1
|
Li M, Zhang J, Fang J, Xin Y, Zhu H, Ding X. Pre-administration of human umbilical cord mesenchymal stem cells has better therapeutic efficacy in rats with D-galactosamine-induced acute liver failure. Int Immunopharmacol 2024; 130:111672. [PMID: 38377851 DOI: 10.1016/j.intimp.2024.111672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/22/2024]
Abstract
BACKGROUND Acute liver failure (ALF) is characterized by an intense systemic inflammatory response, single or multiple organ system failure and high mortality. However, specific and effective treatments for ALF patients are still lacking. According to the current investigation, human umbilical cord mesenchymal stem cells (hUCMSCs) have shown remarkable potential to enhance the functional recovery of injured livers. We aimed to investigate the therapeutic effects of time-differentiated hUCMSCs administration regimens on ALF. METHODS The rat model of ALF was induced by D-galactosamine (D-gal), and hUCMSCs were administered via the tail vein 12 h before or 2 h after induction. The potential mechanisms of hUCMSCs in treatment of ALF, regulation cell subset and secretion of inflammatory factors, were verified by co-culturing with PBMCs in vitro. Liver function indicators were detected by an automatic biochemistry analyzer and inflammatory factors were obtained by ELISA detection. The distribution of hUCMSCs in rats after administration was followed by quantitative real-time fluorescence PCR. RESULTS The findings of the study discovered that administration of hUCMSCs 12 h prior to surgery could significantly improve the survival rate of rats, stabilize various liver function indicators in serum levels of ALT, AST, T-BIL, or ALB diminish inflammatory infiltration in liver tissue, and inhibit the secretion of inflammatory factors. CONCLUSION Our data showed that pre-transplantation of hUCMSCs had a better therapeutic effect on ALF rats, providing empirical evidence for preclinical studies. Thus, the timing of hUCMSCs transplantation is necessary for the optimal clinical treatment effect.
Collapse
Affiliation(s)
- Min Li
- Sinoneural Cell Engineering Group Holdings., Co, Ltd, No.1188, Lianhang Road, Shanghai 201100, PR China
| | - Jigang Zhang
- Clinical Research Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.100 Haining Road, Shanghai 200080, PR China; Shanghai Engineering Research Center of Translational Medicine of Cell Therapy, Shanghai 200080, PR China
| | - Jingmeng Fang
- Sinoneural Cell Engineering Group Holdings., Co, Ltd, No.1188, Lianhang Road, Shanghai 201100, PR China
| | - Yuan Xin
- Sinoneural Cell Engineering Group Holdings., Co, Ltd, No.1188, Lianhang Road, Shanghai 201100, PR China
| | - Hao Zhu
- Sinoneural Cell Engineering Group Holdings., Co, Ltd, No.1188, Lianhang Road, Shanghai 201100, PR China.
| | - Xueying Ding
- Clinical Research Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.100 Haining Road, Shanghai 200080, PR China; Shanghai Engineering Research Center of Translational Medicine of Cell Therapy, Shanghai 200080, PR China.
| |
Collapse
|
2
|
Uder C, Brückner S, Winkler S, Tautenhahn HM, Christ B. Mammalian MSC from selected species: Features and applications. Cytometry A 2017; 93:32-49. [PMID: 28906582 DOI: 10.1002/cyto.a.23239] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal/stem cells (MSC) are promising candidates for cellular therapy of different diseases in humans and in animals. Following the guidelines of the International Society for Cell Therapy, human MSC may be identified by expression of a specific panel of cell surface markers (CD105+, CD73+, CD90+, CD34-, CD14-, or CD11b-, CD79- or CD19-, HLA-DR-). In addition, multiple differentiation potential into at least the osteogenic, adipogenic, and chondrogenic lineage is a main criterion for MSC definition. Human MSC and MSC of a variety of mammals isolated from different tissues meet these criteria. In addition to the abovementioned, they express many more cell surface markers. Yet, these are not uniquely expressed by MSC. The gross phenotypic appearance like marker expression and differentiation potential is similar albeit not identical for MSC from different tissues and species. Similarly, MSC may feature different biological characteristics depending on the tissue source and the isolation and culture procedures. Their versatile