1
|
Wang J, Ye W, Jiang M, Zhou Y, Zheng J. Therapeutic potential of exosome derived from hepatocyte growth factor-overexpressing adipose mesenchymal stem cells in TGFβ1-stimulated hepatic stellate cells. Cytotechnology 2024; 76:217-229. [PMID: 38495297 PMCID: PMC10940570 DOI: 10.1007/s10616-023-00611-0] [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: 07/18/2023] [Accepted: 12/14/2023] [Indexed: 03/19/2024] Open
Abstract
Cirrhosis is a familiar end-stage of multiple chronic liver diseases. The gene-modified mesenchymal stem cells (MSCs) have become one of the most promising schemes for the treatment of cirrhosis. MSCs exhibit their therapeutic role mainly by secreting hepatocyte growth factor (HGF). The aim of this research was to probe the anti-fibrosis role of exosomes secreted by HGF modified-mouse adipose MSCs (ADMSCs) on activated hepatic stellate cells (HSCs) and to preliminarily explore the possible mechanism. Firstly, mouse ADMSCs were isolated and identified. Quantitative real-time polymerase chain reaction verified the transfection efficiency of ADMSC transfected with HGF lentivirus. Exosomes derived from ADMSC transfecting negative control/HGF (ADMSCNC-Exo/ADMSCHGF-Exo) were extracted by density gradient centrifugation. HSCs were allocated to the control, TGF-β, TGF-β + ADMSC-Exo, TGF-β + ADMSCNC-Exo, and TGF-β + ADMSCHGF-Exo groups. Moreover, all mice were distributed to the control, CCl4 (40% CCl4 in olive oil), CCl4+ADMSC-Exo, CCl4+ADMSCNC-Exo, and CCl4+ADMSCHGF-Exo groups. Exosomes derived from ADMSCs with or without HGF transfection suppressed HSC activation, as evidenced by attenuating cell viability and cell cycle arrest at S phase but inducing apoptosis. Moreover, ADMSC-Exo, ADMSCNC-Exo, and ADMSCHGF-Exo effectively repressed the gene and protein levels of α-SMA, Col-I, Rho A, Cdc42, and Rac1 in TGF-β-treated HSCs, and ADMSCHGF-Exo had the best effect. ADMSCHGF-Exo had a stronger regulatory effect on serum liver index than ADMSCNC-Exo in CCl4-induced mice. In conclusion, ADMSCHGF-Exo alleviated liver fibrosis by weakening the Rho pathway, thus reducing collagen production.
Collapse
Affiliation(s)
- Jin Wang
- Department of Pancreatology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People′s Hospital, No. 100 Minjiang Avenue, Kecheng District, 324000 Quzhou, Zhejiang China
| | - Weikang Ye
- Department of Pancreatology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People′s Hospital, No. 100 Minjiang Avenue, Kecheng District, 324000 Quzhou, Zhejiang China
| | - Ming Jiang
- Department of Pancreatology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People′s Hospital, No. 100 Minjiang Avenue, Kecheng District, 324000 Quzhou, Zhejiang China
| | - Yinong Zhou
- Department of Pancreatology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People′s Hospital, No. 100 Minjiang Avenue, Kecheng District, 324000 Quzhou, Zhejiang China
| | - Jie Zheng
- Department of Pancreatology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People′s Hospital, No. 100 Minjiang Avenue, Kecheng District, 324000 Quzhou, Zhejiang China
| |
Collapse
|
2
|
Liu Q, Lv C, Jiang Y, Luo K, Gao Y, Liu J, Zhang X, Mohammad Omar J, Jin S. From hair to liver: emerging application of hair follicle mesenchymal stem cell transplantation reverses liver cirrhosis by blocking the TGF-β/Smad signaling pathway to inhibit pathological HSC activation. PeerJ 2022; 10:e12872. [PMID: 35186473 PMCID: PMC8855721 DOI: 10.7717/peerj.12872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/11/2022] [Indexed: 01/10/2023] Open
Abstract
Liver cirrhosis (LC) involves multiple systems throughout the body, and patients with LC often die of multiple organ failure. However, few drugs are useful to treat LC. Hair follicle mesenchymal stem cells (HF-MSCs) are derived from the dermal papilla and the bulge area of hair follicles and are pluripotent stem cells in the mesoderm with broad prospects in regenerative medicine. As an emerging seed cell type widely used in skin wound healing and plastic surgery, HF-MSCs show considerable prospects in the treatment of LC due to their proliferation and multidirectional differentiation capabilities. We established an LC model in C57BL/6J mice by administering carbon tetrachloride (CCl4) and injected HF-MSCs through the tail vein to explore the therapeutic effects and potential mechanisms of HF-MSCs on LC. Here, we found that HF-MSCs improved liver function and ameliorated the liver pathology of LC. Notably, PKH67-labeled HF-MSCs were detected in the injured liver and expressed the hepatocyte-specific markers cytokeratin 18 (CK18) and albumin (ALB). In addition, in contrast to that in the LC group, the α-SMA expression showed a decreasing trend in the treatment group in vitro and in vivo, indicating that the pathological activation of hepatic stellate cells (HSCs) was inhibited by HF-MSC treatment. Moreover, the levels of transforming growth factor β (TGF-β1) and p-Smad3, a signaling molecule downstream of TGF-β1, were increased in mice with LC, while HF-MSC treatment reversed these changes in vivo and in vitro. Based on these findings, HF-MSCs may reverse LC by blocking the TGF-β/Smad pathway and inhibiting the pathological activation of HSCs, which may provide evidence for the application of HF-MSCs to treat LC.
Collapse
Affiliation(s)
- Qi Liu
- Department of Gastroenterology and Hepatology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chengqian Lv
- Department of Gastroenterology and Hepatology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanan Jiang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy of Harbin Medical University, Harbin, China,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Kunpeng Luo
- Department of Gastroenterology and Hepatology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yang Gao
- Department of Gastroenterology and Hepatology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jingyang Liu
- Department of Gastroenterology and Hepatology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xu Zhang
- Department of Gastroenterology and Hepatology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jan Mohammad Omar
- Department of Gastroenterology and Hepatology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shizhu Jin
- Department of Gastroenterology and Hepatology, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
3
|
Ye S, Boeter JWB, Penning LC, Spee B, Schneeberger K. Hydrogels for Liver Tissue Engineering. Bioengineering (Basel) 2019; 6:E59. [PMID: 31284412 PMCID: PMC6784004 DOI: 10.3390/bioengineering6030059] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 06/28/2019] [Accepted: 07/03/2019] [Indexed: 12/14/2022] Open
Abstract
Bioengineered livers are promising in vitro models for drug testing, toxicological studies, and as disease models, and might in the future be an alternative for donor organs to treat end-stage liver diseases. Liver tissue engineering (LTE) aims to construct liver models that are physiologically relevant. To make bioengineered livers, the two most important ingredients are hepatic cells and supportive materials such as hydrogels. In the past decades, dozens of hydrogels have been developed to act as supportive materials, and some have been used for in vitro models and formed functional liver constructs. However, currently none of the used hydrogels are suitable for in vivo transplantation. Here, the histology of the human liver and its relationship with LTE is introduced. After that, significant characteristics of hydrogels are described focusing on LTE. Then, both natural and synthetic materials utilized in hydrogels for LTE are reviewed individually. Finally, a conclusion is drawn on a comparison of the different hydrogels and their characteristics and ideal hydrogels are proposed to promote LTE.
Collapse
Affiliation(s)
- Shicheng Ye
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Jochem W B Boeter
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Louis C Penning
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Bart Spee
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands
| | - Kerstin Schneeberger
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CT Utrecht, The Netherlands.
| |
Collapse
|
4
|
The Pharmabiotic Approach to Treat Hyperammonemia. Nutrients 2018; 10:nu10020140. [PMID: 29382084 PMCID: PMC5852716 DOI: 10.3390/nu10020140] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/22/2018] [Accepted: 01/26/2018] [Indexed: 12/12/2022] Open
Abstract
Ammonia is constantly produced as a metabolic waste from amino acid catabolism in mammals. Ammonia, the toxic waste metabolite, is resolved in the liver where the urea cycle converts free ammonia to urea. Liver malfunctions cause hyperammonemia that leads to central nervous system (CNS) dysfunctions, such as brain edema, convulsions, and coma. The current treatments for hyperammonemia, such as antibiotics or lactulose, are designed to decrease the intestinal production of ammonia and/or its absorption into the body and are not effective, besides being often accompanied by side effects. In recent years, increasing evidence has shown that modifications of the gut microbiota could be used to treat hyperammonemia. Considering the role of the gut microbiota and the physiological characteristics of the intestine, the removal of ammonia from the intestine by modulating the gut microbiota would be an ideal approach to treat hyperammonemia. In this review, we discuss the significance of hyperammonemia and its related diseases and the efficacy of the current management methods for hyperammonemia to understand the mechanism of ammonia transport in the human body. The possibility to use the gut microbiota as pharmabiotics to treat hyperammonemia and its related diseases is also explored.
Collapse
|
5
|
Ramanathan R, Rupert S, Selvaraj S, Satyanesan J, Vennila R, Rajagopal S. Role of Human Wharton's Jelly Derived Mesenchymal Stem Cells (WJ-MSCs) for Rescue of d-Galactosamine Induced Acute Liver Injury in Mice. J Clin Exp Hepatol 2017; 7:205-214. [PMID: 28970707 PMCID: PMC5620364 DOI: 10.1016/j.jceh.2017.03.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 03/14/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND/AIM Mesenchymal stem cells (MSCs) are multipotent precursor cells having self-renewal ability making them a candidate for use in regenerative medicine. Acute liver injury results in sudden loss of hepatic function leading to organ failure. Liver transplantation is often required to salvage patients with acute liver failure. Due to shortage of organs, identification of alternate method is the need of the hour. In view of this, an attempt has been made to check the regenerative ability of WJ-MSCs (wharton's jelly derived MSC) in mice models for acute liver injury. METHODS Swiss albino mice weighing 25 ± 5 g were used in this study. The control mice (Group I), was given saline. Group II mice received d-Galactosamine (d-GalN-800 mg/kg; i.p). Group III mice similar with Group II, received WJ-MSCs (5 × 105 cells/0.5 ml DMEM) through tail vein, 24 h after d-GalN administration and Group IV mice received MSC alone. RESULTS Parameters, indicative of hepatotoxicity and oxidative stress were analyzed. A two-fold elevation in the marker enzymes of liver toxicity such as aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphatase (SAP), and total serum bilirubin (TBIL) confirms hepatocellular injury, while a greater than four-fold increase in malondialdehyde (MDA) formation, along with around 40% fall in superoxide-dis-mutase (SOD) activity was indicative of oxidative stress and loss of hepatocellular membrane integrity induced by d-GalN. The above biochemical and pathological changes were significantly restored in mice that received WJ-MSCs indicating hepatoprotective and probable regenerative property. CONCLUSION The present study showed that WJ-MSC treatment is able to rescue/ameliorate the hepatotoxicity induced by d-GalN in mice.
