1
|
Sun Y, Zhao H, Yang S, Wang G, Zhu L, Sun C, An Y. Urine-derived stem cells: Promising advancements and applications in regenerative medicine and beyond. Heliyon 2024; 10:e27306. [PMID: 38509987 PMCID: PMC10951541 DOI: 10.1016/j.heliyon.2024.e27306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 03/22/2024] Open
Abstract
Currently, stem cells are a prominent focus of regenerative engineering research. However, due to the limitations of commonly used stem cell sources, their application in therapy is often restricted to the experimental stage and constrained by ethical considerations. In contrast, urine-derived stem cells (USCs) offer promising advantages for clinical trials and applications. The noninvasive nature of the collection process allows for repeated retrieval within a short period, making it a more feasible option. Moreover, studies have shown that USCs have a protective effect on organs, promoting vascular regeneration, inhibiting oxidative stress, and reducing inflammation in various acute and chronic organ dysfunctions. The application of USCs has also been enhanced by advancements in biomaterials technology, enabling better targeting and controlled release capabilities. This review aims to summarize the current state of research on USCs, providing insights for future applications in basic and clinical settings.
Collapse
Affiliation(s)
| | | | - Shuguang Yang
- Department of Critical Care Medicine, Peking University People's Hospital, PR China
| | - Guangjie Wang
- Department of Critical Care Medicine, Peking University People's Hospital, PR China
| | - Leijie Zhu
- Department of Critical Care Medicine, Peking University People's Hospital, PR China
| | - Chang Sun
- Department of Critical Care Medicine, Peking University People's Hospital, PR China
| | - Youzhong An
- Department of Critical Care Medicine, Peking University People's Hospital, PR China
| |
Collapse
|
2
|
Wong CY. Current advances of stem cell-based therapy for kidney diseases. World J Stem Cells 2021; 13:914-933. [PMID: 34367484 PMCID: PMC8316868 DOI: 10.4252/wjsc.v13.i7.914] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/10/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023] Open
Abstract
Kidney diseases are a prevalent health problem around the world. Multidrug therapy used in the current routine treatment for kidney diseases can only delay disease progression. None of these drugs or treatments can reverse the progression to an end-stage of the disease. Therefore, it is crucial to explore novel therapeutics to improve patients’ quality of life and possibly cure, reverse, or alleviate the kidney disease. Stem cells have promising potentials as a form of regenerative medicine for kidney diseases due to their unlimited replication and their ability to differentiate into kidney cells in vitro. Mounting evidences from the administration of stem cells in an experimental kidney disease model suggested that stem cell-based therapy has therapeutic or renoprotective effects to attenuate kidney damage while improving the function and structure of both glomerular and tubular compartments. This review summarises the current stem cell-based therapeutic approaches to treat kidney diseases, including the various cell sources, animal models or in vitro studies. The challenges of progressing from proof-of-principle in the laboratory to widespread clinical application and the human clinical trial outcomes reported to date are also highlighted. The success of cell-based therapy could widen the scope of regenerative medicine in the future.
Collapse
Affiliation(s)
- Chee-Yin Wong
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang 43000, Selangor, Malaysia
- Research Department, Cytopeutics, Cyberjaya 63000, Selangor, Malaysia
| |
Collapse
|
3
|
Shemies RS, Gaber TZ, Baiomy A, Aladle DA, Mosbah A, Abdel-Hady ES, Sayed-Ahmed N, Sobh M. Angiogenic markers predict kidney injury and obstetric complications in women with preeclampsia and pregnancy-related acute kidney injury. Ther Apher Dial 2021; 26:306-315. [PMID: 33533567 DOI: 10.1111/1744-9987.13633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/25/2021] [Accepted: 02/01/2021] [Indexed: 11/28/2022]
Abstract
Pregnancy-related acute kidney injury (PRAKI) particularly on top of preeclampsia (PE) represents a major cause of maternal and fetal morbidity and mortality. Reliable diagnostic tools are needed to further evaluate the diagnosis and prognosis of PRAKI. Our objective was to study the diagnostic and prognostic value of angiogenic markers (e.g., stromal cell-derived factor 1 (SDF-1), vascular endothelial growth factor (VEGF), alarmins as uric acid) in women with PE and PRAKI. This prospective study included three groups; PRAKI, PE patients, and healthy controls that were compared regarding serum levels of the studied markers correlated to renal, maternal, and fetal outcomes. SDF-1, VEGF, and uric acid levels were significantly different between the three included groups and predicted PRAKI diagnosis. Patients with hemolysis, elevated liver enzymes, and low platelet (HELLP) syndrome exhibited the highest titers of SDF-1 and VEGF. A positive correlation was found between SDF-1 and renal recovery. Conclusively, serum assays of SDF-1, VEGF, and uric acid may add a diagnostic value in PRAKI and PE.
Collapse
Affiliation(s)
| | - Tamer Zaki Gaber
- Mansoura Nephrology and Dialysis Unit, Mansoura University, Mansoura, Egypt
| | - Azza Baiomy
- Clinical Pathology Department, Mansoura University, Mansoura, Egypt
| | - Doaa A Aladle
- Clinical Pathology Department, Mansoura University, Mansoura, Egypt
| | - Alaa Mosbah
- Obstetrics and Gynecology Departments, Mansoura University, Mansoura, Egypt
| | - El-Said Abdel-Hady
- Obstetrics and Gynecology Departments, Mansoura University, Mansoura, Egypt
| | - Nagy Sayed-Ahmed
- Mansoura Nephrology and Dialysis Unit, Mansoura University, Mansoura, Egypt
| | - Mohammed Sobh
- Mansoura Nephrology and Dialysis Unit, Mansoura University, Mansoura, Egypt
| |
Collapse
|
4
|
Majka SM, Rojas M, Petrache I, Foronjy RF. Mesenchymal Regulation of the Microvascular Niche in Chronic Lung Diseases. Compr Physiol 2019; 9:1431-1441. [PMID: 31688970 DOI: 10.1002/cphy.c180043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The adult lung is comprised of diverse vascular, epithelial, and mesenchymal progenitor cell populations that reside in distinct niches. Mesenchymal progenitor cells (MPCs) are intimately associated with both the epithelium and the vasculature, and new evidence is emerging to describe their functional roles in these niches. Also emerging, following lineage analysis and single cell sequencing, is a new understanding of the diversity of mesenchymal cell subpopulations in the lung. However, several gaps in knowledge remain, including how newly defined MPC lineages interact with cells in the vascular niche and the role of adult lung MPCs during lung repair and regeneration following injury, especially in chronic lung diseases. Here we summarize how the current evidence on MPC regulation of the microvasculature during tissue homeostasis and injury may inform studies on understanding their role in chronic lung disease pathogenesis or repair. © 2019 American Physiological Society. Compr Physiol 9:1431-1441, 2019.
