1
|
Kazeminia S, Zhu XY, Tang H, Jordan KL, Saadiq IM, Herrmann SM, Chade AR, Irazabal MV, Lerman LO, Eirin A. Renal ischemia alters the transcriptomic and epigenetic profile of inflammatory genes in swine scattered tubular-like cells. Clin Sci (Lond) 2023; 137:1265-1283. [PMID: 37606084 PMCID: PMC10644845 DOI: 10.1042/cs20230555] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/31/2023] [Accepted: 08/11/2023] [Indexed: 08/23/2023]
Abstract
BACKGROUND Scattered tubular-like cells (STCs) are differentiated renal tubular cells that during recovery from ischemic injury dedifferentiate to repair other injured renal cells. Renal artery stenosis (RAS), often associated with chronic inflammatory injury, compromises the integrity and function of STCs, but the underlying mechanisms remain unknown. We hypothesized that RAS alters the transcriptomic and epigenetic profile of inflammatory genes in swine STCs. METHODS STCs were harvested from pig kidneys after 10 weeks of RAS or sham (n=6 each). STC mRNA profiles of inflammatory genes were analyzed using high-throughput mRNA-sequencing (seq) and their DNA methylation (5mC) and hydroxymethylation (5hmC) profiles by DNA immunoprecipitation and next-generation sequencing (MeDIP-seq) (n=3 each), followed by an integrated (mRNA-seq/MeDIP-seq) analysis. STC protein expression of candidate differentially expressed (DE) genes and common proinflammatory proteins were subsequently assessed in vitro before and after epigenetic (Bobcat339) modulation. RESULTS mRNA-seq identified 57 inflammatory genes up-regulated in RAS-STCs versus Normal-STCs (>1.4 or <0.7-fold, P<0.05), of which 14% exhibited lower 5mC and 5% higher 5hmC levels in RAS-STCs versus Normal-STCs, respectively. Inflammatory gene and protein expression was higher in RAS-STCs compared with Normal-STCs but normalized after epigenetic modulation. CONCLUSIONS These observations highlight a novel modulatory mechanism of this renal endogenous repair system and support development of epigenetic or anti-inflammatory therapies to preserve the reparative capacity of STCs in individuals with RAS.
Collapse
Affiliation(s)
- Sara Kazeminia
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Xiang-Yang Zhu
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Hui Tang
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Kyra L. Jordan
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Ishran M. Saadiq
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Sandra M. Herrmann
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Alejandro R. Chade
- Department of Medical Pharmacology and Physiology and Department of Medicine, University of Missouri-Columbia
| | - Maria V. Irazabal
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Lilach O. Lerman
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States
| | - Alfonso Eirin
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
2
|
Veys K, Berlingerio SP, David D, Bondue T, Held K, Reda A, van den Broek M, Theunis K, Janssen M, Cornelissen E, Vriens J, Diomedi-Camassei F, Gijsbers R, van den Heuvel L, Arcolino FO, Levtchenko E. Urine-Derived Kidney Progenitor Cells in Cystinosis. Cells 2022; 11:cells11071245. [PMID: 35406807 PMCID: PMC8997687 DOI: 10.3390/cells11071245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/14/2022] [Accepted: 03/31/2022] [Indexed: 12/10/2022] Open
Abstract
Nephropathic cystinosis is an inherited lysosomal storage disorder caused by pathogenic variants in the cystinosin (CTNS) gene and is characterized by the excessive shedding of proximal tubular epithelial cells (PTECs) and podocytes into urine, development of the renal Fanconi syndrome and end-stage kidney disease (ESKD). We hypothesized that in compensation for epithelial cell losses, cystinosis kidneys undertake a regenerative effort, and searched for the presence of kidney progenitor cells (KPCs) in the urine of cystinosis patients. Urine was cultured in a specific progenitor medium to isolate undifferentiated cells. Of these, clones were characterized by qPCR, subjected to a differentiation protocol to PTECs and podocytes and assessed by qPCR, Western blot, immunostainings and functional assays. Cystinosis patients voided high numbers of undifferentiated cells in urine, of which various clonal cell lines showed a high capacity for self-renewal and expressed kidney progenitor markers, which therefore were assigned as cystinosis urine-derived KPCs (Cys-uKPCs). Cys-uKPC clones showed the capacity to differentiate between functional PTECs and/or podocytes. Gene addition with wild-type CTNS using lentiviral vector technology resulted in significant reductions in cystine levels. We conclude that KPCs present in the urine of cystinosis patients can be isolated, differentiated and complemented with CTNS in vitro, serving as a novel tool for disease modeling.
