1
|
Shang Z, Jiang Y, Guan X, Wang A, Ma B. Therapeutic Effects of Stem Cells From Different Source on Renal Ischemia- Reperfusion Injury: A Systematic Review and Network Meta-analysis of Animal Studies. Front Pharmacol 2021; 12:713059. [PMID: 34539400 PMCID: PMC8444551 DOI: 10.3389/fphar.2021.713059] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/17/2021] [Indexed: 12/20/2022] Open
Abstract
Objective: Although stem cell therapy for renal ischemia-reperfusion injury (RIRI) has made immense progress in animal studies, conflicting results have been reported by the investigators. Therefore, we aimed to systematically evaluate the effects of different stem cells on renal function of animals with ischemia-reperfusion injury and to compare the efficacies of stem cells from various sources. Methods: PubMed, Web of Science, Embase, Cochrane, CNKI, VIP, CBM, and WanFang Data were searched for records until April 2021. Two researchers independently conducted literature screening, data extraction, and literature quality evaluation. Results and conclusion: Seventy-two animal studies were included for data analysis. Different stem cells significantly reduced serum creatinine and blood urea nitrogen levels in the early and middle stages (1 and 7 days) compared to the negative control group, however there was no significant difference in the late stage among all groups (14 days); In the early stage (1 day), the renal histopathological score in the stem cell group was significantly lower than that in the negative control group, and there was no significant difference among these stem cells. In addition, there was no significant difference between stem cell and negative control in proliferation of resident cells, however, significantly less apoptosis of resident cells than negative control. In conclusion, the results showed that stem cells from diverse sources could improve the renal function of RIRI animals. ADMSCs and MDMSCs were the most-researched stem cells, and they possibly hold the highest therapeutic potential. However, the quality of evidence included in this study is low, and there are many risks of bias. The exact efficacy of the stem cells and the requirement for further clinical studies remain unclear.
Collapse
Affiliation(s)
- Zhizhong Shang
- Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China.,The Second Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Yanbiao Jiang
- Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China.,The Second Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Xin Guan
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Anan Wang
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Bin Ma
- Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China
| |
Collapse
|
2
|
Packialakshmi B, Stewart IJ, Burmeister DM, Chung KK, Zhou X. Large animal models for translational research in acute kidney injury. Ren Fail 2021; 42:1042-1058. [PMID: 33043785 PMCID: PMC7586719 DOI: 10.1080/0886022x.2020.1830108] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
While extensive research using animal models has improved the understanding of acute kidney injury (AKI), this knowledge has not been translated into effective treatments. Many promising interventions for AKI identified in mice and rats have not been validated in subsequent clinical trials. As a result, the mortality rate of AKI patients remains high. Inflammation plays a fundamental role in the pathogenesis of AKI, and one reason for the failure to translate promising therapeutics may lie in the profound difference between the immune systems of rodents and humans. The immune systems of large animals such as swine, nonhuman primates, sheep, dogs and cats, more closely resemble the human immune system. Therefore, in the absence of a basic understanding of the pathophysiology of human AKI, large animals are attractive models to test novel interventions. However, there is a lack of reviews on large animal models for AKI in the literature. In this review, we will first highlight differences in innate and adaptive immunities among rodents, large animals, and humans in relation to AKI. After illustrating the potential merits of large animals in testing therapies for AKI, we will summarize the current state of the evidence in terms of what therapeutics have been tested in large animal models. The aim of this review is not to suggest that murine models are not valid to study AKI. Instead, our objective is to demonstrate that large animal models can serve as valuable and complementary tools in translating potential therapeutics into clinical practice.
Collapse
Affiliation(s)
| | - Ian J Stewart
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - David M Burmeister
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Kevin K Chung
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Xiaoming Zhou
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
3
|
Alvites RD, Branquinho MV, Sousa AC, Lopes B, Sousa P, Mendonça C, Atayde LM, Maurício AC. Small Ruminants and Its Use in Regenerative Medicine: Recent Works and Future Perspectives. BIOLOGY 2021; 10:biology10030249. [PMID: 33810087 PMCID: PMC8004958 DOI: 10.3390/biology10030249] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 12/16/2022]
Abstract
Simple Summary Small ruminants such as sheep and goats have been increasingly used as animal models due to their dimensions, physiology and anatomy identical to those of humans. Their low costs, ease of accommodation, great longevity and easy handling make them advantageous animals to be used in a wide range of research work. Although there is already a lot of scientific literature describing these species, their use still lacks some standardization. The purpose of this review is to summarize the general principles related to the use of small ruminants as animal models for scientific research. Abstract Medical and translational scientific research requires the use of animal models as an initial approach to the study of new therapies and treatments, but when the objective is an exploration of translational potentialities, classical models fail to adequately mimic problems in humans. Among the larger animal models that have been explored more intensely in recent decades, small ruminants, namely sheep and goats, have emerged as excellent options. The main advantages associated to the use of these animals in research works are related to their anatomy and dimensions, larger than conventional laboratory animals, but very similar to those of humans in most physiological systems, in addition to their low maintenance and feeding costs, tendency to be docile, long life expectancies and few ethical complications raised in society. The most obvious disadvantages are the significant differences in some systems such as the gastrointestinal, and the reduced amount of data that limits the comparison between works and the validation of the characterization essays. Despite everything, recently these species have been increasingly used as animal models for diseases in different systems, and the results obtained open doors for their more frequent and advantageous use in the future. The purpose of this review is to summarize the general principles related to the use of small ruminants as animal models, with a focus on regenerative medicine, to group the most relevant works and results published recently and to highlight the potentials for the near future in medical research.
Collapse
Affiliation(s)
- Rui Damásio Alvites
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Mariana Vieira Branquinho
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Ana Catarina Sousa
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Bruna Lopes
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Patrícia Sousa
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Carla Mendonça
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Luís Miguel Atayde
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Ana Colette Maurício
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
- Correspondence: ; Tel.: +351-919-071-286 or +351-220-428-000
| |
Collapse
|
4
|
Ahmadi A, Rad NK, Ezzatizadeh V, Moghadasali R. Kidney Regeneration: Stem Cells as a New Trend. Curr Stem Cell Res Ther 2020; 15:263-283. [DOI: 10.2174/1574888x15666191218094513] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 12/23/2022]
Abstract
Renal disease is a major worldwide public health problem that affects one in ten people.
Renal failure is caused by the irreversible loss of the structural and functional units of kidney (nephrons)
due to acute and chronic injuries. In humans, new nephrons (nephrogenesis) are generated until
the 36th week of gestation and no new nephron develops after birth. However, in rodents, nephrogenesis
persists until the immediate postnatal period. The postnatal mammalian kidney can partly repair
their nephrons. The kidney uses intrarenal and extra-renal cell sources for maintenance and repair.
Currently, it is believed that dedifferentiation of surviving tubular epithelial cells and presence of resident
stem cells have important roles in kidney repair. Many studies have shown that stem cells obtained
from extra-renal sites such as the bone marrow, adipose and skeletal muscle tissues, in addition
to umbilical cord and amniotic fluid, have potential therapeutic benefits. This review discusses the
main mechanisms of renal regeneration by stem cells after a kidney injury.