Collapse
Affiliation(s)
- Susan M Majka
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, National Jewish Health, Denver, Colorado, USA
| | - Mauricio Rojas
- McGowan Institute for Regenerative Medicine, Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Irina Petrache
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, National Jewish Health, Denver, Colorado, USA
| | - Robert F Foronjy
- Division of Pulmonary and Critical Care Medicine, SUNY Downstate Medical Center, Brooklyn, New York, USA
| |
Collapse
|
5
|
Acellular Mouse Kidney ECM can be Used as a Three-Dimensional Substrate to Test the Differentiation Potential of Embryonic Stem Cell Derived Renal Progenitors. Stem Cell Rev Rep 2018; 13:513-531. [PMID: 28239758 PMCID: PMC5493730 DOI: 10.1007/s12015-016-9712-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The development of strategies for tissue regeneration and bio-artificial organ development is based on our understanding of embryogenesis. Differentiation protocols attempt to recapitulate the signaling modalities of gastrulation and organogenesis, coupled with cell selection regimens to isolate the cells of choice. This strategy is impeded by the lack of optimal in vitro culture systems since traditional culture systems do not allow for the three-dimensional interaction between cells and the extracellular matrix. While artificial three-dimensional scaffolds are available, using the natural extracellular matrix scaffold is advantageous because it has a distinct architecture that is difficult to replicate. The adult extracellular matrix is predicted to mediate signaling related to tissue repair not embryogenesis but existing similarities between the two argues that the extracellular matrix will influence the differentiation of stem and progenitor cells. Previous studies using undifferentiated embryonic stem cells grown directly on acellular kidney ECM demonstrated that the acellular kidney supported cell growth but limited differentiation occurred. Using mouse kidney extracellular matrix and mouse embryonic stem cells we report that the extracellular matrix can support the development of kidney structures if the stem cells are first differentiated to kidney progenitor cells before being applied to the acellular organ.
Collapse
|
6
|
Antifibrotic Actions of Serelaxin – New Roles for an Old Player. Trends Pharmacol Sci 2016; 37:485-497. [DOI: 10.1016/j.tips.2016.02.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/16/2016] [Accepted: 02/19/2016] [Indexed: 12/25/2022]
|
7
|
Yuan L, Liu HQ, Wu MJ. Human embryonic mesenchymal stem cells participate in differentiation of renal tubular cells in newborn mice. Exp Ther Med 2016; 12:641-648. [PMID: 27446255 PMCID: PMC4950250 DOI: 10.3892/etm.2016.3383] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 04/25/2016] [Indexed: 12/13/2022] Open
Abstract
Stem cells are used with increasing success in the treatment of renal tubular injury. However, whether mesenchymal stem cells (MSC) differentiate into renal tubular epithelial cells remains controversial. The aims of the present study were to observe the localization of human embryonic MSCs (hMSCs) in the kidneys of newborn mice, and to investigate hMSC differentiation into tubular epithelium. Primary culture hMSCs were derived from 4–7-week-old embryos and labeled with the cell membrane fluorescent dye PKH-26. The degree of apoptosis, cell growth, differentiation and localization of hMSCs with and without this label were then determined using immunohistochemical methods and flow cytometry. hMSCs and PKH26-labeled hMSCs were revealed to differentiate into chondrocytes and adipocytes, and were demonstrated to have similar proliferative capability. In the two cell types, the antigens CD34 and CD45, indicative of hematopoietic lineages, were not expressed; however, the expression of the mesenchymal markers CD29 and CD90 in MSCs, was significantly increased. During a 4-week culture period, laser confocal microscopy revealed that PKH26-labeled hMSCs in the kidneys of newborn mice gradually dispersed. Two weeks after the injection of the PKH26-labeled cells, the percentage of PKH26-labeled hMSCs localized to the renal tubules was 10±2.1%. In conclusion, PKH26 labeling has no effect on hMSC differentiation, proliferation and mesenchymal cell surface features, and hMSCs injected into the kidneys of newborn mice may transform to renal tubule epithelium.
Collapse
Affiliation(s)
- Li Yuan
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Hou-Qi Liu
- Department of Histology and Embryology, Research Center of Developmental Biology, Second Military Medical University, Shanghai 200433, P.R. China
| | - Min-Juan Wu
- Department of Histology and Embryology, Research Center of Developmental Biology, Second Military Medical University, Shanghai 200433, P.R. China
| |
Collapse
|
8
|
D'Addio F, Trevisani A, Ben Nasr M, Bassi R, El Essawy B, Abdi R, Secchi A, Fiorina P. Harnessing the immunological properties of stem cells as a therapeutic option for diabetic nephropathy. Acta Diabetol 2014; 51:897-904. [PMID: 24894496 DOI: 10.1007/s00592-014-0603-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Accepted: 05/17/2014] [Indexed: 12/27/2022]
Abstract
Diabetic nephropathy is the leading and possibly the most devastating complication of diabetes, with a prevalence ranging from 25 to 40 % in diabetic individuals, and as such represents an important challenge for public health worldwide. As a major cause of end-stage renal disease, diabetic nephropathy also accounts for a large proportion of deaths in diabetic individuals. To date, therapeutic options for overt diabetic nephropathy include medical interventions to reduce blood glucose levels and to control blood pressure and proteinuria. Recent evidence suggests a strong role for inflammation in the development and progression of diabetic nephropathy. Various immune cells, cytokines and chemokines have been implicated in the onset of diabetic nephropathy, while immune-related transcription factors and adhesion molecules have been correlated with the establishment of a renal proinflammatory microenvironment. Both inflammation and immune activation may promote severe distress in the kidney, with subsequent increased local fibrosis, ultimately leading to the development of end-stage renal disease. Stem cells are undifferentiated cells capable of regenerating virtually any organ or tissue and bearing important immunoregulatory and anti-inflammatory properties. Due to the aforementioned considerations, significant interest has been ignited with regard to the use of stem cells as novel therapeutics for diabetic nephropathy. Here, we will be examining in detail how anti-inflammatory properties of different populations of stem cells may offer novel therapy for the treatment of diabetic nephropathy.