Collapse
Affiliation(s)
- Raghu Ramanathan
- Centre for Advanced Research, Stem Cell Research Centre, Govt. Stanley Medical College and Hospital, Chennai, Tamilnadu, India
| | - Secunda Rupert
- Centre for Advanced Research, Stem Cell Research Centre, Govt. Stanley Medical College and Hospital, Chennai, Tamilnadu, India
| | - Sakthivel Selvaraj
- Centre for Advanced Research, Stem Cell Research Centre, Govt. Stanley Medical College and Hospital, Chennai, Tamilnadu, India
| | - Jeswanth Satyanesan
- Centre for Advanced Research, Stem Cell Research Centre, Govt. Stanley Medical College and Hospital, Chennai, Tamilnadu, India
| | - Rosy Vennila
- Centre for Advanced Research, Stem Cell Research Centre, Govt. Stanley Medical College and Hospital, Chennai, Tamilnadu, India
| | - Surendran Rajagopal
- Director, Hepato-Pancreato-Biliary Centre for Surgery & Transplantation, MIOT International, Chennai, Tamilnadu, India
| |
Collapse
|
6
|
Tao Y, Wang M, Chen E, Tang H. Liver Regeneration: Analysis of the Main Relevant Signaling Molecules. Mediators Inflamm 2017; 2017:4256352. [PMID: 28947857 PMCID: PMC5602614 DOI: 10.1155/2017/4256352] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/19/2017] [Accepted: 08/10/2017] [Indexed: 02/06/2023] Open
Abstract
Liver regeneration is a highly organized tissue regrowth process and is the most important reaction of the liver to injury. The overall process of liver regeneration includes three phases: priming stage, proliferative phase, and termination phase. The initial step aims to induce hepatocytes to be sensitive to growth factors with the aid of some cytokines, including TNF-α and IL-6. The proliferation phase promotes hepatocytes to re-enter G1 with the stimulation of growth factors. While during the termination stage, hepatocytes will discontinue to proliferate to maintain normal liver mass and function. Except for cytokine- and growth factor-mediated pathways involved in regulating liver regeneration, new substances and technologies emerge to influence the regenerative process. Here, we reviewed novel and important signaling molecules involved in the process of liver regeneration to provide a cue for further research.
Collapse
Affiliation(s)
- Yachao Tao
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Menglan Wang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Enqiang Chen
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Gajalakshmi P, Majumder S, Viebahn CS, Swaminathan A, Yeoh GC, Chatterjee S. Interleukin-6 secreted by bipotential murine oval liver stem cells induces apoptosis of activated hepatic stellate cells by activating NF-κB-inducible nitric oxide synthase signaling. Biochem Cell Biol 2016; 95:263-272. [PMID: 28177770 DOI: 10.1139/bcb-2016-0011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Liver fibrosis is now well recognized as the causative factor for increased mortality from complications associated with liver pathologies. Activated hepatic stellate cells (HSCs) play a critical role in the progression of liver fibrosis. Therefore, targeting these activated HSCs to prevent and (or) treat liver disease is a worthwhile approach to explore. In the present in vitro study, we investigated the use of bipotential murine oval liver cells (BMOL) in regulating the functions of activated HSCs to prevent progression of liver fibrosis. We used a conditioned medium-based approach to study the effect of BMOL cells on activated HSC survival and function. Our data showed that BMOL cells block the contraction of activated HSCs by inducing apoptosis of these cells. We demonstrated that BMOL cells secrete soluble factors, such as interleukin-6 (IL-6), which induced apoptosis of activated HSCs. Using both pharmacological and molecular inhibitor approaches, we further identified that IL-6-mediated activation of NF-κB-iNOS-NO-ROS signaling in activated HSCs plays a critical role in BMOL-cell-mediated apoptosis of activated HSCs. Thus, the present study provides an alternative cell-based therapeutic approach to treat liver fibrosis.
Collapse
Affiliation(s)
| | - Syamantak Majumder
- b Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Cornelia S Viebahn
- c Centre for Medical Research, Harry Perkins Institute of Medical Research, University of Western Australia, Nedlands, Australia
| | - Akila Swaminathan
- a Life Sciences Division, AU-KBC Research Centre, Anna University, Chennai, India
| | - George C Yeoh
- c Centre for Medical Research, Harry Perkins Institute of Medical Research, University of Western Australia, Nedlands, Australia
| | - Suvro Chatterjee
- a Life Sciences Division, AU-KBC Research Centre, Anna University, Chennai, India.,d Department of Biotechnology, Anna University, Chennai, India
| |
Collapse
|
8
|
Kim HJ, Cho YA, Lee YM, Lee SY, Bae WJ, Kim EC. PIN1 Suppresses the Hepatic Differentiation of Pulp Stem Cells via Wnt3a. J Dent Res 2016; 95:1415-1424. [PMID: 27439725 DOI: 10.1177/0022034516659642] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
This study aimed to investigate the role of PIN1 on the hepatic differentiation of human dental pulp stem cells (hDPSCs) and its signaling pathway, as well as the potential therapeutic effects of hDPSC transplantation and PIN1 inhibition on CCl4 (carbon tetrachloride)-induced liver fibrosis in mice. The in vitro results showed that hepatic differentiation was suppressed by infection with adenovirus-PIN1 and promoted by PIN1 inhibitor juglone via the downregulation of Wnt3a and β-catenin. Compared with treatment with either hDPSC transplantation or juglone alone, the combination of hDPSCs and juglone into CCl4-injured mice significantly suppressed liver fibrosis and restored serum levels of alanine transaminase, aspartate transaminase, and ammonia. Collectively, the present study shows for the first time that PIN1 inhibition promotes hepatic differentiation of hDPSCs through the Wnt/β-catenin pathway. Furthermore, juglone in combination with hDPSC transplantation effectively treats liver fibrosis, suggesting that hDPSC transplantation with PIN1 inhibition may be a novel therapeutic candidate for the treatment of liver injury.
Collapse
Affiliation(s)
- H J Kim
- 1 Department of Oral Physiology, BK21 PLUS Project, and Institute of Translational Dental Sciences, School of Dentistry, Pusan National University, Yangsan, Republic of Korea
| | - Y A Cho
- 2 Department of Oral and Maxillofacial Pathology, Research Center for Tooth and Periodontal Regeneration, and School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Y M Lee
- 2 Department of Oral and Maxillofacial Pathology, Research Center for Tooth and Periodontal Regeneration, and School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - S Y Lee
- 2 Department of Oral and Maxillofacial Pathology, Research Center for Tooth and Periodontal Regeneration, and School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - W J Bae
- 2 Department of Oral and Maxillofacial Pathology, Research Center for Tooth and Periodontal Regeneration, and School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - E C Kim
- 2 Department of Oral and Maxillofacial Pathology, Research Center for Tooth and Periodontal Regeneration, and School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
9
|
Lee PH, Tu CT, Hsiao CC, Tsai MS, Ho CM, Cheng NC, Hung TM, Shih DTB. Antifibrotic Activity of Human Placental Amnion Membrane-Derived CD34+ Mesenchymal Stem/Progenitor Cell Transplantation in Mice With Thioacetamide-Induced Liver Injury. Stem Cells Transl Med 2016; 5:1473-1484. [PMID: 27405780 DOI: 10.5966/sctm.2015-0343] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 04/18/2016] [Indexed: 12/31/2022] Open
Abstract
: Liver fibrosis represents the end stage of chronic liver inflammatory diseases and is defined by the abnormal accumulation of extracellular matrix in the liver. Advanced liver fibrosis results in cirrhosis, liver failure, and portal hypertension. Liver transplantation has been the most effective treatment for these diseases, but the procedure is limited by the shortage of suitable donors. Mesenchymal stromal cells (MSCs) have shown great potential in the treatment of chronic inflammatory diseases associated with fibrosis. This study aimed to evaluate the therapeutic effect of MSC-based cell transplantation as an alternative treatment for liver fibrosis. A CD34-positive subpopulation of human placental amnion membrane-derived stem/progenitor cells (CD34+ AMSPCs) was isolated through the depletion of CD34-negative stromal fibroblasts (CD34- AMSFCs) facilitated by CD34 fluorescence-activated cell sorting, enriched and expanded ex vivo. These cells express pluripotency markers and demonstrate multidirectional differentiation potentials. Comparative analysis was made between CD34+ AMSPCs and CD34- AMSFCs in terms of the expressions of stemness surface markers, embryonic surface antigens, and multilineage differentiation potentials. A mouse model of liver fibrosis was established by thioacetamide (TAA) administration. When injected into the spleen of TAA-injured mice, human placental amnion membrane-derived MSCs (hAM-MSCs) can engraft into the injury site, ameliorate liver fibrosis, and restore liver function, as shown by pathological and blood biochemical analysis and downregulated gene expressions associated with liver damage. CD34+ AMSPCs represent a more primitive subset of hAM-MSCs and could be a suitable candidate with a potentially better safety profile for cell-based therapy in treatment of liver diseases associated with fibrosis. SIGNIFICANCE In this study, a CD34+ subpopulation of stem/progenitor cells derived from neonatal placental amnion membrane, denoted as CD34+ AMSPCs, were identified, enriched, and characterized. These cells are highly proliferative, express mesenchymal stromal cells and pluripotent stem cell markers, and demonstrate multidirectional differentiation potentials, indicating their promising application in clinical regenerative therapies. CD34+ AMSPC transplantation ameliorated liver fibrosis in mice with drug-induced liver injury. These cells represent a potential therapeutic agent for treating liver diseases associated with fibrosis.