Collapse
Affiliation(s)
- Amin Ahmadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Niloofar K. Rad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Vahid Ezzatizadeh
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Reza Moghadasali
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
5
|
Lau RWK, Al‐Rubaie A, Saini S, Wise AF, Ricardo SD. Percutaneous intrarenal transplantation of differentiated induced pluripotent stem cells into newborn mice. Anat Rec (Hoboken) 2020; 303:2603-2612. [DOI: 10.1002/ar.24371] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/22/2019] [Accepted: 12/07/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Ricky W. K. Lau
- Department of Anatomy and Developmental BiologyBiomedical Discovery Institute, Monash University Clayton Victoria Australia
| | - Ali Al‐Rubaie
- Department of Anatomy and Developmental BiologyBiomedical Discovery Institute, Monash University Clayton Victoria Australia
| | - Sheetal Saini
- Department of Anatomy and Developmental BiologyBiomedical Discovery Institute, Monash University Clayton Victoria Australia
| | - Andrea F. Wise
- Department of Anatomy and Developmental BiologyBiomedical Discovery Institute, Monash University Clayton Victoria Australia
| | - Sharon D. Ricardo
- Department of Anatomy and Developmental BiologyBiomedical Discovery Institute, Monash University Clayton Victoria Australia
| |
Collapse
|
6
|
Osman Y, Hamed SM, Barakat NM, Khater S, Gabr M, Mosbah A, Gaballah MA, Shaaban A. Prophylaxis against renal ischemia-reperfusion injury in canine model: Stem cell approach. INDIAN JOURNAL OF UROLOGY : IJU : JOURNAL OF THE UROLOGICAL SOCIETY OF INDIA 2020; 36:44-49. [PMID: 31983826 PMCID: PMC6961430 DOI: 10.4103/iju.iju_114_19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Introduction Stem cell therapy at the time of ischemia/reperfusion (I/R) injury has been hypothesized to attenuate the severity of acute kidney injury and to accelerate the regeneration process in lower animal models. Data in higher animal models is limited and discordant. We aimed to explore the reno-protective effects of stem cells on I/R related renal injury in a canine model. Materials and Methods Twenty-seven dogs that were treated with bone marrow-derived mesenchymal stem cells (BM-MSCs) were compared with another 27 dogs treated with adipose tissue-derived MSCs (AT-MSCs) following 90 min of warm ischemia to assess IR injury. Each group was divided into three subgroups (nine dogs each), according to the stem cell dose (5, 10, 15 × 106 in 500 μl volume) injected directly into the renal cortex after reperfusion. All dogs were re-evaluated by renogram, histopathology, and pro-inflammatory markers at 2 weeks, 2, and 3 months. Results In Group I, there was a mean reduction of creatinine clearance by 78%, 64%, and 74% at the three used doses, respectively, at 2 weeks. At 3 months, these kidneys regained a mean of 84%, 92%, and 72%, respectively, of its basal function. In Group II, the reduction of clearance was much more modest with mean of 14%, 6%, and 24% respectively at 2 weeks with more intense recovery of renal function by mean of 90%, 100%, and 76%, respectively, at 3 months. Group I had significantly more tubular necrosis and delayed regeneration compared with the Group II. Expressions of pro-inflammatory markers were upregulated in both the groups with a higher and more sustained expression in Group I. Conclusion Stem cells protected against ischemic reperfusion injury in a canine model. AT-MSCs provided better protection than BM-MSCs.
Collapse
Affiliation(s)
- Yasser Osman
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Sahar M Hamed
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Nashwa M Barakat
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Sherry Khater
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Mahmoud Gabr
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Ahmed Mosbah
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | | | - Atallah Shaaban
- Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| |
Collapse
|
7
|
Sierra-Parraga JM, Munk A, Andersen C, Lohmann S, Moers C, Baan CC, Ploeg RJ, Pool M, Keller AK, Møller BK, Leuvenink H, Hoogduijn MJ, Jespersen B, Eijken M. Mesenchymal Stromal Cells Are Retained in the Porcine Renal Cortex Independently of Their Metabolic State After Renal Intra-Arterial Infusion. Stem Cells Dev 2019; 28:1224-1235. [PMID: 31280676 DOI: 10.1089/scd.2019.0105] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The regenerative capacities of mesenchymal stromal cells (MSCs) make them suitable for renal regenerative therapy. The most common delivery route of MSC is through intravenous infusion, which is associated with off-target distribution. Renal intra-arterial delivery offers a targeted therapy, but limited knowledge is available regarding the fate of MSCs delivered through this route. Therefore, we studied the efficiency and tissue distribution of MSCs after renal intra-arterial delivery to a porcine renal ischemia-reperfusion model. MSCs were isolated from adipose tissue of healthy male pigs, fluorescently labeled and infused into the renal artery of female pigs. Flow cytometry allowed MSC detection and quantification in tissue and blood. In addition, quantitative polymerase chain reaction was used to trace MSCs by their Y-chromosome. During infusion, a minor number of MSCs left the kidney through the renal vein, and no MSCs were identified in arterial blood. Ischemic and healthy renal tissues were analyzed 30 min and 8 h after infusion, and 1-4 × 104 MSCs per gram of tissue were detected, predominantly, in the renal cortex, with a viability >70%. Confocal microscopy demonstrated mainly glomerular localization of MSCs, but they were also observed in the capillary network around tubuli. The infusion of heat-inactivated (HI) MSCs, which are metabolically inactive, through the renal artery showed that HI-MSCs were distributed in the kidney in a similar manner to regular MSCs, suggesting a passive retention mechanism. Long-term MSC survival was analyzed by Y-chromosome tracing, and demonstrated that a low percentage of the infused MSCs were present in the kidney 14 days after administration, while HI-MSCs were completely undetectable. In conclusion, renal intra-arterial MSC infusion limited off-target engraftment, leading to efficient MSC delivery to the kidney, most of them being cleared within 14 days. MSC retention was independent of the metabolic state of MSC, indicating a passive mechanism.
Collapse
Affiliation(s)
- Jesus M Sierra-Parraga
- Nephrology and Transplantation, Internal Medicine Department, University Medical Center Rotterdam, Erasmus MC, Rotterdam, the Netherlands.,Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Anders Munk
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Stine Lohmann
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark.,Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Cyril Moers
- Department of Surgery-Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Carla C Baan
- Nephrology and Transplantation, Internal Medicine Department, University Medical Center Rotterdam, Erasmus MC, Rotterdam, the Netherlands
| | - Rutger J Ploeg
- Nuffield Department of Surgical Sciences and Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Merel Pool
- Department of Surgery-Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anna K Keller
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Bjarne K Møller
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Henri Leuvenink
- Department of Surgery-Organ Donation and Transplantation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Martin J Hoogduijn
- Nephrology and Transplantation, Internal Medicine Department, University Medical Center Rotterdam, Erasmus MC, Rotterdam, the Netherlands
| | - Bente Jespersen
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Marco Eijken
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
8
|
Gugjoo MB, Amarpal. Mesenchymal stem cell research in sheep: Current status and future prospects. Small Rumin Res 2018. [DOI: 10.1016/j.smallrumres.2018.08.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
9
|
Marcheque J, Bussolati B, Csete M, Perin L. Concise Reviews: Stem Cells and Kidney Regeneration: An Update. Stem Cells Transl Med 2018; 8:82-92. [PMID: 30302937 PMCID: PMC6312445 DOI: 10.1002/sctm.18-0115] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 08/03/2018] [Indexed: 02/06/2023] Open
Abstract
Significant progress has been made to advance stem cell products as potential therapies for kidney diseases: various kinds of stem cells can restore renal function in preclinical models of acute and chronic kidney injury. Nonetheless this literature contains contradictory results, and for this reason, we focus this review on reasons for apparent discrepancies in the literature, because they contribute to difficulty in translating renal regenerative therapies. Differences in methodologies used to derive and culture stem cells, even those from the same source, in addition to the lack of standardized renal disease animal models (both acute and chronic), are important considerations underlying contradictory results in the literature. We propose that harmonized rigorous protocols for characterization, handling, and delivery of stem cells in vivo could significantly advance the field, and present details of some suggested approaches to foster translation in the field of renal regeneration. Our goal is to encourage coordination of methodologies (standardization) and long‐lasting collaborations to improve protocols and models to lead to reproducible, interpretable, high‐quality preclinical data. This approach will certainly increase our chance to 1 day offer stem cell therapeutic options for patients with all‐too‐common renal diseases. Stem Cells Translational Medicine2019;8:82–92
Collapse
Affiliation(s)
- Julia Marcheque
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics, Children's Hospital Los Angeles, Division of Urology, Saban Research Institute, University of Southern California, Los Angeles, California
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Italy
| | - Marie Csete
- Medical Engineering, California Institute of Technology, Los Angeles, California.,Department of Anesthesiology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Laura Perin
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics, Children's Hospital Los Angeles, Division of Urology, Saban Research Institute, University of Southern California, Los Angeles, California
| |
Collapse
|
10
|
Lee SJ, Ryu MO, Seo MS, Park SB, Ahn JO, Han SM, Kang KS, Bhang DH, Youn HY. Mesenchymal Stem Cells Contribute to Improvement of Renal Function in a Canine Kidney Injury Model. ACTA ACUST UNITED AC 2018; 31:1115-1124. [PMID: 29102933 DOI: 10.21873/invivo.11177] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 09/09/2017] [Accepted: 09/15/2017] [Indexed: 12/27/2022]
Abstract
BACKGROUND/AIM The kidney excretes waste materials and regulates important metabolic functions, and renal disorders constitute a significant medical problem and can result in fatalities. In the present study, mesenchymal stem cells derived from canine umbilical cord blood (cUCB-MSCs) were isolated and evaluated for their ability to improve renal function in a canine model of acute kidney injury (AKI). MATERIALS AND METHODS The canine AKI model was developed by i.v. injection of cisplatin and gentamycin into 14 male beagle dogs. cUCB-MSCs were administered into the renal corticomedullary junction following AKI induction. Survival time, clinical signs, blood analysis and histological parameters were analyzed. RESULTS The group treated with AKI plus cUCB-MSCs had decreased blood urea nitrogen and creatinine levels, and showed an extended life-span and improved histological manifestations. MSCs were detected around the tubules of these kidneys at the histological level. CONCLUSION Taken together, our findings suggest that cUCB-MSCs could be an alternative therapeutic agent for canine AKI.