Collapse
|
9
|
Gregorini M, Bosio F, Rocca C, Corradetti V, Valsania T, Pattonieri EF, Esposito P, Bedino G, Collesi C, Libetta C, Frassoni F, Canton AD, Rampino T. Mesenchymal stromal cells reset the scatter factor system and cytokine network in experimental kidney transplantation. BMC Immunol 2014; 15:44. [PMID: 25277788 PMCID: PMC4193986 DOI: 10.1186/s12865-014-0044-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 09/25/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND In former studies we showed in a rat model of renal transplantation that Mesenchymal Stromal Cells (MSC) prevent acute rejection in an independent way of their endowing in the graft. In this study we investigated whether MSC operate by resetting cytokine network and Scatter Factor systems, i.e. Hepatocyte Growth Factor (HGF), Macrophage Stimulating Protein (MSP) and their receptors Met and RON, respectively. METHODS MSC were injected into the renal artery soon after reperfusion. Controls were grafted untreated and normal rats. Rats were sacrificed 7 days after grafting. Serum and renal tissue levels of IFN-γ, IL-1, IL-2, IL-4, IL-6, IL-10, MSP/RON, HGF/Met systems, Treg lymphocytes were investigated. RESULTS In grafted untreated rats IFN-γ increased in serum and renal tissue and IL-6 rose in serum. MSC prevented both the phenomena, increased IL-10 serum levels and Treg number in the graft. Furthermore MSC increased serum and tissue HGF levels, Met tubular expression and prevented the suppression of tubular MSP/RON expression. CONCLUSIONS Our results demonstrate that MSC modify cytokine network to a tolerogenic setting, they suppress Th1 cells, inactivate monocytes/macrophage, recruit Tregs. In addition, MSC sustain the expression of the Scatter Factor systems expression, i.e. systems that are committed to defend survival and stimulate regeneration of tubular cells.
Collapse
Affiliation(s)
- Marilena Gregorini
- Unit of Nephrology, Dialysis and Transplantation, Fondazione, IRCCS Policlinico San Matteo and University of Pavia, viale Golgi 19, 27100 Pavia, Italy
| | - Francesca Bosio
- Unit of Nephrology, Dialysis and Transplantation, Fondazione, IRCCS Policlinico San Matteo and University of Pavia, viale Golgi 19, 27100 Pavia, Italy
| | - Chiara Rocca
- Unit of Nephrology, Dialysis and Transplantation, Fondazione, IRCCS Policlinico San Matteo and University of Pavia, viale Golgi 19, 27100 Pavia, Italy
| | - Valeria Corradetti
- Unit of Nephrology, Dialysis and Transplantation, Fondazione, IRCCS Policlinico San Matteo and University of Pavia, viale Golgi 19, 27100 Pavia, Italy
| | - Teresa Valsania
- Unit of Nephrology, Dialysis and Transplantation, Fondazione, IRCCS Policlinico San Matteo and University of Pavia, viale Golgi 19, 27100 Pavia, Italy
| | - Eleonora Francesca Pattonieri
- Unit of Nephrology, Dialysis and Transplantation, Fondazione, IRCCS Policlinico San Matteo and University of Pavia, viale Golgi 19, 27100 Pavia, Italy
| | - Pasquale Esposito
- Unit of Nephrology, Dialysis and Transplantation, Fondazione, IRCCS Policlinico San Matteo, viale Golgi 19, 27100 Pavia, Italy
| | - Giulia Bedino
- Unit of Nephrology, Dialysis and Transplantation, Fondazione, IRCCS Policlinico San Matteo and University of Pavia, viale Golgi 19, 27100 Pavia, Italy
| | - Chiara Collesi
- ICGEB, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Carmelo Libetta
- Unit of Nephrology, Dialysis and Transplantation, Fondazione, IRCCS Policlinico San Matteo and University of Pavia, viale Golgi 19, 27100 Pavia, Italy
| | - Francesco Frassoni
- Stem Cells Therapy and Hemato-Oncology, S.Martino Hospital, 16100 Genoa, Italy
| | - Antonio Dal Canton
- Unit of Nephrology, Dialysis and Transplantation, Fondazione, IRCCS Policlinico San Matteo and University of Pavia, viale Golgi 19, 27100 Pavia, Italy
| | - Teresa Rampino
- Unit of Nephrology, Dialysis and Transplantation, Fondazione, IRCCS Policlinico San Matteo, viale Golgi 19, 27100 Pavia, Italy
| |
Collapse
|
10
|
Morales EE, Wingert RA. Renal stem cell reprogramming: Prospects in regenerative medicine. World J Stem Cells 2014; 6:458-466. [PMID: 25258667 PMCID: PMC4172674 DOI: 10.4252/wjsc.v6.i4.458] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Revised: 08/21/2014] [Accepted: 09/01/2014] [Indexed: 02/06/2023] Open
Abstract
Stem cell therapy is a promising future enterprise for renal replacement in patients with acute and chronic kidney disease, conditions which affect millions worldwide and currently require patients to undergo lifelong medical treatments through dialysis and/or organ transplant. Reprogramming differentiated renal cells harvested from the patient back into a pluripotent state would decrease the risk of tissue rejection and provide a virtually unlimited supply of cells for regenerative medicine treatments, making it an exciting area of current research in nephrology. Among the major hurdles that need to be overcome before stem cell therapy for the kidney can be applied in a clinical setting are ensuring the fidelity and relative safety of the reprogrammed cells, as well as achieving feasible efficiency in the reprogramming processes that are utilized. Further, improved knowledge about the genetic control of renal lineage development is vital to identifying predictable and efficient reprogramming approaches, such as the expression of key modulators or the regulation of gene activity through small molecule mimetics. Here, we discuss several recent advances in induced pluripotent stem cell technologies. We also explore strategies that have been successful in renal progenitor generation, and explore what these methods might mean for the development of cell-based regenerative therapies for kidney disease.
Collapse
|
11
|
Alvarez-Viejo M, Menendez-Menendez Y, Blanco-Gelaz MA, Ferrero-Gutierrez A, Fernandez-Rodriguez MA, Gala J, Otero-Hernandez J. Quantifying mesenchymal stem cells in the mononuclear cell fraction of bone marrow samples obtained for cell therapy. Transplant Proc 2013; 45:434-9. [PMID: 23375334 DOI: 10.1016/j.transproceed.2012.05.091] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Revised: 12/14/2011] [Accepted: 05/04/2012] [Indexed: 12/15/2022]
Abstract
AIMS The use of bone marrow mononuclear cells (BMMNCs) as a source of mesenchymal stem cells (MSCs) for therapy has recently attracted the attention of researchers because BMMNCs can be easily obtained and do not require in vitro expansion before their use. This study was designed to quantify the MSC population in bone marrow (BM) samples obtained for cell therapy using flow cytometry to detect the CD271 antigen. MATERIAL AND METHODS Autologous BM was obtained by posterior superior iliac crest aspiration under topical anesthesia. Mononuclear cells isolated from the BM aspirate on a Ficoll density gradient were used to treat patients with pressure ulcer (n = 13) bone nonunions (n = 3) or diabetic foot ulcers (n = 5). RESULTS Our flow cytometry data revealed a low percentage as well as a high variability among patients of CD271(+)CD45(-) cells (range, 0.0017 to 0.0201%). All cultured MSC adhered to plastic dishes showing a capacity to differentiate into adipogenic and osteogenic lineages. CONCLUSIONS Our findings suggested that the success of cell therapy was independent of the number of MSCs present in the BM aspirate used for autologous cell therapy.
