201
|
Jung KH, Song SU, Yi T, Jeon MS, Hong SW, Zheng HM, Lee HS, Choi MJ, Lee DH, Hong SS. Human bone marrow-derived clonal mesenchymal stem cells inhibit inflammation and reduce acute pancreatitis in rats. Gastroenterology 2011; 140:998-1008. [PMID: 21130088 DOI: 10.1053/j.gastro.2010.11.047] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2010] [Revised: 10/26/2010] [Accepted: 11/16/2010] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Acute pancreatitis (AP) has a high mortality rate; repetitive AP induces chronic AP and pancreatic adenocarcinoma. Mesenchymal stem cells (MSCs) have immunoregulatory effects and reduce inflammation. We developed a protocol to isolate human bone marrow-derived clonal MSCs (hcMSCs) from bone marrow aspirate and investigated the effects of these cells in rat models of mild and severe AP. METHODS Mild AP was induced in Sprague-Dawley rats by 3 intraperitoneal injections of cerulein (100 μg/kg), given at 2-hour intervals; severe AP was induced by intraparenchymal injection of 3% sodium taurocholate solution. hcMSCs were labeled with CM-1,1'-dioctadecyl-3,3,3'-tetramethylindo-carbocyanine perchloride and administered to rats through the tail vein. RESULTS hcMSCs underwent self-renewal and had multipotent differentiation capacities and immunoregulatory functions. Greater numbers of infused hcMSCs were detected in pancreas of rats with mild and severe AP than of control rats. Infused hcMSCs reduced acinar-cell degeneration, pancreatic edema, and inflammatory cell infiltration in each model of pancreatitis. The hcMSCs reduced expression of inflammation mediators and cytokines in rats with mild and severe AP. hcMSCs suppressed the mixed lymphocyte reaction and increased expression of Foxp3(+) (a marker of regulatory T cells) in cultured rat lymph node cells. Rats with mild or severe AP that were given infusions of hcMSCs had reduced numbers of CD3(+) T cells and increased expression of Foxp3(+) in pancreas tissues. CONCLUSIONS hcMSCs reduced inflammation and damage to pancreatic tissue in a rat model of AP; they reduced levels of cytokines and induced numbers of Foxp3(+) regulatory T cells. hcMSCs might be developed as a cell therapy for pancreatitis.
Collapse
Affiliation(s)
- Kyung Hee Jung
- Department of Biomedical Sciences, College of Medicine, Inha University, Sinheung-dong, Jung-gu, Incheon, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
202
|
Abstract
Acute kidney injury (AKI) as a consequence of ischemia is a common clinical event leading to unacceptably high morbidity and mortality, development of chronic kidney disease (CKD), and transition from pre-existing CKD to end-stage renal disease. Data indicate a close interaction between the many cell types involved in the pathophysiology of ischemic AKI, which has critical implications for the treatment of this condition. Inflammation seems to be the common factor that links the various cell types involved in this process. In this Review, we describe the interactions between these cells and their response to injury following ischemia. We relate these events to patients who are at high risk of AKI, and highlight the characteristics that might predispose these patients to injury. We also discuss how therapy targeting specific cell types can minimize the initial and subsequent injury following ischemia, thereby limiting the extent of acute changes and, hopefully, long-term structural and functional alterations to the kidney.
Collapse
|
203
|
Li JY, Yong TY, Michael MZ, Gleadle JM. Review: The role of microRNAs in kidney disease. Nephrology (Carlton) 2011; 15:599-608. [PMID: 20883280 DOI: 10.1111/j.1440-1797.2010.01363.x] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
MicroRNAs (miRNAs) are short non-coding RNAs that modulate physiological and pathological processes by inhibiting target gene expression via blockade of protein translation or by inducing mRNA degradation. These miRNAs potentially regulate the expression of thousands of proteins. As a result, miRNAs have emerged rapidly as a major new area of biomedical research with relevance to kidney disease. MiRNA expression has been shown to differ between the kidney and other organs as well as between different kidney regions. Furthermore, miRNAs have been found to be functionally important in models of podocyte development, diabetic nephropathy and polycystic kidney disease. Of particular interest, podocyte-specific deletion of Dicer, a key enzyme in the biogenesis of miRNA, results in proteinuria and severe renal impairment in mice. One miRNA (miR-192) can also act as an effector of transforming growth factor-β activity in the high-glucose environment of diabetic nephropathy. Differential expression of miRNAs has been reported in kidney allograft rejection. It is anticipated that future studies involving miRNAs will generate new insights into the complex pathophysiology underlying various kidney diseases, generate diagnostic biomarkers and might be of value as therapeutic targets for progressive kidney diseases. The purpose of this review is to highlight key miRNA developments in kidney diseases and how this might influence the diagnosis and management of patients with kidney disease in the future.
Collapse
Affiliation(s)
- Jordan Yz Li
- Departments of Renal Medicine, Flinders Medical Centre, Flinders University, Adelaide, South Australia, Australia
| | | | | | | |
Collapse
|
204
|
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; 20:1193-1208. [PMID: 21092414 DOI: 10.3727/096368910x543394] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [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
|
205
|
Pleniceanu O, Harari-Steinberg O, Dekel B. Concise review: Kidney stem/progenitor cells: differentiate, sort out, or reprogram? Stem Cells 2010; 28:1649-60. [PMID: 20652959 PMCID: PMC2996087 DOI: 10.1002/stem.486] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
End-stage renal disease (ESRD) is defined as the inability of the kidneys to remove waste products and excess fluid from the blood. ESRD progresses from earlier stages of chronic kidney disease (CKD) and occurs when the glomerular filtration rate (GFR) is below 15 ml/minute/1.73 m2. CKD and ESRD are dramatically rising due to increasing aging population, population demographics, and the growing rate of diabetes and hypertension. Identification of multipotential stem/progenitor populations in mammalian tissues is important for therapeutic applications and for understanding developmental processes and tissue homeostasis. Progenitor populations are ideal targets for gene therapy, cell transplantation, and tissue engineering. The demand for kidney progenitors is increasing due to severe shortage of donor organs. Because dialysis and transplantation are currently the only successful therapies for ESRD, cell therapy offers an alternative approach for kidney diseases. However, this approach may be relevant only in earlier stages of CKD, when kidney function and histology are still preserved, allowing for the integration of cells and/or for their paracrine effects, but not when small and fibrotic end-stage kidneys develop. Although blood- and bone marrow-derived stem cells hold a therapeutic promise, they are devoid of nephrogenic potential, emphasizing the need to seek kidney stem cells beyond known extrarenal sources. Moreover, controversies regarding the existence of a true adult kidney stem cell highlight the importance of studying cell-based therapies using pluripotent cells, progenitor cells from fetal kidney, or dedifferentiated/reprogrammed adult kidney cells. Stem Cells 2010; 28:1649–1660.
