1
|
Dawood RM, El-Meguid MA, Salum GM, El Awady MK. Key Players of Hepatic Fibrosis. J Interferon Cytokine Res 2020; 40:472-489. [PMID: 32845785 DOI: 10.1089/jir.2020.0059] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Affiliation(s)
- Reham M. Dawood
- Genetic Engineering Division, Department of Microbial Biotechnology, National Research Centre, Giza, Egypt
| | - Mai A. El-Meguid
- Genetic Engineering Division, Department of Microbial Biotechnology, National Research Centre, Giza, Egypt
| | - Ghada Maher Salum
- Genetic Engineering Division, Department of Microbial Biotechnology, National Research Centre, Giza, Egypt
| | - Mostafa K. El Awady
- Genetic Engineering Division, Department of Microbial Biotechnology, National Research Centre, Giza, Egypt
| |
Collapse
|
2
|
Dong W, Kong M, Zhu Y, Shao Y, Wu D, Lu J, Guo J, Xu Y. Activation of TWIST Transcription by Chromatin Remodeling Protein BRG1 Contributes to Liver Fibrosis in Mice. Front Cell Dev Biol 2020; 8:340. [PMID: 32478075 PMCID: PMC7237740 DOI: 10.3389/fcell.2020.00340] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 04/17/2020] [Indexed: 12/12/2022] Open
Abstract
Liver fibrosis is a complex pathophysiological process to which many different cell types contribute. Endothelial cells play versatile roles in the regulation of liver fibrosis. The underlying epigenetic mechanism is not fully appreciated. In the present study, we investigated the role of BRG1, a chromatin remodeling protein, in the modulation of endothelial cells in response to pro-fibrogenic stimuli in vitro and liver fibrosis in mice. We report that depletion of BRG1 by siRNA abrogated TGF-β or hypoxia induced down-regulation of endothelial marker genes and up-regulation of mesenchymal marker genes in cultured endothelial cells. Importantly, endothelial-specific BRG1 deletion attenuated CCl4 induced liver fibrosis in mice. BRG1 knockdown in vitro or BRG1 knockout in vivo was accompanied by the down-regulation of TWIST, a key regulator of endothelial phenotype. Mechanistically, BRG1 interacted with and was recruited to the TWIST promoter by HIF-1α to activate TWIST transcription. BRG1 silencing rendered a more repressive chromatin structure surrounding the TWIST promoter likely contributing to TWIST down-regulation. Inhibition of HIF-1α activity dampened liver fibrosis in mice. Similarly, pharmaceutical inhibition of TWIST alleviated liver fibrosis in mice. In conclusion, our data suggest that epigenetic activation of TWIST by BRG1 contributes to the modulation of endothelial phenotype and liver fibrosis. Therefore, targeting the HIF1α-BRG1-TWIST axis may yield novel therapeutic solutions to treat liver fibrosis.
Collapse
Affiliation(s)
- Wenhui Dong
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Ming Kong
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Yuwen Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Yang Shao
- Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research and Key Laboratory of Emergency and Trauma of Ministry of Education, Institute of Cardiovascular Research of the First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Dongmei Wu
- Key Laboratory of Biotechnology on Medical Plants of Jiangsu Province and School of Life Sciences, Jiangsu Normal University, Xuzhou, China
| | - Jun Lu
- Key Laboratory of Biotechnology on Medical Plants of Jiangsu Province and School of Life Sciences, Jiangsu Normal University, Xuzhou, China
| | - Junli Guo
- Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research and Key Laboratory of Emergency and Trauma of Ministry of Education, Institute of Cardiovascular Research of the First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| |
Collapse
|
3
|
Chen J, Li HY, Wang D, Guo XZ. Delphinidin protects β2m-/Thy1+ bone marrow-derived hepatocyte stem cells against TGF-β1-induced oxidative stress and apoptosis through the PI3K/Akt pathway in vitro. Chem Biol Interact 2018; 297:109-118. [PMID: 30365941 DOI: 10.1016/j.cbi.2018.10.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/15/2018] [Accepted: 10/22/2018] [Indexed: 12/12/2022]
Abstract
β2m-/Thy1+ bone marrow-derived hepatocyte stem cells (BDHSCs) have a potential to be applied for cellular treatment in liver cirrhosis. However, the resultant tissue regeneration is restricted by transplanted cells' death. The accumulation of transforming growth factor beta 1 (TGF-β1) in liver fibrosis local microenvironment may play an essential role in the rapid cell death of implanted β2m-/Thy1+ BDHSCs. The main mechanism of poor survival of the target stem cells is still unknown. Delphinidin, an anthocyanidin, has potent antioxidant and anti-inflammatory activities. However, whether this bio-active ingredient can substantially contribute to β2m-/Thy1+ BDHSCs' protection from TGF-β1 induced apoptosis in vitro remains to be elucidated. In the present research, we determined whether delphinidin pretreatment can improve the survival of β2m-/Thy1+ BDHSCs during exposure to TGF-β1 and elucidated its underlying mechanisms. By using TGF-β1, we induced the apoptosis of β2m-/Thy1+ BDHSCs and assessed the apoptotic rates up to 24 h by flow cytometry. β2m-/Thy1+ BDHSC proliferation was gauged using 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl- 2H-tetrazolium bromide (MTT) assay. The expression grades of Bcl-2, Akt, caspase-3, and Bax were observed through Western blot analysis. We found that delphinidin can significantly impede TGF-β1-induced apoptosis dose-dependently, scavenge reactive oxygen species (ROS), and inhibit the discharge of caspase-3 in β2m-/Thy1+ BDHSCs. We also demonstrated that delphinidin can activate the phosphatidylinositol-3-kinase (PI3K)/Akt signaling pathway. The suppression of ROS and succeeding apoptosis was achieved by pretreatment with LY294002, a PI3K/Akt pathway inhibitor. In summary, our findings revealed that delphinidin can protect β2m-/Thy1+ BDHSCs from apoptosis and ROS-dependent oxidative stress induced by the TGF-β1 via PI3K/Akt signaling pathway. On the basis of these data, delphinidin can be regarded as a promising anti-apoptotic agent for enhancing β2m-/Thy1+ BDHSC survival during cell transplantation in liver cirrhosis patients.