Collapse
Affiliation(s)
- M Alvarez-Viejo
- Transplant and Cell Therapy Unit, Hospital Universitario Central de Asturias, Oviedo, Spain.
| | | | | | | | | | | | | |
Collapse
|
12
|
Liu N, Han G, Cheng J, Huang J, Tian J. Erythropoietin promotes the repair effect of acute kidney injury by bone-marrow mesenchymal stem cells transplantation. Exp Biol Med (Maywood) 2013; 238:678-86. [PMID: 23918879 DOI: 10.1177/1535370213489486] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Bone-marrow mesenchymal stem cells (BMSCs) transplantation is effective for acute kidney injury (AKI) repair but with limited efficiency. In the present study, BMSCs marked by bromodeoxyuridine (BrdU) were transplanted to the AKI mouse model with erythropoietin (EPO) being subcutaneously injected. The blood urea nitrogen (BUN) and serum creatinine (Scr) levels, pathological changes, distribution of BMSCs, expressions of the cytokeratin 18 (CK18) and the stromal cell-derived factor 1 (SDF-1) in the nephridial tissues were measured. The directional migration of BMSCs to the AKI microenvironment in vitro was also tested. The results showed that BMSCs transplantation or EPO injection alone decreased the BUN and Scr levels and the acute tubular necrosis (ATN) scoring in varied degrees. The combination of these decreased the above indicators’ levels significantly. BrdU+ cells (BMSCs) were observed in the AKI nephridial tissues, and CK18 expressed in the cytoplasm of these cells. EPO injection increased the proportion of BrdU+ cells with the enhanced expression of SDF-1 in the AKI nephridial tissues. EPO increased the migrating number of BMSCs to the AKI microenvironment in vitro, and additional anti-SDF-1 treatment with SDF-1 antibody neutralized this effect. Our results showed that EPO increased the number of the transplanted BMSCs in the injured nephridial tissues and enhanced the AKI repair effect of BMSCs transplantation. The enhanced kidney-homing efficiency for BMSCs mediated by the SDF-1/CXCR4 pathway is one of the possible mechanisms for EPO performance.
Collapse
Affiliation(s)
- Nanmei Liu
- Department of Nephrology, 455th hospital of PLA, Shanghai 200052, China
| | - Guofeng Han
- Department of Nephrology, 455th hospital of PLA, Shanghai 200052, China
| | - Jin Cheng
- Department of Nephrology, 455th hospital of PLA, Shanghai 200052, China
| | - Jian Huang
- Department of Nephrology, 455th hospital of PLA, Shanghai 200052, China
| | - Jun Tian
- Department of Nephrology, 455th hospital of PLA, Shanghai 200052, China
| |
Collapse
|
13
|
Liu N, Tian J, Cheng J, Zhang J. Effect of erythropoietin on the migration of bone marrow-derived mesenchymal stem cells to the acute kidney injury microenvironment. Exp Cell Res 2013; 319:2019-2027. [DOI: 10.1016/j.yexcr.2013.04.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 04/10/2013] [Accepted: 04/11/2013] [Indexed: 01/16/2023]
|
14
|
Quimby JM, Webb TL, Habenicht LM, Dow SW. Safety and efficacy of intravenous infusion of allogeneic cryopreserved mesenchymal stem cells for treatment of chronic kidney disease in cats: results of three sequential pilot studies. Stem Cell Res Ther 2013; 4:48. [PMID: 23632128 PMCID: PMC3707049 DOI: 10.1186/scrt198] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 04/23/2013] [Indexed: 01/05/2023] Open
Abstract
Introduction Administration of mesenchymal stem cells (MSCs) has been shown to improve renal function in rodent models of chronic kidney disease (CKD), in part by reducing intrarenal inflammation and suppressing fibrosis. CKD in cats is characterized by tubulointerstitial inflammation and fibrosis, and thus treatment with MSCs might improve renal function and urinary markers of inflammation in this disease. Therefore, a series of pilot studies was conducted to assess the safety and efficacy of intravenous administration of allogeneic adipose-derived MSCs (aMSCs) in cats with naturally occurring CKD. Methods Cats enrolled in these studies received an intravenous infusion of allogeneic aMSCs every 2 weeks collected from healthy, young, specific pathogen-free cats. Cats in pilot study 1 (six cats) received 2 × 106 cryopreserved aMSCs per infusion, cats in pilot study 2 (five cats) received 4 × 106 cryopreserved aMSCs per infusion, and cats in pilot study 3 (five cats) received 4 × 106 aMSCs cultured from cryopreserved adipose. Serum biochemistry, complete blood count, urinalysis, urine protein, glomerular filtration rate, and urinary cytokine concentrations were monitored during the treatment period. Changes in clinical parameters were compared statistically by means of repeated measures analysis of variance (ANOVA) followed by Bonferroni’s correction. Results Cats in pilot study 1 had few adverse effects from the aMSC infusions and there was a statistically significant decrease in serum creatinine concentrations during the study period, however the degree of decrease seems unlikely to be clinically relevant. Adverse effects of the aMSC infusion in cats in pilot study 2 included vomiting (2/5 cats) during infusion and increased respiratory rate and effort (4/5 cats). Cats in pilot study 3 did not experience any adverse side effects. Serum creatinine concentrations and glomerular filtration rates did not change significantly in cats in pilot studies 2 and 3. Conclusions Administration of cryopreserved aMSCs was associated with significant adverse effects and no discernible clinically relevant improvement in renal functional parameters. Administration of aMSCs cultured from cryopreserved adipose was not associated with adverse effects, but was also not associated with improvement in renal functional parameters.