Collapse
Affiliation(s)
- Oren Pleniceanu
- Pediatric Stem Cell Research Institute, Sheba Medical Center, Tel Hashomer, Israel
| | | | | |
Collapse
|
206
|
Petrovic V, Jovanovic I, Pesic I, Stefanovic V. Role of stem cells in kidney repair. Ren Fail 2010; 32:1237-44. [DOI: 10.3109/0886022x.2010.517352] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
207
|
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
|
208
|
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: 6.9] [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
|
209
|
Abstract
Microvesicles (MVs) are circular fragments of membrane released from the endosomal compartment as exosomes or shed from the surface membranes of most cell types. An increasing body of evidence indicates that they play a pivotal role in cell-to-cell communication. Indeed, they may directly stimulate target cells by receptor-mediated interactions or may transfer from the cell of origin to various bioactive molecules including membrane receptors, proteins, mRNAs, microRNAs, and organelles. In this review we discuss the pleiotropic biologic effects of MVs that are relevant for communication among cells in physiological and pathological conditions. In particular, we discuss their potential involvement in inflammation, renal disease, and tumor progression, and the evidence supporting a bidirectional exchange of genetic information between stem and injured cells. The transfer of gene products from injured cells may explain stem cell functional and phenotypic changes without the need of transdifferentiation into tissue cells. On the other hand, transfer of gene products from stem cells may reprogram injured cells to repair damaged tissues.
Collapse
|
210
|
Garwood S. Cardiac surgery-associated acute renal injury: new paradigms and innovative therapies. J Cardiothorac Vasc Anesth 2010; 24:990-1001. [PMID: 20702119 DOI: 10.1053/j.jvca.2010.05.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Indexed: 01/02/2023]
Affiliation(s)
- Susan Garwood
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT 06520-8051, USA.
| |
Collapse
|
211
|
Li K, Han Q, Yan X, Liao L, Zhao RC. Not a Process of Simple Vicariousness, the Differentiation of Human Adipose-Derived Mesenchymal Stem Cells to Renal Tubular Epithelial Cells Plays an Important Role in Acute Kidney Injury Repairing. Stem Cells Dev 2010; 19:1267-75. [DOI: 10.1089/scd.2009.0196] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- Kanghua Li
- Institute of Basic Medical Sciences and School of Basic Medicine, Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Qin Han
- Institute of Basic Medical Sciences and School of Basic Medicine, Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Xi Yan
- Institute of Basic Medical Sciences and School of Basic Medicine, Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Lianming Liao
- Institute of Basic Medical Sciences and School of Basic Medicine, Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Robert Chunhua Zhao
- Institute of Basic Medical Sciences and School of Basic Medicine, Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|
212
|
Asanuma H, Vanderbrink BA, Campbell MT, Hile KL, Zhang H, Meldrum DR, Meldrum KK. Arterially delivered mesenchymal stem cells prevent obstruction-induced renal fibrosis. J Surg Res 2010; 168:e51-9. [PMID: 20850784 DOI: 10.1016/j.jss.2010.06.022] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2010] [Revised: 05/12/2010] [Accepted: 06/14/2010] [Indexed: 11/30/2022]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) hold promise for the treatment of renal disease. While MSCs have been shown to accelerate recovery and prevent acute renal failure in multiple disease models, the effect of MSC therapy on chronic obstruction-induced renal fibrosis has not previously been evaluated. MATERIALS AND METHODS Male Sprague-Dawley rats underwent renal artery injection of vehicle or fluorescent-labeled human bone marrow-derived MSCs immediately prior to sham operation or induction of left ureteral obstruction (UUO). One or 4 wk later, the kidneys were harvested and the renal cortex analyzed for evidence of stem cell infiltration, epithelial-mesenchymal transition (EMT) as evidenced by E-cadherin/α-smooth muscle actin (α-SMA) expression and fibroblast specific protein (FSP+) staining, renal fibrosis (collagen content, Masson's trichrome staining), and cytokine and growth factor activity (ELISA and real time RT-PCR). RESULTS Fluorescent-labeled MSCs were detected in the interstitium of the kidney up to 4 wk post-obstruction. Arterially delivered MSCs significantly reduced obstruction-induced α-SMA expression, FSP+ cell accumulation, total collagen content, and tubulointerstitial fibrosis, while simultaneously preserving E-cadherin expression, suggesting that MSCs prevent obstruction-induced EMT and renal fibrosis. Exogenous MSCs reduced obstruction-induced tumor necrosis factor-α (TNF-α) levels, but did not alter transforming growth factor-β1 (TGF-β1), vascular endothelial growth factor (VEGF), interleukin-10 (IL-10), fibroblast growth factor (FGF), or hepatocyte growth factor (HGF) expression. CONCLUSIONS Human bone marrow-derived MSCs remain viable several weeks after delivery into the kidney and provide protection against obstruction-induced EMT and chronic renal fibrosis. While the mechanism of MSCs-induced renal protection during obstruction remains unclear, our results demonstrate that alterations in TNF-α production may be involved.
Collapse
Affiliation(s)
- Hiroshi Asanuma
- Department of Urology, Indiana University School of Medicine, Indianapolis, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | | | |
Collapse
|
213
|
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
|
214
|
Kumar R, Sharma A, Pattnaik AK, Varadwaj PK. Stem cells: An overview with respect to cardiovascular and renal disease. J Nat Sci Biol Med 2010; 1:43-52. [PMID: 22096336 PMCID: PMC3217290 DOI: 10.4103/0976-9668.71674] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
In recent years, there has been a tremendous increase in the understanding of stem cell biology. Stem cells have clonogenic and self-renewing capabilities, and under certain conditions, can differentiate into multiple lineages of mature cells. Recent studies have shown that adult stem cells can be isolated from a wide variety of tissues, including bone marrow, peripheral blood, muscle, and adipose tissue. The potential clinical applications lead to an extended interest in the use of stem cells in many medical disciplines. In this article, we present an overview of stem cells with special reference to cardiovascular and renal diseases treatments by stem cells.