Collapse
Affiliation(s)
- Po-Huang Lee
- National Taiwan University Hospital, Taipei City, Taiwan, Republic of China
- E-Da Hospital/I-Shou University, Yan-Chau Shiang, Kaohsiung County, Taiwan, Republic of China
| | - Chi-Tang Tu
- National Taiwan University Hospital, Taipei City, Taiwan, Republic of China
| | - Chih-Chiang Hsiao
- Taipei Medical University Hospital, Taipei City, Taiwan, Republic of China
| | - Ming-Song Tsai
- Prenatal Diagnosis Center, Cathay General Hospital, Taipei City, Taiwan, Republic of China
| | - Cheng-Maw Ho
- National Taiwan University Hospital, Taipei City, Taiwan, Republic of China
| | - Nai-Chen Cheng
- National Taiwan University Hospital, Taipei City, Taiwan, Republic of China
| | - Tzu-Min Hung
- National Taiwan University Hospital, Taipei City, Taiwan, Republic of China
- E-Da Hospital/I-Shou University, Yan-Chau Shiang, Kaohsiung County, Taiwan, Republic of China
| | - Daniel Tzu-Bi Shih
- E-Da Hospital/I-Shou University, Yan-Chau Shiang, Kaohsiung County, Taiwan, Republic of China
- Taipei Medical University Hospital, Taipei City, Taiwan, Republic of China
- Innovation Incubation Center, National Taiwan University Hospital, Taipei City, Taiwan, Republic of China
| |
Collapse
|
10
|
Font-Burgada J, Shalapour S, Ramaswamy S, Hsueh B, Rossell D, Umemura A, Taniguchi K, Nakagawa H, Valasek MA, Ye L, Kopp JL, Sander M, Carter H, Deisseroth K, Verma IM, Karin M. Hybrid Periportal Hepatocytes Regenerate the Injured Liver without Giving Rise to Cancer. Cell 2016; 162:766-79. [PMID: 26276631 DOI: 10.1016/j.cell.2015.07.026] [Citation(s) in RCA: 375] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 03/25/2015] [Accepted: 06/26/2015] [Indexed: 12/24/2022]
Abstract
Compensatory proliferation triggered by hepatocyte loss is required for liver regeneration and maintenance but also promotes development of hepatocellular carcinoma (HCC). Despite extensive investigation, the cells responsible for hepatocyte restoration or HCC development remain poorly characterized. We used genetic lineage tracing to identify cells responsible for hepatocyte replenishment following chronic liver injury and queried their roles in three distinct HCC models. We found that a pre-existing population of periportal hepatocytes, located in the portal triads of healthy livers and expressing low amounts of Sox9 and other bile-duct-enriched genes, undergo extensive proliferation and replenish liver mass after chronic hepatocyte-depleting injuries. Despite their high regenerative potential, these so-called hybrid hepatocytes do not give rise to HCC in chronically injured livers and thus represent a unique way to restore tissue function and avoid tumorigenesis. This specialized set of pre-existing differentiated cells may be highly suitable for cell-based therapy of chronic hepatocyte-depleting disorders.
Collapse
Affiliation(s)
- Joan Font-Burgada
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| | - Shabnam Shalapour
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Suvasini Ramaswamy
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Brian Hsueh
- Departments of Bioengineering, Psychiatry, and Behavioral Sciences, Neurosciences Program, Howard Hughes Medical Institute, Stanford University, 318 Campus Drive West, Clark Center W080, Stanford, CA 94305, USA
| | - David Rossell
- Department of Statistics, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
| | - Atsushi Umemura
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Koji Taniguchi
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Hayato Nakagawa
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Gastroenterology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655
| | - Mark A Valasek
- Department of Pathology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Li Ye
- Departments of Bioengineering, Psychiatry, and Behavioral Sciences, Neurosciences Program, Howard Hughes Medical Institute, Stanford University, 318 Campus Drive West, Clark Center W080, Stanford, CA 94305, USA
| | - Janel L Kopp
- Department of Pediatrics and Department of Cellular & Molecular Medicine, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Cellular & Physiological Sciences, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Maike Sander
- Department of Pediatrics and Department of Cellular & Molecular Medicine, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Hannah Carter
- Department of Medicine, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Karl Deisseroth
- Departments of Bioengineering, Psychiatry, and Behavioral Sciences, Neurosciences Program, Howard Hughes Medical Institute, Stanford University, 318 Campus Drive West, Clark Center W080, Stanford, CA 94305, USA
| | - Inder M Verma
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Pathology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
11
|
Deng Q, Cai T, Zhang S, Hu A, Zhang X, Wang Y, Huang J. Autologous Peripheral Blood Stem Cell Transplantation Improves Portal Hemodynamics in Patients with Hepatitis B Virus-related Decompensated Cirrhosis. HEPATITIS MONTHLY 2015; 15:e32498. [PMID: 26977164 PMCID: PMC4774339 DOI: 10.5812/hepatmon.32498] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 11/08/2015] [Accepted: 11/08/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Chronic hepatitis B virus (HBV) infection may eventually lead to decompensated liver cirrhosis, which is a terminal illness. OBJECTIVES The aim of this study was to investigate the therapeutic efficacy of autologous peripheral blood stem cell (APBSC) transplantation to improve portal vein hemodynamics in patients with HBV-related decompensated cirrhosis. PATIENTS AND METHODS This prospective study included 68 hospitalized patients who were diagnosed with HBV-related decompensated cirrhosis. These patients were divided into two groups: the transplantation group included 33 patients, while the control group included 35. Both groups received conventional medical treatment simultaneously, and APBSC transplantation was performed on the patients in the transplantation group. We evaluated the effects of APBSC transplantation on postoperative liver function using the following indices: total bilirubin, serum prothrombin and albumin, spleen size, and portal vein hemodynamics. Postoperatively, all of the patients were followed up at 24, 36, and 48 weeks. RESULTS The transplantation group had no serious reactions. Compared with the control group, albumin and prothrombin activity in the transplantation group was significantly improved at 24, 36, and 48 weeks after the procedure, and spleen length and portal vein diameter were substantially reduced at 48 weeks. The velocity of peak portal vein blood flow and mean maximum portal vein blood flow were greatly increased in the APBSC transplantation group at 36 and 48 weeks, respectively; however, there was also decreased portal vein diameter, which reduced portal vein pressure in patients with HBV-related decompensated cirrhosis. CONCLUSIONS APBSC transplantation greatly benefits HBV-linked decompensated cirrhosis patients and should be recommended in clinical practice.
Collapse
Affiliation(s)
- Qinzhi Deng
- Department of Liver Disease, Ningbo No. 2 Hospital, Ningbo, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ting Cai
- Department of Liver Disease, Ningbo No. 2 Hospital, Ningbo, China
| | - Shun Zhang
- Department of Liver Disease, Ningbo No. 2 Hospital, Ningbo, China
| | - Airong Hu
- Department of Liver Disease, Ningbo No. 2 Hospital, Ningbo, China
| | - Xingfen Zhang
- Department of Liver Disease, Ningbo No. 2 Hospital, Ningbo, China
| | - Yinyin Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianrong Huang
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Corresponding Author: Jianrong Huang, State Key Laboratory for Diagnosis and Treatment of Infectious Disease, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China. Tel: +86-57187236743, Fax: +86-57187236743, E-mail:
| |
Collapse
|
12
|
Zhu XQ, Pan XH, Yao L, Li W, Cui J, Wang G, Mrsny RJ, Hoffman AR, Hu JF. Converting Skin Fibroblasts into Hepatic-like Cells by Transient Programming. J Cell Biochem 2015; 117:589-98. [PMID: 26312781 DOI: 10.1002/jcb.25355] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Accepted: 08/25/2015] [Indexed: 12/30/2022]
Affiliation(s)
- Xiang-Qing Zhu
- Research Center of Stem Cell, Tissue and Organ Engineering; Kunming Army General Hospital; Kunming Yunnan P. R. China
| | - Xing-Hua Pan
- Research Center of Stem Cell, Tissue and Organ Engineering; Kunming Army General Hospital; Kunming Yunnan P. R. China
| | - Ling Yao
- Stanford University Medical School; Palo Alto California
| | - Wei Li
- Stem Cell and Cancer Center; The First Affiliated Hospital; Jilin University; Changchun P. R. China
| | - Jiuwei Cui
- Stem Cell and Cancer Center; The First Affiliated Hospital; Jilin University; Changchun P. R. China
| | - Guanjun Wang
- Stem Cell and Cancer Center; The First Affiliated Hospital; Jilin University; Changchun P. R. China
| | - Randall J. Mrsny
- GMR Epigenetics; Palo Alto California
- Department of Pharmacy & Pharmacology; University of Bath; Bath England
| | | | - Ji-Fan Hu
- Stanford University Medical School; Palo Alto California
- Stem Cell and Cancer Center; The First Affiliated Hospital; Jilin University; Changchun P. R. China
| |
Collapse
|
13
|
Irfan A, Ahmed I. Could Stem Cell Therapy be the Cure in Liver Cirrhosis? J Clin Exp Hepatol 2015; 5:142-6. [PMID: 26155042 PMCID: PMC4491605 DOI: 10.1016/j.jceh.2014.03.042] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 03/03/2014] [Indexed: 12/12/2022] Open
Abstract
Over the past five decades, liver cirrhosis has become an increasingly prevalent disease and one that will often require considerable medical intervention. However, current treatment options have demonstrated severe problems that have prompted research to provide a suitable alternative. These treatments are scarcely available, very expensive and present at a huge cost to the patient's quality of life. The introduction of stem cell therapy into liver disease has been heralded as the future of personalized medicine and may be the alternative that the healthcare system desperately seeks. To truly determine the scientific basis surrounding this excitement, a literature search was carried out in January 2013 to determine all the data that was present in this topic area. All articles also underwent full cross-referencing to ensure no data was missed. 11 clinical trials were found to meet this criteria and trials were included in both English and non-English languages. The sporadic nature of the data across the trials, with various methods and stem cell types, made comparisons difficult. The basic trends from the data were positive and the majority deemed the use of stem cells safe and feasible in patients presenting with cirrhotic liver disease. However, there is a clear requirement for more research, not only to determine the most efficacious technique and stem cell type but also to further understand stem cells to enhance progress. There may also be a requirement for a framework that future stem cell trials can be based on, which would allow future data to be comparative and allow valid conclusions to be drawn which may propel this therapy into standard clinical practice.
Collapse
Affiliation(s)
- Ahmer Irfan
- University of Edinburgh, Edinburgh, United Kingdom,Address for correspondence: Ahmer Irfan, University of Edinburgh Medical School, 47 Little France Crescent, Edinburgh, EH16 4TJ, United Kingdom.
| | - Irfan Ahmed
- University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|
14
|
Alejandra MR, Juan AB, Ana SR. Cell therapy for liver diseases: current medicine and future promises. Expert Rev Gastroenterol Hepatol 2015; 9:837-50. [PMID: 25747732 DOI: 10.1586/17474124.2015.1016913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Liver diseases are a major health problem worldwide since they usually represent the main causes of death in most countries, causing excessive costs to public health systems. Nowadays, there are no efficient current therapies for most hepatic diseases and liver transplant is infrequent due to the availability of organs, cost and risk of transplant rejection. Therefore, alternative therapies for liver diseases have been developed, including cell-based therapies. Stem cells (SCs) are characterized by their self-renewing capacity, unlimited proliferation and differentiation under certain conditions into tissue- or organ-specific cells with special functions. Cell-based therapies for liver diseases have been successful in experimental models, showing anti-inflammatory, antifibrogenic and regenerative effects. Nowadays, clinical trials using SCs for liver pathologies are increasing in number, and those that have reached publication have achieved favorable effects, encouraging us to think that SCs will have a potential clinical use in a short time.