Collapse
|
15
|
Wang WW, Wang W, Jiang Y, Han GF, Lu S, Li G, Zhang J. Reprogramming of mouse renal tubular epithelial cells to induced pluripotent stem cells. Cytotherapy 2013; 15:578-85. [PMID: 23415920 DOI: 10.1016/j.jcyt.2013.01.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Revised: 01/05/2013] [Accepted: 01/07/2013] [Indexed: 02/07/2023]
Abstract
Kidney disease has reached epidemic proportions and is associated with high mortality and morbidity rates. Stem cell-based therapy may effectively treat kidney damage by cell transplantation. The breakthrough discovery using a combination of four transcription factors to reprogram genetically somatic cells into induced pluripotent stem (iPS) cells was a milestone in stem cell therapy. The lentivirus was packaged containing OCT4, SOX2, c-MYC and KLF4 transcription factors and then transfected mouse renal tubular epithelial cells (RTECs). The colonies were picked up at 21 days and were tested by cytochemistry, immunofluorescence assay and quantitative real-time polymerase chain reaction. Viral transgene expression levels were also assessed by quantitative analysis. Additionally, embryoid bodies from iPS cells were formed, and immunofluorescence and teratoma assays were performed. Karyotype analysis of mouse RTEC-derived iPS cells was also performed. The iPS cells were indistinguishable from mouse embryonic stem cells with respect to colony morphology, the expression of pluripotency-associated transcription factors and surface markers, embryoid body-mediated differentiation potential and teratoma assays. Quantitative polymerase chain reaction demonstrated that the lentiviral transgenes were largely silenced. The mouse RTEC-derived iPS cells exhibited a normal karyotype of 40,XY. iPS cells can be produced using mouse RTECs, which would be helpful in investigations to ameliorate the symptoms of kidney disease and to slow the progression of kidney disease by in vitro and in vivo animal studies.
Collapse
Affiliation(s)
- Wei-Wei Wang
- Department of Nephrology, Jimin Hospital, Shanghai, PR China
| | | | | | | | | | | | | |
Collapse
|
16
|
El-Ansary M, Saadi G, Abd El-Hamid SM. Mesenchymal stem cells are a rescue approach for recovery of deteriorating kidney function. Nephrology (Carlton) 2013; 17:650-7. [PMID: 22640266 DOI: 10.1111/j.1440-1797.2012.01622.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
AIM Stem cell (SC) therapy for chronic kidney disease (CKD) is urgently needed. The use of mesenchymal stem cells (MSC) is a possible new therapeutic modality. Our work aimed to isolate human MSC from adult bone marrow to improve kidney functions in CKD patients. METHODS In our study 30 patients with impaired kidney function were included, their ages ranged from 22 to 68 years. They included 10 inactive glomerulonephritis patients due to systemic lupus erythromatosus (SLE) (group I), 10 renal transplantation cases (group II) and 10 patients of other aetiologies as the control group. Fifty millilitres of bone marrow was aspirated from the iliac bone, for separation of MSC. RESULTS There was a highly statistically significant difference between both CD271 and CD29 before and after culture with increase of both markers at end of culture, P < 0.01. Finally 50-70 million MSC in 10 mL saline (0.7-1.0 × 10(6) MSC/kg body weight) were infused intravenously in two divided doses one week apart. There was a highly statistically significant difference between each of serum creatinine and creatinine clearance levels before and after MSC injection at 1, 3 and 6 months post-infusion with SLE cases showing a greater decline of their serum creatinine and elevation of mean creatinine clearance levels after injection than transplantation and control groups, P < 0.05. CONCLUSION Mesenchymal stem cells therapy is a potential therapeutic modality for early phases of CKD.
Collapse
Affiliation(s)
- Mervat El-Ansary
- Departments of Clinical Pathology, Internal Medicine and Nephrology, Kasr El-Aini, Cairo University, Cairo, Egypt
| | | | | |
Collapse
|
17
|
Tomasoni S, Longaretti L, Rota C, Morigi M, Conti S, Gotti E, Capelli C, Introna M, Remuzzi G, Benigni A. Transfer of growth factor receptor mRNA via exosomes unravels the regenerative effect of mesenchymal stem cells. Stem Cells Dev 2012; 22:772-80. [PMID: 23082760 DOI: 10.1089/scd.2012.0266] [Citation(s) in RCA: 257] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Bone marrow-mesenchymal stem cells (BM-MSC) ameliorate renal dysfunction and repair tubular damage of acute kidney injury by locally releasing growth factors, including the insulin-like growth factor-1 (IGF-1). The restricted homing of BM-MSC at the site of injury led us to investigate a possible gene-based communication mechanism between BM-MSC and tubular cells. Human BM-MSC (hBM-MSC) released microparticles and exosomes (Exo) enriched in mRNAs. A selected pattern of transcripts was detected in Exo versus parental cells. Exo expressed the IGF-1 receptor (IGF-1R), but not IGF-1 mRNA, while hBM-MSC contained both mRNAs. R- cells lacking IGF-1R exposed to hBM-MSC-derived Exo acquired the human IGF-1R transcript that was translated in the corresponding protein. Transfer of IGF-1R mRNA from Exo to cisplatin-damaged proximal tubular cells (proximal tubular epithelial cell [PTEC]) increased PTEC proliferation. Coincubation of damaged PTEC with Exo and soluble IGF-1 further enhanced cell proliferation. These findings suggest that horizontal transfer of the mRNA for IGF-1R to tubular cells through Exo potentiates tubular cell sensitivity to locally produced IGF-1 providing a new mechanism underlying the powerful renoprotection of few BM-MSC observed in vivo.
Collapse
Affiliation(s)
- Susanna Tomasoni
- Mario Negri Institute for Pharmacological Research, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Liu H, Zhang J, Liu CY, Hayashi Y, Kao WWY. Bone marrow mesenchymal stem cells can differentiate and assume corneal keratocyte phenotype. J Cell Mol Med 2012; 16:1114-24. [PMID: 21883890 PMCID: PMC4365890 DOI: 10.1111/j.1582-4934.2011.01418.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
It remains elusive as to what bone marrow (BM) cell types infiltrate into injured and/or diseased tissues and subsequently differentiate to assume the phenotype of residential cells, for example, neurons, cardiac myocytes, keratocytes, etc., to repair damaged tissue. Here, we examined the possibility of whether BM cell invasion via circulation into uninjured and injured corneas could assume a keratocyte phenotype, using chimeric mice generated by transplantation of enhanced green fluorescent protein (EGFP)+ BM cells into keratocan null (Kera−/−) and lumican null (Lum−/−) mice. EGFP+ BM cells assumed dendritic cell morphology, but failed to synthesize corneal-specific keratan sulfate proteoglycans, that is KS-lumican and KS-keratocan. In contrast, some EGFP+ BM cells introduced by intrastromal transplantation assumed keratocyte phenotypes. Furthermore, BM cells were isolated from Kera-Cre/ZEG mice, a double transgenic mouse line in which cells expressing keratocan become EGFP+ due to the synthesis of Cre driven by keratocan promoter. Three days after corneal and conjunctival transplantations of such BM cells into Kera−/− mice, green keratocan positive cells were found in the cornea, but not in conjunctiva. It is worthy to note that transplanted BM cells were rejected in 4 weeks. MSC isolated from BM were used to examine if BM mesenchymal stem cells (BM-MSC) could assume keratocyte phenotype. When BM-MSC were intrastromal-transplanted into Kera−/− mice, they survived in the cornea without any immune and inflammatory responses and expressed keratocan in Kera−/− mice. These observations suggest that corneal intrastromal transplantation of BM-MSC may be an effective treatment regimen for corneal diseases involving dysfunction of keratocytes.