Collapse
Affiliation(s)
- Rajnish Kumar
- Department of Biotechnology, Amity University, Lucknow, Uttar Pradesh, India
| | | | | | | |
Collapse
|
215
|
Kelly KJ, Kluve-Beckerman B, Zhang J, Dominguez JH. Intravenous cell therapy for acute renal failure with serum amyloid A protein-reprogrammed cells. Am J Physiol Renal Physiol 2010; 299:F453-64. [PMID: 20534870 DOI: 10.1152/ajprenal.00050.2010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Serum amyloid A protein (SAA), a prominent component of the acute-phase response, is strongly expressed in developing and repairing kidneys and promotes tubulogenesis. Accordingly, we reprogrammed relatively undifferentiated NRK52E cells with the mouse SAA1.1 gene and transplanted SAA-positive and -negative cells into rats with acute renal failure. We found that SAA-positive cells accelerated renal recovery in three models of acute renal failure: gentamicin nephrotoxicity, cisplatin-mediated renal injury, and ischemia-reperfusion renal injury. The dramatic improvement of renal failure was demonstrable within 2 days, consistent with an early paracrine effect. However, abundant donor cells were also found integrated in the healing tubular architecture after 7 days. We conclude that infusions of SAA-positive cells promote renal recovery after acute renal failure and offer a potentially powerful and novel therapy of renal failure.
Collapse
|
216
|
Ivanova L, Hiatt MJ, Yoder MC, Tarantal AF, Matsell DG. Ontogeny of CD24 in the human kidney. Kidney Int 2010; 77:1123-31. [DOI: 10.1038/ki.2010.39] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
217
|
Sharma AK, Hota PV, Matoka DJ, Fuller NJ, Jandali D, Thaker H, Ameer GA, Cheng EY. Urinary bladder smooth muscle regeneration utilizing bone marrow derived mesenchymal stem cell seeded elastomeric poly(1,8-octanediol-co-citrate) based thin films. Biomaterials 2010; 31:6207-17. [PMID: 20488535 DOI: 10.1016/j.biomaterials.2010.04.054] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Accepted: 04/22/2010] [Indexed: 02/08/2023]
Abstract
Bladder regeneration studies have yielded inconclusive results possibly due to the use of unfavorable cells and primitive scaffold design. We hypothesized that human mesenchymal stem cells seeded onto poly(1,8-octanediol-co-citrate) elastomeric thin films would provide a suitable milieu for partial bladder regeneration. POCfs were created by reacting citric acid with 1,8-octanediol and seeded on opposing faces with human MSCs and urothelial cells; normal bladder smooth muscle cells and UCs, or unseeded POCfs. Partial cystectomized nude rats were augmented with the aforementioned POCfs, enveloped with omentum and sacrificed at 4 and 10 weeks. Isolated bladders were subjected to Trichrome and anti-human gamma-tubulin, calponin, caldesmon, smooth muscle gamma-actin, and elastin stainings. Mechanical testing of POCfs revealed a Young's modulus of 138 kPa with elongation 137% its initial length without permanent deformation demonstrating its high uniaxial elastic potential. Trichrome and immunofluorescent staining of MSC/UC POCf augmented bladders exhibited typical bladder architecture with muscle bundle formation and the expression and retention of bladder smooth muscle contractile proteins of human derivation. Quantitative morphometry of MSC/UC samples revealed muscle/collagen ratios approximately 1.75x greater than SMC/UC controls at 10 weeks. Data demonstrate MSC seeded POCfs support partial regeneration of bladder tissue in vivo.
Collapse
Affiliation(s)
- Arun K Sharma
- Children's Memorial Hospital Chicago, Division of Pediatric Urology, 2300 Children's Plaza, Chicago, IL 60614, USA.
| | | | | | | | | | | | | | | |
Collapse
|
218
|
Semedo P, Donizetti-Oliveira C, Burgos-Silva M, Cenedeze MA, Avancini Costa Malheiros DM, Pacheco-Silva A, Câmara NOS. Bone marrow mononuclear cells attenuate fibrosis development after severe acute kidney injury. J Transl Med 2010; 90:685-95. [PMID: 20308984 DOI: 10.1038/labinvest.2010.45] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
One of the early phases that lead to fibrosis progression is inflammation. Once this stage is resolved, fibrosis might be prevented. Bone marrow mononuclear cells (BMMCs) are emerging as a new therapy for several pathologies, including autoimmune diseases, because they enact immunosuppression. In this study we aimed to evaluate the role of BMMC administration in a model of kidney fibrosis induced by an acute injury. C57Bl6 mice were subjected to unilateral severe ischemia by clamping the left renal pedicle for 1h. BMMCs were isolated from femurs and tibia, and after 6h of reperfusion, 1 x 10(6) cells were administrated intraperitoneally. At 24h after surgery, treated animals showed a significant decrease in creatinine and urea levels when compared with untreated animals. Different administration routes were tested. Moreover, interferon (IFN) receptor knockout BMMCs were used, as this receptor is necessary for BMMC activation. Labeled BMMCs were found in ischemic kidney on FACS analysis. This improved outcome was associated with modulation of inflammation in the kidney and systemic modulation, as determined by cytokine expression profiling. Despite non-amelioration of functional parameters, kidney mRNA expression of interleukin (IL)-6 at 6 weeks was lower in BMMC-treated animals, as were levels of collagen 1, connective tissue growth factor (CTGF), transforming growth factor-beta (TGF-beta) and vimentin. Protective molecules, such as IL-10, heme oxygenase 1 (HO-1) and bone morphogenetic 7 (BMP-7), were increased in treated animals after 6 weeks. Moreover, Masson and Picrosirius red staining analyses showed less fibrotic areas in the kidneys of treated animals. Thus, early modulation of inflammation by BMMCs after an ischemic injury leads to reduced fibrosis through modulation of early inflammation.
Collapse
Affiliation(s)
- Patricia Semedo
- Experimental and Clinical Immunology Laboratory, Division of Nephrology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, Brazil
| | | | | | | | | | | | | |
Collapse
|
219
|
Semedo P, Correa-Costa M, Antonio Cenedeze M, Maria Avancini Costa Malheiros D, Antonia dos Reis M, Shimizu MH, Seguro AC, Pacheco-Silva A, Saraiva Camara NO. Mesenchymal stem cells attenuate renal fibrosis through immune modulation and remodeling properties in a rat remnant kidney model. Stem Cells 2010; 27:3063-73. [PMID: 19750536 DOI: 10.1002/stem.214] [Citation(s) in RCA: 192] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells (MSCs) have regenerative properties in acute kidney injury, but their role in chronic kidney diseases is still unknown. More specifically, it is not known whether MSCs halt fibrosis. The purpose of this work was to investigate the role of MSCs in fibrogenesis using a model of chronic renal failure. MSCs were obtained from the tibias and femurs of male Wistar-EPM rats. Female Wistar rats were subjected to the remnant model, and 2|x|10(5) MSCs were intravenously administrated to each rat every other week for 8 weeks or only once and followed for 12 weeks. SRY gene expression was observed in female rats treated with male MSCs, and immune localization of CD73(+)CD90(+) cells at 8 weeks was also assessed. Serum and urine analyses showed an amelioration of functional parameters in MSC-treated animals at 8 weeks, but not at 12 weeks. Masson's trichrome and Sirius red staining demonstrated reduced levels of fibrosis in MSC-treated animals. These results were corroborated by reduced vimentin, type I collagen, transforming growth factor beta, fibroblast specific protein 1 (FSP-1), monocyte chemoattractant protein 1, and Smad3 mRNA expression and alpha smooth muscle actin and FSP-1 protein expression. Renal interleukin (IL)-6 and tumor necrosis factor alpha mRNA expression levels were significantly decreased after MSC treatment, whereas IL-4 and IL-10 expression levels were increased. All serum cytokine expression levels were decreased in MSC-treated animals. Taken together, these results suggested that MSC therapy can indeed modulate the inflammatory response that follows the initial phase of a chronic renal injury. The immunosuppressive and remodeling properties of MSCs may be involved in the decreased fibrosis in the kidney.
