51
|
Nakamura T, Torimura T, Iwamoto H, Kurogi J, Inoue H, Hori Y, Sumie S, Fukushima N, Sakata M, Koga H, Abe M, Ikezono Y, Hashimoto O, Ueno T, Oho K, Okamura T, Okuda S, Kawamoto A, Ii M, Asahara T, Sata M. CD34(+) cell therapy is safe and effective in slowing the decline of hepatic reserve function in patients with decompensated liver cirrhosis. J Gastroenterol Hepatol 2014; 29:1830-8. [PMID: 24731186 DOI: 10.1111/jgh.12622] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/28/2014] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND AIM Preclinical studies in rodent models of chronic liver fibrosis have shown that transplantation of peripheral blood (PB) CD34(+) cells leads to hepatic regeneration and a reduction of liver fibrosis by suppressing hepatic stellate cell activity and increasing matrix metalloproteinase activity. The aim of this study was to examine the safety and clinical efficacy of intrahepatic transplantation of autologous granulocyte colony-stimulating factor (G-CSF)-mobilized PB-CD34(+) cells in patients with decompensated liver cirrhosis. METHODS PB-CD34(+) cells were isolated from G-CSF-mobilized apheresis products. Ten patients were treated with G-CSF-mobilized PB-CD34(+) cells (treatment group) and seven patients were treated with standard medical therapy. For mobilization, patients in the treatment group received subcutaneous injections of 10 μg G-CSF/kg/day for 5 days. The cells were then injected at three different doses (5 × 10(5) , 1 × 10(6) and 2 × 10(6) cells/kg) through the hepatic artery. Thereafter, all patients were followed up for 24 months. RESULTS G-CSF treatment and leukapheresis were well tolerated, and no serious adverse events were observed. Patients in the treatment group had a significant but transient splenomegaly. After 24 weeks, serum albumin was significantly increased in patients who had received middle or high doses of CD34(+) cells compared with baseline. Doppler ultrasound showed a significant increase in hepatic blood flow velocity and blood flow volume after CD34(+) cell therapy. The hepatic vein pressure gradient decreased in two patients who received high-dose CD34(+) cells at week 16. CONCLUSIONS CD34(+) cell therapy is feasible, safe and effective in slowing the decline of hepatic reserve function.
Collapse
Affiliation(s)
- Toru Nakamura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan; Liver Cancer Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
52
|
Behbahan IS, Keating A, Gale RP. Concise review: bone marrow autotransplants for liver disease? Stem Cells 2014; 31:2313-29. [PMID: 23939914 DOI: 10.1002/stem.1510] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 07/08/2013] [Accepted: 07/15/2013] [Indexed: 12/11/2022]
Abstract
There are increasing reports of using bone marrow-derived stem cells to treat advanced liver disease. We consider several critical issues that underlie this approach. For example, are there multipotent stem cell populations in human adult bone marrow? Can they develop into liver cells or supporting cell types? What are stromal stem/progenitor cells, and can they promote tissue repair without replacing hepatocytes? Does reversal of end-stage liver disease require new hepatocytes, a new liver microenvironment, both, neither or something else? Although many of these questions are unanswered, we consider the conceptual and experimental bases underlying these issues and critically analyze results of clinical trials of stem cell therapy of end-stage liver disease.
Collapse
Affiliation(s)
- Iman Saramipoor Behbahan
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | | | | |
Collapse
|
53
|
Singh V, Sharma AK, Narasimhan RL, Bhalla A, Sharma N, Sharma R. Granulocyte colony-stimulating factor in severe alcoholic hepatitis: a randomized pilot study. Am J Gastroenterol 2014; 109:1417-23. [PMID: 24935272 DOI: 10.1038/ajg.2014.154] [Citation(s) in RCA: 177] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 04/17/2014] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Severe alcoholic hepatitis has high short-term mortality. The aim of this study was to test the hypothesis that treatment of patients with alcoholic hepatitis with granulocyte colony-stimulating factor (G-CSF) might mobilize bone marrow-derived stem cells and promote hepatic regeneration and thus improve survival. METHODS Forty-six patients with severe alcoholic hepatitis were prospectively randomized in an open study to standard medical therapy (SMT) plus G-CSF (group A; n=23) at a dose of 5 μg/kg subcutaneously every 12 h for 5 consecutive days or to SMT alone (group B; n=23) at a tertiary care center. We assessed the mobilization of CD34(+) cells on day 6, Child-Turcotte-Pugh (CTP), model for end-stage liver disease (MELD), and modified Maddrey's discriminant function (mDF) scores, and survival until day 90. RESULTS There was a statistically significant increase in the number of CD34(+) cells in peripheral blood in group A as compared with group B (P=0.019) after 5 days of G-GSF therapy. There was a significant reduction in median Δ change% in CTP, MELD, and mDF at 1, 2, and 3 months in group A as compared with group B (P<0.05). There was marked improvement in survival in group A as compared with group B (78.3% vs. 30.4%; P=0.001) at 90 days. CONCLUSIONS G-CSF is safe and effective in the mobilization of hematopoietic stem cells and improves liver function as well as survival in patients with severe alcoholic hepatitis.
Collapse
Affiliation(s)
- Virendra Singh
- Department of Hepatology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Arun K Sharma
- Department of Hepatology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - R Lakshmi Narasimhan
- Department of Internal Medicine, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Ashish Bhalla
- Department of Internal Medicine, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Navneet Sharma
- Department of Internal Medicine, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Ratiram Sharma
- Department of Transfusion Medicine, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| |
Collapse
|
54
|
Margini C, Vukotic R, Brodosi L, Bernardi M, Andreone P. Bone marrow derived stem cells for the treatment of end-stage liver disease. World J Gastroenterol 2014; 20:9098-9105. [PMID: 25083082 PMCID: PMC4112892 DOI: 10.3748/wjg.v20.i27.9098] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 02/27/2014] [Accepted: 04/15/2014] [Indexed: 02/06/2023] Open
Abstract
End-stage disease due to liver cirrhosis is an important cause of death worldwide. Cirrhosis results from progressive, extensive fibrosis and impaired hepatocyte regeneration. The only curative treatment is liver transplantation, but due to the several limitations of this procedure, the interest in alternative therapeutic strategies is increasing. In particular, the potential of bone marrow stem cell (BMSC) therapy in cirrhosis has been explored in different trials. In this article, we evaluate the results of 18 prospective clinical trials, and we provide a descriptive overview of recent advances in the research on hepatic regenerative medicine. The main message from the currently available data in the literature is that BMSC therapy is extremely promising in the context of liver cirrhosis. However, its application should be further explored in randomized, controlled trials with large cohorts and long follow-ups.
Collapse
|
55
|
Abstract
Liver disease is a rising cause of mortality and morbidity, and treatment options remain limited. Liver transplantation is curative but limited by donor organ availability, operative risk and long-term complications. The contribution of bone marrow (BM)-derived stem cells to tissue regeneration has been recognised and there is considerable interest in the potential benefits of BM stem cells in patients with liver disease. In chronic liver disease, deposition of fibrous scar tissue inhibits hepatocyte proliferation and leads to portal hypertension. Although initial reports had suggested transdifferentiation of stem cells into hepatocytes, the beneficial effects of BM stem cells are more likely derived from the ability to breakdown scar tissue and stimulate hepatocyte proliferation. Studies in animal models have yielded promising results, although the exact mechanisms and cell type responsible have yet to be determined. Small-scale clinical studies have quickly followed and, although primarily designed to examine safety and feasibility of this approach, have reported improvements in liver function in treated patients. Well-designed, controlled studies are required to fully determine the benefits of BM stem cell therapy.
Collapse
Affiliation(s)
- Andrew King
- NIHR Liver Biomedical Research Unit and Centre for Liver Research, University of Birmingham, Birmingham, UK
| | | |
Collapse
|
56
|
Tsolaki E, Athanasiou E, Gounari E, Zogas N, Siotou E, Yiangou M, Anagnostopoulos A, Yannaki E. Hematopoietic stem cells and liver regeneration: differentially acting hematopoietic stem cell mobilization agents reverse induced chronic liver injury. Blood Cells Mol Dis 2014; 53:124-32. [PMID: 24923531 DOI: 10.1016/j.bcmd.2014.05.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 02/02/2014] [Accepted: 02/03/2014] [Indexed: 12/20/2022]
Abstract
Bone marrow (BM) could serve as a source of cells facilitating liver repopulation in case of hepatic damage. Currently available hematopoietic stem cell (HSC) mobilizing agents, were comparatively tested for healing potential in liver fibrosis. Carbon tetrachloride (CCl4)-injured mice previously reconstituted with Green Fluorescent Protein BM were mobilized with Granulocyte-Colony Stimulating Factor (G-CSF), Plerixafor or G-CSF+Plerixafor. Hepatic fibrosis, stellate cell activation and oval stem cell frequency were measured by Gomori and by immunohistochemistry for a-Smooth Muscle Actin and Cytokeratin-19, respectively. Angiogenesis was evaluated by ELISA and immunohistochemistry. Quantitative real-time PCR was used to determine the mRNA levels of liver Peroxisome Proliferator-Activated Receptor gamma (PPAR-γ), Interleukin-6 (IL-6) and Tumor Necrosis-alpha (TNFα). BM-derived cells were tracked by double immunofluorescence. The spontaneous migration of mobilized HSCs towards injured liver and its cytokine secretion profile was determined in transwell culture systems. Either single-agent mobilization or the combination of agents significantly ameliorated hepatic damage by decreasing fibrosis and restoring the abnormal vascular network in the liver of mobilized mice compared to CCl4-only mice. The degree of fibrosis reduction was similar among all mobilized mice despite that G-CSF+Plerixafor yielded significantly higher numbers of circulating HSCs over other agents. The liver homing potential of variously mobilized HSCs differed among the agents. An extended G-CSF treatment provided the highest anti-fibrotic effect over all tested modalities, induced by the proliferation of hepatic stem cells and decreased hepatic inflammation. Plerixafor-mobilized HSCs, despite their reduced liver homing potential, reversed fibrosis mainly by increasing hepatic PPAR-γ and VEGF expression. In all groups, BM-derived mature hepatocytes as well as liver-committed BM stem cells were detected only at low frequencies, further supporting the concept that alternative mechanisms rather than direct HSC effects regulate liver recovery. Overall, our data suggest that G-CSF, Plerixafor and G-CSF+Plerixafor act differentially during the wound healing process, ultimately providing a potent anti-fibrotic effect.