Collapse
Affiliation(s)
- Koenraad Veys
- Department of Pediatrics, University Hospitals Leuven Campus Gasthuisberg, B-3000 Leuven, Belgium;
- Laboratory of Pediatric Nephrology, Department of Development & Regeneration, KU Leuven Campus Gasthuisberg, B-3000 Leuven, Belgium; (S.P.B.); (T.B.); (A.R.); (L.v.d.H.); (F.O.A.)
| | - Sante Princiero Berlingerio
- Laboratory of Pediatric Nephrology, Department of Development & Regeneration, KU Leuven Campus Gasthuisberg, B-3000 Leuven, Belgium; (S.P.B.); (T.B.); (A.R.); (L.v.d.H.); (F.O.A.)
| | - Dries David
- Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven Campus Gasthuisberg, B-3000 Leuven, Belgium; (D.D.); (R.G.)
| | - Tjessa Bondue
- Laboratory of Pediatric Nephrology, Department of Development & Regeneration, KU Leuven Campus Gasthuisberg, B-3000 Leuven, Belgium; (S.P.B.); (T.B.); (A.R.); (L.v.d.H.); (F.O.A.)
| | - Katharina Held
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine (LEERM), Department of Development & Regeneration, KU Leuven Campus Gasthuisberg, B-3000 Leuven, Belgium; (K.H.); (J.V.)
| | - Ahmed Reda
- Laboratory of Pediatric Nephrology, Department of Development & Regeneration, KU Leuven Campus Gasthuisberg, B-3000 Leuven, Belgium; (S.P.B.); (T.B.); (A.R.); (L.v.d.H.); (F.O.A.)
| | - Martijn van den Broek
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6524 Nijmegen, The Netherlands;
- Department of Pediatrics, Division of Pediatric Nephrology, Amalia Children’s Hospital, Radboud University Medical Center, 6524 Nijmegen, The Netherlands;
| | - Koen Theunis
- Department of Human Genetics, KU Leuven Campus Gasthuisberg, B-3000 Leuven, Belgium;
| | - Mirian Janssen
- Department of Internal Medicine, Radboud University Medical Center, 6524 Nijmegen, The Netherlands;
| | - Elisabeth Cornelissen
- Department of Pediatrics, Division of Pediatric Nephrology, Amalia Children’s Hospital, Radboud University Medical Center, 6524 Nijmegen, The Netherlands;
| | - Joris Vriens
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine (LEERM), Department of Development & Regeneration, KU Leuven Campus Gasthuisberg, B-3000 Leuven, Belgium; (K.H.); (J.V.)
| | - Francesca Diomedi-Camassei
- Unit of Pathology, Department of Laboratories, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Rik Gijsbers
- Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven Campus Gasthuisberg, B-3000 Leuven, Belgium; (D.D.); (R.G.)
- Leuven Viral Vector Core, KU Leuven, B-3000 Leuven, Belgium
| | - Lambertus van den Heuvel
- Laboratory of Pediatric Nephrology, Department of Development & Regeneration, KU Leuven Campus Gasthuisberg, B-3000 Leuven, Belgium; (S.P.B.); (T.B.); (A.R.); (L.v.d.H.); (F.O.A.)
- Department of Pediatrics, Division of Pediatric Nephrology, Amalia Children’s Hospital, Radboud University Medical Center, 6524 Nijmegen, The Netherlands;
| | - Fanny O. Arcolino
- Laboratory of Pediatric Nephrology, Department of Development & Regeneration, KU Leuven Campus Gasthuisberg, B-3000 Leuven, Belgium; (S.P.B.); (T.B.); (A.R.); (L.v.d.H.); (F.O.A.)
| | - Elena Levtchenko
- Department of Pediatrics, University Hospitals Leuven Campus Gasthuisberg, B-3000 Leuven, Belgium;
- Laboratory of Pediatric Nephrology, Department of Development & Regeneration, KU Leuven Campus Gasthuisberg, B-3000 Leuven, Belgium; (S.P.B.); (T.B.); (A.R.); (L.v.d.H.); (F.O.A.)