Collapse
Affiliation(s)
- Jiang Chen
- Department of Gastroenterology, Shenyang General Hospital of PLA, No. 83 Wenhua Road Shenyang City, 110016, Liaoning, PR China
| | - Hong-Yu Li
- Department of Gastroenterology, Shenyang General Hospital of PLA, No. 83 Wenhua Road Shenyang City, 110016, Liaoning, PR China
| | - Di Wang
- Department of Gastroenterology, Shenyang General Hospital of PLA, No. 83 Wenhua Road Shenyang City, 110016, Liaoning, PR China
| | - Xiao-Zhong Guo
- Department of Gastroenterology, Shenyang General Hospital of PLA, No. 83 Wenhua Road Shenyang City, 110016, Liaoning, PR China.
| |
Collapse
|
4
|
Du C, Jiang M, Wei X, Qin J, Xu H, Wang Y, Zhang Y, Zhou D, Xue H, Zheng S, Zeng W. Transplantation of human matrix metalloproteinase-1 gene-modified bone marrow-derived mesenchymal stem cell attenuates CCL4-induced liver fibrosis in rats. Int J Mol Med 2018; 41:3175-3184. [PMID: 29512750 PMCID: PMC5881841 DOI: 10.3892/ijmm.2018.3516] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 02/12/2018] [Indexed: 12/18/2022] Open
Abstract
It has been reported that bone marrow-derived mesenchymal stem cells (BMSCs) alleviated liver fibrosis. We investigated whether BMSCs transfected with human matrix metalloproteinase 1 (BMSCs/MMP1) would improve their therapeutic effect in liver fibrosis induced by carbon tetrachloride (CCl4) in rats. BMSCs were transfected with an adenovirus carrying enhanced green fluorescence protein (GFP) and human MMP1 gene. BMSCs or BMSCs/MMP1 were directly injected into fibrotic rats via the tail vein. GFP-labeled cells appeared in the fibrotic liver after BMSC transplantation. The expression of BMSCs/MMP1 elevated levels of MMP1 in vitro. Although BMSC administration reduced liver fibrosis, transplantation of BMSCs/MMP1 enhanced the reduction of liver fibrosis to a higher level. Treatment with BMSCs/MMP1 not only decreased collagen content but also suppressed activation of hepatic stellate cells (HSCs) in fibrotic liver, which led to subsequent improvement of both liver injury and fibrosis. Treatment with BMSCs/MMP1 resulted in an improved therapeutic effect compared with BMSCs alone, which is probably because of the sustainably expressed MMP1 level in the liver. BMSCs/MMP1 transplantation not only improved biochemical parameters but also attenuated progression of liver fibrosis, suggesting that BMSCs may be a potential cell source in preventing liver fibrosis and MMP1 gene may enhance the anti-fibrotic effect of BMSCs.
Collapse
Affiliation(s)
- Chao Du
- Department of Gastroenterology and Hepatology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Mingde Jiang
- Department of Gastroenterology and Hepatology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Xiaolong Wei
- Department of Gastroenterology and Hepatology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Jianpin Qin
- Department of Gastroenterology and Hepatology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Hui Xu
- Department of Gastroenterology and Hepatology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Yunxia Wang
- Department of Gastroenterology and Hepatology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Yong Zhang
- Department of Gastroenterology and Hepatology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Dejiang Zhou
- Department of Gastroenterology and Hepatology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Hongli Xue
- Department of Gastroenterology and Hepatology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Shumei Zheng
- Department of Gastroenterology and Hepatology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Weizheng Zeng
- Department of Gastroenterology and Hepatology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| |
Collapse
|
5
|
Lan L, Liu R, Qin LY, Cheng P, Liu BW, Zhang BY, Ding SZ, Li XL. Transplantation of bone marrow-derived endothelial progenitor cells and hepatocyte stem cells from liver fibrosis rats ameliorates liver fibrosis. World J Gastroenterol 2018; 24:237-247. [PMID: 29375209 PMCID: PMC5768942 DOI: 10.3748/wjg.v24.i2.237] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/06/2017] [Accepted: 11/21/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To explore the effectiveness for treating liver fibrosis by combined transplantation of bone marrow-derived endothelial progenitor cells (BM-EPCs) and bone marrow-derived hepatocyte stem cells (BDHSCs) from the liver fibrosis environment.