Collapse
Affiliation(s)
- Eleftheria Tsolaki
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece; Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evangelia Athanasiou
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece
| | - Eleni Gounari
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece; Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Zogas
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece; Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eleni Siotou
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece
| | - Minas Yiangou
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Achilles Anagnostopoulos
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece
| | - Evangelia Yannaki
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki, Greece.
| |
Collapse
|
57
|
Abstract
The liver has a unique capacity to repair following injury, which is largely achieved by proliferation of hepatocytes. However, in situations of chronic or overwhelming liver injury, additional repair mechanisms, namely liver progenitor or oval cells, are activated. These cells, located in the canals of Hering, express markers for both hepatocytes and biliary cells and have the capacity to differentiate down both hepatocyte and biliary lineages. Previous work has suggested that the administration of autologous or allogeneic cell therapies such as haematopoietic or mesenchymal stem cells can augment liver repair by either stimulating endogenous repair mechanisms or by suppressing ongoing damage. A better understanding of how cell therapies can promote liver regeneration will lead to the refinement of these therapeutic approaches and also develop new pharmacological agents for liver repair.
Collapse
Affiliation(s)
- N N Than
- From the Centre for Liver Research & NIHR Biomedical Research Unit in Liver Diseases, University of Birmingham, Birmingham, UK
| | - P N Newsome
- From the Centre for Liver Research & NIHR Biomedical Research Unit in Liver Diseases, University of Birmingham, Birmingham, UK
| |
Collapse
|
58
|
Kedarisetty CK, Anand L, Khanam A, Kumar A, Rastogi A, Maiwall R, Sarin SK. Growth factors enhance liver regeneration in acute-on-chronic liver failure. Hepatol Int 2014. [PMID: 26201333 DOI: 10.1007/s12072-014-9538-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Acute-on-chronic liver failure is a distinct syndrome characterized by a rapid progression of liver disease culminating in organ failure and death. The only definitive treatment is liver transplantation. However, there is a possible element of reversibility and hepatic regeneration if the acute insult can be tided over. Exogenously administered growth factors may stimulate hepatocytes, hepatic progenitor cells and bone marrow-derived cells to supplement hepatic regeneration. The proposed review is intended to provide an in-depth analysis of the individual components of hepatic and bone marrow niches and highlight the growing role of various growth factors in liver regeneration in health and in liver failure.
Collapse
Affiliation(s)
| | - Lovkesh Anand
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Arshi Khanam
- Department of Research, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Anupam Kumar
- Department of Research, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Archana Rastogi
- Department of Pathology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Rakhi Maiwall
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Shiv Kumar Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India.
| |
Collapse
|
59
|
Ryu KH. Liver stem cells derived from the bone marrow and umbilical cord blood. Int J Stem Cells 2014; 2:97-101. [PMID: 24855527 DOI: 10.15283/ijsc.2009.2.2.97] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2009] [Indexed: 02/06/2023] Open
Affiliation(s)
- Kyung Ha Ryu
- Department of Pediatrics, College of Medicine, Ewha womans University, Seoul, Korea
| |
Collapse
|
60
|
Huebert RC, Rakela J. Cellular therapy for liver disease. Mayo Clin Proc 2014; 89:414-24. [PMID: 24582199 PMCID: PMC4212517 DOI: 10.1016/j.mayocp.2013.10.023] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 10/28/2013] [Accepted: 10/30/2013] [Indexed: 12/19/2022]
Abstract
Regenerative medicine is energizing and empowering basic science and has the potential to dramatically transform health care in the future. Given the remarkable intrinsic regenerative properties of the liver, as well as widespread adoption of regenerative strategies for liver disease (eg, liver transplant, partial hepatectomy, living donor transplant), hepatology has always been at the forefront of clinical regenerative medicine. However, an expanding pool of patients awaiting liver transplant, a limited pool of donor organs, and finite applicability of the current surgical approaches have created a need for more refined and widely available regenerative medicine strategies. Although cell-based therapies have been used extensively for hematologic malignant diseases and other conditions, the potential application of cellular therapy for acute and chronic liver diseases has only more recently been explored. New understanding of the mechanisms of liver regeneration and repair, including activation of local stem/progenitor cells and contributions from circulating bone marrow-derived stem cells, provide the theoretical underpinnings for the rational use of cell-based therapies in clinical trials. In this review, we dissect the scientific rationale for various modalities of cell therapy for liver diseases being explored in animal models and review those tested in human clinical trials. We also attempt to clarify some of the important ongoing questions that need to be addressed in order to bring these powerful therapies to clinical translation. Discussions will cover transplant of hepatocytes and liver stem/progenitor cells as well as infusion or stimulation of bone marrow-derived stem cells. We also highlight tremendous scientific advances on the horizon, including the potential use of induced pluripotent stem cells and their derivatives as individualized regenerative therapy for liver disease.
Collapse
Affiliation(s)
| | - Jorge Rakela
- Division of Hepatology, Mayo Clinic, Phoenix, AZ
| |
Collapse
|
61
|
Clinical Outcome of Autologous Hematopoietic Stem Cell Infusion via Hepatic Artery or Portal Vein in Patients with End-stage Liver Diseases. ACTA ACUST UNITED AC 2014; 29:15-22. [DOI: 10.1016/s1001-9294(14)60018-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
62
|
Zheng L, Chu J, Shi Y, Zhou X, Tan L, Li Q, Cui L, Han Z, Han Y, Fan D. Bone marrow-derived stem cells ameliorate hepatic fibrosis by down-regulating interleukin-17. Cell Biosci 2013; 3:46. [PMID: 24314294 PMCID: PMC3882099 DOI: 10.1186/2045-3701-3-46] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Accepted: 10/28/2013] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Accumulating evidences have identified the immunoregulatory features of stem cells. In this study, the immunoregulation of bone marrow-derived stem cells (BMSCs) transplanted into patients with HBV-related decompensated cirrhosis and mouse model of liver injury induced by carbon tetrachloride (CCl4) administration was observed. RESULTS Compared with healthy controls, patients with HBV-related decompensated cirrhosis showed significantly higher levels of TNF-alpha, IL-12, TGF-beta1, IL-17, and IL-8. However, only IL-17 was markedly decreased after autologous BMSCs transplantation during their follow-up. The same results were found in the CCl4-treated mice. Furthermore, we found that exogenous IL-17 partly abolished the therapeutic effect of BMSCs whereas IL-17-specific antibody promoted improvement of liver injury in CCl4-treated mice, resembling the therapeutic effect of BMSCs transplantation. CONCLUSIONS These data suggested that BMSCs transplantation induces a decrease of IL-17 level, which at least in part delineates the mechanisms of stem cells-mediated therapeutic benefit on liver disease.
Collapse
Affiliation(s)
- Linhua Zheng
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Disease, Xijing Hospital, Fourth Military Medical University, 127 Changle Western Road, Xi'an 710032, Shaanxi Province, China
| | - Jindong Chu
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Disease, Xijing Hospital, Fourth Military Medical University, 127 Changle Western Road, Xi'an 710032, Shaanxi Province, China
| | - Yongquan Shi
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Disease, Xijing Hospital, Fourth Military Medical University, 127 Changle Western Road, Xi'an 710032, Shaanxi Province, China
| | - Xinmin Zhou
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Disease, Xijing Hospital, Fourth Military Medical University, 127 Changle Western Road, Xi'an 710032, Shaanxi Province, China
| | - Ling Tan
- Department of Pediatric Dentistry, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Qiang Li
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Disease, Xijing Hospital, Fourth Military Medical University, 127 Changle Western Road, Xi'an 710032, Shaanxi Province, China
| | - Lina Cui
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Disease, Xijing Hospital, Fourth Military Medical University, 127 Changle Western Road, Xi'an 710032, Shaanxi Province, China
| | - Zheyi Han
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Disease, Xijing Hospital, Fourth Military Medical University, 127 Changle Western Road, Xi'an 710032, Shaanxi Province, China
| | - Ying Han
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Disease, Xijing Hospital, Fourth Military Medical University, 127 Changle Western Road, Xi'an 710032, Shaanxi Province, China
| | - Daiming Fan
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Disease, Xijing Hospital, Fourth Military Medical University, 127 Changle Western Road, Xi'an 710032, Shaanxi Province, China
| |
Collapse
|
63
|
Gaia S, Olivero A, Smedile A, Ruella M, Abate ML, Fadda M, Rolle E, Omedè P, Bondesan P, Passera R, Risso A, Aragno M, Marzano A, Ciancio A, Rizzetto M, Tarella C. Multiple courses of G-CSF in patients with decompensated cirrhosis: consistent mobilization of immature cells expressing hepatocyte markers and exploratory clinical evaluation. Hepatol Int 2013. [PMID: 26202037 DOI: 10.1007/s12072-013-9473-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Bone marrow-derived cells (BMCs) include stem cells capable of self-renewal and differentiation into a variety of cell types. Administration of granulocyte colony-stimulating factor (G-CSF) induces the circulation of BMCs in the peripheral blood. A phase II prospective trial was carried out for evaluation of BMC mobilization induced by multiple courses of G-CSF in cirrhotic patients. PATIENTS AND METHODS Fifteen patients with advanced liver cirrhosis (Child-Pugh score ≥6 points) were enrolled and treated with a 3-day G-CSF course, administered at 3-month intervals for a total of four courses. BMC mobilization was assessed by evaluating CD34+ve cells using flow cytometry. Expressions of multiple hepatic and stem markers were assessed on mobilized CD34+ve cells. Feasibility and safety were explored; clinical and adverse events were compared to those of a control group. Telomere length was monitored to rule out early cell aging caused by G-CSF. RESULTS A significant increase in G-CSF-induced circulating CD34+ve cells was consistently observed, although a progressive reduction of peak values was documented from cycle I to IV (p < 0.005). Mobilized CD34+ve cells expressed both stem and multiple hepatocyte markers, including mRNA of albumin and CYP2B6 (cytochrome P2 B6). Treatment was well tolerated, with no severe adverse events and no significant telomere length shortening following G-CSF. The procedure was safe. Overall, ten patients had either improved or had stable liver function tests (such as the Child-Pugh score), whereas five worsened and died from liver-related causes. CONCLUSION This study demonstrates that G-CSF can be safely administrated up to four times over a 1-year period in decompensated cirrhotic patients. The repeated BMC mobilization favors the circulation of stem cells coexpressing hepatic markers and mRNA of liver-related genes.