- Correspondence: ; Tel.: +32-16-34-13-62
| |
Collapse
|
3
|
Bradley JR, Wang J, Bardsley V, Broecker V, Thiru S, Pober JS, Al-Lamki RS. Signaling through tumor necrosis receptor 2 induces stem cell marker in CD133 + regenerating tubular epithelial cells in acute cell-mediated rejection of human renal allografts. Am J Transplant 2020; 20:2380-2391. [PMID: 32167668 DOI: 10.1111/ajt.15846] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 01/25/2023]
Abstract
Tumor necrosis factor receptor 2 (TNFR2) is strongly upregulated on renal tubular epithelial cells by acute cell-mediated rejection (ACR. In human kidney organ culture, TNFR2 signaling both upregulates TNFR2 expression and promotes cell cycle entry of tubular epithelial cells. We find significantly more cells express CD133 mRNA and protein, a putative stem cell marker, in allograft biopsy samples with ACR compared to acute tubular injury without rejection or pretransplant "normal kidney" biopsy samples. Of CD133+ cells, ~85% are within injured tubules and ~15% are interstitial. Both populations express stem cell marker TRA-1-60 and TNFR2, but only tubular CD133+ cells express proximal tubular markers megalin and aquaporin-1. TNFR2+ CD133+ cells in tubules express proliferation marker phospho-histone H3S10 (pH3S10 ). Tubular epithelial cells in normal kidney organ cultures respond to TNFR2 signaling by expressing CD133 mRNA and protein, stem cell marker TRA-1-60, and pH3S10 within 3 hours of treatment. This rapid response time suggests that CD133+ cells in regenerating tubules of kidneys undergoing ACR represent proliferating tubular epithelial cells with TNFR2-induced stem cell markers rather than expansion of resident stem cells. Infiltrating host mononuclear cells are a likely source of TNF as these changes are absent in acute tubular injury .
Collapse
Affiliation(s)
- John R Bradley
- Department of Medicine, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Jun Wang
- Department of Medicine, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Victoria Bardsley
- Department of Histopathology, Addenbrookes Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Verena Broecker
- Department of Clinical Pathology, Sahlgrenska University Hospital Gothenburg, Gothenburg, Sweden
| | - Sathia Thiru
- Department of Histopathology, Addenbrookes Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Jordan S Pober
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
| | - Rafia S Al-Lamki
- Department of Medicine, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|
4
|
Andrianova NV, Buyan MI, Zorova LD, Pevzner IB, Popkov VA, Babenko VA, Silachev DN, Plotnikov EY, Zorov DB. Kidney Cells Regeneration: Dedifferentiation of Tubular Epithelium, Resident Stem Cells and Possible Niches for Renal Progenitors. Int J Mol Sci 2019; 20:ijms20246326. [PMID: 31847447 PMCID: PMC6941132 DOI: 10.3390/ijms20246326] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/10/2019] [Accepted: 12/12/2019] [Indexed: 12/11/2022] Open
Abstract
A kidney is an organ with relatively low basal cellular regenerative potential. However, renal cells have a pronounced ability to proliferate after injury, which undermines that the kidney cells are able to regenerate under induced conditions. The majority of studies explain yielded regeneration either by the dedifferentiation of the mature tubular epithelium or by the presence of a resident pool of progenitor cells in the kidney tissue. Whether cells responsible for the regeneration of the kidney initially have progenitor properties or if they obtain a “progenitor phenotype” during dedifferentiation after an injury, still stays the open question. The major stumbling block in resolving the issue is the lack of specific methods for distinguishing between dedifferentiated cells and resident progenitor cells. Transgenic animals, single-cell transcriptomics, and other recent approaches could be powerful tools to solve this problem. This review examines the main mechanisms of kidney regeneration: dedifferentiation of epithelial cells and activation of progenitor cells with special attention to potential niches of kidney progenitor cells. We attempted to give a detailed description of the most controversial topics in this field and ways to resolve these issues.