Collapse
Affiliation(s)
- Meza-Ríos Alejandra
- Department of Molecular Biology and Genomics, Health Sciences University Center, Institute for Molecular Biology and Gene Therapy, University of Guadalajara, Sierra Mojada 950, Colonia Independencia, Guadalajara, Jalisco 44340, México
| | | | | |
Collapse
|
15
|
Cho YA, Noh K, Jue SS, Lee SY, Kim EC. Melatonin promotes hepatic differentiation of human dental pulp stem cells: clinical implications for the prevention of liver fibrosis. J Pineal Res 2015; 58:127-35. [PMID: 25431168 DOI: 10.1111/jpi.12198] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 11/24/2014] [Indexed: 12/21/2022]
Abstract
Melatonin's effect on hepatic differentiation of stem cells remains unclear. The aim of this study was to investigate the action of melatonin on hepatic differentiation as well as its related signaling pathways of human dental pulp stem cells (hDPSCs) and to examine the therapeutic effects of a combination of melatonin and hDPSC transplantation on carbon tetrachloride (CCl4 )-induced liver fibrosis in mice. In vitro hepatic differentiation was assessed by periodic acid-Schiff (PAS) staining and mRNA expression for hepatocyte markers. Liver fibrosis model was established by injecting 0.5 mL/kg CCl4 followed by treatment with melatonin (5 mg/kg, twice a week) and hDPSCs. In vivo therapeutic effects were evaluated by histopathology and by means of liver function tests including measurement of alanine transaminase (ALT), aspartate transaminase (AST), and ammonia levels. Melatonin promoted hepatic differentiation based on mRNA expression of differentiation markers and PAS-stained glycogen-laden cells. In addition, melatonin increased bone morphogenic protein (BMP)-2 expression and Smad1/5/8 phosphorylation, which was blocked by the BMP antagonist noggin. Furthermore, melatonin activated p38, extracellular signal-regulated kinase (ERK), and nuclear factor-κB (NF-κB) in hDPSCs. Melatonin-induced hepatic differentiation was attenuated by inhibitors of BMP, p38, ERK, and NF-κB. Compared to treatment of CCl4 -injured mice with either melatonin or hDPSC transplantation alone, the combination of melatonin and hDPSC significantly suppressed liver fibrosis and restored ALT, AST, and ammonia levels. For the first time, this study demonstrates that melatonin promotes hepatic differentiation of hDPSCs by modulating the BMP, p38, ERK, and NF-κB pathway. Combined treatment of grafted hDPSCs and melatonin could be a viable approach for the treatment of liver cirrhosis.
Collapse
Affiliation(s)
- Young-Ah Cho
- Department of Oral and Maxillofacial Pathology, School of Dentistry and Research Center for Tooth & Periodontal Regeneration (MRC), Kyung Hee University, Seoul, Korea
| | | | | | | | | |
Collapse
|
16
|
Ishkitiev N, Yaegaki K, Imai T, Tanaka T, Fushimi N, Mitev V, Okada M, Tominaga N, Ono S, Ishikawa H. Novel management of acute or secondary biliary liver conditions using hepatically differentiated human dental pulp cells. Tissue Eng Part A 2014; 21:586-93. [PMID: 25234861 DOI: 10.1089/ten.tea.2014.0162] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The current definitive treatment for acute or chronic liver condition, that is, cirrhosis, is liver transplantation from a limited number of donors, which might cause complications after donation. Hence, bone marrow stem cell transplantation has been developed, but the risk of carcinogenesis remains. We have recently developed a protocol for hepatic differentiation of CD117(+) stem cells from human exfoliated deciduous teeth (SHED). In the present study, we examine whether SHED hepatically differentiated (hd) in vitro could be used to treat acute liver injury (ALI) and secondary biliary cirrhosis. The CD117(+) cell fraction was magnetically separated from SHED and then differentiated into hepatocyte-like cells in vitro. The cells were transplanted into rats with either ALI or induced secondary biliary cirrhosis. Engraftment of human liver cells was determined immunohistochemically and by in situ hybridization. Recovery of liver function was examined by means of histochemical and serological tests. Livers of transplanted animals were strongly positive for human immunohistochemical factors, and in situ hybridization confirmed engraftment of human hepatocytes. The tests for recovery of liver function confirmed the presence of human hepatic markers in the animals' blood serum and lack of fibrosis and functional integration of transplanted human cells into livers. No evidence of malignancy was found. We show that in vitro hdSHED engraft morphologically and functionally into the livers of rats having acute injury or secondary biliary cirrhosis. SHED are readily accessible adult stem cells, capable of proliferating in large numbers before differentiating in vitro. This makes SHED an appropriate and safe stem cell source for regenerative medicine.
Collapse
Affiliation(s)
- Nikolay Ishkitiev
- 1 Department of Oral Health, School of Life Dentistry at Tokyo, Nippon Dental University , Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Friedman SL, Quigley EMM, Sharkey KA, Sung JJY, Whitcomb DC. The past 10 years of gastroenterology and hepatology-reflections and predictions. Nat Rev Gastroenterol Hepatol 2014; 11:692-700. [PMID: 25291429 DOI: 10.1038/nrgastro.2014.167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In November 2004, the very first issue of this journal featured articles on the pathogenesis of ulcerative colitis, mechanisms leading to chronic pancreatitis, and treatment of recurrent Clostridium-difficile-associated diarrhoea. Although those topics might seem familiar, much has changed in the intervening years in our understanding, diagnosis and treatment of many different diseases across the field of gastroenterology and hepatology. Nonetheless, many challenges remain. Here, we have asked five of our Advisory Board members-international experts across different subspecialties in gastroenterology and hepatology-to reflect on the progress and frustrations of the past 10 years. They also comment on where effort and money should be invested now, as well as their predictions for progress in the next 10 years.
Collapse
Affiliation(s)
- Scott L Friedman
- Division of Liver Disease, Box 1123, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, Room 11-70C, New York, NY 10029-6574, USA
| | - Eamonn M M Quigley
- Division of Gastroenterology and Hepatology, Houston Methodist Hospital, 6550 Fannin Street, SM 1001, Houston, TX 77030, USA
| | - Keith A Sharkey
- Hotchkiss Brain Institute and Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
| | - Joseph J Y Sung
- University Administration Building, The Chinese University of Hong Kong, 1/F, Room 101, Shatin, NT, Hong Kong SAR, The People's Republic of China
| | - David C Whitcomb
- Department of Medicine, Cell Biology &Molecular Physiology and Human Genetics, University of Pittsburgh and UPMC, Room 401.4, 3708 Fifth Avenue, Pittsburgh, PA 15231, USA
| |
Collapse
|
18
|
Zhao S, Wang Y, Gao C, Zhang J, Bao H, Wang Z, Gong P. Superparamagnetic iron oxide magnetic nanomaterial-labeled bone marrow mesenchymal stem cells for rat liver repair after hepatectomy. J Surg Res 2014; 191:290-301. [DOI: 10.1016/j.jss.2014.03.064] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 03/18/2014] [Accepted: 03/21/2014] [Indexed: 12/14/2022]
|
19
|
Sun L, Fan X, Zhang L, Shi G, Aili M, Lu X, Jiang T, Zhang Y. Bone mesenchymal stem cell transplantation via four routes for the treatment of acute liver failure in rats. Int J Mol Med 2014; 34:987-96. [PMID: 25110277 PMCID: PMC4152144 DOI: 10.3892/ijmm.2014.1890] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 07/30/2014] [Indexed: 12/16/2022] Open
Abstract
In the present study, we assessed the efficiency of four BMSC transplantation methods as a therapy for liver failure. A rat model (80 Sprague-Dawley rats) of D-galactosamine (D-gal)/lipopolysaccharide (LPS)-induced acute liver failure (ALF) was established and the rats were divided into 5 groups: a hepatic artery injection group, a portal vein injection group, a vena caudalis injection group, an intraperitoneal injection group and a control group (16 per group). Following transplantation, the liver tissue and blood samples were collected on days 1, 3 and 7, we detected the EdU (5-ethynyl-2′-deoxyuridine)-labeled cells homing to the liver tissue and assessed the proliferating cell nuclear antigen (PCNA) and cysteine-containing aspartate-specific protease (caspase)-3 expression in the liver tissue and detected the levels of stromal cell-derived factor 1 (SDF-1) and hepatocyte growth factor (HGF) in the liver tissues. Compared with the control group, the levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) and damage to the liver tissue in the hepatic artery group, the portal vein group and the vena caudalis group improved in vivo. The expression of PCNA and HGF in the liver was higher and caspase-3 expression was lower in the hepatic artery injection group, the portal vein injection group and the vena caudalis injection group than that in the intraperitoneal injection and control groups. The EdU-labeled BMSCs were only observed homing to the liver tissue in these three groups. However, no significant differences were observed between these three groups. Liver function in the rats with ALF was improved following BMSC transplantation via 3 endovascular implantation methods (through the hepatic artery, portal vein and vena caudalis). These 3 methods were effective in transplanting BMSCs for the treatment of ALF. However, the selection of blood vessel in the implantation pathway does not affect the transplantation outcome. Transplantation via intraperitoneal injection showed no therapeutic effect in our animal experiments.
Collapse
Affiliation(s)
- Lihua Sun
- Department of Hepatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Xiaotang Fan
- Department of Hepatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Lijuan Zhang
- Department of Hepatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Guixiu Shi
- Department of Hepatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Maimaiti Aili
- Department of Hepatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Xiaobo Lu
- Department of Hepatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Tao Jiang
- Department of Hepatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Yuexin Zhang
- Department of Hepatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| |
Collapse
|
20
|
Park CH, Bae SH, Kim HY, Kim JK, Jung ES, Chun HJ, Song MJ, Lee SE, Cho SG, Lee JW, Choi JY, Yoon SK, Han NI, Lee YS. A pilot study of autologous CD34-depleted bone marrow mononuclear cell transplantation via the hepatic artery in five patients with liver failure. Cytotherapy 2013; 15:1571-1579. [PMID: 23849977 DOI: 10.1016/j.jcyt.2013.05.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 04/15/2013] [Accepted: 05/19/2013] [Indexed: 02/07/2023]
Abstract
BACKGROUND AIMS Many rodent experiments and human studies on stem cell therapy have shown promising therapeutic approaches to liver diseases. We investigated the clinical outcomes of five patients with liver failure of various causes who received autologous CD34-depleted bone marrow-derived mononuclear cell (BM-MNC) transplantation, including mesenchymal stromal cells, through the hepatic artery. METHODS CD34-depleted BM-MNCs were obtained from five patients waiting for liver transplantation by bone marrow aspiration and using the CliniMACS CD34 Reagent System (Miltenyi Biotech, Bergisch Gladbach, Germany), and autologous hepatic artery infusion was performed. The causes of hepatic decompensation were hepatitis B virus (HBV), hepatitis C virus (HCV), propylthiouracil-induced toxic hepatitis and Wilson disease. RESULTS Serum albumin levels improved 1 week after transplantation from 2.8 g/dL, 2.4 g/dL, 2.7 g/dL and 1.9 g/dL to 3.3 g/dL, 3.1 g/dL, 2.8 g/dL and 2.6 g/dL. Transient liver elastography data showed some change from 65 kPa, 33 kPa, 34.8 kPa and undetectable to 46.4 kPa, 19.8 kPa, 29.1 kPa and 67.8 kPa at 4 weeks after transplantation in a patient with Wilson disease, a patient with HCV, and two patients with HBV. Ascites decreased in two patients. One of the patients with HBV underwent liver transplantation 4 months after the infusion, and the hepatic progenitor markers (cytokeratin [CD]-7, CD-8, CD-9, CD-18, CD-19, c-Kit and epithelial cell adhesion molecule [EpCAM]) were highly expressed in the explanted liver. CONCLUSIONS Serum albumin levels, liver stiffness, liver volume, subjective healthiness and quality of life improved in the study patients. Although these findings were observed in a small population, the results may suggest a promising future for autologous CD34-depleted BM-MNC transplantation as a bridge to liver transplantation in patients with liver failure.