Collapse
Affiliation(s)
- Seung-Jun Lee
- Department of Internal Veterinary Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Min-Ok Ryu
- Department of Internal Veterinary Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Min-Soo Seo
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea
| | - Sang-Bum Park
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.,Laboratory of Stem Cell and Tumor Biology, Department of Veterinary Public Health, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jin-Ok Ahn
- Department of Internal Veterinary Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Sei-Myoung Han
- Department of Internal Veterinary Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.,Laboratory of Stem Cell and Tumor Biology, Department of Veterinary Public Health, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Dong-Ha Bhang
- Department of Molecular and Cellular Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Hwa-Young Youn
- Department of Internal Veterinary Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
11
|
Lipowsky HH, Bowers DT, Banik BL, Brown JL. Mesenchymal Stem Cell Deformability and Implications for Microvascular Sequestration. Ann Biomed Eng 2018; 46:640-654. [PMID: 29352448 PMCID: PMC5862759 DOI: 10.1007/s10439-018-1985-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/16/2018] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cells (MSCs) have received considerable attention in regenerative medicine, particularly in light of prospects for targeted delivery by intra-arterial injection. However, little is known about the mechanics of MSC sequestration in the microvasculature and the yield pressure (PY), above which MSCs will pass through microvessels of a given diameter. The objectives of the current study were to delineate the dependency of PY on cell size and the heterogeneity of cell mechanical properties and diameters (DCELL) of cultured MSCs. To this end the transient filtration test was employed to elucidate the mean filtration pressure (〈PY〉) for an ensemble of pores of a given size (DPORE) similar to in vivo microvessels. Cultured MSCs had a log-normal distribution of cell diameters (DCELL) with a mean of 15.8 ± 0.73 SD μm. MSC clearance from track-etched polycarbonate filters was studied for pore diameters of 7.3-15.4 μm. The pressure required to clear cells from filters with 30-85 × 103 pores rose exponentially with the ratio λ = DCELL/DPORE for 1.1 ≤ λ ≤ 2.2. The clearance of cells from each filter was characterized by a log-normal distribution in PY, with a mean filtration pressure of 0.02 ≤ 〈PY〉 ≤ 6.7 cmH2O. For λ ≤ 1.56, the yield pressure (PY) was well represented by the cortical shell model of a cell with a viscous interior encapsulated by a shell under cortical tension τ0 = 0.99 ± 0.42 SD dyn/cm. For λ > 1.56, the 〈PY〉 characteristic of the cell population rose exponentially with λ. Analysis of the mean filtration pressure (〈PY〉) of each sample suggested that the larger diameter cells that skewed the distribution of DCELL contributed to about 20% of the mean filtration pressure. Further, if all cells had the same deformability (i.e., PY as a function of λ) as the average cell population, then 〈PY〉 would have risen an order of magnitude above the average from fivefold at λ = 1.56 to 200-fold at λ = 2.1. Comparison of 〈PY〉 to published microvascular pressures suggested that 〈PY〉 may exceed microvessel pressure drops for λ exceeding 2.1, and rise 14-fold above capillary pressure drop at λ = 3 leading to 100% sequestration. However, due to the large variance of in vivo microvascular pressures entrapment of MSCs may be mitigated. Thus it is suggested that selecting fractions of the MSC population according to cell deformability may permit optimization of entrapment at sites targeted for tissue regeneration.
Collapse
Affiliation(s)
- Herbert H Lipowsky
- Department of Biomedical Engineering, The Pennsylvania State University, 215 Hallowell Bldg, University Park, PA, 16802, USA.
| | - Daniel T Bowers
- Department of Biomedical Engineering, The Pennsylvania State University, 215 Hallowell Bldg, University Park, PA, 16802, USA
| | - Brittany L Banik
- Department of Biomedical Engineering, The Pennsylvania State University, 215 Hallowell Bldg, University Park, PA, 16802, USA
| | - Justin L Brown
- Department of Biomedical Engineering, The Pennsylvania State University, 215 Hallowell Bldg, University Park, PA, 16802, USA
| |
Collapse
|
12
|
Li W, Wang L, Chu X, Cui H, Bian Y. Icariin combined with human umbilical cord mesenchymal stem cells significantly improve the impaired kidney function in chronic renal failure. Mol Cell Biochem 2017; 428:203-212. [PMID: 28116543 DOI: 10.1007/s11010-016-2930-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 12/23/2016] [Indexed: 12/23/2022]
Abstract
At present, the main therapy for chronic renal failure (CRF) is dialysis and renal transplantation, but neither obtains satisfactory results. Human umbilical cord mesenchymal stem cells (huMSCs) are isolated from the fetal umbilical cord which has a high self-renewal and multi-directional differentiation potential. Icariin (ICA), a kidney-tonifying Chinese Medicine can enhance the multipotency of huMSCs. Therefore, this work seeks to employ the use of ICA-treated huMSCs for the treatment of chronic renal failure. Blood urea nitrogen and creatinine (Cr) analyses showed amelioration of functional parameters in ICA-treated huMSCs for the treatment of CRF rats at 3, 7, and 14 days after transplantation. ICA-treated huMSCs can obviously increase the number of cells in injured renal tissues at 3, 7, and 14 days after transplantation by optical molecular imaging system. Hematoxylin-eosin staining demonstrated that ICA-treated huMSCs reduced the levels of fibrosis in CRF rats at 14 days after transplantation. Superoxide dismutase and Malondialdehyde analyses showed that ICA-treated huMSCs reduced the oxidative damage in CRF rats. Moreover, transplantation with ICA-treated huMSCs decreased inflammatory responses, promoted the expression of growth factors, and protected injured renal tissues. Taken together, our findings suggest that ICA-treated huMSCs could improve the kidney function in CRF rats.