Collapse
Affiliation(s)
- Nadezda V. Andrianova
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119992 Moscow, Russia
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Marina I. Buyan
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Ljubava D. Zorova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Irina B. Pevzner
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Vasily A. Popkov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Valentina A. Babenko
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Denis N. Silachev
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Egor Y. Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
- Sechenov First Moscow State Medical University, Institute of Molecular Medicine, 119991 Moscow, Russia
- Correspondence: (E.Y.P.); (D.B.Z.); Tel.: +7-495-939-5944 (E.Y.P.)
| | - Dmitry B. Zorov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
- Correspondence: (E.Y.P.); (D.B.Z.); Tel.: +7-495-939-5944 (E.Y.P.)
| |
Collapse
|
5
|
Nargesi AA, Zhu XY, Conley SM, Woollard JR, Saadiq IM, Lerman LO, Eirin A. Renovascular disease induces mitochondrial damage in swine scattered tubular cells. Am J Physiol Renal Physiol 2019; 317:F1142-F1153. [PMID: 31461348 DOI: 10.1152/ajprenal.00276.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Scattered tubular-like cells (STCs) contribute to repair neighboring injured renal tubular cells. Mitochondria mediate STC biology and function but might be injured by the ambient milieu. We hypothesized that the microenviroment induced by the ischemic and metabolic components of renovascular disease impairs STC mitochondrial structure and function in swine, which can be attenuated with mitoprotection. CD24+/CD133+ STCs were quantified in pig kidneys after 16 wk of metabolic syndrome (MetS) or lean diet (Lean) with or without concurrent renal artery stenosis (RAS) (n = 6 each). Pig STCs were isolated and characterized, and mitochondrial structure, membrane potential, and oxidative stress were assessed in cells untreated or incubated with the mitoprotective drug elamipretide (1 nM for 6 h). STC-protective effects were assessed in vitro by their capacity to proliferate and improve viability of injured pig tubular epithelial cells. The percentage of STCs was higher in MetS, Lean + RAS, and MetS + RAS kidneys compared with Lean kidneys. STCs isolated from Lean + RAS and MetS + RAS pigs showed mitochondrial swelling and decreased matrix density, which were both restored by mitoprotection. In addition, mitochondrial membrane potential and ATP production were reduced and production of reactive oxygen species elevated in MetS, Lean + RAS, and MetS + RAS STCs. Importantly, mitoprotection improved mitochondrial structure and function as well as the capacity of MetS + RAS STCs to repair injured tubular cells in vitro. Renovascular disease in swine is associated with a higher prevalence of STCs but induces structural and functional alterations in STC mitochondria, which impair their reparative potency. These observations suggest a key role for mitochondria in the renal reparative capacity of STCs.
Collapse
Affiliation(s)
- Arash Aghajani Nargesi
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesora
| | - Xiang-Yang Zhu
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesora
| | - Sabena M Conley
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesora
| | - John R Woollard
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesora
| | - Ishran M Saadiq
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesora
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesora
| | - Alfonso Eirin
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesora
| |
Collapse
|
6
|
Burks SR, Nguyen BA, Tebebi PA, Kim SJ, Bresler MN, Ziadloo A, Street JM, Yuen PST, Star RA, Frank JA. Pulsed focused ultrasound pretreatment improves mesenchymal stromal cell efficacy in preventing and rescuing established acute kidney injury in mice. Stem Cells 2016; 33:1241-53. [PMID: 25640064 DOI: 10.1002/stem.1965] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 10/28/2014] [Accepted: 11/08/2014] [Indexed: 12/13/2022]
Abstract
Animal studies have shown that mesenchymal stromal cell (MSC) infusions improve acute kidney injury (AKI) outcomes when administered early after ischemic/reperfusion injury or within 24 hours after cisplatin administration. These findings have spurred several human clinical trials to prevent AKI. However, no specific therapy effectively treats clinically obvious AKI or rescues renal function once advanced injury is established. We investigated if noninvasive image-guided pulsed focused ultrasound (pFUS) could alter the kidney microenvironment to enhance homing of subsequently infused MSC. To examine the efficacy of pFUS-enhanced cell homing in disease, we targeted pFUS to kidneys to enhance MSC homing after cisplatin-induced AKI. We found that pFUS enhanced MSC homing at 1 day post-cisplatin, prior to renal functional deficits, and that enhanced homing improved outcomes of renal function, tubular cell death, and regeneration at 5 days post-cisplatin compared to MSC alone. We then investigated whether pFUS+MSC therapy could rescue established AKI. MSC alone at 3 days post-cisplatin, after renal functional deficits were obvious, significantly improved 7-day survival of animals. Survival was further improved by pFUS and MSC. pFUS prior to MSC injections increased IL-10 production by MSC that homed to kidneys and generated an anti-inflammatory immune cell profile in treated kidneys. This study shows pFUS is a neoadjuvant approach to improve MSC homing to diseased organs. pFUS with MSC better prevents AKI than MSC alone and allows rescue therapy in established AKI, which currently has no meaningful therapeutic options.