Collapse
Affiliation(s)
- Chung-Hwa Park
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
The role of bone marrow mesenchymal stem cells in the treatment of acute liver failure. BIOMED RESEARCH INTERNATIONAL 2013; 2013:251846. [PMID: 24312909 PMCID: PMC3842049 DOI: 10.1155/2013/251846] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 09/13/2013] [Accepted: 09/14/2013] [Indexed: 12/30/2022]
Abstract
Objective. This study is to investigate the effects of bone marrow mesenchymal stem cell (BMSC) transplantation on acute liver failure (ALF). Methods. BMSCs were separated from rat bone marrow, cultured, and identified by flow cytometry. Rat model with ALF was established by injecting D-galactosamine and lipopolysaccharide. Rats were randomly divided into the control group and BMSC transplantation group. The serum levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were measured at 24 h, 120 h, and 168 h after BMSC transplantation. Apoptosis was detected by TUNEL assay. The expression of VEGF and AFP proteins was detected by immunofluorescence. Caspase-1 and IL-18 proteins and mRNA were detected by immunohistochemistry and RT-PCR. Results. Compared with the control group, levels of ALT, AST, caspase-1 and IL-18 proteins, and mRNA in the transplantation group were significantly lower at 120 h and 168 h after BMSCs transplantation. Apoptosis was inhibited by BMSCs transplantation. The VEGF protein levels were increased with the improvement of liver function, and the AFP protein levels were increased with the deterioration of the liver function after BMSCs transplantation. Conclusions. BMSCs transplantation can improve liver function and inhibit hepatocyte apoptosis as well as promote hepatocyte proliferation in rat model with ALF.
Collapse
|
22
|
Transplantation of bone marrow-derived mesenchymal stem cells after regional hepatic irradiation ameliorates thioacetamide-induced liver fibrosis in rats. J Surg Res 2013; 186:408-16. [PMID: 24071025 DOI: 10.1016/j.jss.2013.08.016] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Revised: 08/03/2013] [Accepted: 08/16/2013] [Indexed: 12/20/2022]
Abstract
BACKGROUND Recent studies have demonstrated that bone marrow-derived mesenchymal stem cells (BM-MSCs) can potentially revert liver fibrosis, but it is not known if preparative hepatic irradiation (HIR) contributes to the therapeutic effect of transplanted BM-MSCs. In this study, we investigate the effects of HIR on transplanted BM-MSCs in cirrhotic rats and the underlying mechanism by which mesenchymal stem cells (MSCs) relieve liver fibrosis. MATERIALS AND METHODS The BM-MSCs from male rats were labeled with CM-Dil and injected via portal vein into two groups of thioacetamide-induced cirrhotic rats, and the controls were injected with the same volume of saline. The right hemiliver of one cirrhotic rat group was irradiated (15 Gy) 4 d before transplantation. Liver function tests and histologic experiments were performed, and the liver population of BM-MSCs was estimated. RESULTS The transplantation of MSCs alleviated liver fibrosis and reduced expression of transforming growth factor-β1, Smad2, collagen type I, and α-SMA. HIR preconditioning promoted homing and repopulation of MSCs and resulted in better treatment outcomes. CONCLUSIONS HIR preconditioning enhances the effect of BM-MSCs in improving thioacetamide-induced liver fibrosis in rats by promoting their homing and repopulation. BM-MSCs may function by inhibiting transforming growth factor-β1-Smad signaling pathway in the liver.
Collapse
|
23
|
De Minicis S, Rychlicki C, Agostinelli L, Saccomanno S, Trozzi L, Candelaresi C, Bataller R, Millán C, Brenner DA, Vivarelli M, Mocchegiani F, Marzioni M, Benedetti A, Svegliati-Baroni G. Semaphorin 7A contributes to TGF-β-mediated liver fibrogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:820-830. [PMID: 23850082 PMCID: PMC5819760 DOI: 10.1016/j.ajpath.2013.05.030] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2012] [Revised: 04/15/2013] [Accepted: 05/11/2013] [Indexed: 12/22/2022]
Abstract
Semaphorin7A (SEMA7A) is a membrane-anchored protein involved in immune and inflammatory responses, exerting an effect on pulmonary fibrosis. Thus, we aimed to investigate the role of SEMA7A in hepatic fibrosis. Liver injury was induced in vivo by carbon tetrachloride i.p. injection or bile duct ligation in wild-type and SEMA7A knockout (KO) mice. Human and mouse liver samples and primary mouse hepatic cell populations were used for Western blot analysis, quantitative real-time RT-PCR, and immunohistochemistry. SEMA7A is highly expressed in hepatic stellate cells (HSCs). The expression of SEMA7A and its receptor β1-integrin subunit increase during liver injury and in activated HSCs. Transforming growth factor β-stimulated HSCs showed increased expression of SEMA7A in a SMAD2/3-independent manner, leading to increased expression of fibrogenic and inflammation markers. This pattern was significantly blunted in SEMA7A KO HSCs. Overexpression of SEMA7A in HSCs showed increased fibrogenic and inflammation markers expression. In vivo, SEMA7A KO mice treated with carbon tetrachloride and bile duct ligation developed reduced fibrosis versus wild-type mice. Moreover, SEMA7A expression increased in liver samples of patients with fibrosis versus healthy controls. SEMA7A was expressed in the liver and was increased in the course of liver fibrosis, both in mice and in humans. SEMA7A was mainly expressed in HSCs with respect to other cell types in the liver and plays a critical role in regulating fibrosis.
Collapse
Affiliation(s)
- Samuele De Minicis
- Department of Gastroenterology, Polytechnic University of Marche, Ancona, Italy
| | - Chiara Rychlicki
- Department of Gastroenterology, Polytechnic University of Marche, Ancona, Italy
| | - Laura Agostinelli
- Department of Gastroenterology, Polytechnic University of Marche, Ancona, Italy
| | - Stefania Saccomanno
- Department of Gastroenterology, Polytechnic University of Marche, Ancona, Italy
| | - Luciano Trozzi
- Department of Gastroenterology, Polytechnic University of Marche, Ancona, Italy
| | - Cinzia Candelaresi
- Department of Gastroenterology, Polytechnic University of Marche, Ancona, Italy
| | | | | | - David A. Brenner
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine, San Diego, California
| | - Marco Vivarelli
- Department of Gastroenterology, Polytechnic University of Marche, Ancona, Italy
| | | | - Marco Marzioni
- Department of Gastroenterology, Polytechnic University of Marche, Ancona, Italy
| | - Antonio Benedetti
- Department of Gastroenterology, Polytechnic University of Marche, Ancona, Italy
| | | |
Collapse
|
24
|
Nunes de Carvalho S, Helal-Neto E, de Andrade DC, Costa Cortez EA, Thole AA, Barja-Fidalgo C, de Carvalho L. Bone marrow mononuclear cell transplantation increases metalloproteinase-9 and 13 and decreases tissue inhibitors of metalloproteinase-1 and 2 expression in the liver of cholestatic rats. Cells Tissues Organs 2013; 198:139-48. [PMID: 23886643 DOI: 10.1159/000353215] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2013] [Indexed: 11/19/2022] Open
Abstract
Liver fibrosis results from chronic injury followed by activation of macrophages and fibrogenic cells like myofibroblasts and activated hepatic stellate cells. These fibrogenic cells express α-smooth muscle actin (α-SMA) and produce excessive extracellular matrix (ECM), with disorganization and loss of function of hepatic parenchyma. It is known that increased levels of metalloproteinases (MMPs) in liver fibrosis are associated with reduction of the pathologic ECM and fibrosis resolution. Recently, it has been shown that bone marrow mononuclear cells (BMMNCs) may reduce collagen and α-SMA expression, and ameliorate liver function in cholestatic rats. Therefore, this study aimed to analyze MMP-2, MMP-9 and MMP-13, and tissue inhibitors of MMPs (TIMPs)-1 and TIMP-2 in the liver of cholestatic rats transplanted with BMMNC. Animals were divided into normal rats, cholestatic rats obtained after 14 and 21 days of bile duct ligation (BDL), and rats obtained after 14 days of BDL that received BMMNCs and were killed after 7 days. MMP and TIMP expression was assessed by Western blotting, along with α-SMA, CD68 and CD11b expression by confocal microscopy. Western blotting analysis showed that 14-day BDL animals had significantly reduced amounts of MMP-2 and MMP-13, but increased amounts of MMP-9 compared to normal rats. After 21 days of BDL, overall MMP amounts were decreased and TIMPs were increased. BMMNC transplantation significantly increased MMP-9 and MMP-13, and decreased TIMP expression. Increased MMP activity was confirmed by zymography. MMP-9 and MMP-13 were expressed by macrophages near fibrotic septa, suggesting BMMNC may stimulate MMP production in fibrotic livers, contributing to ECM degradation and hepatic regeneration.
Collapse
Affiliation(s)
- Simone Nunes de Carvalho
- Laboratório Cultura de Células, Departamento de Histologia e Embriologia, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, UERJ, Rio de Janeiro, Brazil
| | | | | | | | | | | | | |
Collapse
|
25
|
Effect of bone marrow mesenchymal stem cells on hepatocellular carcinoma in microcirculation. Tumour Biol 2013; 34:2161-8. [PMID: 23584896 DOI: 10.1007/s13277-013-0749-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 03/13/2013] [Indexed: 12/21/2022] Open
Abstract
This study aims t explore the effect and application of bone marrow mesenchymal stem cells (BMSCs) on hepatocellular carcinoma in microcirculation by observing the angiogenesis of hepatocellular carcinoma in transplanted area. BMSCs were isolated and cultured primarily using the method of whole bone marrow culture and identifying surface antigens of third-generation bone marrow-derived mesenchymal stem cells using flow cytometry. Hepatoma cells cultured with BMSCs-conditioned medium (BMSCs-CM) were assayed using the cell proliferation rate of the MTT method. Nude mice were divided into control group (group A), BMSCs cell transplantation group (group B), HepG-2 cell group (group C), and combined BMSCs and HepG-2 cell cotransplanted group (group D). The result showed that the microvascular density was not significantly different in groups A and B. However, the microvascular density at 14 days was higher than 0 day in group C (P < 0.05). In group D, the microvascular density at 14 days was higher than that of 7 and 0 days (P < 0.05) and 7 days was higher than 0 days (P < 0.05). It was showed that the microvascular density did not get significant difference at 0 and 7 days in the four groups (P > 0.05). But the microvascular density of group C was higher than groups A and B at 14 days (P < 0.05), group D was higher than groups A and B at 14 days (P < 0.05) and group D was higher than group C at 14 days (P < 0.05). BMSCs could promote the growth of microvascular in hepatoma cells in a transplanted area.