biological qualities comprising immunomodulatory, anti-inflammatory, and proregenerative capacities rely largely on the migratory and secretory capabilities of MSC. They are attracted to sites of tissue lesion and secrete factors to promote self-repair of the injured tissue. This is a big perspective for clinical MSC applications in both veterinary and human medicine. Phase I/II clinical trials have been initiated to assess safety and feasibility of MSC therapies in acute and chronic disease settings. Yet, since the mode of MSC action in a specific disease environment is still unknown at large, it is mandatory to unravel the response of MSC from a given source onto a specific disease environment in suitable animal models prior to clinical applications. © 2017 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Christiane Uder
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Applied Molecular Hepatology Laboratory, University Hospital of Leipzig, Liebigstraße 21, Leipzig D-04103, Germany
| | - Sandra Brückner
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Applied Molecular Hepatology Laboratory, University Hospital of Leipzig, Liebigstraße 21, Leipzig D-04103, Germany
| | - Sandra Winkler
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Applied Molecular Hepatology Laboratory, University Hospital of Leipzig, Liebigstraße 21, Leipzig D-04103, Germany
| | - Hans-Michael Tautenhahn
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Applied Molecular Hepatology Laboratory, University Hospital of Leipzig, Liebigstraße 21, Leipzig D-04103, Germany
| | | |
Collapse
|
3
|
Sang JF, Shi XL, Han B, Huang T, Huang X, Ren HZ, Ding YT. Intraportal mesenchymal stem cell transplantation prevents acute liver failure through promoting cell proliferation and inhibiting apoptosis. Hepatobiliary Pancreat Dis Int 2016; 15:602-611. [PMID: 27919849 DOI: 10.1016/s1499-3872(16)60141-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Transplantation of mesenchymal stem cells (MSCs) has been regarded as a potential treatment for acute liver failure (ALF), but the optimal route was unknown. The present study aimed to explore the most effective MSCs transplantation route in a swine ALF model. METHODS The swine ALF model induced by intravenous injection of D-Gal was treated by the transplantation of swine MSCs through four routes including intraportal injection (InP group), hepatic intra-arterial injection (AH group), peripheral intravenous injection (PV group) and intrahepatic injection (IH group). The living conditions and survival time were recorded. Blood samples before and after MSCs transplantation were collected for the analysis of hepatic function. The histology of liver injury was interpreted and scored in terminal samples. Hepatic apoptosis was detected by TUNEL assay. Apoptosis and proliferation related protein expressions including cleaved caspase-3, survivin, AKT, phospho-AKT (Ser473), ERK and phospho-ERK (Tyr204) were analyzed by Western blotting. RESULTS The average survival time of each group was 10.7+/-1.6 days (InP), 6.0+/-0.9 days (AH), 4.7+/-1.4 days (PV), 4.3+/-0.8 days (IH), respectively, when compared with the average survival time of 3.8+/-0.8 days in the D-Gal group. The survival rates between the InP group and D-Gal group revealed a statistically significant difference (P<0.01). Pathological and biochemical analysis showed that liver damage was the worst in the D-Gal group, while less injury in the InP group. Histopathological scores revealed a significant decrease in the InP group (3.17+/-1.04, P<0.01) and AH group (8.17+/-0.76, P<0.05) as compared with that in the D-Gal group (11.50+/-1.32). The apoptosis rate in the InP group (25.0%+/-3.4%, P<0.01) and AH group (40.5%+/-1.0%, P<0.05) was lower than that in the D-Gal group (70.6%+/-8.5%). The expression of active caspase-3 was inhibited, while the expression of survivin, AKT, phospho-AKT (Ser473), ERK and phospho-ERK (Tyr204) was elevated in the InP group. CONCLUSIONS Intraportal injection was superior to other pathways for MSC transplantation. Intraportal MSC transplantation could improve liver function, inhibit apoptosis and prolong the survival time of swine with ALF. The transplanted MSCs may participate in liver regeneration via promoting cell proliferation and suppressing apoptosis during the initial stage of ALF.