METHODS The liver fibrosis rat models were induced with carbon tetrachloride injections for 6 wk. BM-EPCs from rats with liver fibrosis were obtained by different rates of adherence and culture induction. BDHSCs from rats with liver fibrosis were isolated by magnetic bead cell sorting. Tracing analysis was conducted by labeling EPCs with PKH26 in vitro to show EPC location in the liver. Finally, BM-EPCs and/or BDHSCs transplantation into rats with liver fibrosis were performed to evaluate the effectiveness of BM-EPCs and/or BDHSCs on liver fibrosis.
RESULTS Normal functional BM-EPCs from liver fibrosis rats were successfully obtained. The co-expression level of CD133 and VEGFR2 was 63.9% ± 2.15%. Transplanted BM-EPCs were located primarily in/near hepatic sinusoids. The combined transplantation of BM-EPCs and BDHSCs promoted hepatic neovascularization, liver regeneration and liver function, and decreased collagen formation and liver fibrosis degree. The VEGF levels were increased in the BM-EPCs (707.10 ± 54.32) and BM-EPCs/BDHSCs group (615.42 ± 42.96), compared with those in the model group and BDHSCs group (P < 0.05). Combination of BM-EPCs/BDHSCs transplantation induced maximal up-regulation of PCNA protein and HGF mRNA levels. The levels of alanine aminotransferase (AST), aspartate aminotransferase, total bilirubin (TBIL), prothrombin time (PT) and activated partial thromboplastin time in the BM-EPCs/BDHSCs group were significantly improved, to be equivalent to normal levels (P > 0.05) compared with those in the BDHSC (AST, TBIL and PT, P < 0.05) and BM-EPCs (TBIL and PT, P < 0.05) groups. Transplantation of BM-EPCs/BDHSCs combination significantly reduced the degree of liver fibrosis (staging score of 1.75 ± 0.25 vs BDHSCs 2.88 ± 0.23 or BM-EPCs 2.75 ± 0.16, P < 0.05).
CONCLUSION The combined transplantation exhibited maximal therapeutic effect compared to that of transplantation of BM-EPCs or BDHSCs alone. Combined transplantation of autogenous BM-EPCs and BDHSCs may represent a promising strategy for the treatment of liver fibrosis, which would eventually prevent cirrhosis and liver cancer.
Collapse
Affiliation(s)
- Ling Lan
- Department of Gastroenterology and Hepatology, the People’s Hospital of Zhengzhou University (the Henan Provincial People’s Hospital), Zhengzhou 450003, Henan Province, China
| | - Ran Liu
- Department of Oncology, Henan Provincial Rongjun Hospital, Xinxiang 453000, Henan Province, China
| | - Ling-Yun Qin
- Department of Gastroenterology and Hepatology, the Children’s Hospital of Zhengzhou, Zhengzhou 450003, Henan Province, China
| | - Peng Cheng
- Intensive Care Unit, the Second Affiliated Hospital of Luohe Medical College, Luohe 462000, Henan Province, China
| | - Bo-Wei Liu
- Department of Gastroenterology and Hepatology, the People’s Hospital of Zhengzhou University (the Henan Provincial People’s Hospital), Zhengzhou 450003, Henan Province, China
| | - Bing-Yong Zhang
- Department of Gastroenterology and Hepatology, the People’s Hospital of Zhengzhou University (the Henan Provincial People’s Hospital), Zhengzhou 450003, Henan Province, China
| | - Song-Ze Ding
- Department of Gastroenterology and Hepatology, the People’s Hospital of Zhengzhou University (the Henan Provincial People’s Hospital), Zhengzhou 450003, Henan Province, China
| | - Xiu-Ling Li
- Department of Gastroenterology and Hepatology, the People’s Hospital of Zhengzhou University (the Henan Provincial People’s Hospital), Zhengzhou 450003, Henan Province, China
| |
Collapse
|
6
|
Ahn J, Son MK, Jung KH, Kim K, Kim GJ, Lee SH, Hong SS, Park SG. Aminoacyl-tRNA synthetase interacting multi-functional protein 1 attenuates liver fibrosis by inhibiting TGFβ signaling. Int J Oncol 2015; 48:747-55. [PMID: 26692190 DOI: 10.3892/ijo.2015.3303] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 12/07/2015] [Indexed: 11/05/2022] Open
Abstract
The aminoacyl-tRNA synthetase interacting multi-functional protein 1 (AIMP1) participates in a variety of cellular processes, including translation, cell proliferation, inflammation and wound healing. Previously, we showed that the N-terminal peptide of AIMP1 (6-46 aa) induced ERK phosphorylation. Liver fibrosis is characterized by excessive deposition of extracellular matrix, which is induced by TGFβ signaling, and activated ERK is known to induce the phosphorylation of SMAD, thereby inhibiting TGFβ signaling. We assessed whether the AIMP1 peptide can inhibit collagen synthesis in hepatic stellate cells (HSCs) by activating ERK. The AIMP1 peptide induced phosphorylation of SMAD2 via ERK activation, and inhibited the nuclear translocation of SMAD, resulting in a reduction of the synthesis of type I collagen. The AIMP1 peptide attenuated liver fibrosis induced by CCl4, in a dose-dependent manner. Masson-Trichrome staining showed that the AIMP1 peptide reduced collagen deposition. Immunohistochemical staining showed that the levels of α-SMA, TGFβ and type I collagen were all reduced by the AIMP1 peptide. Liver toxicity analysis showed that the AIMP1 peptide improved the levels of relevant biological parameters in the blood. These results suggest that AIMP1 peptide may have potential for development as a therapeutic agent to treat liver fibrosis.