Collapse
Affiliation(s)
- Patricia Semedo
- Nephrology Division, Medicine Department, Federal University of São Paulo, 05508-900 São Paulo, SP, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
220
|
Paracrine/endocrine mechanism of stem cells on kidney repair: role of microvesicle-mediated transfer of genetic information. Curr Opin Nephrol Hypertens 2010; 19:7-12. [PMID: 19823086 DOI: 10.1097/mnh.0b013e328332fb6f] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW The mechanism of stem cell-induced kidney repair remains controversial. Engraftment of bone marrow-derived stem cells is considered a rare event and several studies point to paracrine/endocrine processes. This review focuses on microvesicle-mediated transfer of genetic information between stem cells and injured tissue as a paracrine/endocrine mechanism. RECENT FINDINGS The following findings support a bidirectional exchange of genetic information between stem and injured cells: microvesicles shuttle defined patterns of mRNA and microRNA, are actively released from embryonic and adult stem cells and are internalized by a receptor-mediated mechanism in target cells; transcripts delivered by microvesicles from injured cells may reprogram the phenotype of stem cells to acquire specific features of the tissue; transcripts delivered by microvesicles from stem cells may induce dedifferentiation of cells surviving injury with cell cycle reentry and tissue self-repair. SUMMARY Transfer of genetic information from injured cells may explain stem cell functional and phenotypic changes without the need for transdifferentiation into tissue cells. On the contrary, transfer of genetic information from stem cells may redirect altered functions in target cells suggesting that stem cells may repair damaged tissues without directly replacing parenchymal cells.
Collapse
|
221
|
Park HC, Yasuda K, Kuo MC, Ni J, Ratliff B, Chander P, Goligorsky MS. Renal capsule as a stem cell niche. Am J Physiol Renal Physiol 2010; 298:F1254-62. [PMID: 20200095 DOI: 10.1152/ajprenal.00406.2009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Renal resident stem cells were previously reported within the renal tubules and papillary area. The aim of the present study was to determine whether renal capsules harbor stem cells and whether this pool can be recruited to the renal parenchyma after ischemic injury. We demonstrated the presence of label-retaining cells throughout the renal capsule, at a density of ∼10 cells/mm(2), and their close apposition to the blood vessels. By flow cytometry, in vitro cultured cells derived from the renal capsule were positive for mesenchymal stem cell (MSC) markers (CD29+, vimentin+, Sca-1+, nestin+) but did not express hematopoietic and endothelial stem cell markers. Moreover, renal capsule-derived cells also exhibited self-renewal, clonogenicity, and multipotency in differentiation conditions, all favoring stem cell characteristics and identifying them with MSC. In situ labeling of renal capsules with CM-DiI CellTracker demonstrated in vivo a directed migration of CM-DiI-labeled cells to the ischemic renal parenchyma, with the rate of migration averaging 30 μm/h. Decapsulation of the kidneys during ischemia resulted in a modest, but statistically significant, deceleration of recovery of plasma creatinine compared with ischemic kidneys with intact renal capsule. Comparison of these conditions allows the conclusion that renal capsular cells may contribute ∼25-30% of the recovery from ischemia. In conclusion, the data suggest that the renal capsule may function as a novel stem cell niche harboring MSC capable of participating in the repair of renal injury.
Collapse
Affiliation(s)
- Hyeong-Cheon Park
- Department of Medicine, New York Medical College, Valhalla, New York, USA.
| | | | | | | | | | | | | |
Collapse
|
222
|
Perin L, Sedrakyan S, Giuliani S, Da Sacco S, Carraro G, Shiri L, Lemley KV, Rosol M, Wu S, Atala A, Warburton D, De Filippo RE. Protective effect of human amniotic fluid stem cells in an immunodeficient mouse model of acute tubular necrosis. PLoS One 2010; 5:e9357. [PMID: 20195358 PMCID: PMC2827539 DOI: 10.1371/journal.pone.0009357] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Accepted: 01/29/2010] [Indexed: 01/17/2023] Open
Abstract
Acute Tubular Necrosis (ATN) causes severe damage to the kidney epithelial tubular cells and is often associated with severe renal dysfunction. Stem-cell based therapies may provide alternative approaches to treating of ATN. We have previously shown that clonal c-kitpos stem cells, derived from human amniotic fluid (hAFSC) can be induced to a renal fate in an ex-vivo system. Herein, we show for the first time the successful therapeutic application of hAFSC in a mouse model with glycerol-induced rhabdomyolysis and ATN. When injected into the damaged kidney, luciferase-labeled hAFSC can be tracked using bioluminescence. Moreover, we show that hAFSC provide a protective effect, ameliorating ATN in the acute injury phase as reflected by decreased creatinine and BUN blood levels and by a decrease in the number of damaged tubules and apoptosis therein, as well as by promoting proliferation of tubular epithelial cells. We show significant immunomodulatory effects of hAFSC, over the course of ATN. We therefore speculate that AFSC could represent a novel source of stem cells that may function to modulate the kidney immune milieu in renal failure caused by ATN.