Collapse
Affiliation(s)
- Hongshan Liu
- Department of Ophthalmology, Edith Crawley Vision Research Center, University of Cincinnati, Cincinnati, OH, USA.
| | | | | | | | | |
Collapse
|
19
|
Abstract
The kidney is widely regarded as an organ without regenerative abilities. However, in recent years this dogma has been challenged on the basis of observations of kidney recovery following acute injury, and the identification of renal populations that demonstrate stem cell characteristics in various species. It is currently speculated that the human kidney can regenerate in some contexts, but the mechanisms of renal regeneration remain poorly understood. Numerous controversies surround the potency, behaviour and origins of the cell types that are proposed to perform kidney regeneration. The present review explores the current understanding of renal stem cells and kidney regeneration events, and examines the future challenges in using these insights to create new clinical treatments for kidney disease.
Collapse
|
20
|
Alfarano C, Roubeix C, Chaaya R, Ceccaldi C, Calise D, Mias C, Cussac D, Bascands JL, Parini A. Intraparenchymal injection of bone marrow mesenchymal stem cells reduces kidney fibrosis after ischemia-reperfusion in cyclosporine-immunosuppressed rats. Cell Transplant 2012; 21:2009-19. [PMID: 22525800 DOI: 10.3727/096368912x640448] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Ischemia-reperfusion and immunosuppressive therapy are a major cause of progressive renal failure after kidney transplantation. Recent studies have shown that administration of bone marrow mesenchymal stem cells (MSCs) improves kidney functional recovery in the acute phase of post ischemia-reperfusion injury. In the present study, we used an original model of renal ischemia-reperfusion in immunosuppressed rats (NIRC) to investigate the effects of bone marrow MSCs on progression of chronic renal failure and the mechanisms potentially involved. Left renal ischemia-reperfusion (IR) was induced in unilateral nephrectomized Lewis rats. After IR, rats were treated daily with cyclosporine (10 mg/kg SC) for 28 days. MSCs were injected into the kidney at day 7 after IR. At day 28 after IR, kidneys were removed for histomorphological, biochemical, and gene expression analysis. The effect of conditioned media from MSCs on epithelial-mesenchymal transition was studied in vitro on HK2 cells. Our results show that, as compared to untreated NIRC rats, rats treated by intrarenal injection of MSCs 7 days after IR displayed improvement in renal function, reduction of interstitial fibrosis, and decrease in chronic tubule injury. These effects were associated with a decrease in interstitial α-SMA accumulation and MMP2 activity, markers of fibroblast/fibroblast-like cell activation, and renal remodeling, respectively. Finally, experiments in vitro showed that MSC-conditioned medium prevented epithelial-mesenchymal transition induced by TGF-β in HK2 cells. In conclusion, our results show that, in immunosuppressed animals, a single intrarenal administration of MSCs reduced renal fibrosis and promoted the recovery of renal function.
Collapse
Affiliation(s)
- C Alfarano
- Inserm, UMR 1048, Institute of Metabolic and Cardiovascular Diseases (I2MC), Toulouse, France
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Sun X, Fu X, Han W, Zhao Y, Liu H, Sheng Z. Dedifferentiation of human terminally differentiating keratinocytes into their precursor cells induced by basic fibroblast growth factor. Biol Pharm Bull 2011; 34:1037-45. [PMID: 21720010 DOI: 10.1248/bpb.34.1037] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Reprogramming differentiated cells toward stem cells may have long-term applications in stem-cell research and regenerative medicine. Here we report on the dedifferentiation of human epidermal keratinocytes into their precursor cells in vitro with basic fibroblast growth factor (bFGF) but not external gene intervention. After incubation of human terminally differentiating keratinocytes, some of the surviving keratinocytes reverted from a differentiated to a dedifferentiated state, as evidenced by re-expression of biological markers of native keratinocyte stem cells (nKSCs), including β(1)-integrin, CK19 and CK14. Moreover, these dedifferentiation-derived KSCs (dKSCs) showed an ability for high colony formation correlated with cell cycle analysis showing a marked accumulation in S phases, acquired a similar regional distribution of both α(6)-integrin and CD71 expression at the ultrastructural level, and had a increased proliferative capacity by releasing telomerase from nucleolar sites to nucleoplasmic distribution. However, on comparing dKSCs with nKSCs, 2 points seem noteworthy: (1) the proportion of transit amplifying cells in dKSCs treated with bFGF is much higher than that in nKSCs and (2) regional differences exist in the subcellular localization of telomerase in nKSCs and dKSCs. Most nKSCs showed a prominent nucleolar concentration of human telomerase reverse transcriptase expression, whereas most dKSCs showed a more diffuse intranuclear distribution of telomerase or even signal depletion at nucleoli relative to the general nucleoplasm. These results indicate that bFGF could induce the terminally differentiating epidermal keratinocytes to convert into their precursor cells, which offers a new approach for generating residual healthy stem cells for wound repair and regeneration.
Collapse
Affiliation(s)
- Xiaoyan Sun
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Trauma Center of Postgraduate Medical School, Chinese PLA General Hospital, Beijing 100853, PR China
| | | | | | | | | | | |
Collapse
|
22
|
La Manna G, Bianchi F, Cappuccilli M, Cenacchi G, Tarantino L, Pasquinelli G, Valente S, Della Bella E, Cantoni S, Claudia C, Neri F, Tsivian M, Nardo B, Ventura C, Stefoni S. Mesenchymal stem cells in renal function recovery after acute kidney injury: use of a differentiating agent in a rat model. Cell Transplant 2010; 20:1193-208. [PMID: 21092414 DOI: 10.3727/096368910x543394] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Acute kidney injury (AKI) is a major health care condition with limited current treatment options. Within this context, stem cells may provide a clinical approach for AKI. Moreover, a synthetic compound previously developed, hyaluronan monoesters with butyric acid (HB), able to induce metanephric differentiation, formation of capillary-like structures, and secretion of angiogenic cytokines, was tested in vitro. Thereafter, we investigated the effects of human mesenchymal stem cells from fetal membranes (FMhMSCs), both treated and untreated with HB, after induction of ischemic AKI in a rat model. At reperfusion following 45-min clamping of renal pedicles, each rat was randomly assigned to one of four groups: CTR, PBS, MSC, and MSC-HB. Renal function at 1, 3, 5, and 7 days was assessed. Histological samples were analyzed by light and electron microscopy and renal injury was graded. Cytokine analysis on serum samples was performed. FMhMSCs induced an accelerated renal functional recovery, demonstrated by biochemical parameters and confirmed by histology showing that histopathological alterations associated with ischemic injury were less severe in cell-treated kidneys. HB-treated rats showed a minor degree of inflammation, both at cytokine and TEM analyses. Better functional and morphological recovery were not associated to stem cells' regenerative processes, but possibly suggest paracrine effects on microenvironment that induce retrieval of renal damaged tissues. These results suggest that FMhMSCs could be useful in the treatment of AKI and the utilization of synthetic compounds could enhance the recovery induction ability of cells.