Collapse
Affiliation(s)
- Silvia Gaia
- Department of Gastro-hepatology, A.O. Città della Salute e della Scienza, S. Giovanni Battista Hospital, University of Turin, C. Bramante 88, 10126, Turin, Italy.
| | - Antonella Olivero
- Department of Gastro-hepatology, A.O. Città della Salute e della Scienza, S. Giovanni Battista Hospital, University of Turin, C. Bramante 88, 10126, Turin, Italy
| | - Antonina Smedile
- Department of Gastro-hepatology, A.O. Città della Salute e della Scienza, S. Giovanni Battista Hospital, University of Turin, C. Bramante 88, 10126, Turin, Italy
| | - Marco Ruella
- Division of Hematology and Cell Therapy, University of Torino, Mauriziano Hospital, Turin, Italy
| | - Maria Lorena Abate
- Department of Gastro-hepatology, A.O. Città della Salute e della Scienza, S. Giovanni Battista Hospital, University of Turin, C. Bramante 88, 10126, Turin, Italy
| | - Maurizio Fadda
- Department of Clinical Nutrition, A.O. Città della Salute e della Scienza, S. Giovanni Battista Hospital, University of Turin, C. Bramante 88, 10126, Turin, Italy
| | - Emanuela Rolle
- Department of Gastro-hepatology, A.O. Città della Salute e della Scienza, S. Giovanni Battista Hospital, University of Turin, C. Bramante 88, 10126, Turin, Italy
| | - Paola Omedè
- Division of Hematology 1, A.O. Città della Salute e della Scienza, S. Giovanni Battista Hospital, University of Turin, C. Bramante 88, 10126, Turin, Italy
| | - Paola Bondesan
- Division of Hematology 1, A.O. Città della Salute e della Scienza, S. Giovanni Battista Hospital, University of Turin, C. Bramante 88, 10126, Turin, Italy
| | - Roberto Passera
- Division of Nuclear Medicine 2, A.O. Città della Salute e della Scienza, S. Giovanni Battista Hospital, University of Turin, C. Bramante 88, 10126, Turin, Italy
| | - Alessandra Risso
- Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Manuela Aragno
- Department of Medicine and Experimental Oncology, University of Torino, Turin, Italy
| | - Alfredo Marzano
- Department of Gastro-hepatology, A.O. Città della Salute e della Scienza, S. Giovanni Battista Hospital, University of Turin, C. Bramante 88, 10126, Turin, Italy
| | - Alessia Ciancio
- Department of Gastro-hepatology, A.O. Città della Salute e della Scienza, S. Giovanni Battista Hospital, University of Turin, C. Bramante 88, 10126, Turin, Italy
| | - Mario Rizzetto
- Department of Gastro-hepatology, A.O. Città della Salute e della Scienza, S. Giovanni Battista Hospital, University of Turin, C. Bramante 88, 10126, Turin, Italy
| | - Corrado Tarella
- Division of Hematology and Cell Therapy, University of Torino, Mauriziano Hospital, Turin, Italy
| |
Collapse
|
64
|
Yao CL, Hsu SC, Hwang SM, Lee WC, Chiou TJ. A stromal-free, serum-free system to expand ex vivo hematopoietic stem cells from mobilized peripheral blood of patients with hematologic malignancies and healthy donors. Cytotherapy 2013; 15:1126-35. [PMID: 23768928 DOI: 10.1016/j.jcyt.2013.04.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 03/26/2013] [Accepted: 04/04/2013] [Indexed: 11/26/2022]
Abstract
BACKGROUND AIMS The number of hematopoietic stem cells (HSCs) is critical for transplantation. The ex vivo expansion of mobilized peripheral blood (MPB) HSCs is of clinical value for reconstitution to meet clinical need. METHODS This study proposed a simple, defined, stromal-free and serum-free culture system (SF-HSC medium) for clinical use, which is composed of Iscove's modified Dulbecco's medium, cytokine cocktails and serum substitutes. This study also characterized the cellular properties of expanded MPB CD133(+) HSCs from patients with hematologic malignancies and healthy donors by surface antigen, colony-forming cell, long-term culture-initiating cell, gene expression and in vivo engraftment assays. RESULTS The expanded fold values of CD45(+) white blood cells and CD34(+), CD133(+), CD34(+)CD38(-), CD133(+)CD38(-), CD34(+)CD133(+), colony-forming and long-term culture-initiating cells at the end of 7-day culture from CD133(+) MPB of hematologic malignancies were 9.4-fold, 5.9-fold, 4.0-fold, 35.8-fold, 21.9-fold, 3.8-fold, 11.8-fold and 6.7-fold, and values from healthy donor CD133(+) MPB were 20.7-fold, 14.5-fold, 8.5-fold, 83.8-fold, 37.3-fold, 6.2-fold, 19.1-fold and 14.6-fold. The high enrichment of CD38(-) cells, which were either CD34(+) or CD133(+), sustained the proliferation of early uncommitted HSCs. The expanded cells showed high levels of messenger RNA expression of HOBX4, ABCG2 and HTERT and had the in vivo ability to re-populate NOD/SCID mice. CONCLUSIONS Our results demonstrated that an initial, limited number of MPB CD133(+) HSCs could be expanded functionally in SF-HSC medium. We believe that this serum-free expansion technique can be employed in both basic research and clinical transplantation.
Collapse
Affiliation(s)
- Chao-Ling Yao
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Chung-Li, Taiwan.
| | | | | | | | | |
Collapse
|
65
|
In vitro interactions between rat bone marrow-derived endothelial progenitor cells and hepatic stellate cells: interaction between EPCs and HSCs. In Vitro Cell Dev Biol Anim 2013; 49:537-47. [PMID: 23722413 DOI: 10.1007/s11626-013-9637-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 05/11/2013] [Indexed: 01/22/2023]
Abstract
Transplantation of bone marrow (BM)-derived endothelial progenitor cells (EPCs) has been reported to improve liver fibrosis, but there is no direct evidence for the mechanism of improvement. We investigated the mechanism in vitro by coculturing BM-derived EPCs with activated hepatic stellate cells (HSCs) to mimic the hepatic environment. EPCs and HSCs were cultured alone and indirectly cocultured at a 1:1 ratio in a Transwell system. The characteristics of HSCs and EPCs were examined at different time points. An invasion assay showed the time-dependent effect on degradation of the extracellular matrix (ECM) layer in EPCs cultured alone. Real-time PCR and enzyme-linked immunosorbent assay analysis revealed that EPCs served as a source of matrix metalloproteinase-9 (MMP-9), and MMP-9 expression levels significantly increased during the 2 d of coculture. CFSE labeling showed that EPCs inhibited proliferation of HSCs. Annexin-V/PI staining, erminal deoxynucleotidyl transferase X-dUTP nick end labeling analysis, and (cleaved) caspase-3 activity revealed that EPCs promoted HSC apoptosis. However, the proliferation and apoptosis of EPCs were unaffected by cocultured HSCs. Coculturing increased the expression of inducible nitric oxide synthase, vascular endothelial growth factor, and hepatocyte growth factor (HGF) in EPCs, promoted differentiation of EPCs, and reduced the expression of types I and III collagens and transforming growth factor beta 1. Knockdown of HGF expression attenuated EPC-induced activation of HSC apoptosis and profibrotic ability. These findings demonstrated that BM-derived EPCs could degrade ECM, promoting activated HSC apoptosis, suppressing proliferation and profibrotic ability of activated HSCs. HGF secretion by EPCs plays a key role in inducing activated HSC apoptosis and HSC profibrotic ability.
Collapse
|
66
|
Bird TG, Lu WY, Boulter L, Gordon-Keylock S, Ridgway RA, Williams MJ, Taube J, Thomas JA, Wojtacha D, Gambardella A, Sansom OJ, Iredale JP, Forbes SJ. Bone marrow injection stimulates hepatic ductular reactions in the absence of injury via macrophage-mediated TWEAK signaling. Proc Natl Acad Sci U S A 2013; 110:6542-7. [PMID: 23576749 PMCID: PMC3631632 DOI: 10.1073/pnas.1302168110] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Tissue progenitor cells are an attractive target for regenerative therapy. In various organs, bone marrow cell (BMC) therapy has shown promising preliminary results, but to date no definite mechanism has been demonstrated to account for the observed benefit in organ regeneration. Tissue injury and regeneration is invariably accompanied by macrophage infiltration, but their influence upon the progenitor cells is incompletely understood, and direct signaling pathways may be obscured by the multiple roles of macrophages during organ injury. We therefore examined a model without injury; a single i.v. injection of unfractionated BMCs in healthy mice. This induced ductular reactions (DRs) in healthy mice. We demonstrate that macrophages within the unfractionated BMCs are responsible for the production of DRs, engrafting in the recipient liver and localizing to the DRs. Engrafted macrophages produce the cytokine TWEAK (TNF-like weak inducer of apoptosis) in situ. We go on to show that recombinant TWEAK activates DRs and that BMC mediated DRs are TWEAK dependent. DRs are accompanied by liver growth, occur in the absence of liver tissue injury and hepatic progenitor cells can be isolated from the livers of mice with DRs. Overall these results reveal a hitherto undescribed mechanism linking macrophage infiltration to DRs in the liver and highlight a rationale for macrophage derived cell therapy in regenerative medicine.