Collapse
Affiliation(s)
- Laura Perin
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Childrens Hospital Los Angeles, Los Angeles, California, United States of America
- University of Southern California Institute of Urology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Sargis Sedrakyan
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Childrens Hospital Los Angeles, Los Angeles, California, United States of America
- University of Southern California Institute of Urology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Stefano Giuliani
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Childrens Hospital Los Angeles, Los Angeles, California, United States of America
- University of Southern California Institute of Urology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Stefano Da Sacco
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Childrens Hospital Los Angeles, Los Angeles, California, United States of America
- University of Southern California Institute of Urology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Gianni Carraro
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Childrens Hospital Los Angeles, Los Angeles, California, United States of America
- University of Southern California Institute of Urology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Liron Shiri
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Childrens Hospital Los Angeles, Los Angeles, California, United States of America
- University of Southern California Institute of Urology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Kevin V. Lemley
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Childrens Hospital Los Angeles, Los Angeles, California, United States of America
- University of Southern California Institute of Urology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Michael Rosol
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Childrens Hospital Los Angeles, Los Angeles, California, United States of America
- University of Southern California Institute of Urology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Sam Wu
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Childrens Hospital Los Angeles, Los Angeles, California, United States of America
- University of Southern California Institute of Urology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
| | - David Warburton
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Childrens Hospital Los Angeles, Los Angeles, California, United States of America
- University of Southern California Institute of Urology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Roger E. De Filippo
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute, Childrens Hospital Los Angeles, Los Angeles, California, United States of America
- University of Southern California Institute of Urology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
223
|
Abstract
PURPOSE OF REVIEW Secondary prevention follows identification of acute kidney injury (AKI), in which functional outcome is incomplete. Secondary prevention requires biomarkers for early diagnosis and response to appropriate treatment, on-going injury and repair, and meaningful metrics to monitor outcome. This review summarizes recent research in these areas. RECENT FINDINGS Proteomics and genetic studies have identified new risk factors and biomarkers of AKI. Biomarker performance studies reveal differences in prognostic performance according to population and AKI definition. The first early secondary prevention study utilizing a urinary biomarker of AKI as a triaging tool to randomize to treatment has been completed. Recent creatinine-kinetic modelling has highlighted issues with defining AKI which continue to make comparison of treatment outcomes difficult. Biomarkers to monitor repair are emerging. SUMMARY The prognostic performance of novel biomarkers of AKI in a range of clinical settings is encouraging and critical to effective secondary prevention. Identification of cause and time-course of specific biomarkers are required before biomarker panels for secondary prevention are developed. Agreed standards around reporting of biomarker studies would facilitate comparisons between studies. Phase-specific biomarkers are required to triage to phase-specific treatment.
Collapse
|
224
|
Gheisari Y, Nassiri SM, Arefian E, Ahmadbeigi N, Azadmanesh K, Jamali M, Jahanzad I, Zeinali S, Vasei M, Soleimani M. Severely damaged kidneys possess multipotent renoprotective stem cells. Cytotherapy 2010; 12:303-12. [DOI: 10.3109/14653241003709645] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
225
|
Abstract
The application of stem cells and their use in tissue-engineering approaches is emerging in clinical therapeutic intervention strategies. The use of adult stem cells, either autologous or allogenic, does not raise ethical concerns, in contrast to embryonic stem cells. Mesenchymal stromal cells (MSCs) can be easily obtained from bone marrow or from adipose tissue and further expanded in vitro. Due to their differentiation capacity, MSCs are very attractive for tissue engineering purposes. Furthermore, MSCs secrete a variety of mediators that have beneficial effects on the regenerating tissue. In this review we give an insight into stem cell hierarchy, define the properties of MSCs and summarize recent reports of their administration in urological diseases.
Collapse
Affiliation(s)
- Katrin Montzka
- Department of Urology, RWTH University Aachen, Aachen, Germany.
| | | |
Collapse
|
226
|
Reinders MEJ, Fibbe WE, Rabelink TJ. Multipotent mesenchymal stromal cell therapy in renal disease and kidney transplantation. Nephrol Dial Transplant 2009; 25:17-24. [PMID: 19861311 DOI: 10.1093/ndt/gfp552] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cell therapies aim at differentiation of stem cells into the specific cell type required to repair damaged or destroyed cells or tissues. Over recent years, cell therapy has been introduced in a variety of application areas, including cardiovascular repair, diabetes, musculoskeletal disorders and renal repair. Multipotent mesenchymal stromal cells (MSCs), often referred to as mesenchymal stem cells, are of particular interest as a cell therapy model, as this is one of the few cell types that are on the brink of entering the clinical arena in different areas of application. MSCs can be differentiated in vitro and in vivo into various cell types of mesenchymal origin such as bone, fat and cartilage. They have important effects on the innate and adaptive immune system and possess striking anti-inflammatory properties that make them attractive for potential use in diseases characterized by autoimmunity and inflammation. In addition, MSCs have been shown to migrate to sites of tissue injury and to enhance repair by secreting anti-fibrotic and pro-angiogenic factors. In this review, evidence for the renoprotective mechanisms of MSCs as well as their therapeutic possibilities and potential hazards in acute and chronic renal disease and allograft rejection is summarized.
Collapse
|
227
|
Barzilay R, Sadan O, Melamed E, Offen D. Comparative characterization of bone marrow-derived mesenchymal stromal cells from four different rat strains. Cytotherapy 2009; 11:435-42. [PMID: 19521891 DOI: 10.1080/14653240902849796] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND AIMS Bone marrow (BM) multipotent mesenchymal stromal cells (MSC) hold great potential for cell-based regenerative medicine. Because of the growing use of autologous rat MSC transplantation in various rat models, there is a need to establish minimal criteria for rat MSC characterization independent of the specific strain employed in each study. We aimed to compare the phenotypic and functional traits of BM MSC from the four strains of rats commonly used in research: Fisher, Lewis, Sprague-Dawley and Wistar. METHODS Rat MSC were isolated from the BM of the four different rat strains in an identical fashion. Cells were characterized for their cell-surface phenotype in early and late passage. Functional mesenchymal differentiation capacities were examined following adipogenic and osteogenic inductions. Population doubling times were determined across the four strains throughout 10 passages. In vitro proliferation assays of immune cells were conducted following co-culture of spleen cells and MSC of the four different strains. RESULTS We found that rat MSC from different strains exhibited similar cell-surface phenotype. Expansion rates and differentiation capacities of the MSC were also similar across the different strains. Co-culture of rat MSC with spleen cells obtained from rats of a different strain did not induce proliferation of immune cells. CONCLUSIONS Our findings suggest that BM-derived MSC from different strains share similar characteristics, in contrast to the variations previously described in the characterization of mice MSC from different strains.
