficacy and safety of this treatment in multicenter clinical studies. With the goal of further expanding the applications of ABMi, we first cultured human bone marrow cells and then determined whether a cell fraction found to be effective in improving liver fibrosis can be amplified. Cells harvested after two passages (P2 cells) consistently contained approximately 94% mesenchymal stem cells (MSCs); conversely, the cells harvested after only medium change (P0 cells) contained many macrophages. MSCs (2.8 × 10(8)) in P2 cells were harvested from 3.8 × 10(8) bone marrow-derived mononuclear cells after 22 days. DNA-chip analysis also showed during the culturing step that bone marrow-derived cells decreased with macrophage phenotype. The infused 5 × 10(5) P2 cells significantly improved liver fibrosis in the nonobese diabetic/severe combined immunodeficient (NOD-SCID) mouse carbon tetrachloride (CCl4) liver cirrhosis model and induced the expression of matrix metalloproteinase (MMP)-9 and suppressed expressions of alpha smooth muscle actin (αSMA), tumor necrosis factor alpha (TNFα) and transforming growth factor beta (TGFβ) in the liver. Cultured human bone marrow-derived cells (P2 cells) significantly inhibited liver fibrosis. The increase of MMP-9 and suppressed activation of hepatic stellate cells (HSCs) through the regulation of humoral factors (TNFα and TGFβ) contribute to the improvement of liver fibrosis by MSCs comprising about 94% of P2 cells. MSCs in cultured human bone marrow-derived mono-nuclear cells (BM-MNCs) proliferate sufficiently in cell therapy, so we believe our cultured bone marrow-derived cell therapy can lead to expanded clinical applications and enable outpatient therapy.
Collapse
Affiliation(s)
- Haruko Tanimoto
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Minami-Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | | | | | | | | | | | | |
Collapse
|
50
|
Chen L, Chang WC, Hung YC, Chang YY, Bao BY, Huang HC, Chung WM, Shyr CR, Ma WL. Androgen receptor increases CD133 expression and progenitor-like population that associate with cisplatin resistance in endometrial cancer cell line. Reprod Sci 2013; 21:386-94. [PMID: 23962788 DOI: 10.1177/1933719113497281] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Endometrial cancer (EMC) is a sex steroid hormone-related female malignancy. Androgen and androgen receptor (androgen/AR) signals have been implicated in EMC progression. Cancer stem/progenitor cells (CSPCs) are suspected to link to chemoresistance in patients with EMC. In this study, we examined the androgen/AR roles in cisplatin resistance and CSPC population. We found AR expression increased naive EMC side population, CSPC population, cell migration, and epithelial-mesenchymal transition. Meanwhile, it decreased cisplatin cytotoxic effect on EMC cells. Collaterally, endogenous AR expressions in EMC cells were upregulated in the cisplatin-resisting state. Moreover, AR expression could further enhance CD133 expression, CSPC-related markers, and drug-resistance gene messenger RNA expression in EMC cells. Finally, the AR-associated gene expression might go through indirect regulation. This is the first report revealing AR function on EMC cells' CSPC and cisplatin resistance.
Collapse
Affiliation(s)
- Lumin Chen
- 1Sex Hormone Research Center, Graduate Institution of Clinical Medical Science, School of Medicine, China Medical University, Taichung, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|