Collapse
Affiliation(s)
- Jian-Feng Sang
- Department of General Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China.
| | | | | | | | | | | | | |
Collapse
|
4
|
Kakisaka K, Kataoka K, Onodera M, Suzuki A, Endo K, Tatemichi Y, Kuroda H, Ishida K, Takikawa Y. Alpha-fetoprotein: A biomarker for the recruitment of progenitor cells in the liver in patients with acute liver injury or failure. Hepatol Res 2015; 45:E12-20. [PMID: 25376981 DOI: 10.1111/hepr.12448] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Revised: 10/28/2014] [Accepted: 10/28/2014] [Indexed: 12/12/2022]
Abstract
AIM The optimal conditions for hepatocyte proliferation should be clarified in an attempt to improve the impaired liver regeneration observed in patients with acute liver failure (ALF). In order to evaluate the significance of the serum α-fetoprotein (AFP). level and prothrombin time international normalized ratio (PT-INR) as possible biomarkers of the proliferation of liver stem/progenitor cells (LPC) and mature hepatocytes (MH), respectively, we focused on donors of living donor liver transplantation (LDLT) and patients with acute liver injury (ALI), including ALF. METHODS Seventy-three patients with ALI/ALF and 11 donors for LDLT were evaluated. LPC induction was histologically evaluated using cytokeratin (CK)-7 staining in 45 ALI/ALF patients. RESULTS The AFP level was not apparently elevated during the observation period in any of the LDLT donors, whereas the serum AFP levels were substantially increased in the patients with ALI/ALF and significantly correlated with the number of CK-7 positive LPC in the liver, except for very severe damaged liver. All patients exhibiting an early peak in the AFP level prior to PT-INR elevation died. CONCLUSION The serum AFP level may reflect the induction of LPC in ALI/ALF patients. The substantial and persistent induction of LPC until sufficient regeneration of MH may be needed for a recovery from ALF. We herein demonstrate that the serum AFP level may be a serum marker of LPC in patients with ALI/ALF. A comparison of the serial changes in the AFP levels and PT-INR in our study patients showed impaired proliferation of LPC and delayed recovery of MH in the patients who died.
Collapse
Affiliation(s)
- Keisuke Kakisaka
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Japan
| | - Kojiro Kataoka
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Japan
| | - Mio Onodera
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Japan
| | - Akiko Suzuki
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Japan
| | - Kei Endo
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Japan
| | - Yoshinori Tatemichi
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Japan
| | - Hidekatsu Kuroda
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Japan
| | - Kazuyuki Ishida
- Department of Molecular Diagnostic Pathology, Iwate Medical University, Morioka, Japan
| | - Yasuhiro Takikawa
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University, Morioka, Japan
| |
Collapse
|
5
|
Liu Z, Meng F, Li C, Zhou X, Zeng X, He Y, Mrsny RJ, Liu M, Hu X, Hu JF, Li T. Human umbilical cord mesenchymal stromal cells rescue mice from acetaminophen-induced acute liver failure. Cytotherapy 2015; 16:1207-19. [PMID: 25108650 DOI: 10.1016/j.jcyt.2014.05.018] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 04/25/2014] [Accepted: 05/20/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND AIMS Acute liver failure (ALF), a life-threatening disease characterized by the sudden loss of hepatic function, can occur after an accidental or intentional acetaminophen overdose. METHODS With the use of an ALF mouse model, we examined both the preventive and therapeutic potential of intravenously administered human umbilical cord-derived mesenchymal stromal cells (hUCMSCs). Primary hUCMSCs were purified from freshly collected full-term umbilical cords and intravenously transplanted into BALB/c mice either before and after ALF induced by acetaminophen intoxication. We found that hUCMSCs significantly improved survival rates and relative liver weight of mice in both pre-ALF and post-ALF animals. Correspondingly, serum levels of markers that reflect hepatic injury (ie, aspartate aminotransferase, alanine aminotransferase and total bilirubin) were significantly attenuated in the group receiving hUCMSC therapy. RESULTS Mechanistically, we found that the protective potential of intravenously administered hUCMSCs was mediated by paracrine pathways that involved antioxidants (glutathione, superoxide dismutase), the reduction of inflammatory agents (tumor necrosis factor-α, interleukin-6) and elevated serum levels of hepatocyte growth factor. CONCLUSIONS Through these paracrine effects, intravenously administered hUCMSCs reduced hepatic necrosis/apoptosis and enhanced liver regeneration. Thus, our data demonstrate that intravenously administered hUCMSCs may be useful in the prevention or treatment of acetaminophen-induced ALF.