Collapse
Affiliation(s)
- Thomas G. Bird
- Medical Research Council (MRC), Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom; and
| | - Wei-Yu Lu
- Medical Research Council (MRC), Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom; and
| | - Luke Boulter
- Medical Research Council (MRC), Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom; and
| | - Sabrina Gordon-Keylock
- Medical Research Council (MRC), Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Rachel A. Ridgway
- Beatson Institute for Cancer Research, Glasgow G61 1BD, United Kingdom
| | - Michael J. Williams
- Medical Research Council (MRC), Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom; and
| | - Jessica Taube
- Medical Research Council (MRC), Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom; and
| | - James A. Thomas
- Medical Research Council (MRC), Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom; and
| | - Davina Wojtacha
- Medical Research Council (MRC), Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom; and
| | - Adriana Gambardella
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom; and
| | - Owen J. Sansom
- Beatson Institute for Cancer Research, Glasgow G61 1BD, United Kingdom
| | - John P. Iredale
- Medical Research Council (MRC), Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Stuart J. Forbes
- Medical Research Council (MRC), Centre for Inflammation Research, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom; and
| |
Collapse
|
67
|
Marmotti A, Castoldi F, Rossi R, Marenco S, Risso A, Ruella M, Tron A, Borrè A, Blonna D, Tarella C. Bone marrow-derived cell mobilization by G-CSF to enhance osseointegration of bone substitute in high tibial osteotomy. Knee Surg Sports Traumatol Arthrosc 2013; 21:237-48. [PMID: 22872005 DOI: 10.1007/s00167-012-2150-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Accepted: 07/19/2012] [Indexed: 12/14/2022]
Abstract
PURPOSE To evaluate granulocyte colony-stimulating factor (G-CSF) efficacy in accelerating bone regeneration following opening-wedge high tibial valgus osteotomy for genu varum. METHODS A phase II trial was conducted for evaluating the preoperative administration of G-CSF given at 10 μg/kg/day for 3 consecutive days with an additional half-dose 4 h before the opening-wedge high tibial valgus osteotomy. Overall, 12 patients (Group A) received G-CSF treatment, and the subsequent 12 patients (Group B) underwent surgery without G-CSF. The osteotomy gap was filled by a bone graft substitute. Bone marrow cell (BMC) mobilization was monitored by CD34+ve cell and clonogenic progenitor cell analysis. All patients underwent a clinical (Lysholm Knee Scale and SF-36) and radiographic evaluation preoperatively, as well as at given intervals postsurgery. RESULTS All patients completed the treatment program without major side effects; G-CSF was well tolerated. BMC mobilization occurred in all Group A patients, with median peak values of circulating CD34+ve cells of 110/μL (range 29-256). Circulating clonogenic progenitors paralleled CD34+ve cell levels. A significant improvement in Lysholm Knee Scale was recorded at follow-up in Group A compared to Group B. At the radiographic evaluation, there was a significant increase in osseointegration at the bone-graft junction in Group A at 1, 2, 3 and 6 months postsurgery compared to Group B. The computerized tomography scan of the grafted area at 2 months postsurgery showed no significant difference in the quality of the newly formed bone between the two Groups. CONCLUSIONS Although the limited number of patients does not allow firm conclusions, the study suggests that G-CSF can be safely administered preoperatively in subjects undergoing opening-wedge high tibial valgus osteotomy; in addition, the clinical, radiographic and CT monitoring indicate that G-CSF and/or mobilized BMCs may hasten bone graft substitute osseointegration. LEVEL OF EVIDENCE I.
Collapse
Affiliation(s)
- A Marmotti
- Department of Orthopaedics and Traumatology, Ordine Mauriziano, Umberto I Hospital, University of Torino, Turin, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
68
|
Sharma AD, Iacob R, Cantz T, Manns MP, Ott M. Liver. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
69
|
Aini W, Miyagawa-Hayashino A, Ozeki M, Tsuruyama T, Tamaki K, Uemoto S, Haga H. Frequent hepatocyte chimerism in long-term human liver allografts independent of graft outcome. Transpl Immunol 2012; 28:100-5. [PMID: 23268137 DOI: 10.1016/j.trim.2012.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 11/24/2012] [Accepted: 12/08/2012] [Indexed: 12/13/2022]
Abstract
Microchimerism after liver transplantation is considered to promote graft tolerance or tissue repair, but its significance is controversial. By using multiplex polymerase chain reaction (PCR) of short tandem repeat (STR) loci after laser capture microdissection of hepatocyte nuclei, we compared the proportions of recipient-derived hepatocytes in long-term stable liver allografts and late dysfunctional allografts caused by chronic rejection or idiopathic post-transplantation hepatitis. Through fluorescence in situ hybridization (FISH), we also analyzed the presence of recipient-derived Y-positive hepatocytes in the biopsies of livers transplanted from female donors to male recipients. The study population comprised 24 pediatric liver transplant recipients who survived with the initial graft, whose 10-year protocol biopsy records were available, and who had normal liver function (stable graft, SG; n=13) or a late dysfunctional graft (LDG; n=11) with similar follow-up periods (mean 10.8years in the SG group and 11.2years in the LDG group). STR analysis revealed that hepatocyte chimerism occurred in 7 of 13 (54%) SGs and 5 of 11 (45%) LDGs (p=0.68). The proportion of hepatocyte chimerism was low, with a mean of 3% seen in 2 of 3 female-to-male transplanted livers (one each of SG and LDG). In conclusion, hepatocyte chimerism was a constant event. The extent of engraftment of recipient-derived hepatocytes does not seem to correlate with the degree of hepatic injury in long-term liver allografts.
Collapse
Affiliation(s)
- Wulamujiang Aini
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | | | | | | | | | | | | |
Collapse
|
70
|
Booth C, Soker T, Baptista P, Ross CL, Soker S, Farooq U, Stratta RJ, Orlando G. Liver bioengineering: Current status and future perspectives. World J Gastroenterol 2012; 18:6926-34. [PMID: 23322990 PMCID: PMC3531676 DOI: 10.3748/wjg.v18.i47.6926] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 11/16/2012] [Accepted: 11/24/2012] [Indexed: 02/06/2023] Open
Abstract
The present review aims to illustrate the strategies that are being implemented to regenerate or bioengineer livers for clinical purposes. There are two general pathways to liver bioengineering and regeneration. The first consists of creating a supporting scaffold, either synthetically or by decellularization of human or animal organs, and seeding cells on the scaffold, where they will mature either in bioreactors or in vivo. This strategy seems to offer the quickest route to clinical translation, as demonstrated by the development of liver organoids from rodent livers which were repopulated with organ specific cells of animal and/or human origin. Liver bioengineering has potential for transplantation and for toxicity testing during preclinical drug development. The second possibility is to induce liver regeneration of dead or resected tissue by manipulating cell pathways. In fact, it is well known that the liver has peculiar regenerative potential which allows hepatocyte hyperplasia after amputation of liver volume. Infusion of autologous bone marrow cells, which aids in liver regeneration, into patients was shown to be safe and to improve their clinical condition, but the specific cells responsible for liver regeneration have not yet been determined and the underlying mechanisms remain largely unknown. A complete understanding of the cell pathways and dynamics and of the functioning of liver stem cell niche is necessary for the clinical translation of regenerative medicine strategies. As well, it will be crucial to elucidate the mechanisms through which cells interact with the extracellular matrix, and how this latter supports and drives cell fate.
Collapse
|
71
|
Ross CL, Booth C, Sanders B, Babbar P, Bergman C, Soker T, Sittadjody S, Salvatori M, Al-Shraideh Y, Stratta RJ, Orlando G. Regeneration and bioengineering of transplantable abdominal organs: current status and future challenges. Expert Opin Biol Ther 2012; 13:103-13. [PMID: 23110384 DOI: 10.1517/14712598.2013.732063] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The most critical issue to organ transplantation is the identification of new sources of organs. The present manuscript illustrates the state-of-the-art regenerative medicine (RM) investigations aiming to manufacturing abdominal organs for transplant purposes. AREAS COVERED This manuscript focuses on research in the bioengineering and regeneration of kidneys, insulin-producing cells, livers and small bowel. The main technology currently under development exploits the seeding of cells on supporting scaffolding material. Despite favorable preliminary results obtained with relatively simple, hollow organs, when more complex organs are considered, the scenario changes dramatically. Investigations are still in early stages, and clinical translation is not yet foreseeable based on current knowledge and information. Obstacles are numerous but we believe the critical factor hampering success is lack of in-depth understanding of the extracellular matrix (ECM) and cell-ECM interactions, as well as the mechanisms with which organs develop in utero. EXPERT OPINION The success of RM to generate transplantable abdominal organs relies heavily on progress in (stem) cell therapies, developmental and ECM biology, and in the thorough understanding of the intricate relationship and interplay between cells and the ECM. This will require enormous investments in financial and medical resources, which ideally should be embarked upon by governments, the private sector and academia.
Collapse
Affiliation(s)
- Christina L Ross
- Wake Forest University School of Medicine, Wake Forest Institute for Regenerative Medicine, Winston Salem, NC, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Baldelli CMF, Ruella M, Scuderi S, Monni M, Passera R, Omedè P, Tarella C. A short course of granulocyte–colony-stimulating factor to accelerate wound repair in patients undergoing surgery for sacrococcygeal pilonidal cyst: proof of concept. Cytotherapy 2012; 14:1101-9. [DOI: 10.3109/14653249.2012.697147] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
73
|
Wang PP, Xie DY, Liang XJ, Peng L, Zhang GL, Ye YN, Xie C, Gao ZL. HGF and direct mesenchymal stem cells contact synergize to inhibit hepatic stellate cells activation through TLR4/NF-kB pathway. PLoS One 2012; 7:e43408. [PMID: 22927965 PMCID: PMC3426540 DOI: 10.1371/journal.pone.0043408] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2012] [Accepted: 07/23/2012] [Indexed: 12/31/2022] Open
Abstract
Aims Bone marrow-derived mesenchymal stem cells (BMSCs) can reduce liver fibrosis. Apart from the paracrine mechanism by which the antifibrotic effects of BMSCs inhibit activated hepatic stellate cells (HSCs), the effects of direct interplay and juxtacrine signaling between the two cell types are poorly understood. The purpose of this study was to explore the underlying mechanisms by which BMSCs modulate the function of activated HSCs. Methods We used BMSCs directly and indirectly co-culture system with HSCs to evaluate the anti-fibrosis effect of BMSCs. Cell proliferation and activation were examined in the presence of BMSCs and HGF. c-met was knockdown in HSCs to evaluate the effect of HGF secreted by BMSCs. The TLR4 and Myeloid differentiation primary response gene 88(MyD88) mRNA levels and the NF-kB pathway activation were determined by real-time PCR and western blotting analyses. The effect of BMSCs on HSCs activation was investigated in vitro in either MyD88 silencing or overexpression in HSCs. Liver fibrosis in rats fed CCl4 with and without BMSCs supplementation was compared. Histopathological examinations and serum biochemical tests were compared between the two groups. Results BMSCs remarkably inhibited the proliferation and activation of HSCs by interfering with LPS-TLR4 pathway through a cell–cell contact mode that was partially mediated by HGF secretion. The NF-kB pathway is involved in HSCs activation inhibition by BMSCs. MyD88 over expression reduced the BMSC inhibition of NF-kB luciferase activation. BMSCs protected liver fibrosis in vivo. Conclusion BMSCs modulate HSCs in vitro via TLR4/MyD88/NF-kB signaling pathway through cell–cell contact and secreting HGF. BMSCs have therapeutic effects on cirrhosis rats. Our results provide new insights into the treatment of hepatic fibrosis with BMSCs.