Collapse
Affiliation(s)
- Wen Li
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Nankai University, Tianjin, 300071, China
| | - Li Wang
- Tianjin Second People's Hospital, Tianjin, 300192, China
| | - Xiaoqian Chu
- Tianjin People's Hospital, Tianjin, 300000, China
| | - Huantian Cui
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Yuhong Bian
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| |
Collapse
|
13
|
Stoltz JF, Bensoussan D, De Isla N, Zhang L, Han Z, Magdalou J, Huselstein C, Ye J, Leballe B, Decot V, Reppel L. Stem cells and vascular regenerative medicine: A mini review. Clin Hemorheol Microcirc 2017; 64:613-633. [DOI: 10.3233/ch-168036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- J.-F. Stoltz
- CNRS, UMR 7365, Biopole, Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CHRU de Nancy, Unité de Thérapie Cellulaire et Tissulaire (UTCT) (FR CNRS-INSERM-UHP-CHU), Vandoeuvre-Lès-Nancy, France
| | - D. Bensoussan
- CNRS, UMR 7365, Biopole, Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CHRU de Nancy, Unité de Thérapie Cellulaire et Tissulaire (UTCT) (FR CNRS-INSERM-UHP-CHU), Vandoeuvre-Lès-Nancy, France
| | - N. De Isla
- CNRS, UMR 7365, Biopole, Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
| | - L. Zhang
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- Centre de Recherche, Calmette Hospital, Kunming, China
| | - Z. Han
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- Centre de Recvherche sur les cellules souches, Beijing et Tianjin, China
| | - J. Magdalou
- CNRS, UMR 7365, Biopole, Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
| | - C. Huselstein
- CNRS, UMR 7365, Biopole, Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
| | - J.S. Ye
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- Centre de Recherche, Calmette Hospital, Kunming, China
| | | | - V. Decot
- CNRS, UMR 7365, Biopole, Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CHRU de Nancy, Unité de Thérapie Cellulaire et Tissulaire (UTCT) (FR CNRS-INSERM-UHP-CHU), Vandoeuvre-Lès-Nancy, France
| | - L. Reppel
- CNRS, UMR 7365, Biopole, Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CNRS – GDRI France-Chine « Stem cells and Regenerative medicine », Faculté de Médecine, Vandoeuvre-Lès-Nancy, France
- CHRU de Nancy, Unité de Thérapie Cellulaire et Tissulaire (UTCT) (FR CNRS-INSERM-UHP-CHU), Vandoeuvre-Lès-Nancy, France
| |
Collapse
|
14
|
Lim CY, Han JI, Kim SG, Lee CM, Park HM. Evaluation of autologous bone marrow-derived mesenchymal stem cells on renal regeneration after experimentally induced acute kidney injury in dogs. Am J Vet Res 2016; 77:208-17. [PMID: 27027716 DOI: 10.2460/ajvr.77.2.208] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To evaluate the usefulness of autologous bone marrow-derived mesenchymal stem cell (BM-MSC) therapy for the treatment of dogs with experimentally induced acute kidney injury. ANIMALS 6 healthy dogs. PROCEDURES After induction of kidney injury (day 0) with cisplatin (5 mg/kg, IV), dogs immediately received saline (0.9% NaCl) solution (10 mL; n = 3) or BM-MSCs (1 × 10(6) cells/kg in 10 mL of saline solution; 3) IV. A CBC, serum biochemical analysis, and urinalysis were performed for each dog before administration of cisplatin and on days 1 through 4. Glomerular filtration rate was determined for all dogs on days -7 and 2; BM-MSC tracking by MRI was performed on BM-MSC-treated dogs on days -14 and 4. After sample collection and BM-MSC tracking on day 4, all dogs were euthanized; kidney tissue samples underwent histologic evaluation, immunohistochemical analysis, and cytokine profiling via reverse transcriptase PCR assays. RESULTS Kidney tissue from both groups had mononuclear inflammatory cell infiltration, tubular necrosis, dilated tubules, and glomerular damage. However, there was less fibrotic change and increased proliferation of renal tubular epithelial cells in the BM-MSC-treated dogs, compared with findings for the control dogs. Expressions of tumor necrosis factor-α and transforming growth factor-β were lower in the BM-MSC-treated group, compared with findings for the control group. Laboratory data revealed no improvement in the renal function in BM-MSC-treated dogs. CONCLUSIONS AND CLINICAL RELEVANCE Results of this study suggested that autologous BM-MSCs may accelerate renal regeneration after experimentally induced acute kidney injury in dogs.
Collapse
|
15
|
Fate and Effect of Intravenously Infused Mesenchymal Stem Cells in a Mouse Model of Hepatic Ischemia Reperfusion Injury and Resection. Stem Cells Int 2016; 2016:5761487. [PMID: 26981132 PMCID: PMC4766354 DOI: 10.1155/2016/5761487] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 01/06/2016] [Accepted: 01/12/2016] [Indexed: 01/13/2023] Open
Abstract
Liver ischemia reperfusion injury (IRI) is inevitable during transplantation and resection and is characterized by hepatocellular injury. Therapeutic strategies to reduce IRI and accelerate regeneration could offer major benefits. Mesenchymal stem cells (MSC) are reported to have anti-inflammatory and regeneration promoting properties. We investigated the effect of MSC in a model of combined IRI and partial resection in the mouse. Hepatic IRI was induced by occlusion of 70% of the blood flow during 60 minutes, followed by 30% hepatectomy. 2 × 105 MSC or PBS were infused 2 hours before or 1 hour after IRI. Six, 48, and 120 hours postoperatively mice were sacrificed. Liver damage was evaluated by liver enzymes, histology, and inflammatory markers. Regeneration was determined by liver/body weight ratio, proliferating hepatocytes, and TGF-β levels. Fate of MSC was visualized with 3D cryoimaging. Infusion of 2 × 105 MSC 2 hours before or 1 hour after IRI and resection showed no beneficial effects. Tracking revealed that MSC were trapped in the lungs and did not migrate to the site of injury and many cells had already disappeared 2 hours after infusion. Based on these findings we conclude that intravenously infused MSC disappear rapidly and were unable to induce beneficial effects in a clinically relevant model of IRI and resection.
Collapse
|
16
|
Improving the outcome of kidney transplantation by ameliorating renal ischemia reperfusion injury: lost in translation? J Transl Med 2016; 14:20. [PMID: 26791565 PMCID: PMC4721068 DOI: 10.1186/s12967-016-0767-2] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 12/20/2015] [Indexed: 01/03/2023] Open
Abstract
Kidney transplantation is the treatment of choice in patients with end stage renal disease. During kidney transplantation ischemia reperfusion injury (IRI) occurs, which is a risk factor for acute kidney injury, delayed graft function and acute and chronic rejection. Kidneys from living donors show a superior short- and long-term graft survival compared with deceased donors. However, the shortage of donor kidneys has resulted in expansion of the donor pool by using not only living- and brain death donors but also kidneys from donation after circulatory death and from extended criteria donors. These grafts are associated with an increased sensitivity to IRI and decreased graft outcome due to prolonged ischemia and donor comorbidity. Therefore, preventing or ameliorating IRI may improve graft survival. Animal experiments focus on understanding the mechanism behind IRI and try to find methods to minimize IRI either before, during or after ischemia. This review evaluates the different experimental strategies that have been investigated to prevent or ameliorate renal IRI. In addition, we review the current state of translation to the clinical setting. Experimental research has contributed to the development of strategies to prevent or ameliorate IRI, but promising results in animal studies have not yet been successfully translated to clinical use.
Collapse
|
17
|
Stem Cells and Regenerative Medicine: Myth or Reality of the 21th Century. Stem Cells Int 2015; 2015:734731. [PMID: 26300923 PMCID: PMC4537770 DOI: 10.1155/2015/734731] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 04/22/2015] [Accepted: 05/24/2015] [Indexed: 02/07/2023] Open
Abstract
Since the 1960s and the therapeutic use of hematopoietic stem cells of bone marrow origin, there has been an increasing interest in the study of undifferentiated progenitors that have the ability to proliferate and differentiate into various tissues. Stem cells (SC) with different potency can be isolated and characterised. Despite the promise of embryonic stem cells, in many cases, adult or even fetal stem cells provide a more interesting approach for clinical applications. It is undeniable that mesenchymal stem cells (MSC) from bone marrow, adipose tissue, or Wharton's Jelly are of potential interest for clinical applications in regenerative medicine because they are easily available without ethical problems for their uses. During the last 10 years, these multipotent cells have generated considerable interest and have particularly been shown to escape to allogeneic immune response and be capable of immunomodulatory activity. These properties may be of a great interest for regenerative medicine. Different clinical applications are under study (cardiac insufficiency, atherosclerosis, stroke, bone and cartilage deterioration, diabetes, urology, liver, ophthalmology, and organ's reconstruction). This review focuses mainly on tissue and organ regeneration using SC and in particular MSC.