Collapse
Affiliation(s)
- Gaetano La Manna
- Department of Internal Medicine, Aging and Renal Disease-Section of Nephrology, University of Bologna, Bologna, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Hauser PV, De Fazio R, Bruno S, Sdei S, Grange C, Bussolati B, Benedetto C, Camussi G. Stem cells derived from human amniotic fluid contribute to acute kidney injury recovery. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:2011-21. [PMID: 20724594 DOI: 10.2353/ajpath.2010.091245] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Stem cells isolated from human amniotic fluid are gaining attention with regard to their therapeutic potential. In this work, we investigated whether these cells contribute to tubular regeneration after experimental acute kidney injury. Cells expressing stem cell markers with multidifferentiative potential were isolated from human amniotic fluid. The regenerative potential of human amniotic fluid stem cells was compared with that of bone marrow-derived human mesenchymal stem cells. We found that the intravenous injection of 3.5 × 10(5) human amniotic fluid stem cells into nonimmune-competent mice with glycerol-induced acute kidney injury was followed by rapid normalization of renal function compared with injection of mesenchymal stem cells. Both stem cell types showed enhanced tubular cell proliferation and reduced apoptosis. Mesenchymal stem cells were more efficient in inducing proliferation than amniotic fluid-derived stem cells, which, in contrast, were more antiapoptotic. Both cell types were found to accumulate within the peritubular capillaries and the interstitium, but amniotic fluid stem cells were more persistent than mesenchymal stem cells. In vitro experiments demonstrated that the two cell types produced different cytokines and growth factors, suggesting that a combination of different mediators is involved in their biological actions. These results suggest that the amniotic fluid-derived stem cells may improve renal regeneration in acute kidney injury, but they are not more effective than mesenchymal stem cells.
Collapse
Affiliation(s)
- Peter V Hauser
- Renal and Vascular Physiopathology Laboratory, Department of Internal Medicine, Molecular Biotechnology Centre, University of Torino, Torino, Italy
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Liu H, Zhang J, Liu CY, Wang IJ, Sieber M, Chang J, Jester JV, Kao WWY. Cell therapy of congenital corneal diseases with umbilical mesenchymal stem cells: lumican null mice. PLoS One 2010; 5:e10707. [PMID: 20502663 PMCID: PMC2873411 DOI: 10.1371/journal.pone.0010707] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2010] [Accepted: 04/19/2010] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Keratoplasty is the most effective treatment for corneal blindness, but suboptimal medical conditions and lack of qualified medical personnel and donated cornea often prevent the performance of corneal transplantation in developing countries. Our study aims to develop alternative treatment regimens for congenital corneal diseases of genetic mutation. METHODOLOGY/PRINCIPAL FINDINGS Human mesenchymal stem cells isolated from neonatal umbilical cords were transplanted to treat thin and cloudy corneas of lumican null mice. Transplantation of umbilical mesenchymal stem cells significantly improved corneal transparency and increased stromal thickness of lumican null mice, but human umbilical hematopoietic stem cells failed to do the same. Further studies revealed that collagen lamellae were re-organized in corneal stroma of lumican null mice after mesenchymal stem cell transplantation. Transplanted umbilical mesenchymal stem cells survived in the mouse corneal stroma for more than 3 months with little or no graft rejection. In addition, these cells assumed a keratocyte phenotype, e.g., dendritic morphology, quiescence, expression of keratocyte unique keratan sulfated keratocan and lumican, and CD34. Moreover, umbilical mesenchymal stem cell transplantation improved host keratocyte functions, which was verified by enhanced expression of keratocan and aldehyde dehydrogenase class 3A1 in lumican null mice. CONCLUSIONS/SIGNIFICANCE Umbilical mesenchymal stem cell transplantation is a promising treatment for congenital corneal diseases involving keratocyte dysfunction. Unlike donated corneas, umbilical mesenchymal stem cells are easily isolated, expanded, stored, and can be quickly recovered from liquid nitrogen when a patient is in urgent need.
Collapse
Affiliation(s)
- Hongshan Liu
- Department of Ophthalmology, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Jianhua Zhang
- Department of Ophthalmology, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Chia-Yang Liu
- Department of Ophthalmology, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - I-Jong Wang
- Department of Ophthalmology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | - James V. Jester
- Gavin Herbert Eye Institute, University of California Irvine Medical Center, Orange, California, United States of America
| | - Winston W. Y. Kao
- Department of Ophthalmology, University of Cincinnati, Cincinnati, Ohio, United States of America
| |
Collapse
|
25
|
Chen X, Hofmann TJ, Otsuru S, Jethva R, Lind C, Monos D, Horwitz EM. A strategy for single nucleotide polymorphism analysis of chimerism for somatic cell therapy. Cytotherapy 2010; 12:1035-43. [PMID: 20429790 DOI: 10.3109/14653241003774029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND AIMS Chimerism is an important outcome measure in hematopoietic cell transplantation as well as somatic cell therapy. Commonly used methods to estimate chimerism are restricted by either gender or inefficient sensitivity. In principle, real-time polymerase chain reaction (PCR)-based assays can be used to assess single nucleotide polymorphisms (SNP), which are a vast resource of molecular markers, and such assays demonstrate a substantially higher sensitivity (0.001%), but the specificity is unclear because of a low-level signal from mismatched sequences. METHODS In this study, we cloned 14 pairs of SNP selected from the SNP HapMap database and examined the specificity and sensitivity of their detection by real-time PCR using two primer/fluorescent probe pairs to allow genotyping of the two possible variant alleles. Clinical donor-recipient pairs from 18 families were used to explore the efficacy of using SNP assays to measure chimerism. RESULTS We found that the polymorphic nucleotide influences the ability to distinguish the signal generated by the target and mismatched sequences. Moreover, the specific fluorescent reporter probe can affect the difference in signal intensity between the target and mismatched sequences. Real-time PCR SNP assays can attain a sensitivity of 0.1-0.5% with 100% specificity. When comparing possible clinical donor-recipient pairs, we found an average 3.3 out of 14 SNP were informative. CONCLUSIONS By optimal selection of the polymorphic sequences and fluorescent reporter, the real-time PCR SNP assay is superior to the short-tandem repeat chimerism assay and broadly applicable. This strategy may be applied in future clinical trials of bone marrow cell therapy.