Collapse
Affiliation(s)
- Pei-pei Wang
- Department of Infectious Diseases, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | | | | | | | | | | | | | | |
Collapse
|
74
|
Kisseleva T, Brenner DA. The phenotypic fate and functional role for bone marrow-derived stem cells in liver fibrosis. J Hepatol 2012; 56:965-72. [PMID: 22173163 PMCID: PMC3307836 DOI: 10.1016/j.jhep.2011.09.021] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 08/12/2011] [Accepted: 09/04/2011] [Indexed: 12/13/2022]
Abstract
Liver fibrosis is an outcome of chronic liver injury of any etiology. It is manifested by extensive deposition of extracellular matrix (ECM) proteins that produce a fibrous scar in the injured liver. Bone marrow (BM) cells may play an important role in pathogenesis and resolution of liver fibrosis. BM cells contribute to the inflammatory response by TGF-β1 secretion and activation of liver resident myofibroblasts. Moreover, BM itself can serve as a source of collagen expressing cells, e.g. BM-derived fibrocytes and mesenchymal progenitors, which in turn, have a potential to in situ differentiate into fibrogenic myofibroblasts and facilitate fibrosis. Finally, BM cells play an active part in resolution of liver fibrosis after cessation of fibrogenic stimuli. While natural killer (NK) cells are implicated in apoptosis of activated hepatic stellate cells/myofibroblasts, cells of myelo-monocitic lineage secrete matrix metalloproteinases to actively degrade the fibrous scar. The focus of this review is on the current understanding of the role of different subsets of BM cells in the onset, development and resolution of liver fibrosis.
Collapse
|
75
|
Garg V, Garg H, Khan A, Trehanpati N, Kumar A, Sharma BC, Sakhuja P, Sarin SK. Granulocyte colony-stimulating factor mobilizes CD34(+) cells and improves survival of patients with acute-on-chronic liver failure. Gastroenterology 2012; 142:505-512.e1. [PMID: 22119930 DOI: 10.1053/j.gastro.2011.11.027] [Citation(s) in RCA: 291] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Revised: 11/03/2011] [Accepted: 11/04/2011] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS Acute-on-chronic liver failure (ACLF) develops in patients with chronic liver disease and has high mortality. Mobilization of bone marrow-derived stem cells with granulocyte colony-stimulating factor (G-CSF) could promote hepatic regeneration. METHODS Consecutive patients with ACLF were randomly assigned to groups given 5 μg/kg G-CSF subcutaneously (12 doses; group A, n = 23) or placebo (group B, n = 24) plus standard medical therapy. We assessed survival until day 60; Child-Turcotte-Pugh (CTP), Model for End-Stage Liver Disease (MELD), and Sequential Organ Failure Assessment (SOFA) scores; and the development of other related complications. RESULTS After 1 week of treatment, group A had higher median leukocyte and neutrophil counts than group B (P < .001). Sixteen patients in group A (69.6%) and 7 in group B (29%) survived; the actuarial probability of survival at day 60 was 66% versus 26%, respectively (P = .001). Treatment with G-CSF also reduced CTP scores in group A by a median of 33.3% compared with an increase of 7.1% in group B (P = .001), along with MELD (median reduction of 15.3% compared with an increase of 11.7% in group B; P = .008) and SOFA scores (median reduction of 50% compared with an increase of 50% in group B; P = .001). The percentages of patients who developed hepatorenal syndrome, hepatic encephalopathy, or sepsis were lower in group A than in group B (19% vs 71% [P = .0002], 19% vs 66% [P = .001], and 14% vs 41% [P = .04], respectively). After 1 month of treatment, G-CSF increased the number of CD34(+) cells in the liver (by 45% compared with 27.5% in group B; P = .01). CONCLUSIONS G-CSF therapy more than doubles the percentage of patients with ACLF who survive for 2 months; it also significantly reduces CTP, MELD, and SOFA scores and prevents the development of sepsis, hepatorenal syndrome, and hepatic encephalopathy.
Collapse
Affiliation(s)
- Vishal Garg
- Department of Gastroenterology, GB Pant Hospital, New Delhi, India
| | | | | | | | | | | | | | | |
Collapse
|
76
|
Russo FP, Parola M. Stem cells in liver failure. Best Pract Res Clin Gastroenterol 2012; 26:35-45. [PMID: 22482524 DOI: 10.1016/j.bpg.2012.01.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 01/08/2012] [Indexed: 01/31/2023]
Abstract
Orthotopic liver transplantation (OLT) represents the only reliable therapeutic approach for acute liver failure (ALF), liver failure associated to end-stage chronic liver diseases (CLD) and non-metastatic liver cancer. The clinical impact of liver failure is relevant because of the still high ALF mortality and the increasing worldwide prevalence of cirrhosis that, in turn, is the main predisposing cause for hepatocellular carcinoma (HCC). Moreover, in the next decade because an increased number of patients reaching end-stage disease and requiring OLT may face a shortage of donor livers. This clinical scenario led several laboratories to explore the feasibility and efficiency of alternative approaches, involving cellular therapy, to counteract liver failure. The present chapter overviews results and concepts emerged from recent experimental and clinical studies in which adult or embryonic hepatocytes, hepatic stem/progenitor cells, induced pluripotent stem (iPS) cells as well as extrahepatic stem cells have been used as putative transplantable cell sources.
Collapse
Affiliation(s)
- Francesco P Russo
- Department of Surgical and Gastroenterological Sciences, Gastroenterology Unit, University of Padova, Padova, Italy.
| | | |
Collapse
|
77
|
Allameh A, Kazemnejad S. Safety evaluation of stem cells used for clinical cell therapy in chronic liver diseases; with emphasize on biochemical markers. Clin Biochem 2012; 45:385-96. [PMID: 22306885 DOI: 10.1016/j.clinbiochem.2012.01.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 01/16/2012] [Accepted: 01/18/2012] [Indexed: 12/11/2022]
Abstract
There are several issues to be considered to reduce the risk of rejection and minimize side effects associated with liver cell transplantation in chronic liver diseases. The source and the condition of stem cell proliferation and differentiation ex vivo and the transplantation protocols are important safety considerations for cell based therapy. The biochemical and molecular markers are important tools for safety evaluation of different processes of cell expansion and transplantation. Studies show that hepatocytes differentiated from adult and embryonic stem cells exhibit biochemical and metabolic properties resembling mature hepatocytes. Therefore these assays can help to assess the biological and metabolic performance of hepatocytes and progenitor stem cells. The assays also help in testing the contribution of transplanted hepatocytes in improving the repair and function of damaged liver in the recipient. Here we review the biochemical and metabolic markers, which are implicated in evaluation of safety issues of stem cells used for therapeutic purposes in chronic liver diseases and regeneration of damaged liver. We also highlight application of biochemical tests for assessment of liver cell transplantation.
Collapse
Affiliation(s)
- Abdolamir Allameh
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, PO Box 14115-111, Tehran, Islamic Republic of Iran.
| | | |
Collapse
|
78
|
Zhang M, Zhong Y, Chen J. Model systems and clinical applications of hepatic stem cells for liver regeneration. Hepatol Int 2011. [DOI: 10.1007/s12072-011-9323-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
79
|
Meng YC, Jiang HX, Zhang JH. Clinical applications of bone marrow stem cells to treat liver diseases: recent progress. Shijie Huaren Xiaohua Zazhi 2011; 19:2846-2851. [DOI: 10.11569/wcjd.v19.i27.2846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In recent years, great advances have been made in the treatment of liver diseases, such as fulminant and chronic hepatic failure, end-stage liver disease and inherited metabolic disorders, by bone marrow stem cell transplantation. Stem cell transplantation possesses advantages of low cost, easy obtainment of stem cells, and little or no immune rejection and therefore has good efficacy, safety and tolerability. Although liver transplantation is an effective way for the treatment of end-stage liver disease, it has limited clinical applications due to the shortage of organ donors, complicated operation procedure, severe complications, immunological rejection and high cost. Therefore, bone marrow stem cell transplantation has shed light on the treatment of end-stage liver diseases. In this article we review the clinical applications of bone marrow stem cell transplantation in the treatment of liver diseases.
Collapse
|
80
|
Burra P, Bizzaro D, Ciccocioppo R, Marra F, Piscaglia AC, Porretti L, Gasbarrini A, Russo FP. Therapeutic application of stem cells in gastroenterology: an up-date. World J Gastroenterol 2011; 17:3870-3880. [PMID: 22025875 PMCID: PMC3198016 DOI: 10.3748/wjg.v17.i34.3870] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Revised: 03/07/2011] [Accepted: 03/14/2011] [Indexed: 02/06/2023] Open
Abstract
Adult stem cells represent the self-renewing progenitors of numerous body tissues, and they are currently classified according to their origin and differentiation ability. In recent years, the research on stem cells has expanded enormously and holds therapeutic promises for many patients suffering from currently disabling diseases. This paper focuses on the possible use of stem cells in the two main clinical settings in gastroenterology, i.e., hepatic and intestinal diseases, which have a strong impact on public health worldwide. Despite encouraging results obtained in both regenerative medicine and immune-mediated conditions, further studies are needed to fully understand the biology of stem cells and carefully assess their putative oncogenic properties. Moreover, the research on stem cells arouses fervent ethical, social and political debate. The Italian Society of Gastroenterology sponsored a workshop on stem cells held in Verona during the XVI Congress of the Federation of Italian Societies of Digestive Diseases (March 6-9, 2010). Here, we report on the issues discussed, including liver and intestinal diseases that may benefit from stem cell therapy, the biology of hepatic and intestinal tissue repair, and stem cell usage in clinical trials.