Collapse
|
18
|
Luu NT, McGettrick HM, Buckley CD, Newsome PN, Rainger GE, Frampton J, Nash GB. Crosstalk between mesenchymal stem cells and endothelial cells leads to downregulation of cytokine-induced leukocyte recruitment. Stem Cells 2015; 31:2690-702. [PMID: 23939932 DOI: 10.1002/stem.1511] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 07/22/2013] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSC) have immunomodulatory properties, but their effects on endothelial cells (EC) and recruitment of leukocytes are unknown. We cocultured human bone marrow-derived MSC with EC and found that MSC could downregulate adhesion of flowing neutrophils or lymphocytes and their subsequent transendothelial migration. This applied for EC treated with tumor necrosis factor-α (TNF), interleukin-1β (IL-1), or TNF and interferon-γ combined. Supernatant from cocultures also inhibited endothelial responses. This supernatant had much higher levels of IL-6 than supernatant from cultures of the individual cells, which also lacked inhibitory functions. Addition of neutralizing antibody against IL-6 removed the bioactivity of the supernatant and also the immunomodulatory effects of coculture. Studies using siRNA showed that IL-6 came mainly from the MSC in coculture, and reduction in production in MSC alone was sufficient to impair the protective effects of coculture. Interestingly, siRNA knockdown of IL-6-receptor expression in MSC as well as EC inhibited anti-inflammatory effects. This was explained when we detected soluble IL-6R receptor in supernatants and showed that receptor removal reduced the potency of supernatant. Neutralization of transforming growth factor-β indicated that activation of this factor in coculture contributed to IL-6 production. Thus, crosstalk between MSC and EC caused upregulation of production of IL-6 by MSC which in turn downregulated the response of EC to inflammatory cytokines, an effect potentiated by MSC release of soluble IL-6R. These studies establish a novel mechanism by which MSC might have protective effects against inflammatory pathology and cardiovascular disease.
Collapse
Affiliation(s)
- N Thin Luu
- Centre for Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
19
|
Moghadasali R, Azarnia M, Hajinasrollah M, Arghani H, Nassiri SM, Molazem M, Vosough A, Mohitmafi S, Najarasl M, Ajdari Z, Yazdi RS, Bagheri M, Ghanaati H, Rafiei B, Gheisari Y, Baharvand H, Aghdami N. Intra-renal arterial injection of autologous bone marrow mesenchymal stromal cells ameliorates cisplatin-induced acute kidney injury in a rhesus Macaque mulatta monkey model. Cytotherapy 2014; 16:734-49. [PMID: 24801377 DOI: 10.1016/j.jcyt.2014.01.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 12/26/2013] [Accepted: 01/08/2014] [Indexed: 12/29/2022]
Abstract
BACKGROUND Clinically, acute kidney injury (AKI) is a potentially devastating condition for which no specific therapy improves efficacy of the repair process. Bone marrow mesenchymal stromal cells (BM-MSCs) are proven to be beneficial for the renal repair process after AKI in different experimental rodent models, but their efficacy in large animals and humans remains unknown. This study aims to assess the effect of autologous rhesus Macaque mulatta monkey BM-MSC transplantation in cisplatin-induced AKI. METHODS We chose a model of AKI induced by intravenous administration of 5 mg/kg cisplatin. BM-MSCs were transplanted through intra-arterial injection. The animals were followed for survival, biochemistry analysis and pathology. RESULTS Transplantation of 5 × 10(6) cells/kg ameliorated renal function during the first week, as shown by significantly lower serum creatinine and urea values and higher urine creatinine and urea clearance without hyponatremia, hyperkalemia, proteinuria and polyuria up to 84 d compared with the vehicle and control groups. The superparamagnetic iron oxide nanoparticle-labeled cells were found in both the glomeruli and tubules. BM-MSCs markedly accelerated Foxp3+ T-regulatory cells in response to cisplatin-induced damage, as revealed by higher numbers of Foxp3+ cells within the tubuli of these monkeys compared with cisplatin-treated monkeys in the control and vehicle groups. CONCLUSIONS These data demonstrate that BM-MSCs in this unique large-animal model of cisplatin-induced AKI exhibited recovery and protective properties.
Collapse
Affiliation(s)
- Reza Moghadasali
- Department of Biology, Kharazmi University, Tehran, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenerative Medicine at the Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahnaz Azarnia
- Department of Biology, Kharazmi University, Tehran, Iran
| | - Mostafa Hajinasrollah
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hassan Arghani
- Urology and Nephrology Research Center, Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mahdi Nassiri
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mohammad Molazem
- Department of Veterinary Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ahmad Vosough
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Soroush Mohitmafi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Clinical Sciences, Faculty of Veterinary Medicine, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Mostafa Najarasl
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Zahra Ajdari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Reza Salman Yazdi
- Department of Andrology at the Reproductive Biomedicine Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mohsen Bagheri
- Department of Andrology at the Reproductive Biomedicine Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Hossein Ghanaati
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Sciences, Tehran, Iran
| | - Behrooz Rafiei
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Sciences, Tehran, Iran
| | - Yousof Gheisari
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenerative Medicine at the Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, University of Science and Culture, ACECR, Tehran, Iran
| | - Nasser Aghdami
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenerative Medicine at the Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
20
|
Bruno S, Chiabotto G, Camussi G. Concise review: different mesenchymal stromal/stem cell populations reside in the adult kidney. Stem Cells Transl Med 2014; 3:1451-5. [PMID: 25355731 DOI: 10.5966/sctm.2014-0142] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
During fetal life, mesenchymal stromal/stem cells (MSCs) surround glomeruli and tubules and contribute to the development of the renal interstitium by secretion of growth factors that drive nephron differentiation. In the adult, an MSC-like population has been demonstrated in different compartments of human and murine nephrons. After injury, these cells might provide support for kidney regeneration by recapitulating the role they have in embryonic life. In this short review, we discuss the evidence of an MSC presence within the adult kidney and their potential contribution to the turnover of renal cells and injury repair.
Collapse
Affiliation(s)
- Stefania Bruno
- Departments of Molecular Biotechnology and Health Science and Medical Sciences, University of Torino, Torino, Italy
| | - Giulia Chiabotto
- Departments of Molecular Biotechnology and Health Science and Medical Sciences, University of Torino, Torino, Italy
| | - Giovanni Camussi
- Departments of Molecular Biotechnology and Health Science and Medical Sciences, University of Torino, Torino, Italy
| |
Collapse
|
21
|
Kilpinen L, Impola U, Sankkila L, Ritamo I, Aatonen M, Kilpinen S, Tuimala J, Valmu L, Levijoki J, Finckenberg P, Siljander P, Kankuri E, Mervaala E, Laitinen S. Extracellular membrane vesicles from umbilical cord blood-derived MSC protect against ischemic acute kidney injury, a feature that is lost after inflammatory conditioning. J Extracell Vesicles 2013; 2:21927. [PMID: 24349659 PMCID: PMC3860334 DOI: 10.3402/jev.v2i0.21927] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 10/31/2013] [Accepted: 11/12/2013] [Indexed: 12/21/2022] Open
Abstract
Background Mesenchymal stromal cells (MSC) are shown to have a great therapeutic potential in many immunological disorders. Currently the therapeutic effect of MSCs is considered to be mediated via paracrine interactions with immune cells. Umbilical cord blood is an attractive but still less studied source of MSCs. We investigated the production of extracellular membrane vesicles (MVs) from human umbilical cord blood derived MSCs (hUCBMSC) in the presence (MVstim) or absence (MVctrl) of inflammatory stimulus. Methods hUCBMSCs were cultured in serum free media with or without IFN-γ and MVs were collected from conditioned media by ultracentrifugation. The protein content of MVs were analyzed by mass spectrometry. Hypoxia induced acute kidney injury rat model was used to analyze the in vivo therapeutic potential of MVs and T-cell proliferation and induction of regulatory T cells were analyzed by co-culture assays. Results Both MVstim and MVctrl showed similar T-cell modulation activity in vitro, but only MVctrls were able to protect rat kidneys from reperfusion injury in vivo. To clarify this difference in functionality we made a comparative mass spectrometric analysis of the MV protein contents. The IFN-γ stimulation induced dramatic changes in the protein content of the MVs. Complement factors (C3, C4A, C5) and lipid binding proteins (i.e apolipoproteins) were only found in the MVctrls, whereas the MVstim contained tetraspanins (CD9, CD63, CD81) and more complete proteasome complex accompanied with MHCI. We further discovered that differently produced MV pools contained specific Rab proteins suggesting that same cells, depending on external signals, produce vesicles originating from different intracellular locations. Conclusions We demonstrate by both in vitro and in vivo models accompanied with a detailed analysis of molecular characteristics that inflammatory conditioning of MSCs influence on the protein content and functional properties of MVs revealing the complexity of the MSC paracrine regulation.