Collapse
Affiliation(s)
- Jongchan Ahn
- Department of Biomedical Science, College of Life Science, CHA University, Gyunggido, Republic of Korea
| | - Mi Kwon Son
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Kyung Hee Jung
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Kwangil Kim
- Department of Pathology, Bundang CHA General Hospital, CHA University, Gyunggido, Republic of Korea
| | - Gi Jin Kim
- Department of Biomedical Science, College of Life Science, CHA University, Gyunggido, Republic of Korea
| | - Soo-Hong Lee
- Department of Biomedical Science, College of Life Science, CHA University, Gyunggido, Republic of Korea
| | - Soon-Sun Hong
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Sang Gyu Park
- Department of Pharmacy, College of Pharmacy, Ajou University, Suwon, Gyunggido, Republic of Korea
| |
Collapse
|
7
|
Eun JR. Cellular origin of liver cancer stem cells. Yeungnam Univ J Med 2015. [DOI: 10.12701/yujm.2015.32.1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Jong Ryeol Eun
- Department of Internal Medicine, Myongji Hospital, Seonam University College of Medicine, Goyang, Korea
| |
Collapse
|
8
|
Hu JJ, Sun C, Lan L, Chen YW, Li DG. Therapeutic effect of transplanting beta(2)m(-)/Thy1(+) bone marrow-derived hepatocyte stem cells transduced with lentiviral-mediated HGF gene into CCl(4)-injured rats. J Gene Med 2010; 12:244-54. [PMID: 20143305 DOI: 10.1002/jgm.1439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND beta(2)m(-)/Thy1(+) bone marrow-derived hepatocyte stem cells (BDHSCs) isolated from the bone marrow of cholestatic rats by magnetic bead cell sorting consistently express characteristics of both stem and liver cells. These stem cells may be good vehicles for gene transfer. Administration of exogenous hepatocyte growth factor (HGF) may be potentially useful for the treatment of liver fibrosis. Because lentiviral vectors integrate stably into the host-cell genome of nondividing and dividing cells, it may efficiently transfect beta(2)m(-)/Thy1(+) BDHSCs in vitro and secrete high-level HGF consistently. Transplantation of beta(2)m(-)/Thy1(+) BDHSCs transduced with lentiviral vectors containing the HGF gene may reduce liver fibrosis in rats. METHODS Lentiviral vectors expressing HGF were constructed and used to transduce beta(2)m(-)/Thy1(+) BDHSCs sorted from cholestatic rats in vitro. Transduction efficiency was evaluated and then these cells were transplanted into rats through the portal vein. Liver function as well as histological and immunohistochemical examinations were carried out to assess the therapeutic efficacy on liver fibrosis. RESULTS We demonstrated that high-level exogenous HGF was detected in supernatants after beta(2)m(-)/Thy1(+) BDHSCs were transfected with lentiviral vectors expressing HGF. Transplantation of transduced beta(2)m(-)/Thy1(+) BDHSCs significantly enhanced liver function and attenuated liver fibrosis in vivo. CONCLUSIONS The present study indicates that transplantation of beta(2)m(-)/Thy1(+) BDHSCs overexpressing the HGF gene may offer a novel approach for promoting liver function and reverse liver fibrosis.