Collapse
Affiliation(s)
- Zongcai Liu
- Shenzhen Beike Cell Engineering Research Institute, Yuanxing Science and Technology Building, Nanshan, Shenzhen, PR China
| | - Fanwei Meng
- Shenzhen Beike Cell Engineering Research Institute, Yuanxing Science and Technology Building, Nanshan, Shenzhen, PR China
| | - Chan Li
- Shenzhen Beike Cell Engineering Research Institute, Yuanxing Science and Technology Building, Nanshan, Shenzhen, PR China
| | - Xin Zhou
- Shenzhen Beike Cell Engineering Research Institute, Yuanxing Science and Technology Building, Nanshan, Shenzhen, PR China
| | - Xiaoping Zeng
- Shenzhen Beike Cell Engineering Research Institute, Yuanxing Science and Technology Building, Nanshan, Shenzhen, PR China
| | - Yixin He
- Shenzhen Beike Cell Engineering Research Institute, Yuanxing Science and Technology Building, Nanshan, Shenzhen, PR China
| | - Randall J Mrsny
- GMR Epigenetics, Palo Alto, California, USA; Department of Pharmacy & Pharmacology, University of Bath, Bath, England
| | - Muyun Liu
- Shenzhen Beike Cell Engineering Research Institute, Yuanxing Science and Technology Building, Nanshan, Shenzhen, PR China
| | - Xiang Hu
- Shenzhen Beike Cell Engineering Research Institute, Yuanxing Science and Technology Building, Nanshan, Shenzhen, PR China
| | - Ji-Fan Hu
- VA Palo Alto Health Care System, Stanford University Medical School, Palo Alto, California, USA; GMR Epigenetics, Palo Alto, California, USA.
| | - Tao Li
- Shenzhen Beike Cell Engineering Research Institute, Yuanxing Science and Technology Building, Nanshan, Shenzhen, PR China.
| |
Collapse
|
6
|
Stock P, Brückner S, Winkler S, Dollinger MM, Christ B. Human bone marrow mesenchymal stem cell-derived hepatocytes improve the mouse liver after acute acetaminophen intoxication by preventing progress of injury. Int J Mol Sci 2014; 15:7004-28. [PMID: 24758938 PMCID: PMC4013675 DOI: 10.3390/ijms15047004] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 04/02/2014] [Accepted: 04/09/2014] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells from human bone marrow (hMSC) have the potential to differentiate into hepatocyte-like cells in vitro and continue to maintain important hepatocyte functions in vivo after transplantation into host mouse livers. Here, hMSC were differentiated into hepatocyte-like cells in vitro (hMSC-HC) and transplanted into livers of immunodeficient Pfp/Rag2⁻/⁻ mice treated with a sublethal dose of acetaminophen (APAP) to induce acute liver injury. APAP induced a time- and dose-dependent damage of perivenous areas of the liver lobule. Serum levels of aspartate aminotransferase (AST) increased to similar levels irrespective of hMSC-HC transplantation. Yet, hMSC-HC resided in the damaged perivenous areas of the liver lobules short-term preventing apoptosis and thus progress of organ destruction. Disturbance of metabolic protein expression was lower in the livers receiving hMSC-HC. Seven weeks after APAP treatment, hepatic injury had completely recovered in groups both with and without hMSC-HC. Clusters of transplanted cells appeared predominantly in the periportal portion of the liver lobule and secreted human albumin featuring a prominent quality of differentiated hepatocytes. Thus, hMSC-HC attenuated the inflammatory response and supported liver regeneration after acute injury induced by acetaminophen. They hence may serve as a novel source of hepatocyte-like cells suitable for cell therapy of acute liver diseases.
Collapse
Affiliation(s)
- Peggy Stock
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Applied Molecular Hepatology Laboratory, University Hospital Leipzig, Liebigstraße 21, D-04103 Leipzig, Germany.
| | - Sandra Brückner
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Applied Molecular Hepatology Laboratory, University Hospital Leipzig, Liebigstraße 21, D-04103 Leipzig, Germany.
| | - Sandra Winkler
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Applied Molecular Hepatology Laboratory, University Hospital Leipzig, Liebigstraße 21, D-04103 Leipzig, Germany.
| | - Matthias M Dollinger
- Clinics for Internal Medicine I, University Hospital Ulm, Albert-Einstein-Allee 23, D-89081 Ulm, Germany.
| | - Bruno Christ
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Applied Molecular Hepatology Laboratory, University Hospital Leipzig, Liebigstraße 21, D-04103 Leipzig, Germany.