Collapse
Affiliation(s)
| | - Ulla Impola
- Finnish Red Cross Blood Service, Helsinki, Finland
| | | | - Ilja Ritamo
- Finnish Red Cross Blood Service, Helsinki, Finland
| | - Maria Aatonen
- Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Helsinki, Finland
| | | | | | - Leena Valmu
- Finnish Red Cross Blood Service, Helsinki, Finland
| | | | - Piet Finckenberg
- Department of Pharmacology, Institute of Biomedicine, University of Helsinki, Helsinki, Finland
| | - Pia Siljander
- Division of Biochemistry and Biotechnology, Department of Biosciences, University of Helsinki, Helsinki, Finland ; Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | | | | | | |
Collapse
|
22
|
de Almeida DC, Donizetti-Oliveira C, Barbosa-Costa P, Origassa CST, Câmara NOS. In search of mechanisms associated with mesenchymal stem cell-based therapies for acute kidney injury. Clin Biochem Rev 2013; 34:131-144. [PMID: 24353358 PMCID: PMC3866950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Acute kidney injury (AKI) is classically described as a rapid loss of kidney function. AKI affects more than 15% of all hospital admissions and is associated with elevated mortality rates. Although many advances have occurred, intermittent or continuous renal replacement therapies are still considered the best options for reversing mild and severe AKI syndrome. For this reason, it is essential that innovative and effective therapies, without side effects and complications, be developed to treat AKI and the end-stages of renal disease. Mesenchymal stem cell (MSC) based therapies have numerous advantages in helping to repair inflamed and damaged tissues and are being considered as a new alternative for treating kidney injuries. Numerous experimental models have shown that MSCs can act via differentiation-independent mechanisms to help renal recovery. Essentially, MSCs can secrete a pool of cytokines, growth factors and chemokines, express enzymes, interact via cell-to-cell contacts and release bioagents such as microvesicles to orchestrate renal protection. In this review, we propose seven distinct properties of MSCs which explain how renoprotection may be conferred: 1) anti-inflammatory; 2) pro-angiogenic; 3) stimulation of endogenous progenitor cells; 4) anti-apoptotic; 5) anti-fibrotic; 6) anti-oxidant; and 7) promotion of cellular reprogramming. In this context, these mechanisms, either individually or synergically, could induce renal protection and functional recovery. This review summarises the most important effects and benefits associated with MSC-based therapies in experimental renal disease models and attempts to clarify the mechanisms behind the MSC-related renoprotection. MSCs may prove to be an effective, innovative and affordable treatment for moderate and severe AKI. However, more studies need to be performed to provide a more comprehensive global understanding of MSC-related therapies and to ensure their safety for future clinical applications.
Collapse
Affiliation(s)
- Danilo C de Almeida
- Department of Medicine, Division of Nephrology, Universidade Federal de São Paulo, Brazil
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Science IV, Universidade de São Paulo, Brazil
| | | | | | - Clarice ST Origassa
- Department of Medicine, Division of Nephrology, Universidade Federal de São Paulo, Brazil
| | - Niels OS Câmara
- Department of Medicine, Division of Nephrology, Universidade Federal de São Paulo, Brazil
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Science IV, Universidade de São Paulo, Brazil
| |
Collapse
|
23
|
Calloni R, Viegas GS, Türck P, Bonatto D, Pegas Henriques JA. Mesenchymal stromal cells from unconventional model organisms. Cytotherapy 2013; 16:3-16. [PMID: 24113426 DOI: 10.1016/j.jcyt.2013.07.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 07/22/2013] [Accepted: 07/23/2013] [Indexed: 12/23/2022]
Abstract
Mesenchymal stromal cells (MSCs) are multipotent, plastic, adherent cells able to differentiate into osteoblasts, chondroblasts and adipocytes. MSCs can be isolated from many different body compartments of adult and fetal individuals. The most commonly studied MSCs are isolated from humans, mice and rats. However, studies are also being conducted with the use of MSCs that originate from different model organisms, such as cats, dogs, guinea pigs, ducks, chickens, buffalo, cattle, sheep, goats, horses, rabbits and pigs. MSCs derived from unconventional model organisms all present classic fibroblast-like morphology, the expression of MSC-associated cell surface markers such as CD44, CD73, CD90 and CD105 and the absence of CD34 and CD45. Moreover, these MSCs have the ability to differentiate into osteoblasts, chondroblasts and adipocytes. The MSCs isolated from unconventional model organisms are being studied for their potential to heal different tissue defects and injuries and for the development of scaffold compositions that improve the proliferation and differentiation of MSCs for tissue engineering.
Collapse
Affiliation(s)
- Raquel Calloni
- Centro de Biotecnologia da Universidade Federal do Rio Grande do Sul, Departamento de Biologia Molecular e Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), RS, Brazil
| | - Gabrihel Stumpf Viegas
- Centro de Biotecnologia da Universidade Federal do Rio Grande do Sul, Departamento de Biologia Molecular e Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), RS, Brazil
| | - Patrick Türck
- Centro de Biotecnologia da Universidade Federal do Rio Grande do Sul, Departamento de Biologia Molecular e Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), RS, Brazil
| | - Diego Bonatto
- Centro de Biotecnologia da Universidade Federal do Rio Grande do Sul, Departamento de Biologia Molecular e Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), RS, Brazil.
| | - João Antonio Pegas Henriques
- Centro de Biotecnologia da Universidade Federal do Rio Grande do Sul, Departamento de Biologia Molecular e Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), RS, Brazil
| |
Collapse
|
24
|
Bagul A, Frost JH, Drage M. Stem cells and their role in renal ischaemia reperfusion injury. Am J Nephrol 2013; 37:16-29. [PMID: 23295823 DOI: 10.1159/000345731] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 11/07/2012] [Indexed: 01/03/2023]
Abstract
BACKGROUND Ischaemia-reperfusion injury (IRI) remains one of the leading causes of acute kidney injury (AKI). IRI is an underlying multifactorial pathophysiological process which affects the outcome in both native and transplanted patients. The high morbidity and mortality associated with IRI/AKI and disappointing results from current available clinical therapeutic approaches prompt further research. Stem cells (SC) are undifferentiated cells that can undergo both renewal and differentiation into one or more cell types which can possibly ameliorate IRI. AIM To carry out a detailed literature analysis and construct a comprehensive literature review addressing the role of SC in AKI secondary to IRI. METHODS Evidence favouring the role of SC in renal IRI and evidence showing no benefits of SC in renal IRI are the two main aspects to be studied. The search strategy was based on an extensive search addressing MESH terms and free text terms. RESULTS The majority of studies in the field of renal IRI and stem cell therapy show substantial benefits. CONCLUSIONS Studies were mostly conducted in small animal models, thus underscoring the need for further pre-clinical studies in larger animal models, and results should be taken with caution. SC therapy may be promising though controversy exists in the exact mechanism. Thorough scientific exploration is required to assess mechanism, safety profile, reproducibility and methods to monitor administered SC.
Collapse
Affiliation(s)
- Atul Bagul
- Department of Transplantation, MRC Centre for Transplantation, Guys and St. Thomas' NHS Foundation Trust, London, UK.
| | | | | |
Collapse
|
25
|
Gheisari Y, Azadmanesh K, Ahmadbeigi N, Nassiri SM, Golestaneh AF, Naderi M, Vasei M, Arefian E, Mirab-Samiee S, Shafiee A, Soleimani M, Zeinali S. Genetic modification of mesenchymal stem cells to overexpress CXCR4 and CXCR7 does not improve the homing and therapeutic potentials of these cells in experimental acute kidney injury. Stem Cells Dev 2012; 21:2969-2980. [PMID: 22563951 DOI: 10.1089/scd.2011.0588] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The therapeutic potential of bone marrow mesenchymal stem cells (MSCs) in kidney failure has been examined in some studies. However, recent findings indicate that after transplantation, these cells home to kidneys at very low levels. Interaction of stromal derived factor-1 (SDF-1) with its receptor, CXCR4, is of pivotal importance in migration and homing. Recently, CXCR7 has also been recognized as another SDF-1 receptor that interacts with CXCR4 and modulates its functions. In this study, CXCR4 and CXCR7 were separately and simultaneously overexpressed in BALB/c bone marrow MSCs by using a lentiviral vector system and the homing and renoprotective potentials of these cells were evaluated in a mouse model of cisplatin-induced acute kidney injury. Using flow cytometry, immunohistochemistry, and real-time PCR methods for detection of GFP-labeled MSCs, we found that although considerably entrapped in lungs, native MSCs home very rarely to kidneys and bone marrow and this rate cannot be significantly affected by CXCR4 and/or CXCR7 upregulation. Transplantation of neither native nor genetically engineered MSCs ameliorated kidney failure. We concluded that overexpression of CXCR4 and CXCR7 receptors in murine MSCs cannot improve the homing and therapeutic potentials of these cells and it can be due to severe chromosomal abnormalities that these cells bear during ex vivo expansion.