Collapse
|
81
|
Zhang L, Kang W, Lei Y, Han Q, Zhang G, Lv Y, Li Z, Lou S, Liu Z. Granulocyte colony-stimulating factor treatment ameliorates liver injury and improves survival in rats with D-galactosamine-induced acute liver failure. Toxicol Lett 2011; 204:92-99. [PMID: 21550386 DOI: 10.1016/j.toxlet.2011.04.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Revised: 04/14/2011] [Accepted: 04/19/2011] [Indexed: 12/14/2022]
Abstract
Only liver transplantation is currently available therapy for the patients with acute liver failure (ALF). This study was designed to determine whether administration of granulocyte colony-stimulating factor (G-CSF) has therapeutic efficacy in animals with ALF. Female Sprague-Dawley (SD) rats were intraperitoneally injected with a single dose of d-galactosamine (d-GalN, 1.4g/kg) to induce ALF. After 2h, the rats were randomized to receive G-CSF (50μg/kg/day), or saline vehicle injection for 5 days. Rats were observed for survival and assessed for liver injury by serum alanine transaminase (ALT) measurement and histological analysis. CD34+ cells in bone marrow were assessed by flow cytometry. CD34+ cells and Ki-67+ hepatocytes in liver tissue were evaluated by immunohistochemistry. In the ALF model, 5-day survival after d-GalN injection was 33.3% (10/30), while G-CSF administration following d-GalN resulted in 53.3% (16/30) survival (p=0.027). G-CSF treated rats had lower ALT level and less hepatic injury compared with saline vehicle rats. The increases of CD34+ cells in bone marrow and liver tissue and Ki-67+ cells in liver tissue in G-CSF treated rats were higher than those in saline rats. No correlation was observed between CD34+ cells and Ki-67+ hepatocytes in liver tissue in both G-CSF and vehicle rats. It is suggested that G-CSF increases survival rate, decreases liver injury and enhances hepatocyte proliferation in rats with d-GalN-induced ALF possibly through actions including but not limiting to CD34+ cell mobilization, and that G-CSF may be of potential value in treating ALF.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Infectious Diseases, First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, #277 Yanta West Road, Xi'an 710061, Shaanxi Province, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Trebol Lopez J, Georgiev Hristov T, García-Arranz M, García-Olmo D. Stem Cell Therapy for Digestive Tract Diseases: Current State and Future Perspectives. Stem Cells Dev 2011; 20:1113-29. [DOI: 10.1089/scd.2010.0277] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Jacobo Trebol Lopez
- General and Digestive Tract Surgery Department, University Hospital “La Paz”, Madrid, Spain
- Cell Therapy Laboratory, Investigation Institute IdiPAZ, University Hospital “La Paz”, Madrid, Spain
| | - Tihomir Georgiev Hristov
- General and Digestive Tract Surgery Department, University Hospital “La Paz”, Madrid, Spain
- Cell Therapy Laboratory, Investigation Institute IdiPAZ, University Hospital “La Paz”, Madrid, Spain
| | - Mariano García-Arranz
- Cell Therapy Laboratory, Investigation Institute IdiPAZ, University Hospital “La Paz”, Madrid, Spain
| | - Damián García-Olmo
- General and Digestive Tract Surgery Department, University Hospital “La Paz”, Madrid, Spain
- Cell Therapy Laboratory, Investigation Institute IdiPAZ, University Hospital “La Paz”, Madrid, Spain
- Surgery Department, Autonomous University of Madrid, Madrid, Spain
| |
Collapse
|
83
|
Sancho-Bru P. [Therapeutic possibilities of stem cells in the treatment of liver diseases]. GASTROENTEROLOGIA Y HEPATOLOGIA 2011; 34:701-10. [PMID: 21640436 DOI: 10.1016/j.gastrohep.2011.03.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Accepted: 03/09/2011] [Indexed: 12/19/2022]
Abstract
Cell therapy and the use of stem cells in the treatment of liver diseases is still in the research phase. Nevertheless, the diversity of stem cells in terms of their origin, characteristics and potential for differentiation provides a wide spectrum of possibilities for the treatment of liver diseases. The present article describes the main types of stem cells and their potential for the treatment of liver diseases, as well as the main therapeutic strategies that are currently being explored for the treatment of these diseases through cell therapy. In addition, the main preclinical and clinical studies suggesting that stem cells could become an effective therapeutic alternative in distinct liver diseases are discussed.
Collapse
Affiliation(s)
- Pau Sancho-Bru
- Unidad de Hepatología, Hospital Clínic de Barcelona, Centro de Investigación Biológica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, España.
| |
Collapse
|
84
|
Francipane MG, Cervello M, Vizzini GB, Pietrosi G, Montalto G. Management of Liver Failure: From Transplantation to Cell-Based Therapy. CELL MEDICINE 2011; 2:9-25. [PMID: 26998399 DOI: 10.3727/215517911x575993] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The severe shortage of deceased donor organs has driven a search for alternative methods of treating liver failure. In this context, cell-based regenerative medicine is emerging as a promising interdisciplinary field of tissue repair and restoration, able to contribute to improving health in a minimally invasive fashion. Several cell types have allowed long-term survival in experimental models of liver injury, but their therapeutic potential in humans should be regarded with deep caution, because few clinical trials are currently available and the number of patients enrolled so far is too small to assess benefits versus risks. This review summarizes the current literature on the physiological role of endogenous stem cells in liver regeneration and on the therapeutic benefits of exogenous stem cell administration with specific emphasis on the potential clinical uses of mesenchymal stem cells. Moreover, critical points that still need clarification, such as the exact identity of the stem-like cell population exerting the beneficial effects, as well as the limitations of stem cell-based therapies, are discussed.
Collapse
Affiliation(s)
- Maria Giovanna Francipane
- Institute of Biomedicine and Molecular Immunology "Alberto Monroy," National Research Council (CNR), Palermo, Italy; †Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy
| | - Melchiorre Cervello
- Institute of Biomedicine and Molecular Immunology "Alberto Monroy," National Research Council (CNR) , Palermo , Italy
| | - Giovanni Battista Vizzini
- ‡ Istituto Mediterraneo Trapianti e Terapie ad Alta Specializzazione, University of Pittsburgh Medical Center in Italy , Palermo , Italy
| | - Giada Pietrosi
- ‡ Istituto Mediterraneo Trapianti e Terapie ad Alta Specializzazione, University of Pittsburgh Medical Center in Italy , Palermo , Italy
| | - Giuseppe Montalto
- † Department of Internal Medicine and Specialties, University of Palermo , Palermo , Italy
| |
Collapse
|
85
|
Recombinant human granulocyte colony-stimulating factor (rhG-CSF) could be an effective adjuvant therapy for orthopedic implant-related infections (OIRI). Med Hypotheses 2011; 76:703-5. [DOI: 10.1016/j.mehy.2011.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2010] [Revised: 12/04/2010] [Accepted: 02/01/2011] [Indexed: 11/22/2022]
|
86
|
Kisseleva T, Brenner DA. Anti-fibrogenic strategies and the regression of fibrosis. Best Pract Res Clin Gastroenterol 2011; 25:305-17. [PMID: 21497747 PMCID: PMC3086317 DOI: 10.1016/j.bpg.2011.02.011] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 02/18/2011] [Accepted: 02/23/2011] [Indexed: 01/31/2023]
Abstract
Liver fibrosis is an outcome of many chronic diseases, and often results in cirrhosis, liver failure, and portal hypertension. Liver transplantation is the only treatment available for patients with advanced stage of fibrosis. Therefore, alternative methods are required to develop new strategies for anti-fibrotic therapy. Available treatments are designed to substitute for liver transplantation or bridge the patients, they include inhibitors of fibrogenic cytokines such as TGF-β1 and EGF, inhibitors of rennin angiotensin system, and blockers of TLR4 signalling. Development of liver fibrosis is orchestrated by many cell types. However, activated myofibroblasts remain the primary target for anti-fibrotic therapy. Hepatic stellate cells and portal fibroblasts are considered to play a major role in development of liver fibrosis. Here we discuss the origin of activated myofibroblasts and different aspects of their activation, differentiation and potential inactivation during regression of liver fibrosis.
Collapse
|
87
|
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
|
88
|
Shupe T, Petersen BE. Potential applications for cell regulatory factors in liver progenitor cell therapy. Int J Biochem Cell Biol 2011; 43:214-21. [PMID: 20851776 PMCID: PMC3022095 DOI: 10.1016/j.biocel.2010.09.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2009] [Revised: 08/24/2010] [Accepted: 09/06/2010] [Indexed: 12/18/2022]
Abstract
Orthotopic liver transplant represent the state of the art treatment for terminal liver pathologies such as cirrhosis in adults and hemochromatosis in neonates. A limited supply of transplantable organs in relationship to the demand means that many patients will succumb to disease before an organ becomes available. One promising alternative to liver transplant is therapy based on the transplant of liver progenitor cells. These cells may be derived from the patient, expanded in vitro, and transplanted back to the diseased liver. Inborn metabolic disorders represent the most attractive target for liver progenitor cell therapy, as many of these disorders may be corrected by repopulation of only a portion of the liver by healthy cells. Another potential application for liver progenitor cell therapy is the seeding of bio-artificial liver matrix. These ex vivo bioreactors may someday be used to bridge critically ill patients to other treatments. Conferring a selective growth advantage to the progenitor cell population remains an obstacle to therapy development. Understanding the molecular signaling mechanisms and micro-environmental cues that govern liver progenitor cell phenotype may someday lead to strategies for providing this selective growth advantage. The discovery of a population of cells within the bone marrow possessing the ability to differentiate into hepatocytes may provide an easily accessible source of cells for liver therapies.
Collapse
Affiliation(s)
- Thomas Shupe
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, College of Medicine, Gainesville, FL 32610-0275, USA.
| | | |
Collapse
|
89
|
Dalakas E, Newsome PN, Boyle S, Brown R, Pryde A, McCall S, Hayes PC, Bickmore WA, Harrison DJ, Plevris JN. Bone marrow stem cells contribute to alcohol liver fibrosis in humans. Stem Cells Dev 2011; 19:1417-25. [PMID: 20025456 DOI: 10.1089/scd.2009.0387] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Bone marrow-derived stem cell (BMSC) contribution to liver repair varies considerably and recent evidence suggests these cells may contribute to liver fibrosis. We investigated the mobilization and hepatic recruitment of bone marrow (BM) stem cells in patients with alcohol liver injury and their contribution to parenchymal/non-parenchymal liver cell lineages. Liver biopsies from alcoholic hepatitis (AH) patients and male patients, who received a female liver transplant and developed AH, were analyzed for BM stem cell content by fluorescence in situ hybridization and immunostaining. Y chromosome analysis was performed, along with co-staining for hepatocyte, biliary, myofibroblast, and Ki-67 markers. Blood CD34(+) levels were quantified in AH patients by flow cytometry. AH patients had increased CD34(+) cell counts in liver tissue (1.834% +/- 0.605%; P < 0.05) and in blood (0.195% +/- 0.063%; P < 0.05) as compared with matched controls (0.299% + 0.208% and 0.067% +/- 0.01%). A proportion of hepatic myofibroblasts were BM-derived (7.9%-26.8%) as deemed by the co-localization of Y chromosome/alpha-smooth muscle actin (alpha-SMA) staining. In the cross-sex liver grafts with AH, 5.025% of the myofibroblasts were co-staining for CD34, suggesting that a population of CD34(+) cells were contributing to the hepatic myofibroblast population. There was no evidence of BM contribution to hepatocyte or biliary cell differentiation, nor evidence of increased hepatocyte regeneration. Alcohol liver injury mobilizes CD34(+) stem cells into the circulation and recruits them into the liver. These BMSCs contribute to the hepatic myofibroblast population but not to parenchymal lineages and do not promote hepatocyte repair.