| |
Collapse
|
7
|
Sowa JP, Gerken G, Canbay A. From bedside to bench and back again-molecular mechanisms in acute liver failure. Front Physiol 2014; 5:18. [PMID: 24523697 PMCID: PMC3906497 DOI: 10.3389/fphys.2014.00018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 01/09/2014] [Indexed: 11/25/2022] Open
Affiliation(s)
- Jan-Peter Sowa
- Department of Gastroenterology and Hepatology, University Hospital Essen, University Duisburg-Essen Essen, Germany
| | - Guido Gerken
- Department of Gastroenterology and Hepatology, University Hospital Essen, University Duisburg-Essen Essen, Germany
| | - Ali Canbay
- Department of Gastroenterology and Hepatology, University Hospital Essen, University Duisburg-Essen Essen, Germany
| |
Collapse
|
8
|
Brückner S, Tautenhahn HM, Winkler S, Stock P, Dollinger M, Christ B. A fat option for the pig: hepatocytic differentiated mesenchymal stem cells for translational research. Exp Cell Res 2013; 321:267-75. [PMID: 24200501 DOI: 10.1016/j.yexcr.2013.10.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 10/21/2013] [Accepted: 10/27/2013] [Indexed: 02/07/2023]
Abstract
STUDY BACKGROUND Extended liver resection is the only curative treatment option of liver cancer. Yet, the residual liver may not accomplish the high metabolic and regenerative capacity needed, which frequently leads to acute liver failure. Because of their anti-inflammatory and -apoptotic as well as pro-proliferative features, mesenchymal stem cells differentiated into hepatocyte-like cells might provide functional and regenerative compensation. Clinical translation of basic research requires pre-clinical approval in large animals. Therefore, we characterized porcine mesenchymal stem cells (MSC) from adipose tissue and bone marrow and their hepatocyte differentiation potential for future assessment of functional liver support after surgical intervention in the pig model. METHODS Mesenchymal surface antigens and multi-lineage differentiation potential of porcine MSC isolated by collagenase digestion either from bone marrow or adipose tissue (subcutaneous/visceral) were assessed by flow cytometry. Morphology and functional properties (urea-, glycogen synthesis and cytochrome P450 activity) were determined during culture under differentiation conditions and compared with primary porcine hepatocytes. RESULTS MSC from porcine adipose tissue and from bone marrow express the typical mesenchymal markers CD44, CD29, CD90 and CD105 but not haematopoietic markers. MSC from both sources displayed differentiation into the osteogenic as well as adipogenic lineage. After hepatocyte differentiation, expression of CD105 decreased significantly and cells adopted the typical polygonal morphology of hepatocytes. Glycogen storage was comparable in adipose tissue- and bone marrow-derived cells. Urea synthesis was about 35% lower in visceral than in subcutaneous adipose tissue-derived MSC. Cytochrome P450 activity increased significantly during differentiation and was twice as high in hepatocyte-like cells generated from bone marrow as from adipose tissue. CONCLUSION The hepatocyte differentiation of porcine adipose tissue-derived MSC was shown for the first time yielding hepatocyte-like cells with specific functions similar in bone marrow and subcutaneous adipose tissue-derived MSC. That makes them good pre-clinical candidates for supportive approaches after liver resection in the pig.
Collapse
Affiliation(s)
- Sandra Brückner
- University Hospital Leipzig, Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Liebigstraße 21, Leipzig D-04103, Germany.
| | - Hans-Michael Tautenhahn
- University Hospital Leipzig, Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Liebigstraße 21, Leipzig D-04103, Germany; TRM, Translational Centre for Regenerative Medicine, Philipp-Rosenthal-Str. 55, Leipzig D-04103, Germany.
| | - Sandra Winkler
- University Hospital Leipzig, Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Liebigstraße 21, Leipzig D-04103, Germany.
| | - Peggy Stock
- University Hospital Leipzig, Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Liebigstraße 21, Leipzig D-04103, Germany.
| | - Matthias Dollinger
- University Hospital Ulm, First Department of Medicine, Albert-Einstein-Allee 23, Ulm D-89081, Germany.
| | - Bruno Christ
- University Hospital Leipzig, Department of Visceral, Transplantation, Thoracic and Vascular Surgery, Liebigstraße 21, Leipzig D-04103, Germany; TRM, Translational Centre for Regenerative Medicine, Philipp-Rosenthal-Str. 55, Leipzig D-04103, Germany.