Collapse
Affiliation(s)
- Xiaohua Chen
- Department of Pediatrics/Oncology, The Children's Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Miess C, Glashauser A, Denk L, deVries U, Minuth WW. The interface between generating renal tubules and a polyester fleece in comparison to the interstitium of the developing kidney. Ann Biomed Eng 2010; 38:2197-209. [PMID: 20309733 DOI: 10.1007/s10439-010-0006-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Accepted: 03/09/2010] [Indexed: 01/08/2023]
Abstract
An increasing number of investigations is dealing with the repair of acute and chronic renal failure by the application of stem/progenitor cells. However, accurate data concerning the cell biological mechanisms controlling the process of regeneration are scarce. For that reason new implantation techniques, advanced biomaterials and morphogens supporting regeneration of renal parenchyma are under research. Special focus is directed to structural and functional features of the interface between generating tubules and the surrounding interstitial space. The aim of the present experiments was to investigate structural features of the interstitium during generation of tubules. Stem/progenitor cells were isolated from neonatal rabbit kidney and mounted between layers of a polyester fleece to create an artificial interstitium. Perfusion culture was performed for 13 days in chemically defined Iscove's Modified Dulbecco's Medium containing aldosterone (1 x 10(-7) M) as tubulogenic factor. Recordings of the artificial interstitium in comparison to the developing kidney were performed by morphometric analysis, scanning and transmission electron microscopy. The degree of differentiation was registered by immunohistochemistry. The data reveal that generated tubules are embedded in a complex network of fibers consisting of newly synthesized extracellular matrix proteins. Morphometric analysis further shows that the majority of tubules within the artificial interstitium develops in a surprisingly close distance between 5 and 25 mum to each other. The abundance of synthesized extracellular matrix acts obviously as a spacer keeping generated tubules in distance. For comparison, the same principle of construction is found in the developing parenchyma of the neonatal kidney. Most astonishingly, scanning electron microscopy reveals that the composition of interstitial matrix is not homogeneous but differs along a cortico-medullary axis of proceeding tubule development.
Collapse
Affiliation(s)
- C Miess
- Department of Molecular and Cellular Anatomy, University of Regensburg, University Street 31, D-93053 Regensburg, Germany
| | | | | | | | | |
Collapse
|
27
|
Li N, Sarojini H, An J, Wang E. Prosaposin in the secretome of marrow stroma-derived neural progenitor cells protects neural cells from apoptotic death. J Neurochem 2009; 112:1527-38. [PMID: 20050969 DOI: 10.1111/j.1471-4159.2009.06565.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Functionally, adult stem cells not only participate in replication and differentiation to various cell lineages, but also may be involved in rescuing cells from apoptosis. Identifying functional factors secreted by stem cells, as well as their target cells, may advance our understanding of stem cells' multifaceted physiologic functions. Here, we report that mouse bone marrow stromal cell-derived neuroprogenitor cells (mMSC-NPC) provide a protective function by secreting a key factor, prosaposin (PSAP), capable of rescuing mature neurons from apoptotic death. This factor is identified as the lead protein in the secretome of mMSC-NPC cultures by tandem mass spectroscopic profiling, and further validated by western blotting and immunocytochemistry. The secretome of MSC-NPC reduces toxin-induced cell death in cultures of rat pheochromocytoma neuronal cells, human ReNcell CX neurons, and rat cortical primary neurons; removal of PSAP by immunodepletion annuls this protective effect. This neuronal protection against toxin treatment was validated further by the recombinant PSAP peptide. Interestingly, the secretome of neuronal culture does not possess such a self-protective action. We suggest that upon injury, a subgroup of MSCs differentiates into neural/neuronal progenitor cells, and remains in this intermediate stem cell-like stage, defending injured neighboring mature neurons from apoptosis by secreting PSAP.
Collapse
Affiliation(s)
- Na Li
- Department of Biochemistry and Molecular Biology, University of Louisville School of Medicine, Gheens Center on Aging, Louisville, Kentucky 40202, USA
| | | | | | | |
Collapse
|
28
|
Bai K, Huang Y, Jia X, Fan Y, Wang W. Endothelium oriented differentiation of bone marrow mesenchymal stem cells under chemical and mechanical stimulations. J Biomech 2009; 43:1176-81. [PMID: 20022602 DOI: 10.1016/j.jbiomech.2009.11.030] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2009] [Revised: 11/19/2009] [Accepted: 11/24/2009] [Indexed: 10/20/2022]
Abstract
Bone marrow mesenchymal stem cells (MSCs) have multi-differentiation capability. Their endothelial cell (EC) oriented differentiation is the key to vasculogenesis, in which both mechanical and chemical stimulations play important roles. Most previous studies reported individual effects of VEGF or fluid shear stress (SS), when MSCs were subjected to shear stress of 10-15 dyn/cm(2) over 24hr. In this paper, we investigated responses of MSCs from young Sprague Dawley rats to shear stress, VEGF and the combination of the two stimuli. Our study showed that the combined stimulation of shear stress and VEGF resulted in more profound EC oriented differentiation of MSCs in comparison to any individual stimulation. Furthermore, we subjected MSCs to prolonged period of fluid shear stimulation, i.e. 48 hr rather than 24hr, and increased the magnitude of the shear stress from 10 dyn/cm(2) to 15, 20 and 25 dyn/cm(2). We found that without VEGF, the endothelium oriented differentiation of MSCs that was seen following 24hr of shear stimulation was largely abolished if we extended the shear stimulation to 48hr. A similar sharp decrease in MSC differentiation was also observed when the magnitude of the shear stress was increased from 10-15 dyn/cm(2) to 20-25 dyn/cm(2) in 24hr shear stimulation studies. However, with combined VEGF and fluid shear stimulation, most of the endothelial differentiation was retained following an extended period, i.e. at 48 hr, of shear stimulation. Our study demonstrates that chemical and mechanical stimulations work together in determining MSC differentiation dynamics.
Collapse
Affiliation(s)
- Ke Bai
- School of Biological Science and Medical Engineering, Beijing University of Aeronautics and Astronautics, No 37 Xueyuan Road, Beijing 100191, China
| | | | | | | | | |
Collapse
|