Collapse
|
90
|
Abstract
Patients with acute liver failure are a particularly challenging group, with unique difficulties faced in treatment decisions. Life-saving therapy is available, but organ shortage, delays in transplantation, and complications in management result in a high mortality in this group of patients even after transplant. Any pharmacologic intervention that improved outcomes in this population of critically ill patients would be of great benefit. Based on available evidence, different scenarios of participation of HSCs in liver recovery are conceivable. Encouraging HSCs to differentiate into hepatocytes or supply paracrine and cellular level support to accelerate ongoing local repair mechanisms and assist a failing liver with inadequate mass and functional capacity might be directed to occur effectively in humans. Evidence within small animal models of liver injury and observations within the human population suggest that this might also be encouraged. The use of pharmacologic agents to mobilize hematopoietic stem cells is well established and effectively used in a different population of patients. As such, extending the use of these drugs, such as plerixafor, to the human population has a sound basis. However, there is a need for clarification of the mechanisms by which these cells exert their effect as well as which specific population of cells is involved in the regenerative process. To be clinically relevant in scenarios of acute liver failure, stem cell mobilizing strategies would have to impact survival when administered well after injury. Applications in other settings may also prove useful. Limits to liver resection exist where the size of the future liver remnant governs the extent of resection possible. Preexisting functional impairment may be restrictive, and strategies involving stem cells may assist the future liver remnant in both normal and functionally impaired livers. Benefit has already been reported from treatment with G-CSF in other injured tissues, including the injured myocardium and acutely injured kidney. However, as yet no clinical trial exists to assess the effects of stem cell mobilization in humans with acute liver failure. The familiarity in the use of and success demonstrated in the clinical and experimental use of plerixafor and G-CSF make exploration of hematopoietic stem cells as therapy in patients with acute liver failure appealing.
Collapse
Affiliation(s)
- Russell N Wesson
- Department of Surgery, Johns Hopkins Medical Institutions, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | | |
Collapse
|
91
|
Sharma AD, Iacob R, Bock M, Cantz T, Manns MP, Ott M. Liver. Regen Med 2011. [DOI: 10.1007/978-90-481-9075-1_33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
92
|
Mannheimer EG, Quintanilha LF, Carvalho AB, Paredes BD, Gonçalves de Carvalho F, Takyia CM, Resende CMC, Ferreira da Motta Rezende G, Campos de Carvalho AC, Schanaider A, dos Santos Goldenberg RC. Bone marrow cells obtained from cirrhotic rats do not improve function or reduce fibrosis in a chronic liver disease model. Clin Transplant 2011; 25:54-60. [PMID: 20047614 DOI: 10.1111/j.1399-0012.2009.01191.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND The objective of this study was to evaluate the therapeutic potential of bone marrow cells (BMCs) obtained from cirrhotic donors in a model of chronic liver disease. METHODS Chronic liver injury was induced in female Wistar rats by the association of an alcoholic diet with intraperitoneal injections of carbon tetrachloride. BMCs obtained from cirrhotic donors or placebo were injected through the portal vein. Blood analysis of alanine aminotransferase (ALT) and albumin levels, ultrasound assessment including the measurement of the portal vein diameter (PVD) and liver echogenicity, histologic evaluation with hematoxylin and eosin and Sirius red staining, and quantification of collagen deposition were performed. RESULTS ALT and albumin blood levels showed no significant differences between the experimental groups two months after injection. Additionally, no significant variation in PVD and liver echogenicity was found. Histological analysis also showed no significant variation in collagen deposition two months after placebo or BMC injection. CONCLUSION This study suggests that, even though BMC therapy using cells from healthy donors has previously shown to be effective, this is not the case when BMCs are obtained from cirrhotic animals. This result has major clinical implications when considering the use of autologous BMCs from patients with chronic liver diseases.
Collapse
Affiliation(s)
- Elida Gripp Mannheimer
- Centro de Cirurgia Experimental e Programa de Pós-Graduação, Departamento de Cirurgia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Jin SZ, Meng XW, Sun X, Han MZ, Liu BR, Wang XH, Sun LY, Huang Q, Zhao RB, Ban X, Yu HY, Yu HW. Granulocyte colony-stimulating factor enhances bone marrow mononuclear cell homing to the liver in a mouse model of acute hepatic injury. Dig Dis Sci 2010; 55:2805-13. [PMID: 20130994 DOI: 10.1007/s10620-009-1117-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Accepted: 12/28/2009] [Indexed: 12/11/2022]
Abstract
BACKGROUND Experiments have reported that granulocyte colony stimulating factor (G-CSF) can mobilize stem cells. However, few studies have examined the effect of G-CSF on bone marrow mononuclear cell (BMMC) mobilization, in particular regarding their capability to home to acutely injured liver. AIMS The aim of this study was to evaluate the effort of G-CSF on BMMC homing to the liver following chemically-induced hepatic failure. METHODS BMMC were isolated from mice, pre-labeled with PKH26 and infused into the mice in which hepatic injury had been induced followed by administration of G-CSF or vehicle. Livers were studied by fluorescent microscopy after transplantation of pre-labeled BMMC. RESULTS PKH26 labeled cells were found in liver tissue at 102 ± 10 cells/high power field in the BMMC+G-CSF group and 30 ± 5 cells/high power field in the BMMC group, but none in the G-CSF group and the control group (P < 0.05). In the former two groups the majority of PKH26 labeled cells colocalized with proliferative cell nuclear antigen (PCNA). The number of PCNA positive cells in the BMMC+G-CSF group was 20 ± 4 cells/high power field, while in the BMMC group it was 14 ± 2 cells/high power field, in the G-CSF group 12 ± 2 cells/high power field, and 8 ± 1 cells/high power field in the control group. Moreover, albumin expression was increased in the BMMC+G-CSF treated group (149 ± 7/high power field) relative to the BMMC group (48 ± 6/high power field), the G-CSF group (44 ± 5/high power field) and the vehicle group (30 ± 6/high power field), with the former three groups showing elevated levels as compared to vehicle control (30 ± 6) (P < 0.05). CONCLUSION Transplanted BMMC may home to injured liver, which appears to be enhanced by G-CSF administration.
Collapse
Affiliation(s)
- Shi-Zhu Jin
- Department of Gastroenterology, First Hospital, Jilin University, 130021, Changchun, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Mark AL, Sun Z, Warren DS, Lonze BE, Knabel MK, Williams GM, Locke JE, Montgomery RA, Cameron AM. Stem cell mobilization is life saving in an animal model of acute liver failure. Ann Surg 2010; 252:591-596. [PMID: 20881764 PMCID: PMC5283053 DOI: 10.1097/sla.0b013e3181f4e479] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE No therapy except liver transplantation currently exists for patients with acute liver failure (ALF). The aim of this study was to determine whether pharmacologic mobilization of endogenous hematopoietic stem cells (HSCs) can aid in liver repair and improve survival in an animal model of ALF. METHODS Rodents were treated with a single near-lethal intraperitoneal injection of carbon tetrachloride (CCl4). After 12 hours, animals were randomized to receive plerixafor and granulocyte colony-stimulating factor (G-CSF), agents known to mobilize marrow-derived stem cells, or saline vehicle injection. Mice were observed for survival, and serial assessment of liver injury by serum transaminase measurements, and histologic analysis was performed. RESULTS In our ALF model, 7-day survival after injection of CCl4 was 25%. Administration of plerixafor and G-CSF following CCl4 resulted in 87% survival (n = 8, P < 0.05). On serial histopathologic analysis, animals treated with plerixafor and G-CSF demonstrated less hepatic injury compared with control animals. Evaluation of peripheral blood demonstrated an increase in circulating HSCs in response to plerixafor and G-CSF, and immunostaining suggested the infiltration of HSCs into the hepatic parenchyma after stem cell mobilization. CONCLUSIONS Our results suggest a possible new treatment strategy for patients with ALF, a group for whom either liver transplantation or death is frequently the outcome. Pharmacologic agents that mobilize HSCs may lead to an infiltration of the injured liver with cells that may participate in or expedite liver regeneration. This therapy has the potential to avert liver transplantation in some patients with ALF and may be of benefit in a wide variety of medical and surgical patients with liver injury.
Collapse
Affiliation(s)
- Anthony L. Mark
- Department of Surgery, Walter Reed Army Hospital, Bethesda, Maryland
| | - Zhaoli Sun
- Department of Surgery, Division of Transplantation, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Daniel S. Warren
- Department of Surgery, Division of Transplantation, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Bonnie E. Lonze
- Department of Surgery, Division of Transplantation, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Matthew K. Knabel
- Department of Human Genetics, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - G. Melville Williams
- Department of Surgery, Division of Transplantation, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jayme E. Locke
- Department of Surgery, Division of Transplantation, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert A. Montgomery
- Department of Surgery, Division of Transplantation, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Andrew M. Cameron
- Department of Surgery, Division of Transplantation, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
95
|
Wang J, Zhou X, Cui L, Yan L, Liang J, Cheng X, Qiao L, Shi Y, Han Z, Cao Y, Han Y, Fan D. The significance of CD14+ monocytes in peripheral blood stem cells for the treatment of rat liver cirrhosis. Cytotherapy 2010; 12:1022-34. [PMID: 20849360 DOI: 10.3109/14653249.2010.515578] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND AIMS Circulating monocytes have been exploited as an important progenitor cell resource for hepatocytes in vitro and are instrumental in the removal of fibrosis. We investigated the significance of monocytes in peripheral blood stem cells (PBSC) for the treatment of liver cirrhosis. METHODS Rat CD14+ monocytes in PBSC were mobilized with granulocyte-colony-stimulating factor (G-CSF) and harvested by magnetic cell sorting (MACS). Female rats with carbon tetrachloride (CCl₄-induced liver cirrhosis were injected CM-DiI-labeled monocytes, CD14⁻ cells (1 x 10⁷ cells/rat) or saline via the portal vein. RESULTS Rat CD14+ and CD11b+ monocytes in PBSC were partly positive for CD34, CD45, CD44, Oct3/4 and Sox2, suggesting monocytes with progenitor capacity. Compared with CD14⁻ cell-infused and saline-injected rats, rats undergoing monocyte transplantation showed a gradually increased serum albumin level and decreased portal vein pressure, resulting in a significantly improved survival rate. Meanwhile, monocyte transplantation apparently attenuated liver fibrosis by analysis for fibronectin, α2-(1)-procollagen, α-smooth muscle aorta (SMA) and transforming growth factor (TGF)-β. Transplanted monocytes mainly clustered in periportal areas of liver, in which 1.8% cells expressed hepatocyte marker albumin and CK18. The expression level of hepatocyte growth factor (HGF), TGF-α, extracellular matrix (EGF) and vascular endothelial growth factor (VEGF) increased, while monocyte transplantation enhanced hepatocyte proliferation. On the other hand, the activities and expression of matrix metalloproteinases (MMP) increased while tissue inhibitor of metalloproteinase (TIMP)-1 expression significantly reduced in monocyte-transplanted livers. Some transplanted monocytes expressed MMP-9 and -13. CONCLUSIONS The data suggest that CD14+ monocytes in PBSC contribute to hepatocyte regeneration and extracellular matrix (ECM) remodeling in rat liver cirrhosis much more than CD14⁻ cells, and might offer a therapeutic alternative for patients with liver cirrhosis.