Collapse
Affiliation(s)
- Ran Barzilay
- Felsenstein Medical Research Center, Rabin Medical Center and Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | |
Collapse
|
228
|
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
|
229
|
Abstract
The kidney has a remarkable capacity to regenerate after injury, as it is not a terminally differentiated organ. This regenerative potential is somehow incomplete, however, and as the insult continues, progressive and irreversible scarring results in chronic renal disease. Dialysis and organ transplantation are nonspecific and incomplete methods of renal replacement therapy. Stem cells may provide a more efficacious method for both prevention and amelioration of renal disease of many etiologies. Although many reports have claimed the existence of renal-specific stem or progenitor cells isolated and characterized by various methods, the results have been diverse and debatable. The bone marrow stem cells seem to play a minor role in renal regeneration after acute ischemia in mice through transdifferentiation and cell fusion, but their immediate paracrine effects result in considerable improvements in renal function. Therefore, as in stem cell therapy for the heart, bone marrow-derived stem cells show promise in regeneration of the kidney. Although more research is needed in the basic science of renal regeneration, clinical research in animals has demonstrated the versatility of stem cell therapy. The first phase of clinical trials of bone marrow mesenchymal cells in protection against acute kidney injury may begin shortly. This will enable further exploration of stem cell therapy in renal patients with multiple comorbidities.
Collapse
|
230
|
Bonventre JV. Microvesicles from mesenchymal stromal cells protect against acute kidney injury. J Am Soc Nephrol 2009; 20:927-8. [PMID: 19389839 DOI: 10.1681/asn.2009030322] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
231
|
Bruno S, Grange C, Deregibus MC, Calogero RA, Saviozzi S, Collino F, Morando L, Busca A, Falda M, Bussolati B, Tetta C, Camussi G. Mesenchymal stem cell-derived microvesicles protect against acute tubular injury. J Am Soc Nephrol 2009; 20:1053-67. [PMID: 19389847 DOI: 10.1681/asn.2008070798] [Citation(s) in RCA: 1014] [Impact Index Per Article: 63.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Administration of mesenchymal stem cells (MSCs) improves the recovery from acute kidney injury (AKI). The mechanism may involve paracrine factors promoting proliferation of surviving intrinsic epithelial cells, but these factors remain unknown. In the current study, we found that microvesicles derived from human bone marrow MSCs stimulated proliferation in vitro and conferred resistance of tubular epithelial cells to apoptosis. The biologic action of microvesicles required their CD44- and beta1-integrin-dependent incorporation into tubular cells. In vivo, microvesicles accelerated the morphologic and functional recovery of glycerol-induced AKI in SCID mice by inducing proliferation of tubular cells. The effect of microvesicles on the recovery of AKI was similar to the effect of human MSCs. RNase abolished the aforementioned effects of microvesicles in vitro and in vivo, suggesting RNA-dependent biologic effects. Microarray analysis and quantitative real time PCR of microvesicle-RNA extracts indicate that microvesicles shuttle a specific subset of cellular mRNA, such as mRNAs associated with the mesenchymal phenotype and with control of transcription, proliferation, and immunoregulation. These results suggest that microvesicles derived from MSCs may activate a proliferative program in surviving tubular cells after injury via a horizontal transfer of mRNA.
Collapse
Affiliation(s)
- Stefania Bruno
- Department of Internal Medicine, Research Center for Experimental Medicine, University of Torino, Torino, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
232
|
Liang M, Liu Y, Mladinov D, Cowley AW, Trivedi H, Fang Y, Xu X, Ding X, Tian Z. MicroRNA: a new frontier in kidney and blood pressure research. Am J Physiol Renal Physiol 2009; 297:F553-8. [PMID: 19339633 DOI: 10.1152/ajprenal.00045.2009] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
MicroRNA (miRNA) has emerged rapidly as a major new direction in many fields of research including kidney and blood pressure research. A mammalian genome encodes several hundred miRNAs. These miRNAs potentially regulate the expression of thousands of proteins. miRNA expression profiles differ substantially between the kidney and other organs as well as between kidney regions. miRNAs may be functionally important in models of diabetic nephropathy, podocyte development, and polycystic disease. miRNAs may be involved in the regulation of arterial blood pressure, including possible involvement in genetic elements of hypertension. Studies of miRNAs could generate diagnostic biomarkers for kidney disease and new mechanistic insights into the complex regulatory networks underlying kidney disease and hypertension. Further progress in the understanding of miRNA biogenesis and action and technical improvements for target identification and miRNA manipulation will be important for studying miRNAs in renal function and blood pressure regulation.
Collapse
Affiliation(s)
- Mingyu Liang
- Dept. of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
233
|
Abstract
Acute kidney injury (AKI) is a very common condition encountered in a hospital setting. AKI is an independent risk factor for in-hospital mortality. In this review, we discuss in detail about the tubular, inflammatory and vascular molecular targets of AKI which may result in therapies to improve mortality and biomarkers for earlier diagnosis of AKI.
Collapse
|
234
|
Abstract
Chronic kidney disease (CKD) is increasing at the rate of 6-8% per annum in the US alone. At present, dialysis and transplantation remain the only treatment options. However, there is hope that stem cells and regenerative medicine may provide additional regenerative options for kidney disease. Such new treatments might involve induction of repair using endogenous or exogenous stem cells or the reprogramming of the organ to reinitiate development. This review addresses the current state of understanding with respect to the ability of non-renal stem cell sources to influence renal repair, the existence of endogenous renal stem cells and the biology of normal renal repair in response to damage. It also examines the remaining challenges and asks the question of whether there is one solution for all forms of renal disease.
Collapse
Affiliation(s)
- C Hopkins
- Institute for Molecular Bioscience, University of Queensland, St. Lucia, Australia
| | | | | | | |
Collapse
|
235
|
Semedo P, Palasio CG, Oliveira CD, Feitoza CQ, Gonçalves GM, Cenedeze MA, Wang PMH, Teixeira VPA, Reis MA, Pacheco-Silva A, Câmara NOS. Early modulation of inflammation by mesenchymal stem cell after acute kidney injury. Int Immunopharmacol 2009; 9:677-82. [PMID: 19146993 DOI: 10.1016/j.intimp.2008.12.008] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2008] [Revised: 11/17/2008] [Accepted: 12/10/2008] [Indexed: 12/29/2022]
Abstract
Therapy with stem cells has showed to be promising for acute kidney injury (AKI), although how it works is still controversial. Modulation of the inflammatory response is one possible mechanism. Most of published data relies on early time and whether the protection is still maintained after that is not known. Here, we analyzed whether immune modulation continues after 24 h of reperfusion. MSC were obtained from male Wistar rats. After 3-5 passages, cells were screened for CD73, CD90, CD44, CD45, CD29 and CD 31. In addition, MSC were submitted to differentiation in adipocyte and in osteocyte. AKI was induced by bilaterally clamping of renal pedicles for 60 min. Six hours after injury, MSC (2 x 10(5) cells) were administered intravenously. MSC-treated animals presented the lowest serum creatinine compared to non-treated animals (24 h: 1.3+/-0.21 vs. 3.23+/-0.89 mg/dl, p<0.05). The improvement in renal function was followed by a lower expression of IL-1b, IL-6 and TNF-alpha and higher expression of IL-4 and IL-10. However, 48 h after reperfusion, this cytokine profile has changed. The decrease in Th1 cytokines was less evident and IL-6 was markedly up regulated. PCNA analysis showed that regeneration occurs faster in kidney tissues of MSC-treated animals than in controls at 24 h. And also ratio of Bcl-2/Bad was higher at treated animals after 24 and 48 h. Our data demonstrated that the immunomodulatory effects of MSC occur at very early time point, changing the inflammation profile toward a Th2 profile.