Collapse
|
26
|
Liu WH, Liu ZC, You N, Zhang N, Wang T, Gong ZB, Liu HB, Dou KF. Several important in vitro improvements in the amplification, differentiation and tracing of fetal liver stem/progenitor cells. PLoS One 2012; 7:e47346. [PMID: 23056632 PMCID: PMC3467257 DOI: 10.1371/journal.pone.0047346] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 09/11/2012] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVE We previously isolated fetal liver stem/progenitor cells (FLSPCs), but there is an urgent need to properly amplify FLSPCs, effectively induce FLSPCs differentiation, and steadily trace FLSPCs for in vivo therapeutic investigation. METHODS FLSPCs were maintained in vitro as adherent culture or soft agar culture for large-scale amplification. To direct the differentiation of FLSPCs into hepatocytes, FLSPCs were randomly divided into four groups: control, 1% DMSO-treated, 20 ng/ml HGF-treated and 1% DMSO+20 ng/ml HGF-treated. To trace FLSPCs, the GFP gene was introduced into FLSPCs by liposome-mediated transfection. RESULTS For amplifying FLSPCs, the soft agar culture were more suitable than the adherent culture, because the soft agar culture obtained more homogeneous cells. These cells were with high nuclear:cytoplasmic ratio, few cell organelles, high expression of CD90.1 and CD49f, and strong alkaline phosphatase staining. For inducing FLSPCs differentiation, treatment with HGF+DMSO was most effective (P<0.05), which was strongly supported by the typical morphological change and the significant decrease of OV-6 positive cells (P<0.01). In addition, the time of indocyanine green elimination, the percentage of glycogen synthetic cells, and the expressions of ALB, G-6-P, CK-8, CK-18 and CYP450-3A1 in HGF+DMSO-treated group were higher than in any other group. For tracing FLSPCs, after the selection of stable FLSPC transfectants, GFP expression continued over successive generations. CONCLUSIONS FLSPCs can properly self-renew in soft agar culture and effectively differentiate into hepatocyte-like cells by HGF+DMSO induction, and they can be reliably traced by GFP expression.
Collapse
Affiliation(s)
- Wei-hui Liu
- PLA Center of General Surgery, General Hospital of Chengdu Army Region, Chengdu, Sichuan Province, People's Republic of China
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Zheng-cai Liu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Nan You
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Ning Zhang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Tao Wang
- PLA Center of General Surgery, General Hospital of Chengdu Army Region, Chengdu, Sichuan Province, People's Republic of China
| | - Zhen-bin Gong
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Hong-bao Liu
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Ke-feng Dou
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| |
Collapse
|
27
|
Zhao C, Tan A, Pastorin G, Ho HK. Nanomaterial scaffolds for stem cell proliferation and differentiation in tissue engineering. Biotechnol Adv 2012; 31:654-68. [PMID: 22902273 DOI: 10.1016/j.biotechadv.2012.08.001] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 07/31/2012] [Accepted: 08/01/2012] [Indexed: 02/08/2023]
Abstract
Tissue engineering is a clinically driven field and has emerged as a potential alternative to organ transplantation. The cornerstone of successful tissue engineering rests upon two essential elements: cells and scaffolds. Recently, it was found that stem cells have unique capabilities of self-renewal and multilineage differentiation to serve as a versatile cell source, while nanomaterials have lately emerged as promising candidates in producing scaffolds able to better mimic the nanostructure in natural extracellular matrix and to efficiently replace defective tissues. This article, therefore, reviews the key developments in tissue engineering, where the combination of stem cells and nanomaterial scaffolds has been utilized over the past several years. We consider the high potential, as well as the main issues related to the application of stem cells and nanomaterial scaffolds for a range of tissues including bone, cartilage, nerve, liver, eye etc. Promising in vitro results such as efficient attachment, proliferation and differentiation of stem cells have been compiled in a series of examples involving different nanomaterials. Furthermore, the merits of the marriage of stem cells and nanomaterial scaffolds are also demonstrated in vivo, providing early successes to support subsequent clinical investigations. This progress simultaneously drives mechanistic research into the mechanotransduction process responsible for the observations in order to optimize the process further. Current understanding is chiefly reported to involve the interaction of stem cells and the anchoring nanomaterial scaffolds by activating various signaling pathways. Substrate surface characteristics and scaffold bulk properties are also reported to influence not only short term stem cell adhesion, spreading and proliferation, but also longer term lineage differentiation, functionalization and viability. It is expected that the combination of stem cells and nanomaterials will develop into an important tool in tissue engineering for the innovative treatment of many diseases.
Collapse
Affiliation(s)
- Chunyan Zhao
- NanoCore, Engineering Block A, EA, Faculty of Engineering, National University of Singapore, 117576, Singapore
| | | | | | | |
Collapse
|
28
|
Cho JW, Lee CY, Ko Y. Therapeutic potential of mesenchymal stem cells overexpressing human forkhead box A2 gene in the regeneration of damaged liver tissues. J Gastroenterol Hepatol 2012; 27:1362-70. [PMID: 22432472 PMCID: PMC3492917 DOI: 10.1111/j.1440-1746.2012.07137.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIM Although a liver transplantation is considered to be the only effective long-term treatment in many cases of liver diseases, it is limited by a lack of donor organs and immune rejection. As an autologous stem cell approach, this study was conducted to assess whether forkhead box A2 (Foxa2) gene overexpression in bone marrow-derived mesenchymal stem cells (MSC) could protect the liver from hepatic diseases by stimulating tissue regeneration after cell transplantation. METHODS Rat MSC (rMSC) were isolated, characterized, and induced to hepatocytes that expressed liver-specific markers. Four different treatments (control [phosphate-buffered saline], rMSC alone, rMSC/pIRES-enhanced green fluorescent protein (EGFP) vector, and rMSC/pIRES-EGFP/human Foxa2) were injected into the spleen of carbon tetrachloride-injured rats. Biochemical and histological analyses on days 30, 60, and 90 post-transplantation were performed to evaluate the therapeutic capacities of MSC overexpressing hFoxa2. RESULTS rMSC transfected with hFoxa2 were induced into hepatogenic linage and expressed several liver-specific genes, such as, Foxa2, α-fetoprotein, cytokeratin-18, hepatocyte nuclear factor-1α, and hepatocyte growth factor. A group of animals treated with MSC/hFoxa2 showed significant recovery of liver-specific enzyme expressions to normal levels at the end of the study (90 days). Furthermore, when compared to the fibrotic areas of the samples treated with MSC alone or MSC/vector, the fibrotic area of the samples treated with rMSC/hFoxa2 for 90 days significantly decreased, until they were completely gone. CONCLUSIONS Human Foxa2 efficiently promoted the incorporation of MSC into liver grafts, suggesting that hFoxa2 genes could be used for the structural or functional recovery of damaged liver cells.
Collapse
Affiliation(s)
- Jong-Woo Cho
- Department of Biotechnology, Korea UniversitySeoul, Korea
| | - Chul-Young Lee
- Department of Animal Material Engineering, College of Science and Natural Resource, Gyeongnam National University of Science and TechnologyJinju, Korea
| | - Yong Ko
- Department of Biotechnology, Korea UniversitySeoul, Korea
| |
Collapse
|
29
|
Saiman Y, Friedman SL. The role of chemokines in acute liver injury. Front Physiol 2012; 3:213. [PMID: 22723782 PMCID: PMC3379724 DOI: 10.3389/fphys.2012.00213] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 05/30/2012] [Indexed: 11/25/2022] Open
Abstract
Chemokines are small molecular weight proteins primarily known to drive migration of immune cell populations. In both acute and chronic liver injury, hepatic chemokine expression is induced resulting in inflammatory cell infiltration, angiogenesis, and cell activation and survival. During acute injury, massive parenchymal cell death due to apoptosis and/or necrosis leads to chemokine production by hepatocytes, cholangiocytes, Kupffer cells, hepatic stellate cells, and sinusoidal endothelial cells. The specific chemokine profile expressed during injury is dependent on both the type and course of injury. Hepatotoxicity by acetaminophen for example leads to cellular necrosis and activation of Toll-like receptors while the inciting insult in ischemia reperfusion injury produces reactive oxygen species and subsequent production of pro-inflammatory chemokines. Chemokine expression by these cells generates a chemoattractant gradient promoting infiltration by monocytes/macrophages, NK cells, NKT cells, neutrophils, B cells, and T cells whose activity are highly regulated by the specific chemokine profiles within the liver. Additionally, resident hepatic cells express chemokine receptors both in the normal and injured liver. While the role of these receptors in normal liver has not been well described, during injury, receptor up-regulation, and chemokine engagement leads to cellular survival, proliferation, apoptosis, fibrogenesis, and expression of additional chemokines and growth factors. Hepatic-derived chemokines can therefore function in both paracrine and autocrine fashions further expanding their role in liver disease. More recently it has been appreciated that chemokines can have diverging effects depending on their temporal expression pattern and the type of injury. A better understanding of chemokine/chemokine receptor axes will therefore pave the way for development of novel targeted therapies for the treatment of liver disease.
Collapse
Affiliation(s)
- Yedidya Saiman
- Division of Liver Diseases, Department of Medicine, Mount Sinai School of Medicine New York, NY, USA
| | | |
Collapse
|
30
|
Treska V, Safranek J, Lysak D, Mirka H, Skalicky T, Slauf F, Hes O. A complex oncosurgical approach to increasing the resectability of colorectal cancer metastases - a case report. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2012; 158:154-7. [PMID: 22660223 DOI: 10.5507/bp.2012.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Accepted: 01/03/2012] [Indexed: 11/23/2022] Open
Abstract
AIM In this case report, the authors aim to demonstrate the success of recent methods in the radical treatment of a patient with primary inoperable liver and subsequent colorectal cancer pulmonary metastases. METHODS A 75 year old patient with inoperable bulky metastasis in the right hepatic lobe and insufficient future remnant liver volume was indicated for a stage procedure in the liver parenchyma. Embolization of the right branch of the portal vein was first performed with subsequent administration of stem cells into the contralateral liver lobe. Following compensatory growth of the left liver lobe, right-sided hepatectomy was performed with subsequent adjuvant oncological treatment. Six months after the surgery, a metastasis developed in the right pulmonary lobe which was solved by metastasectomy. RESULTS The patient, one year after the diagnosis of inoperable liver metastasis, is completely healthy and free of signs of disease recurrence. CONCLUSION A comprehensive oncosurgical approach using up-to-date diagnostic and treatment options may offer patients with metastatic colorectal cancer, radical treatment with the hope of long-term quality survival.