| |
Collapse
|
9
|
Esrefoglu M. Role of stem cells in repair of liver injury: Experimental and clinical benefit of transferred stem cells on liver failure. World J Gastroenterol 2013; 19:6757-6773. [PMID: 24187451 PMCID: PMC3812475 DOI: 10.3748/wjg.v19.i40.6757] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 07/23/2013] [Accepted: 08/20/2013] [Indexed: 02/06/2023] Open
Abstract
Although the liver has a high regenerative capacity, as a result of massive hepatocyte death, liver failure occurs. In addition to liver failure, for acute, chronic and hereditary diseases of the liver, cell transplantation therapies can stimulate regeneration or at least ensure sufficient function until liver transplantation can be performed. The lack of donor organs and the risks of rejection have prompted extensive experimental and clinical research in the field of cellular transplantation. Transplantation of cell lineages involved in liver regeneration, including mature hepatocytes, fetal hepatocytes, fetal liver progenitor cells, fetal stem cells, hepatic progenitor cells, hepatic stem cells, mesenchymal stem cells, hematopoietic stem cells, and peripheral blood and umbilical cord blood stem cells, have been found to be beneficial in the treatment of liver failure. In this article, the results of experimental and clinical cell transplantation trials for liver failure are reviewed, with an emphasis on regeneration.
Collapse
|
10
|
Martínez-Herrero S, Larráyoz IM, Ochoa-Callejero L, García-Sanmartín J, Martínez A. Adrenomedullin as a growth and cell fate regulatory factor for adult neural stem cells. Stem Cells Int 2012; 2012:804717. [PMID: 23049570 PMCID: PMC3462413 DOI: 10.1155/2012/804717] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 08/16/2012] [Accepted: 08/21/2012] [Indexed: 01/10/2023] Open
Abstract
The use of stem cells as a strategy for tissue repair and regeneration is one of the biomedical research areas that has attracted more interest in the past few years. Despite the classic belief that the central nervous system (CNS) was immutable, now it is well known that cell turnover occurs in the mature CNS. Postnatal neurogenesis is subjected to tight regulation by many growth factors, cell signals, and transcription factors. An emerging molecule involved in this process is adrenomedullin (AM). AM, a 52-amino acid peptide which exerts a plethora of physiological functions, acts as a growth and cell fate regulatory factor for adult neural stem and progenitor cells. AM regulates the proliferation rate and the differentiation into neurons, astrocytes, and oligodendrocytes of stem/progenitor cells, probably through the PI3K/Akt pathway. The active peptides derived from the AM gene are able to regulate the cytoskeleton dynamics, which is extremely important for mature neural cell morphogenesis. In addition, a defective cytoskeleton may impair cell cycle and migration, so AM may contribute to neural stem cell growth regulation by allowing cells to pass through mitosis. Regulation of AM levels may contribute to program stem cells for their use in medical therapies.
Collapse
Affiliation(s)
| | - Ignacio M. Larráyoz
- Oncology Area, Center for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, Spain
| | - Laura Ochoa-Callejero
- Oncology Area, Center for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, Spain
| | | | - Alfredo Martínez
- Oncology Area, Center for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, Spain
| |
Collapse
|
11
|
Christ B, Stock P. Mesenchymal stem cell-derived hepatocytes for functional liver replacement. Front Immunol 2012; 3:168. [PMID: 22737154 PMCID: PMC3381218 DOI: 10.3389/fimmu.2012.00168] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 06/04/2012] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells represent an alternate cell source to substitute for primary hepatocytes in hepatocyte transplantation because of their multiple differentiation potential and nearly unlimited availability. They may differentiate into hepatocyte-like cells in vitro and maintain specific hepatocyte functions also after transplantation into the regenerating livers of mice or rats both under injury and non-injury conditions. Depending on the underlying liver disease their mode of action is either to replace the diseased liver tissue or to support liver regeneration through their anti-inflammatory and anti-apoptotic as well as their pro-proliferative action.
Collapse
Affiliation(s)
- Bruno Christ
- Applied Molecular Hepatology Laboratory, Department of Visceral, Transplantation, Thoracic and Vascular Surgery, University Hospital Leipzig Leipzig, Germany
| | | |
Collapse
|