Collapse
Affiliation(s)
- Jingbo Wang
- Division of Hepatology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shannxi Province, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Piscaglia AC, Campanale M, Gasbarrini A, Gasbarrini G. Stem cell-based therapies for liver diseases: state of the art and new perspectives. Stem Cells Int 2010; 2010:259461. [PMID: 21048845 PMCID: PMC2963137 DOI: 10.4061/2010/259461] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Accepted: 07/04/2010] [Indexed: 12/19/2022] Open
Abstract
Millions of patients worldwide suffer from end-stage liver pathologies, whose only curative therapy is liver transplantation (OLT). Given the donor organ shortage, alternatives to OLT have been evaluated, including cell therapies. Hepatocyte transplantation has been attempted to cure metabolic liver disorders and end-stage liver diseases. The evaluation of its efficacy is complicated by the shortage of human hepatocytes and their difficult expansion and cryopreservation. Recent advances in cell biology have led to the concept of "regenerative medicine", based on the therapeutic potential of stem cells (SCs). Different types of SCs are theoretically eligible for liver cell replacement. These include embryonic and fetal SCs, induced pluripotent cells, annex SCs, endogenous liver SCs, and extrahepatic adult SCs. Aim of this paper is to critically analyze the possible sources of SCs suitable for liver repopulation and the results of the clinical trials that have been published until now.
Collapse
Affiliation(s)
- Anna Chiara Piscaglia
- “Gastrointestinal and Liver Stem Cell Research Group” (GILSteR), Department of Internal Medicine, Gemelli Hospital, Catholic University of Rome, Largo A. Gemelli 8-00168 Roma, Italy
| | - Mariachiara Campanale
- “Gastrointestinal and Liver Stem Cell Research Group” (GILSteR), Department of Internal Medicine, Gemelli Hospital, Catholic University of Rome, Largo A. Gemelli 8-00168 Roma, Italy
| | - Antonio Gasbarrini
- “Gastrointestinal and Liver Stem Cell Research Group” (GILSteR), Department of Internal Medicine, Gemelli Hospital, Catholic University of Rome, Largo A. Gemelli 8-00168 Roma, Italy
| | - Giovanni Gasbarrini
- “Gastrointestinal and Liver Stem Cell Research Group” (GILSteR), Department of Internal Medicine, Gemelli Hospital, Catholic University of Rome, Largo A. Gemelli 8-00168 Roma, Italy
| |
Collapse
|
97
|
Stutchfield BM, Forbes SJ, Wigmore SJ. Prospects for stem cell transplantation in the treatment of hepatic disease. Liver Transpl 2010; 16:827-36. [PMID: 20583084 DOI: 10.1002/lt.22083] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Stem cell therapy has the potential to provide a valuable adjunct to the management of hepatic disease. Preclinical studies have demonstrated a range of endogenous repair processes that can be exploited through stem cell therapy. 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. This article reviews the potential for stem cell therapies to enhance hepatic regeneration in acute and chronic hepatic disease and is based on a MEDLINE and PubMed search for English language articles investigating mechanisms of hepatic regeneration and delivery of cell therapies. Two main mechanisms of potential stem cell therapy delivery have emerged: (1) a direct contribution to the functional hepatocyte population with embryonic, induced pluripotent, or adult stem cells and (2) the promotion of endogenous regenerative processes with bone marrow-derived stem cells. Bioartificial hepatic support systems may be proven to be an effective method of using ex vivo differentiated hepatocytes and be indicated as a bridging therapy to definitive surgery in acute liver failure. The administration of bone marrow-derived stem cells may enhance liver regeneration in chronic liver disease after portal vein embolization and could facilitate regeneration after partial hepatic resection. Ultimately, the most appropriate hepatic disease targets for stem cell therapies will become apparent as mechanisms of stem involvement in hepatic regeneration are further elucidated.
Collapse
|
98
|
Abstract
PURPOSE OF REVIEW Patients with liver cirrhosis often require liver transplantation, which remains the only effective treatment of the end-stage cirrhosis. Here we briefly summarize the current concepts in treatment of liver diseases based on the transplantation of intrahepatic liver cells, capable of repopulating the injured liver. These cells include hepatocytes, oval cells (bipotential intrahepatic progenitor cells), bone marrow hematopoietic and mesenchymal stem cells, and induced pluripotent stem (iPS) cells. RECENT FINDINGS Although liver transplantation remains the only conventional treatment, liver cell transplantation is an experimental procedure which has been successfully used in clinical trials in patients with acute liver failure, chronic liver disease with end-stage cirrhosis. Extraordinary progress has been made in the field of hepatic progenitors and iPS. Liver precursor cells (oval cells) are recognized as bipotential precursor cells in the damaged liver. They can rapidly proliferate, change their cellular composition, and differentiate into hepatocytes and cholangiocytes to compensate for the cellular loss and maintain liver homeostasis in animal models of liver injury. Similarly, iPS are somatic cells obtained from patients and differentiated into hepatocytes in vitro. Future studies of iPS are designed to develop of specific conditions to expand and in vitro differentiate somatic cells into functionally mature liver cells. SUMMARY The current review defines and discusses different populations of hepatic cells which can be potentially used for liver cell transplantation to advance the therapy of hepatic cirrhosis.
Collapse
|
99
|
Yannaki E, Psatha N, Athanasiou E, Karponi G, Constantinou V, Papadopoulou A, Tasouli A, Kaloyannidis P, Batsis I, Arsenakis M, Anagnostopoulos A, Fassas A. Mobilization of hematopoietic stem cells in a thalassemic mouse model: implications for human gene therapy of thalassemia. Hum Gene Ther 2010; 21:299-310. [PMID: 19795976 DOI: 10.1089/hum.2009.077] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Granulocyte colony-stimulating factor (G-CSF)-mobilized blood stem cells may become the preferable source of hematopoietic stem cells (HSCs) for gene therapy because of the higher yield of cells compared with conventional bone marrow harvesting. A G-CSF-associated risk of splenic rupture has been recognized in normal donors of HSCs, but limited information is available about the G-CSF effect in the presence of splenomegaly and extramedullary hematopoiesis. We investigated the G-CSF effect in a thalassemic mouse model (HBB(th-3)) as compared with a normal strain (C57BL/6), in terms of safety, mobilization efficacy, and distribution of stem cells among hematopoietic compartments. There was no death or clinical sequelae of splenic rupture in G-CSF-treated animals of either strain; however, hemorrhagic infarcts in the spleen were detected with low frequency in G-CSF-treated HBB(th-3) mice (12.5%). HBB(th-3) mice mobilized less effectively than C57BL/6 mice (Lin(-)Sca-1(+)c-Kit(+) cells/microl of peripheral blood mononuclear cells [PBMCs]: 90 +/- 55 vs. 255 +/- 174, respectively, p = 0.01; CFU-GM/ml PBMCs: 390 +/- 262 vs. 1131 +/- 875, p = 0.01) because of increased splenic trapping of hematopoietic stem and progenitor cells (Lin(-)Sca-1(+)c-Kit(+) cells per spleen (x10(5)): 487 +/- 35 vs. 109 +/- 19.6, p = 0.01; CFU-GM per spleen (x10(2)): 1470 +/- 347 vs. 530 +/- 425, p = 0.0006). Splenectomy restored the mobilization proficiency of thalassemic mice at comparable levels to normal mice and resulted in the development of a hematopoietic compensatory mechanism in the thalassemic liver that protected splenectomized mice from severe anemia. Our data imply that, in view of human gene therapy for thalassemia, either multiple cycles or alternative ways of mobilization may be required for a sufficient yield of transplantable HSCs. In addition, strategies to minimize the risk of G-CSF-induced splenic infarcts should be explored in a clinical setting.
Collapse
Affiliation(s)
- Evangelia Yannaki
- Gene and Cell Therapy Center, Hematology Department-BMT Unit, George Papanicolaou Hospital, Thessaloniki 57010, Greece.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Li B, Cohen A, Hudson TE, Motlagh D, Amrani DL, Duffield JS. Mobilized human hematopoietic stem/progenitor cells promote kidney repair after ischemia/reperfusion injury. Circulation 2010; 121:2211-20. [PMID: 20458011 PMCID: PMC2919223 DOI: 10.1161/circulationaha.109.928796] [Citation(s) in RCA: 131] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Understanding the mechanisms of repair and regeneration of the kidney after injury is of great interest because there are currently no therapies that promote repair, and kidneys frequently do not repair adequately. We studied the capacity of human CD34(+) hematopoietic stem/progenitor cells (HSPCs) to promote kidney repair and regeneration using an established ischemia/reperfusion injury model in mice, with particular focus on the microvasculature. METHODS AND RESULTS Human HSPCs administered systemically 24 hours after kidney injury were selectively recruited to injured kidneys of immunodeficient mice (Jackson Labs, Bar Harbor, Me) and localized prominently in and around vasculature. This recruitment was associated with enhanced repair of the kidney microvasculature, tubule epithelial cells, enhanced functional recovery, and increased survival. HSPCs recruited to kidney expressed markers consistent with circulating endothelial progenitors and synthesized high levels of proangiogenic cytokines, which promoted proliferation of both endothelial and epithelial cells. Although purified HSPCs acquired endothelial progenitor markers once recruited to the kidney, engraftment of human endothelial cells in the mouse capillary walls was an extremely rare event, indicating that human stem cell mediated renal repair is by paracrine mechanisms rather than replacement of vasculature. CONCLUSIONS These studies advance human HSPCs as a promising therapeutic strategy for promoting renal repair after injury.
Collapse
Affiliation(s)
- Bing Li
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|