Collapse
Affiliation(s)
- Vladislav Treska
- Department of Surgery, University Hospital and Medical Faculty in Pilsen, Czech Republic
| | | | | | | | | | | | | |
Collapse
|
31
|
Abd El Aziz DH, Metwally HG. Effect of stem cell therapy on amiodarone-induced liver injury in albino rats. THE EGYPTIAN JOURNAL OF HISTOLOGY 2012; 35:272-282. [DOI: 10.1097/01.ehx.0000414591.19244.e3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
32
|
Liver tissue engineering: Recent advances in the development of a bio-artificial liver. BIOTECHNOL BIOPROC E 2012. [DOI: 10.1007/s12257-012-0047-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
33
|
Chistiakov DA. Liver regenerative medicine: advances and challenges. Cells Tissues Organs 2012; 196:291-312. [PMID: 22572238 DOI: 10.1159/000335697] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2011] [Indexed: 12/16/2022] Open
Abstract
Liver transplantation is the standard care for many end-stage liver diseases. However, donor organs are scarce and some people succumb to liver failure before a donor is found. Liver regenerative medicine is a special interdisciplinary field of medicine focused on the development of new therapies incorporating stem cells, gene therapy and engineered tissues in order to repair or replace the damaged organ. In this review we consider the emerging progress achieved in the hepatic regenerative medicine within the last decade. The review starts with the characterization of liver organogenesis, fetal and adult stem/progenitor cells. Then, applications of primary hepatocytes, embryonic and adult (mesenchymal, hematopoietic and induced pluripotent) stem cells in cell therapy of liver diseases are considered. Current advances and challenges in producing mature hepatocytes from stem/progenitor cells are discussed. A section about hepatic tissue engineering includes consideration of synthetic and natural biomaterials in engineering scaffolds, strategies and achievements in the development of 3D bioactive matrices and 3D hepatocyte cultures, liver microengineering, generating bioartificial liver and prospects for fabrication of the bioengineered liver.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- Department of Medical Nanobiotechnology, Pirogov State Medical University, Moscow, Russia.
| |
Collapse
|
34
|
|
35
|
Abstract
The refinement of radiation therapy and radioembolization techniques has led to a resurgent interest in radiation-induced liver disease (RILD). The awareness of technical and clinical parameters that influence the chance of RILD is important to guide patient selection and toxicity minimization strategies. "Classic" RILD is characterized by anicteric ascites and hepatomegaly and is unlikely to occur after a mean liver dose of approximately 30 Gy in conventional fractionation. By maintaining a low mean liver dose and sparing a "critical volume" of liver from radiation, stereotactic delivery techniques allow for the safe administration of higher tumor doses. Caution must be exercised for patients with hepatocellular carcinoma or pre-existing liver disease (eg, Child-Pugh score of B or C) because they are more susceptible to RILD that can manifest in a nonclassic pattern. Although no pharmacologic interventions have yet been proven to mitigate RILD, preclinical research shows the potential for therapies targeting transforming growth factor-β and for the transplantation of stem cells, hepatocytes, and liver progenitor cells as strategies that may restore liver function. Also, in the clinical setting of veno-occlusive liver disease after high-dose chemotherapy, agents with fibrinolytic and antithrombotic properties can reverse liver failure, suggesting a possible role in the setting of RILD.
Collapse
Affiliation(s)
- Chandan Guha
- Department of Radiation Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10467, USA.
| | | |
Collapse
|
36
|
Therapeutic implications of mesenchymal stem cells in liver injury. J Biomed Biotechnol 2012. [PMID: 22228987 DOI: 10.1155/2011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs), represent an attractive tool for the establishment of a successful stem-cell-based therapy of liver diseases. A number of different mechanisms contribute to the therapeutic effects exerted by MSCs, since these cells can differentiate into functional hepatic cells and can also produce a series of growth factors and cytokines able to suppress inflammatory responses, reduce hepatocyte apoptosis, regress liver fibrosis, and enhance hepatocyte functionality. To date, the infusion of MSCs or MSC-conditioned medium has shown encouraging results in the treatment of fulminant hepatic failure and in end-stage liver disease in experimental settings. However, some issues under debate hamper the use of MSCs in clinical trials. This paper summarizes the biological relevance of MSCs and the potential benefits and risks that can result from translating the MSC research to the treatment of liver diseases.
Collapse
|
37
|
Abstract
Liver fibrosis is the result of the entire organism responding to a chronic injury. Every cell type in the liver contributes to the fibrosis. This paper first discusses key intracellular signaling pathways that are induced during liver fibrosis. The paper then examines the effects of these signaling pathways on the major cell types in the liver. This will provide insights into the molecular pathophysiology of liver fibrosis and should identify therapeutic targets.
Collapse
|
38
|
Therapeutic implications of mesenchymal stem cells in liver injury. J Biomed Biotechnol 2011; 2011:860578. [PMID: 22228987 PMCID: PMC3250695 DOI: 10.1155/2011/860578] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 10/17/2011] [Accepted: 10/17/2011] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs), represent an attractive tool for the establishment of a successful stem-cell-based therapy of liver diseases. A number of different mechanisms contribute to the therapeutic effects exerted by MSCs, since these cells can differentiate into functional hepatic cells and can also produce a series of growth factors and cytokines able to suppress inflammatory responses, reduce hepatocyte apoptosis, regress liver fibrosis, and enhance hepatocyte functionality. To date, the infusion of MSCs or MSC-conditioned medium has shown encouraging results in the treatment of fulminant hepatic failure and in end-stage liver disease in experimental settings. However, some issues under debate hamper the use of MSCs in clinical trials. This paper summarizes the biological relevance of MSCs and the potential benefits and risks that can result from translating the MSC research to the treatment of liver diseases.
Collapse
|
39
|
Assimakopoulos SF, Tsamandas AC, Alexandris IH, Georgiou C, Vagianos CE, Scopa CD. Stimulation of oval cell and hepatocyte proliferation by exogenous bombesin and neurotensin in partially hepatectomized rats. World J Gastrointest Pathophysiol 2011; 2:146-54. [PMID: 22180848 PMCID: PMC3240906 DOI: 10.4291/wjgp.v2.i6.146] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 11/07/2011] [Accepted: 11/14/2011] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of the neuropeptides bombesin (BBS) and neurotensin (NT) on oval cell proliferation in partially hepatectomized rats not pretreated with a known hepatocyte inhibitor.
METHODS: Seventy male Wistar rats were randomly divided into five groups: I = controls, II = sham operated, III = partial hepatectomy 70% (PHx), IV = PHx + BBS (30 μg/kg per day), V = PHx + NT (300 μg/kg per day). Forty eight hours after liver resection, portal endotoxin levels and hepatic glutathione redox state were determined. α-fetoprotein (AFP) mRNA (in situ hybridisation), cytokeratin-19 and Ki67 antigen expression (immunohistochemistry) and apoptosis (TUNEL) were evaluated on liver tissue samples. Cells with morphological features of oval cells that were cytokeratin-19 (+) and AFP mRNA (+) were scored in morphometric analysis and their proliferation was recorded. In addition, the proliferation and apoptotic rates of hepatocytes were determined.
RESULTS: In the control and sham operated groups, oval cells were significantly less compared to groups III, IV and V (P < 0.001). The neuropeptides BBS and NT significantly increased the proliferation of oval cells compared to group III (P < 0.001). In addition, BBS and NT induced a significant increase of hepatocyte proliferation (P < 0.001), whereas it decreased their apoptotic activity (P < 0.001) compared to group III. BBS and NT significantly decreased portal endotoxemia (P < 0.001) and increased the hepatic GSH: GSSG ratio (P < 0.05 and P < 0.001, respectively) compared to group III.
CONCLUSION: BBS and NT stimulated oval cell proliferation in a model of liver regeneration, without use of concomitant suppression of hepatocyte proliferation as oval cell activation stimuli, and improved the hepatocyte regenerative response. This peptides-induced combined stimulation of oval cell and hepatocyte proliferation might serve as a possible treatment modality for several liver diseases.
Collapse
|
40
|
Fetal liver cell transplantation as a potential alternative to whole liver transplantation? J Gastroenterol 2011; 46:953-65. [PMID: 21698354 DOI: 10.1007/s00535-011-0427-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 05/02/2011] [Indexed: 02/04/2023]
Abstract
Because organ shortage is the fundamental limitation of whole liver transplantation, novel therapeutic options, especially the possibility of restoring liver function through cell transplantation, are urgently needed to treat end-stage liver diseases. Groundbreaking in vivo studies have shown that transplanted hepatocytes are capable of repopulating the rodent liver. The two best studied models are the urokinase plasminogen activator (uPA) transgenic mouse and the fumarylacetoacetate hydrolase (FAH)-deficient mouse, in which genetic modifications of the recipient liver provide a tissue environment in which there is extensive liver injury and selection pressure favoring the proliferation and survival of transplanted hepatocytes. Because transplanted hepatocytes do not significantly repopulate the (near-)normal liver, attention has been focused on finding alternative cell types, such as stem or progenitor cells, that have a higher proliferative potential than hepatocytes. Several sources of stem cells or stem-like cells have been identified and their potential to repopulate the recipient liver has been evaluated in certain liver injury models. However, rat fetal liver stem/progenitor cells (FLSPCs) are the only cells identified to date that can effectively repopulate the (near-)normal liver, are morphologically and functionally fully integrated into the recipient liver, and remain viable long-term. Even though primary human fetal liver cells are not likely to be routinely used for clinical liver cell repopulation in the future, using or engineering candidate cells exhibiting the characteristics of FLSPCs suggests a new direction in developing cell transplantation strategies for therapeutic liver replacement. This review will give a brief overview concerning the existing animal models and cell sources that have been used to restore normal liver structure and function, and will focus specifically on the potential of FLSPCs to repopulate the liver.