Collapse
Affiliation(s)
- Jun-Jie Hu
- Department of Gastroenterology, Xinhua Hospital, College of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | |
Collapse
|
9
|
Muriel P. Role of free radicals in liver diseases. Hepatol Int 2009; 3:526-36. [PMID: 19941170 DOI: 10.1007/s12072-009-9158-6] [Citation(s) in RCA: 258] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Revised: 09/23/2009] [Accepted: 11/11/2009] [Indexed: 12/16/2022]
Abstract
Reactive oxygen and nitrogen species (ROS and RNS) are produced by metabolism of normal cells. However, in liver diseases, redox is increased thereby damaging the hepatic tissue; the capability of ethanol to increase both ROS/RNS and peroxidation of lipids, DNA, and proteins was demonstrated in a variety of systems, cells, and species, including humans. ROS/RNS can activate hepatic stellate cells, which are characterized by the enhanced production of extracellular matrix and accelerated proliferation. Cross-talk between parenchymal and nonparenchymal cells is one of the most important events in liver injury and fibrogenesis; ROS play an important role in fibrogenesis throughout increasing platelet-derived growth factor. Most hepatocellular carcinomas occur in cirrhotic livers, and the common mechanism for hepatocarcinogenesis is chronic inflammation associated with severe oxidative stress; other risk factors are dietary aflatoxin B(1) consumption, cigarette smoking, and heavy drinking. Ischemia-reperfusion injury affects directly on hepatocyte viability, particularly during transplantation and hepatic surgery; ischemia activates Kupffer cells which are the main source of ROS during the reperfusion period. The toxic action mechanism of paracetamol is focused on metabolic activation of the drug, depletion of glutathione, and covalent binding of the reactive metabolite N-acetyl-p-benzoquinone imine to cellular proteins as the main cause of hepatic cell death; intracellular steps critical for cell death include mitochondrial dysfunction and, importantly, the formation of ROS and peroxynitrite. Infection with hepatitis C is associated with increased levels of ROS/RNS and decreased antioxidant levels. As a consequence, antioxidants have been proposed as an adjunct therapy for various liver diseases.
Collapse
Affiliation(s)
- Pablo Muriel
- Department of Pharmacology, Cinvestav-I.P.N., Apdo. Postal 14-740, Mexico, 07000 D.F. Mexico
| |
Collapse
|
10
|
Eckersley-Maslin MA, Warner FJ, Grzelak CA, McCaughan GW, Shackel NA. Bone marrow stem cells and the liver: are they relevant? J Gastroenterol Hepatol 2009; 24:1608-16. [PMID: 19788602 DOI: 10.1111/j.1440-1746.2009.06004.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The contribution of bone marrow stem cell responses to liver homeostasis, injury and malignancy is discussed in this review. Pluripotent stem cells or their more committed progenitor progeny are essential to tissue development, regeneration and repair and are widely implicated in the pathogenesis of malignancy. Stem cell responses to injury are the focus of intense research efforts in the hope of future therapeutic manipulation. Stem cells occur within tissues, such as the liver, or arise from extrahepatic sites, in particular, the bone marrow. As the largest reservoir of stem cells in the adult, the bone marrow has been implicated in the stem cell response associated with liver injury. However, in liver injury, the relative contribution of bone marrow stem cells compared to intrahepatic progenitor responses is poorly characterized. Intrahepatic progenitor responses have been recently reviewed elsewhere. In this review, we have summarized liver-specific extrahepatic stem cell responses originating from the bone marrow. The physiological relevance of bone marrow stem cell responses to adult liver homeostasis, injury and malignancy is discussed with emphasis on mechanisms of bone marrow stem cell recruitment to sites of liver injury and its contribution to intrahepatic malignancy.
Collapse
|
11
|
Lan L, Chen YW, Sun C, Liu BW, Sun QL. Transplantation of interleukin 10-modified bone marrow-derived liver stem cells reduces accumulation of extracellular matrix in fibrotic liver in rats. Shijie Huaren Xiaohua Zazhi 2009; 17:2231-2236. [DOI: 10.11569/wcjd.v17.i22.2231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effects of transplanting interleukin 10 (IL-10)-modified bone marrow-derived liver stem cells (BDLSCs) on accumulation of extracellular matrix in fibrotic liver in rats.
METHODS: Rat beta-2 microglobulin (β2m)-/Thy-1+ BDLSCs were isolated by magnetic bead cell sorting (MACS), and transduced with adenovirus-mediated IL-10 gene. The level of IL-10 protein secretion by BDLSCs was assessed by ELISA. The rats were divided randomly into four groups: normal group, model group, BDLSCs group and BDLSCs plus IL-10 group. IL-10 gene-modified BDLSCs from male rats were transplanted into female liver fibrosis rats via a branch of the portal vein. Sry gene was amplified by PCR to evaluate the implantation of BDLSCs in liver. Collagen area in liver tissues was detected by Van Gieson's (VG) staining. Expression of α-smooth muscle actin (α-SMA) protein in liver tissue was determined by Western blot. Hydroxyproline (Hyp) in liver tissues was quantified by the alkaline hydrolysis method. Extracellular matrix (ECM) proteins in serum were quantified by ELISA.