Collapse
Affiliation(s)
- Patricia Semedo
- Division of Nephrology, Federal University of São Paulo, São Paulo (UNIFESP), Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
236
|
Tögel F, Zhang P, Hu Z, Westenfelder C. VEGF is a mediator of the renoprotective effects of multipotent marrow stromal cells in acute kidney injury. J Cell Mol Med 2008; 13:2109-2114. [PMID: 19397783 DOI: 10.1111/j.1582-4934.2008.00641.x] [Citation(s) in RCA: 128] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Adult stem cell treatment of complex disorders is a promising therapeutic approach and multipotent marrow stromal cells (MSCs) have been shown to be effective in various animal models of diseases. Acute kidney injury (AKI) is a common and serious problem in hospitalized patients and bone marrow derived multipotent MSCs have been shown to be effective in different models of AKI. The mechanism of action of MSCs is complex but involves paracrine actions including growth factor secretion. Knockdown of vascular enthothelial growth factor (VEGF) by siRNA reduced effectiveness of MSCs in the treatment of ischemic AKI in a rat model. Animals treated with MSCs had increased renal microvessel density compared to VEGF knockdown MSC-treated and vehicle-treated animals. These results show that VEGF is an important mediator of the early and late phase of renoprotective action after AKI in the context of stem cell treatment.
Collapse
Affiliation(s)
- Florian Tögel
- University of Utah, Division of Nephrology, Department of Medicine, Salt Lake City, UT, USA.,VA Medical Center, Section of Nephrology, Salt Lake City, UT, USA
| | - Ping Zhang
- University of Utah, Division of Nephrology, Department of Medicine, Salt Lake City, UT, USA.,VA Medical Center, Section of Nephrology, Salt Lake City, UT, USA
| | - Zhuma Hu
- University of Utah, Division of Nephrology, Department of Medicine, Salt Lake City, UT, USA.,VA Medical Center, Section of Nephrology, Salt Lake City, UT, USA
| | - Christof Westenfelder
- University of Utah, Division of Nephrology, Department of Medicine, Salt Lake City, UT, USA.,VA Medical Center, Section of Nephrology, Salt Lake City, UT, USA.,Department of Physiology, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
237
|
Ronconi E, Sagrinati C, Angelotti ML, Lazzeri E, Mazzinghi B, Ballerini L, Parente E, Becherucci F, Gacci M, Carini M, Maggi E, Serio M, Vannelli GB, Lasagni L, Romagnani S, Romagnani P. Regeneration of glomerular podocytes by human renal progenitors. J Am Soc Nephrol 2008; 20:322-32. [PMID: 19092120 DOI: 10.1681/asn.2008070709] [Citation(s) in RCA: 406] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Depletion of podocytes, common to glomerular diseases in general, plays a role in the pathogenesis of glomerulosclerosis. Whether podocyte injury in adulthood can be repaired has not been established. Here, we demonstrate that in the adult human kidney, CD133+CD24+ cells consist of a hierarchical population of progenitors that are arranged in a precise sequence within Bowman's capsule and exhibit heterogeneous potential for differentiation and regeneration. Cells localized to the urinary pole that expressed CD133 and CD24, but not podocyte markers (CD133+CD24+PDX- cells), could regenerate both tubular cells and podocytes. In contrast, cells localized between the urinary pole and vascular pole that expressed both progenitor and podocytes markers (CD133+CD24+PDX+) could regenerate only podocytes. Finally, cells localized to the vascular pole did not exhibit progenitor markers, but displayed phenotypic features of differentiated podocytes (CD133-CD24-PDX+ cells). Injection of CD133+CD24+PDX- cells, but not CD133+CD24+PDX+ or CD133-CD24- cells, into mice with adriamycin-induced nephropathy reduced proteinuria and improved chronic glomerular damage, suggesting that CD133+CD24+PDX- cells could potentially treat glomerular disorders characterized by podocyte injury, proteinuria, and progressive glomerulosclerosis.
Collapse
Affiliation(s)
- Elisa Ronconi
- Excellence Center for Research, Transfer and High Education Denothe, University of Florence, 50139, Firenze, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
238
|
Abdallah BM, Kassem M. The use of mesenchymal (skeletal) stem cells for treatment of degenerative diseases: current status and future perspectives. J Cell Physiol 2008; 218:9-12. [PMID: 18726996 DOI: 10.1002/jcp.21572] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Human bone marrow derived-mesenchymal (skeletal) stem (MSC) cells are a group of non-hematopoietic stem cells residing in the perivascular niches in bone marrow. These cells have the capacity to differentiate mainly into mesoderm-type cells such as osteoblasts, chondrocytes and adipocytes and possibly but not proven to non-mesodermal cell types. Recently, there has been an increased interest in understanding the biology of MSC due to their potential use in cell-based therapy for multiple degenerative diseases. Here, we will provide an update on the current status of these novel therapeutic opportunities. J. Cell. Physiol. 218: 9-12, 2009. (c) 2008 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- Basem M Abdallah
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital & Medical Biotechnology Centre, University of Southern Denmark, Odense, Denmark.
| | | |
Collapse
|
239
|
Jin JD, Wang HX, Xiao FJ, Wang JS, Lou X, Hu LD, Wang LS, Guo ZK. A novel rich source of human mesenchymal stem cells from the debris of bone marrow samples. Biochem Biophys Res Commun 2008; 376:191-5. [PMID: 18774774 DOI: 10.1016/j.bbrc.2008.08.131] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2008] [Accepted: 08/22/2008] [Indexed: 02/07/2023]
Abstract
The debris from human bone marrow (BM) samples is generally filtered out and discarded prior to isolation of mesenchymal stem cells (MSCs). The purpose of this study is to develop a method to harvest MSCs from the debris and investigate their biological characteristics compared with the marrow counterparts. The BM tissue fragments were digested with collagenase and this treatment yielded mononuclear cells half to those from the corresponding filtered BM. The frequencies of colony-forming unit-fibroblast in these two cell populations were not significantly different. MSCs of two origins exhibited similar morphological and phenotypic features. Fluorescent dye-dilution assay showed that they grew at comparable rates both in the primary and passaging cultures. Further, they could be induced into osteoblasts, chondroblasts and adipocytes, as revealed by histological and molecular examinations. Thus, BM tissue fragments may serve as a new source of MSCs in the settings of bench experiments and clinical trials.