Collapse
|
41
|
Kumar A, Pati NT, Sarin SK. Use of stem cells for liver diseases-current scenario. J Clin Exp Hepatol 2011; 1:17-26. [PMID: 25755306 PMCID: PMC3940313 DOI: 10.1016/s0973-6883(11)60114-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 06/04/2011] [Indexed: 12/12/2022] Open
Abstract
End-stage liver disease and liver failure are major health problems worldwide leading to high mortality and morbidity and high healthcare costs. Currently, orthotropic liver transplantation is the only effective treatment available to the patients of end-stage liver disease. However, a serious shortage of liver donors, high cost, and risk of organ rejection are the major obstacles to liver transplantation. Because of the ability of stem cells for differentiation into any tissue type, they have huge potential in therapy of various end-stage or degenerative diseases and traumatic injuries. Stem cell therapy has the potential to provide a valuable adjunct and alternative to liver transplantation and has immense potential in the management of end stage liver disease and liver failure. Stem cell therapy can be mediated by either a direct contribution to the functional hepatocyte population with embryonic, induced pluripotent, or adult stem cells or by promotion of endogenous regenerative processes with bone marrow-derived stem cells. Initial translational studies have been encouraging and have suggested improved liver function in advanced chronic liver disease and enhanced liver regeneration after portal vein embolization and partial hepatic resection. Stem cells infusion in cirrhotic patients has improved liver parameters and could form a viable bridge to transplantation. The present review summarizes basic of stem cell biology relevant to clinicians and an update on recent advances on the management of liver diseases using stem cells.
Collapse
Key Words
- AFP, alpha (α)-fetoprotein
- BM, bone marrow
- EPCAM, epithelial cell adhesion molecule
- ES, embryonic stem
- FSCs, fetal stem cells
- HPC, hepatic progenitor cells
- HSC, hematopoietic stem cells
- Hepatocyte transplantation
- ICAM, intercellular adhesion molecule
- MSCs, mesenchymal stem cells
- NCAM, neural cell adhesion molecule
- UCB, umbilical cord blood
- hAECs, human amniotic epithelial cells
- iPSCs, induced pluripotent stem cells
- liver transplantation
- stem cell
Collapse
Affiliation(s)
- Ashish Kumar
- Department of Hepatology, Institute of Liver and Biliary Sciences (ILBS), New Delhi, India
- Special Center for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
- Address for correspondence: Dr Ashish Kumar MD DM, Associate Professor, Department of Hepatology, Institute of Liver and Biliary Sciences (ILBS), D-1, Vasant Kunj, New Delhi-110070, India
| | - Nirupama Trehan Pati
- Department of Research, Institute of Liver and Biliary Sciences (ILBS), New Delhi, India
| | - Shiv Kumar Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences (ILBS), New Delhi, India
- Special Center for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
42
|
Zhang D, Jiang M, Miao D. Transplanted human amniotic membrane-derived mesenchymal stem cells ameliorate carbon tetrachloride-induced liver cirrhosis in mouse. PLoS One 2011; 6:e16789. [PMID: 21326862 PMCID: PMC3033905 DOI: 10.1371/journal.pone.0016789] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2010] [Accepted: 01/14/2011] [Indexed: 01/19/2023] Open
Abstract
Background Human amniotic membrane-derived mesenchymal stem cells (hAMCs) have the potential to reduce heart and lung fibrosis, but whether could reduce liver fibrosis remains largely unknown. Methodology/Principal Findings Hepatic cirrhosis model was established by infusion of CCl4 (1 ml/kg body weight twice a week for 8 weeks) in immunocompetent C57Bl/6J mice. hAMCs, isolated from term delivered placenta, were infused into the spleen at 4 weeks after mice were challenged with CCl4. Control mice received only saline infusion. Animals were sacrificed at 4 weeks post-transplantation. Blood analysis was performed to evaluate alanine aminotransferase (ALT) and aspartate aminotransferase (AST). Histological analysis of the livers for fibrosis, hepatic stellate cells activation, hepatocyte apoptosis, proliferation and senescence were performed. The donor cell engraftment was assessed using immunofluorescence and polymerase chain reaction. The areas of hepatic fibrosis were reduced (6.2%±2.1 vs. control 9.6%±1.7, p<0.05) and liver function parameters (ALT 539.6±545.1 U/dl, AST 589.7±342.8 U/dl,vs. control ALT 139.1±138.3 U/dl, p<0.05 and AST 212.3±110.7 U/dl, p<0.01) were markedly ameliorated in the hAMCs group compared to control group. The transplantation of hAMCs into liver-fibrotic mice suppressed activation of hepatic stellate cells, decreased hepatocyte apoptosis and promoted liver regeneration. More interesting, hepatocyte senescence was depressed significantly in hAMCs group compared to control group. Immunofluorescence and polymerase chain reaction revealed that hAMCs engraftment into host livers and expressed the hepatocyte-specific markers, human albumin and α-fetoproteinran. Conclusions/Significance The transplantation of hAMCs significantly decreased the fibrosis formation and progression of CCl4-induced cirrhosis, providing a new approach for the treatment of fibrotic liver disease.
Collapse
Affiliation(s)
- DingGuo Zhang
- The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, China
| | - MinYue Jiang
- The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, China
| | - DengShun Miao
- The Research Center for Bone and Stem Cells, Nanjing Medical University, Nanjing, China
- * E-mail:
| |
Collapse
|
43
|
Abstract
Mammalian liver has a unique capacity to regenerate following resection or injury, and recovery of liver mass is mainly through proliferation of remaining adult hepatocytes. However, in pathologic conditions, especially during acute liver failure (ALF) and advanced stages of chronic liver disease (CLD), regeneration eventually fails and orthothopic liver transplantation (OLT) represents the only curative approach. The clinical scenario of a world-wide increasing incidence of end-stage CLD and an associated lack of organ availability has led several laboratories to explore the feasibility and efficiency of experimental alternatives to OLT involving cellular therapy. This review presents experimental and clinical studies performed in the last 10-15 years where adult and embryonic hepatocytes, hepatic stem/progenitor cells and extrahepatic stem cells have been used as transplantable cell sources.
Collapse
Affiliation(s)
- Francesco Paolo Russo
- Department of Surgical and Gastroenterological Sciences, Gastroenterology Unit, University of Padova, Padova, Italy
| | | |
Collapse
|
44
|
Abstract
The unique ability of the liver to regenerate itself has fascinated biologists for years and has made it the prototype for mammalian organ regeneration. Harnessing this process has great potential benefit in the treatment of liver failure and has been the focus of intense research over the past 50 years. Not only will detailed understanding of cell proliferation in response to injury be applicable to other dysfunction of organs, it may also shed light on how cancer develops in a cirrhotic liver, in which there is intense pressure on cells to regenerate. Advances in molecular techniques over the past few decades have led to the identification of many regulatory intermediates, and pushed us onto the verge of an explosive era in regenerative medicine. To date, more than 10 clinical trials have been reported in which augmented regeneration using progenitor cell therapy has been attempted in human patients. This review traces the path that has been taken over the last few decades in the study of liver regeneration, highlights new concepts in the field, and discusses the challenges that still stand between us and clinical therapy.
Collapse
Affiliation(s)
| | - Yock Young Dan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - Nelson Fausto
- Department of Pathology, University of Washington, Seattle, WA
| |
Collapse
|
45
|
Assimakopoulos SF, Tsamandas AC, Georgiou CD, Vagianos CE, Scopa CD. Bombesin and neurotensin exert antiproliferative effects on oval cells and augment the regenerative response of the cholestatic rat liver. Peptides 2010; 31:2294-303. [PMID: 20833216 DOI: 10.1016/j.peptides.2010.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 09/02/2010] [Accepted: 09/02/2010] [Indexed: 12/30/2022]
Abstract
The regenerative capacity of the cholestatic liver is significantly attenuated. Oval cells are hepatic stem cells involved in liver's regeneration following diverse types of injury. The present study investigated the effect of the neuropeptides bombesin (BBS) and neurotensin (NT) on oval cell proliferation as well as on hepatocyte and cholangiocyte proliferation and apoptosis in the cholestatic rat liver. Seventy male Wistar rats were randomly divided into five groups: controls, sham operated, bile duct ligated (BDL), BDL+BBS (30 μg/kg/d), BDL+NT (300 μg/kg/d). Ten days later, alpha-fetoprotein (AFP) mRNA (in situ hybridization), cytokeratin-19 and Ki67 antigen expression (immunohistochemistry) and apoptosis (TUNEL) were evaluated on liver tissue samples. Cells with morphologic features of oval cells that were cytokeratin-19(+) and AFP mRNA(+) were scored in morphometric analysis and their proliferation was recorded. In addition, the proliferation and apoptotic rates of hepatocytes and cholangiocytes were determined. Alanine aminotransferase (ALT) levels and hepatic oxidative stress (lipid peroxidation and glutathione redox state) were also estimated. The neuropeptides BBS and NT significantly reduced ALT levels and hepatic oxidative stress. Both agents exerted similar and cell type-specific effects on oval cells, hepatocytes and cholangiocytes: (a) oval cell proliferation and accumulation in the cholestatic liver was attenuated, (b) hepatocyte proliferation was increased along with a decreased rate of their apoptosis and (c) cholangiocyte proliferation was attenuated and their apoptosis was increased. These observations might be of potential value in patients with extrahepatic cholestasis.
Collapse
|
46
|
Tuleuova N, Lee JY, Lee J, Ramanculov E, Zern MA, Revzin A. Using growth factor arrays and micropatterned co-cultures to induce hepatic differentiation of embryonic stem cells. Biomaterials 2010; 31:9221-31. [PMID: 20832855 DOI: 10.1016/j.biomaterials.2010.08.050] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Accepted: 08/23/2010] [Indexed: 12/21/2022]
Abstract
The success in driving embryonic stem cells towards hepatic lineage has been confounded by the complexity and cost of differentiation protocols that employ large quantities of expensive growth factors (GFs). Instead of supplementing culture media with soluble GFs, we investigated cultivation and differentiation of mouse embryonic stem cells (mESCs) on printed arrays of GFs. Hepatocyte growth factor (HGF), basic fibroblast growth factor (bFGF) and bone morphogenetic protein (BMP4) were mixed in solution with fibronectin and collagen (I) and then printed onto silane-modified glass slides to form 500 μm diameter protein spots. mESCs were cultured on top of GF spots for up to 12 days and analyzed by RT-PCR and immunostaining at different time points. The stem cells residing on HGF-containing combinations of GFs exhibited requisite features of hepatic differentiation including pronounced loss in pluripotency (Oct4), transient (up and down) expression of endoderm (Sox17) and upregulation of early hepatic markers--albumin and alpha-fetoprotein. The hepatic differentiation was enhanced further by adding hepatic stellate cells to surfaces that already contained mESCs on GF spots. A combination of co-culture with non-parenchymal liver cells and the optimal GF stimulation was found to induce endoderm and hepatic phenotype earlier and to a much greater extent than the GF arrays or micropatterned co-cultures used individually. While this paper investigated hepatic differentiation of mouse ESCs, our findings and stem cell culture approaches are likely to be relevant for human ESC cultivation. Overall, the platform combining printed GF arrays and heterotypic co-cultures will be broadly applicable for identifying the composition of the microenvironment niche for ESC differentiation into various tissue types.
Collapse
Affiliation(s)
- Nazgul Tuleuova
- Department of Biomedical Engineering, University of California-Davis, 451 Health Sciences Drive #2519, Davis, CA 95616, USA
| | | | | | | | | | | |
Collapse
|
47
|
|