RESULTS: BDLSCs were successfully isolated by MACS. IL-10 secreted by IL-10 gene-modified BDLSCs presented persistently at a high level. Transduced BDLSCs were implanted successfully into impaired liver. Transplantation of IL-10 gene-modified BDLSCs lessened deposition of collagen, decreased α-SMA expression and thereby suppressed activation of hepatic stellate cells. Compared with the BDLSCs group, the level of Hyp in liver tissue decreased markedly in the BDLSCs plus IL-10 group (255.0 ± 50.5 μg/g vs 373.0 ± 26.7 μg/g, P < 0.01), and the levels of ECM proteins in serum also decreased (40.5 ± 7.7 μg/L vs 79.4 ± 10.3 μg/L, 61.5 ± 16.4 μg/L vs 77.7 ± 12.6 μg/L, 14.3 ± 0.8 μg/L vs 14.9 ± 1.5 μg/L, P < 0.01 or 0.05).
CONCLUSION: Transplantation of IL-10 gene-modified BDLSCs can effectively decrease accumulation of ECM in fibrotic liver in rats, suggesting the potential utility of this novel combined strategy of cell transplantation with gene therapy for the treatment of liver fibrosis.
Collapse
|
12
|
Li JT, Liao ZX, Ping J, Xu D, Wang H. Molecular mechanism of hepatic stellate cell activation and antifibrotic therapeutic strategies. J Gastroenterol 2008; 43:419-28. [PMID: 18600385 DOI: 10.1007/s00535-008-2180-y] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2007] [Accepted: 02/25/2008] [Indexed: 02/06/2023]
Abstract
Activation of hepatic stellate cells (HSCs) is the dominant event in liver fibrosis. The early events in the organization of HSC activation have been termed initiation. Initiation encompasses rapid changes in gene expression and phenotype that render the cells responsive to cytokines and other local stimuli. Cellular responses following initiation are termed perpetuation, which encompasses those cellular events that amplify the activated phenotype through enhanced growth factor expression and responsiveness. Multiple cells and cytokines play a part in the regulation of HSC activation. HSC activation consists of discrete phenotype responses, mainly proliferation, contractility, fibrogenesis, matrix degradation, chemotaxis and retinoid loss. Currently, antifibrotic therapeutic strategies include inhibition of HSC proliferation or stimulation of HSC apoptosis, downregulation of collagen production or promotion of its degradation, administration of cytokines, and infusion of mesenchymal stem cells. In this review, we summarize the latest advances in our understanding of the mechanisms of HSC activation and possible antifibrotic therapeutic strategies.
Collapse
Affiliation(s)
- Jing-Ting Li
- Department of Pharmacology, Basic Medical School of Wuhan University, Luojia Hill, Wuhan 430071, China
| | | | | | | | | |
Collapse
|
13
|
An efficient two-step method to purify very small embryonic-like (VSEL) stem cells from umbilical cord blood (UCB). Folia Histochem Cytobiol 2008; 46:239-43. [PMID: 18519244 DOI: 10.2478/v10042-008-0036-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The identification in murine bone marrow (BM) of very small embryonic-like (VSEL) stem cells, possessing several features of pluripotent stem cells, encouraged us to investigate if similar population of cells could be also isolated from the human umbilical cord blood (UCB). Here our approach to purify VSEL from human UCB is described by employing a two step isolation strategy based on i) hypotonic lysis of erythrocytes followed ii) by multi-parameter FACS sorting. Accordingly, first, erythrocytes are removed from the UCB samples by hypotonic ammonium chloride solution and next, the UCB mononuclear cells (UCB MNC) are stained with monoclonal antibodies against all hematopoietic lineages including the common leukocyte antigen CD45. The cells carrying these markers (lin+CD45+) are eliminated from the sort by electronic gating. At the same time the antibodies against CXCR4, CD34 and CD133 are employed as positive markers to enrich the UCB MNC for VSEL. This combined two step approach enables to purify VSEL stem cells, which are small and express mRNA for pluripotent stem cells (PSC) (Oct-4 and Nanog) and tissue-committed stem cells (TCSC) (Nkx2.5/Csx, VE-cadherin and GFAP) similarly to those isolated from the adult BM (3-5 microm cells with large nuclei).