Collapse
Affiliation(s)
- Yousof Gheisari
- Department of Molecular Medicine, Pasteur Institute of Iran, Tehran, Iran
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
He J, Wang Y, Sun S, Yu M, Wang C, Pei X, Zhu B, Wu J, Zhao W. Bone marrow stem cells-derived microvesicles protect against renal injury in the mouse remnant kidney model. Nephrology (Carlton) 2012; 17:493-500. [PMID: 22369283 DOI: 10.1111/j.1440-1797.2012.01589.x] [Citation(s) in RCA: 158] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
AIMS Several studies have demonstrated administration of mesenchymal stem cells (MSC) could reverse kidney injury by paracrine mechanisms rather than by MSC transdifferentiation. Recently, a few researchers found microvesicles (MV) derived from MSC might be a paracrine mechanism for cell-to-cell communication. The aim of this study was to investigate the repair effects of MV in a 5/6 subtotal nephrectomy (Nx) mice model. METHODS The animals were randomly divided into four groups: Control, Nx, Nx + MSC and Nx + MV group. MSC were injected (1 × 10(6) /mouse) through caudal vein in Nx + MSC group at the second day after the surgery and MV were injected (30 µg/mouse) through caudal vein in Nx + MV group on alternate days. Mice were killed on day 7 after the first time of administration. Blood urea nitrogen (BUN), serum creatinine (Scr), uric acid (UA) and proteinuria were evaluated. Histopathology of kidney was analysed. RESULTS In Nx mice, the levels of Scr, UA and proteinuria were significantly decreased with administration of MV and MSC (P < 0.05). The remnant kidneys of MV and MSC-treated Nx mice showed less fibrosis, interstitial lymphocyte infiltrates and less or absent tubular atrophy compared with the untreated Nx group. The Histological Score of Kidney in untreated mice was 3.13 ± 0.74, while in the MSC-treated group it was 1.67 ± 0.47 and in the MV-treated group it was 1.80 ± 0.44, nearly preserving normal morphology of the kidney (P < 0.01). CONCLUSION This study showed MV protects against renal injury induced by 5/6 Nx, which could mimic the role of MSC in kidney repair. The research showed a newly potential therapeutic approach to kidney diseases.
Collapse
Affiliation(s)
- Juan He
- Department of Geriatrics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Shi Q, Hodara V, Simerly CR, Schatten GP, VandeBerg JL. Ex vivo reconstitution of arterial endothelium by embryonic stem cell-derived endothelial progenitor cells in baboons. Stem Cells Dev 2012; 22:631-42. [PMID: 22931470 DOI: 10.1089/scd.2012.0313] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
There is an increasing need for an animal model that can be used to translate basic research into clinical therapy. We documented the differentiation and functional competence of embryonic stem cell (ESC)-derived endothelial cells in baboons. Baboon angioblasts were sequentially differentiated from embryoid body cultures for 9 days in an angioblast differentiation medium with varying concentrations of BMP-4, FLT-3 ligand, stem cell factor, thrombopoietin, basic fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), and knockout serum replacement. Real-time polymerase chain reaction results showed that ESC-derived angioblasts downregulated NANOG and OCT3/4, upregulated T-brachyury and GATA2, and moderately expressed CD34; they did not express CD144, TEK, or VWF, and varied in levels of CD31 expression. Several populations of putative angioblasts appeared 3 days and 9 days after differentiation, as identified by flow cytometry. Angioblasts at this stage exhibited dual paths of differentiation toward hematopoietic and vascular fates. To examine whether derived angioblasts could reconstitute the endothelium, we built an ex vivo culture system and seeded fluorescently labeled angioblast cultures onto a denuded segment of the femoral artery. We found that the seeded cells were able to grow into the endothelium on the interior surface of denuded artery segments within 5 days after seeding. After 14 days of ex vivo culture, the transplanted cells expressed CD31, an endothelial marker. The control arteries, seeded with vehicle only, did not harbor cells with endothelial markers. We conclude that ESC-derived angioblasts are promising therapeutic agents for repairing damaged vasculature, and that the baboon model will be vital for optimizing therapies for human clinical studies.
Collapse
Affiliation(s)
- Qiang Shi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas 78245-0549, USA.
| | | | | | | | | |
Collapse
|
28
|
Niu SH, Jian LG, Zhang LH. Protective Effects of Mesenchymal Stem Cells with Transient Overexpression of Hmgb1 on Balloon-Induced Carotid Artery Injury. EUR J INFLAMM 2012. [DOI: 10.1177/1721727x1201000310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Mesenchymal stem cells (MSC) play a crucial role in endothelial repair after artery injury. The high mobility group box 1 (HMGB1) is a key modulator of the homing of MSC to impaired artery and endothelialization. This study was aimed to determine whether balloon-induced carotid artery injury could be improved by transplantation with MSC modified by HMGB1. MSC were infected by adenoviral serotype 5 encoding recombinant green fluorescent protein (GFP) gene and HMGB1 (ad5GFP-HMGB1). The expression of HMGB1, vascular endothelial growth factor (VEGF) and proliferating cell nuclear antigen (PCNA) was detected in MSC using Real-time PCR, Western blot and semi-quantitative immunohistochemical assays. In vivo, reendothelialization was examined in rats subjected to carotid artery injury. The homing of MSC was observed under fluorescence microscopy, and the levels of serum tumor necrosis factor-α (TNF-α) and C-reactive protein (CRP) was assessed by ELISA assay. As a result, compared with the MSC group, the expression of HMGB1, VEGF and PCNA was markedly increased, vascular reendothelialization was accelerated, and the levels of serum TNF-α and CRP were decreased in group ad5GFP and ad5GFP-HMGB1. Transplantation of MSC infected with adGFP-HMGB1 strengthened the MSC effect. Taken together, modification of HMGB1 can enhance the protective effects of MSC on balloon-induced carotid artery injury through up-regulation of VEGF and PCNA expression and inhibition of the inflammatory response. HMGB1 in MSC may represent a novel therapeutic target for the treatment of endothelial repair.
Collapse
Affiliation(s)
- S-H. Niu
- Department of Cardiology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - L-G. Jian
- Department of Cardiology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - L-H. Zhang
- Department of Cardiology, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
29
|
Stem cells and regenerative medicine: accomplishments to date and future promise. Ther Deliv 2012; 1:693-705. [PMID: 21113422 DOI: 10.4155/tde.10.57] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
More than 50 years have passed since the first allogeneic hematopoietic stem cell transplant in patients; however, the promise of other stem cell populations for tissue replacement and repair remains unachieved. When considering cell-based interventions for personalized medicine, the factors influencing therapeutic success and safety are more complicated than for traditional small-molecule pharmacological agents and protein biologics. Failure to progress personalized stem cell therapies to the clinic has resulted from complications that include an incomplete understanding of developmental programs and the diversity of host-donor interactions. In order to more rapidly extend the use of stem cells to the clinic, a better understanding of the different stem cell sources and the implications of their host interactions is required. In this review, we introduce the currently available sources and highlight recent literature that instructs the potential and limitations of their use.