Collapse
Affiliation(s)
- Ji-De Jin
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| | | | | | | | | | | | | | | |
Collapse
|
240
|
Bussolati B, Bruno S, Grange C, Ferrando U, Camussi G. Identification of a tumor-initiating stem cell population in human renal carcinomas. FASEB J 2008; 22:3696-705. [PMID: 18614581 DOI: 10.1096/fj.08-102590] [Citation(s) in RCA: 250] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The purpose of the present study was to search for the presence of a tumor-initiating stem cell population in renal carcinomas. Based on the recent identification of mesenchymal stem cells in normal kidneys, we sorted cells expressing the mesenchymal stem cell marker CD105 from 5 human renal carcinomas. Because the CD105(+) but not the CD105(-) population showed enhanced tumorigenicity when injected in severely compromised immunodeficient (SCID) mice, we cloned and characterized CD105(+) cells and evaluated their stemness, differentiative ability, and serial tumor generation. Characterization of the phenotype of CD105(+) clones revealed several stem cell properties: 1) clonogenic ability, 2) expression of nestin, Nanog, Oct4 stem cell markers, and lack of differentiative epithelial markers, 3) ability to grow in nonadhesive spheroids, 4) in vitro differentiation into epithelial and endothelial cell types, and 5) generation in vivo of serially transplantable carcinomas containing an undifferentiated CD105(+) tumorigenic and a differentiated CD105(-) nontumorigenic population. In addition, some vessels present in carcinomas generated from CD105(+) clones were of human origin, suggesting the capability of tumor-initiating stem cells to in vivo differentiate also in endothelial cells. In conclusion, we demonstrate that CD105(+) cells and clones derived from renal carcinomas were enriched in tumor-initiating cells with stem characteristics.
Collapse
Affiliation(s)
- Benedetta Bussolati
- Department of Internal Medicine, Center for Molecular Biotechnology, Turin, Italy
| | | | | | | | | |
Collapse
|
241
|
Tögel F, Yang Y, Zhang P, Hu Z, Westenfelder C. Bioluminescence imaging to monitor the in vivo distribution of administered mesenchymal stem cells in acute kidney injury. Am J Physiol Renal Physiol 2008; 295:F315-21. [PMID: 18480180 DOI: 10.1152/ajprenal.00098.2008] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Effective and targeted delivery of cells to injured organs is critical to the development of cell therapies. However, currently available in vivo cell tracking methods still lack sufficient sensitivity and specificity. We examined, therefore, whether a highly sensitive and specific bioluminescence method is suitable to noninvasively image the organ distribution of administered mesenchymal stem cells (MSCs) in vivo. MSCs were transfected with a luciferase/neomycin phosphotransferase construct (luc/neo-MSC). Bioluminescence of these cells was measured (charge-coupled device camera) after treatment with luciferin, showing a linear increase of photon emission with rising cell numbers. To track these cells in vivo, groups of mice were injected with 1 x 10(5) luc/neo-MSCs/animal and imaged with bioluminescence imaging at various time points. Injection of cells in the suprarenal aorta showed diffuse distribution of cells in normal animals, whereas distinct localization to the kidneys was observed in mice with ischemia- and reperfusion-induced acute kidney injury (AKI). Intrajugular infusion of MSCs demonstrated predominant accumulation of cells in both lungs. In animals with AKI, detectable cell numbers declined over time, as assessed by bioluminescence imaging and confirmed by PCR, a process that was associated with low apoptosis levels of intrarenally located MSCs. In conclusion, the described bioluminescence technology provides a sensitive and safe tool for the repeated in vivo tracking of infused luc/neo-MSCs in all major organs. This method will be of substantial utility in the preclinical testing and design of cell therapeutic strategies in kidney and other diseases.
Collapse
Affiliation(s)
- Florian Tögel
- Department of Medicine/Nephrology, University of Utah, Salt Lake City, UT, USA.
| | | | | | | | | |
Collapse
|
242
|
Perin L, Giuliani S, Sedrakyan S, DA Sacco S, De Filippo RE. Stem cell and regenerative science applications in the development of bioengineering of renal tissue. Pediatr Res 2008; 63:467-71. [PMID: 18427289 DOI: 10.1203/pdr.0b013e3181660653] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
A rising number of patients with acute and chronic renal failure worldwide have created urgency for clinicians and investigators to search out alternative therapies other than chronic renal dialysis and/or organ transplantation. This review focuses on the recent achievements in this area, and discusses the various approaches in the development of bioengineering of renal tissue including recent discoveries in the field of regenerative medicine research and stem cells. A variety of stem cells, ranging from embryonic, bone marrow, endogenous, and amniotic fluid, have been investigated and may prove useful as novel alternatives for organ regeneration both in vitro and in vivo. Tissue engineering, developmental biology, and therapeutic cloning techniques have significantly contributed to our understanding of some of the molecular mechanisms involved in renal regeneration and have demonstrated that renal tissue can be generated de novo with similar physiologic functions as native tissue. Ultimately all of these emerging technologies may provide viable therapeutic options for regenerative medicine applications focused on the bioengineering of renal tissue for the future.
Collapse
Affiliation(s)
- Laura Perin
- Children's Hospital Los Angeles, Division of Urology, Saban Research Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA
| | | | | | | | | |
Collapse
|
243
|
Abstract
Acute kidney injury (AKI) is a common condition with a high risk of death. The standard metrics used to define and monitor the progression of AKI, such as serum creatinine and blood urea nitrogen levels, are insensitive, nonspecific, and change significantly only after significant kidney injury and then with a substantial time delay. This delay in diagnosis not only prevents timely patient management decisions, including administration of putative therapeutic agents, but also significantly affects the preclinical evaluation of toxicity thereby allowing potentially nephrotoxic drug candidates to pass the preclinical safety criteria only to be found to be clinically nephrotoxic with great human costs. Studies to establish effective therapies for AKI will be greatly facilitated by two factors: (a) development of sensitive, specific, and reliable biomarkers for early diagnosis/prognosis of AKI in preclinical and clinical studies, and (b) development and validation of high-throughput innovative technologies that allow rapid multiplexed detection of multiple markers at the bedside.
Collapse
Affiliation(s)
- Vishal S Vaidya
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | | | | |
Collapse
|