Collapse
|
14
|
Lan L, Chen Y, Sun C, Sun Q, Hu J, Li D. Transplantation of bone marrow-derived hepatocyte stem cells transduced with adenovirus-mediated IL-10 gene reverses liver fibrosis in rats. Transpl Int 2008; 21:581-92. [PMID: 18282246 DOI: 10.1111/j.1432-2277.2008.00652.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Bone marrow stem cells (BMSCs) transplantation alone may not be sufficient for treatment of liver fibrosis because of complicated histopathologic changes in the liver. Interleukin-10 (IL-10) is an anti-fibrosis cytokine. IL-10 gene transfer of beta2m(-)/Thy-1+ bone marrow-derived hepatocyte stem cells (BDHSCs) may be useful for treating liver fibrosis. To determine the effect of liver fibrosis in rats by transplanting BDHSCs transduced with adenovirus-mediated IL-10 gene (AdIL-10), rat BDHSCs were isolated by magnetic bead cell sorting, characterized for liver-associated phenotypes, transduced with AdIL-10, and transplanted into liver-fibrotic rats. We show that BDHSCs secreted high-level IL-10 and retained their albumin expression after AdIL-10 transfer in vitro. Intra-portal-infused BDHSCs were implanted into the liver 2 weeks after transplantation. Transplanting AdIL-10-transduced BDHSCs into liver-fibrotic rats downregulated inflammatory response, promoted liver regeneration, suppressed activation of hepatic stellate cells and improved liver histopathology and liver function. These findings demonstrated the potential utility of this novel combined strategy of IL-10 gene and BDHSCs for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Ling Lan
- Digestive Disease Laboratory and Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
15
|
Abstract
The hepatic stellate cell has surprised and engaged physiologists, pathologists, and hepatologists for over 130 years, yet clear evidence of its role in hepatic injury and fibrosis only emerged following the refinement of methods for its isolation and characterization. The paradigm in liver injury of activation of quiescent vitamin A-rich stellate cells into proliferative, contractile, and fibrogenic myofibroblasts has launched an era of astonishing progress in understanding the mechanistic basis of hepatic fibrosis progression and regression. But this simple paradigm has now yielded to a remarkably broad appreciation of the cell's functions not only in liver injury, but also in hepatic development, regeneration, xenobiotic responses, intermediary metabolism, and immunoregulation. Among the most exciting prospects is that stellate cells are essential for hepatic progenitor cell amplification and differentiation. Equally intriguing is the remarkable plasticity of stellate cells, not only in their variable intermediate filament phenotype, but also in their functions. Stellate cells can be viewed as the nexus in a complex sinusoidal milieu that requires tightly regulated autocrine and paracrine cross-talk, rapid responses to evolving extracellular matrix content, and exquisite responsiveness to the metabolic needs imposed by liver growth and repair. Moreover, roles vital to systemic homeostasis include their storage and mobilization of retinoids, their emerging capacity for antigen presentation and induction of tolerance, as well as their emerging relationship to bone marrow-derived cells. As interest in this cell type intensifies, more surprises and mysteries are sure to unfold that will ultimately benefit our understanding of liver physiology and the diagnosis and treatment of liver disease.
Collapse
Affiliation(s)
- Scott L Friedman
- Division of Liver Diseases, Mount Sinai School of Medicine, New York, New York 10029-6574, USA.
| |
Collapse
|
16
|
Higashiyama R, Inagaki Y, Hong YY, Kushida M, Nakao S, Niioka M, Watanabe T, Okano H, Matsuzaki Y, Shiota G, Okazaki I. Bone marrow-derived cells express matrix metalloproteinases and contribute to regression of liver fibrosis in mice. Hepatology 2007; 45:213-22. [PMID: 17187438 DOI: 10.1002/hep.21477] [Citation(s) in RCA: 205] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
UNLABELLED Liver fibrosis is usually progressive, but it can occasionally be reversible if the causative agents are adequately removed or if patients are treated effectively. However, molecular mechanisms responsible for this reversibility of liver fibrosis have been poorly understood. To reveal the contribution of bone marrow (BM)-derived cells to the spontaneous regression of liver fibrosis, mice were treated with repeated carbon tetrachloride injections after hematopoietic reconstitution with enhanced green fluorescent protein (EGFP)-expressing BM cells. The distribution and characteristics of EGFP-positive (EGFP(+)) cells present in fibrotic liver tissue were examined at different time points after cessation of carbon tetrachloride intoxication. A large number of EGFP(+) cells were observed in liver tissue at peak fibrosis, which decreased during the recovery from liver fibrosis. Some of them, as well as EGFP-negative (EGFP(-)) liver resident cells, expressed matrix metalloproteinase (MMP)-13 and MMP-9. Whereas MMP-13 was transiently expressed mainly in the cells clustering in the periportal areas, MMP-9 expression and enzymatic activity were detected over the resolution process in several different kinds of cells located in the portal areas and along the fibrous septa. Therapeutic recruitment of BM cells by granulocyte colony-stimulating factor (G-CSF) treatment significantly enhanced migration of BM-derived cells into fibrotic liver and accelerated the regression of liver fibrosis. Experiments using transgenic mice overexpressing hepatocyte growth factor (HGF) indicated that G-CSF and HGF synergistically increased MMP-9 expression along the fibrous septa. CONCLUSION Autologous BM cells contribute to the spontaneous regression of liver fibrosis, and their therapeutic derivation could be a new treatment strategy for intractable liver fibrosis.