Collapse
|
30
|
Doorn J, Moll G, Le Blanc K, van Blitterswijk C, de Boer J. Therapeutic applications of mesenchymal stromal cells: paracrine effects and potential improvements. TISSUE ENGINEERING PART B-REVIEWS 2011; 18:101-15. [PMID: 21995703 DOI: 10.1089/ten.teb.2011.0488] [Citation(s) in RCA: 236] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Among the various types of cell-to-cell signaling, paracrine signaling comprises those signals that are transmitted over short distances between different cell types. In the human body, secreted growth factors and cytokines instruct, among others, proliferation, differentiation, and migration. In the hematopoietic stem cell (HSC) niche, stromal cells provide instructive cues to stem cells via paracrine signaling and one of these cell types, known to secrete a broad panel of growth factors and cytokines, is mesenchymal stromal cells (MSCs). The factors secreted by MSCs have trophic, immunomodulatory, antiapoptotic, and proangiogenic properties, and their paracrine profile varies according to their initial activation by various stimuli. MSCs are currently studied as treatment for inflammatory diseases such as graft-versus-host disease and Crohn's disease, but also as treatment for myocardial infarct and solid organ transplantation. In addition, MSCs are investigated for their use in tissue engineering applications, in which their differentiation plays an important role, but as we have recently demonstrated, their trophic factors may also be involved. Furthermore, a functional improvement of MSCs might be obtained after preconditioning or tailoring the cells themselves. Also, the way the cells are clinically administered may be specialized for specific therapeutic scenarios. In this review we will first discuss the HSC niche, in which MSCs were recently identified and are thought to play an instructive and supportive role. We will then evaluate therapeutic applications that currently try to utilize the trophic and/or immunomodulatory properties of MSCs, and we will also discuss new options to enhance their therapeutic effects.
Collapse
Affiliation(s)
- Joyce Doorn
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | | | | | | | | |
Collapse
|
31
|
Harari-Steinberg O, Pleniceanu O, Dekel B. Selecting the optimal cell for kidney regeneration: fetal, adult or reprogrammed stem cells. Organogenesis 2011; 7:123-34. [PMID: 21519195 DOI: 10.4161/org.7.2.15783] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Chronic kidney disease (CKD) is a progressive loss in renal function over a period of months or years. End-stage renal disease (ESRD) or stage 5 CKD ensues when renal function deteriorates to under 15% of the normal range. ESRD requires either dialysis or, preferentially, a kidney organ allograft, which is severely limited due to organ shortage for transplantation. To combat this situation, one needs to either increase supply of organs or decrease their demand. Two strategies therefore exist: for those that have completely lost their kidney function (ESRD), we will need to supply new kidneys. Taking into account the kidneys' extremely complex structure, this may prove to be impossible in the near future. In contrast, for those patients that are in the slow progression route from CKD to ESRD but still have functional kidneys, we might be able to halt progression by introducing stem cell therapy to diseased kidneys to rejuvenate or regenerate individual cell types. Multiple cell compartments that fall into three categories are likely to be worthy targets for cell repair: vessels, stroma (interstitium) and nephron epithelia. Different stem/progenitor cells can be linked to regeneration of specific cell types; hematopoietic progenitors and hemangioblastic cell types have specific effects on the vascular niche (vasculogenesis and angiogenesis). Multipotent stromal cells (MSC), whether derived from the bone marrow or isolated from the kidney's non-tubular compartment, may, in turn, heal nephron epithelia via paracrine mechanisms. Nevertheless, as we now know that all of the above lack nephrogenic potential, we should continue our quest to derive genuine nephron (epithelial) progenitors from differentiated pluripotent stem cells, from fetal and adult kidneys and from directly reprogrammed somatic cells.
Collapse
Affiliation(s)
- Orit Harari-Steinberg
- The Pediatric Stem Cell Research Institute, Sackler School of Medicine; Tel Aviv University, Israel
| | | | | |
Collapse
|
32
|
Brunswig-Spickenheier B, Boche J, Westenfelder C, Peimann F, Gruber AD, Jaquet K, Krause K, Zustin J, Zander AR, Lange C. Limited immune-modulating activity of porcine mesenchymal stromal cells abolishes their protective efficacy in acute kidney injury. Stem Cells Dev 2010; 19:719-29. [PMID: 20143956 DOI: 10.1089/scd.2009.0494] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
We demonstrated previously that administration of mesenchymal stromal cells (MSCs) after renal ischemia/reperfusion injury (IRI) in rats protected renal function and hastened repair through complex paracrine mechanisms. Here we investigated kidney-protective actions of MSCs in a porcine IRI model that may have relevance to human acute kidney injury (AKI). Groups of female pigs with bilateral IRI were infused with autologous or male allogeneic MSCs. No acute or late complications were observed, but unexpectedly, MSC therapy also had no beneficial effects on kidney function and histology. In vitro, we demonstrated substantial functional and phenotypic overlaps between rodent, human, and porcine MSCs, all of which exhibited trilineage differentiation, characteristic antigen profiles, and secretion of renoprotective vascular endothelial growth factor (VEGF)-A and insulin-like growth factor-1 (IGF-1). However, in striking contrast to human MSCs, porcine MSCs failed to inhibit the mixed lymphocyte reaction (MLR) and induced robust production of proinflammatory interleukin-6 (IL-6). In summary, in contrast to rodent models, treatment of porcine IRI with MSCs was not kidney-protective. This, we conclude, is due to the fact that porcine MSCs exert inadequate immune-modulating effects, further demonstrating that successful therapy of IRI with MSCs critically depends on their anti-inflammatory actions. As a consequence, treatment of AKI with MSCs is not informative regarding the investigation of the underlying mechanisms in this large animal model. We expect, however, that the treatment of human IRI of the kidney with immune-modulating MSCs will be as effective as in rodent models.
Collapse
|
33
|
Asanuma H, Meldrum DR, Meldrum KK. Therapeutic Applications of Mesenchymal Stem Cells to Repair Kidney Injury. J Urol 2010; 184:26-33. [DOI: 10.1016/j.juro.2010.03.050] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Indexed: 11/30/2022]
Affiliation(s)
- Hiroshi Asanuma
- Department of Urology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Daniel R. Meldrum
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Kirstan K. Meldrum
- Department of Urology, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
34
|
Motaln H, Schichor C, Lah TT. Human mesenchymal stem cells and their use in cell-based therapies. Cancer 2010; 116:2519-30. [PMID: 20301117 DOI: 10.1002/cncr.25056] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The human population is increasingly facing various diseases, including types of cancer, that cannot be cured with conventional drugs. Advanced drug targeting of tumor cells is also often impossible when treating highly invasive and infiltrative tumors such as glioblastoma or pulmonary cancer, because of tumor cells' high migration and invasiveness. Pluripotent human mesenchymal stem cells (hMSCs) have been extensively studied, and strategies are being proposed for treating "incurable" cancers and injury/disease-affected organs. Because of their own intrinsic properties, involving homing and immunomodulatory potency, hMSCs could be used as an excellent cell/drug delivery vehicle in those cell-based therapies. Their unprecedented use has been shadowed, however, by their spontaneous transformation, which links them to cancer-initiating cells during tumor development. How malignant initiation proceeds in vivo, and what are the exact characteristics of the cancer-initiating cells, still remain to be investigated. In the present review, the authors summed up the most recent knowledge about hMSC characteristics, their malignant transformation, and outlined the possibilities of their safe use in novel cell-based therapies.
Collapse
Affiliation(s)
- Helena Motaln
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia.
| | | | | |
Collapse
|
35
|
|
36
|
Mezey E, Mayer B, Németh K. Unexpected roles for bone marrow stromal cells (or MSCs): a real promise for cellular, but not replacement, therapy. Oral Dis 2009; 16:129-35. [PMID: 19656313 DOI: 10.1111/j.1601-0825.2009.01605.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Adult and embryonic stem cells have drawn a lot of attention in the last decade as new tools in regenerative medicine. A variety of such cells have been discovered and put forward as candidates for use in cell replacement therapy. Investigators hope that some, if not all, of our organs can be replaced or restored to function; that new livers, kidneys, and brain cells can be produced. Many reviews have already been written about stem cells and their potential use in regenerating tissues. In this study, we would like to call attention to a different application of a special group of adult stem cells, the stromal cells in the bone marrow (also called mesenchymal stem cells or MSCs). These cells have been discovered to modulate immune function. They can easily be expanded in culture and surprisingly, they also seem not to be immunogenic. Thus, they can be removed from donors, expanded, stored in freezers, and used as allogeneic transplants in a variety of diseases in everyday medicine.
Collapse
Affiliation(s)
- E Mezey
- Adult Stem Cell Unit, NIH, NIDCR, CSDB, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|