Collapse
Affiliation(s)
- Reiichi Higashiyama
- Liver Fibrosis Research Unit, Department of Community Health, Tokai University School of Medicine, Isehara, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Kucia M, Reca R, Campbell FR, Zuba-Surma E, Majka M, Ratajczak J, Ratajczak MZ. A population of very small embryonic-like (VSEL) CXCR4(+)SSEA-1(+)Oct-4+ stem cells identified in adult bone marrow. Leukemia 2006; 20:857-69. [PMID: 16498386 DOI: 10.1038/sj.leu.2404171] [Citation(s) in RCA: 468] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
By employing multiparameter sorting, we identified in murine bone marrow (BM) a homogenous population of rare (approximately 0.02% of BMMNC) Sca-1(+)lin(-)CD45- cells that express by RQ-PCR and immunohistochemistry markers of pluripotent stem cells (PSC) such as SSEA-1, Oct-4, Nanog and Rex-1. The direct electronmicroscopical analysis revealed that these cells are small (approximately 2-4 microm), posses large nuclei surrounded by a narrow rim of cytoplasm, and contain open-type chromatin (euchromatin) that is typical for embryonic stem cells. In vitro cultures these cells are able to differentiate into all three germ-layer lineages. The number of these cells is highest in BM from young (approximately 1-month-old) mice and decreases with age. It is also significantly diminished in short living DBA/2J mice as compared to long living B6 animals. These cells in vitro respond strongly to SDF-1, HGF/SF and LIF and express CXCR4, c-met and LIF-R, respectively, and since they adhere to fibroblasts they may be coisolated with BM adherent cells. We hypothesize that this population of Sca-1(+)lin(-)CD45- very small embryonic-like (VSEL) stem cells is deposited early during development in BM and could be a source of pluripotent stem cells for tissue/organ regeneration.
Collapse
Affiliation(s)
- M Kucia
- Stem Cell Biology Program at James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Ueno T, Nakamura T, Torimura T, Sata M. Angiogenic cell therapy for hepatic fibrosis. Med Mol Morphol 2006; 39:16-21. [PMID: 16575510 DOI: 10.1007/s00795-006-0311-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2006] [Accepted: 01/19/2006] [Indexed: 01/01/2023]
Abstract
Progression of liver fibrosis has been linked with injuries associated with hypoxia and neovascularization. Neovascularization consists of angiogenesis and vasculogenesis, representing formation of blood vessels by differentiation of endothelial progenitor cells (EPCs). We investigated antifibrogenic and regenerative effects of EPC transplantation in chronic liver injury. Rat EPCs were isolated from bone marrow cells and examined in vitro for lineage markers. Recipient rats were injected intraperitoneally with dimethylnitrosamine (DMN) three times weekly for 4 weeks, plus EPC transplantation once weekly for 4 weeks. Transplanted rats showed suppression of liver fibrogenesis. Expression of growth factors promoting liver regeneration such as hepatocyte growth factor (HGF), transforming growth factor (TGF)-alpha, epidermal growth factor (EGF), and vascular endothelial growth factor (VEGF) was increased in transplanted rats, together with hepatocyte proliferation. Normal liver function parameters such as transaminase, total bilirubin, total protein, and albumin were maintained in transplanted rats. EPC transplantation is effective not only for preventing liver fibrosis but also for promoting regeneration in chronically damaged livers. Also, recently it has been reported that green fluorescent protein-positive bone marrow cells contribute to the liver tissue repair of fibrosis model rats. EPC transplantation might become an alternative if further preclinical investigation finds it to be effective in severely cirrhotic livers.
Collapse
Affiliation(s)
- Takato Ueno
- Research Center for Innovative Cancer Therapy and Center of the 21st Century COE Program for Medical Science, Kurume University School of Medicine, Japan.
| | | | | | | |
Collapse
|
19
|
Kucia M, Reca R, Jala VR, Dawn B, Ratajczak J, Ratajczak MZ. Bone marrow as a home of heterogenous populations of nonhematopoietic stem cells. Leukemia 2005; 19:1118-27. [PMID: 15902288 DOI: 10.1038/sj.leu.2403796] [Citation(s) in RCA: 131] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Evidence is presented that bone marrow (BM) in addition to CD45(positive) hematopoietic stem cells contains a rare population of heterogenous CD45(negative) nonhematopoietic tissue committed stem cells (TCSC). These nonhematopoietic TCSC (i) are enriched in population of CXCR4(+) CD34(+) AC133(+) lin(-) CD45(-) and CXCR4(+) Sca-1(+) lin(-) CD45(-) in humans and mice, respectively, (ii) display several markers of pluripotent stem cells (PSC) and (iii) as we envision are deposited in BM early in development. Thus, since BM contains versatile nonhematopoietic stem cells, previous studies on plasticity trans-dedifferentiation of BM-derived hematopoietic stem cells (HSC) that did not include proper controls to exclude this possibility could lead to wrong interpretations. Therefore, in this spotlight review we present this alternative explanation of 'plasticity' of BM-derived stem cells based on the assumption that BM stem cells are heterogenous. We also discuss a potential relationship of TCSC/PSC identified by us with other BM-derived CD45(negative) nonhematopoietic stem cells that were recently identified by other investigators (eg MSC, MAPC, USSC and MIAMI cells). Finally, we discuss perspectives and pitfalls in potential application of these cells in regenerative medicine.
Collapse
Affiliation(s)
- M Kucia
- Stem Cell Biology Program at James Graham Brown Cancer Center, University of Louisville, KY 40202, USA
| | | | | | | | | | | |
Collapse
|