Collapse
Affiliation(s)
- Scott R Burks
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA; Imaging Sciences Training Program, Clinical Center and National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Bussolati B, Camussi G. Therapeutic use of human renal progenitor cells for kidney regeneration. Nat Rev Nephrol 2015; 11:695-706. [PMID: 26241019 DOI: 10.1038/nrneph.2015.126] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The ability of the human kidney to repair itself is limited. Consequently, repeated injury can trigger a maladaptive response that is characterized by fibrosis and loss of renal function. The transcription patterns that characterize nephrogenesis in fetal renal progenitor cells (RPCs) are only partially activated during renal repair in adults. Nevertheless, evidence suggests that segment-restricted progenitor resident cells support renal healing in adults. In this Review, we discuss the evidence for the existence of functional human RPCs in adults and their role in renal repair, and consider the controversial issue of whether RPCs are a fixed population or arise through phenotypical plasticity of tubular cells that is mediated by the microenvironment. We also discuss the strategies for generating renal progenitor cells from pluripotent stem cells or differentiated cells and their use in therapy. Finally, we examine preclinical data on the therapeutic use of human fetal cells, adult progenitor cells and adult renal cells.
Collapse
Affiliation(s)
- Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, Torino 10126, Italy
| | - Giovanni Camussi
- Department of Medical Sciences, University of Torino, Via Nizza 52, Torino 10126, Italy
| |
Collapse
|
8
|
Becherucci F, Lazzeri E, Romagnani P. A Road to Chronic Kidney Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:2072-5. [DOI: 10.1016/j.ajpath.2015.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 05/11/2015] [Accepted: 05/13/2015] [Indexed: 02/09/2023]
|
9
|
Podocyte Regeneration Driven by Renal Progenitors Determines Glomerular Disease Remission and Can Be Pharmacologically Enhanced. Stem Cell Reports 2015; 5:248-63. [PMID: 26235895 PMCID: PMC4618832 DOI: 10.1016/j.stemcr.2015.07.003] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 07/06/2015] [Accepted: 07/06/2015] [Indexed: 12/25/2022] Open
Abstract
Podocyte loss is a general mechanism of glomerular dysfunction that initiates and drives the progression of chronic kidney disease, which affects 10% of the world population. Here, we evaluate whether the regenerative response to podocyte injury influences chronic kidney disease outcome. In models of focal segmental glomerulosclerosis performed in inducible transgenic mice where podocytes are tagged, remission or progression of disease was determined by the amount of regenerated podocytes. When the same model was established in inducible transgenic mice where renal progenitors are tagged, the disease remitted if renal progenitors successfully differentiated into podocytes, while it persisted if differentiation was ineffective, resulting in glomerulosclerosis. Treatment with BIO, a GSK3s inhibitor, significantly increased disease remission by enhancing renal progenitor sensitivity to the differentiation effect of endogenous retinoic acid. These results establish renal progenitors as critical determinants of glomerular disease outcome and a pharmacological enhancement of their differentiation as a possible therapeutic strategy.
Collapse
|
10
|
Lombardi D, Becherucci F, Romagnani P. How much can the tubule regenerate and who does it? An open question. Nephrol Dial Transplant 2015; 31:1243-50. [PMID: 26175143 PMCID: PMC4967725 DOI: 10.1093/ndt/gfv262] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 05/22/2015] [Indexed: 01/09/2023] Open
Abstract
The tubular compartment of the kidney is the primary site of a wide range of insults that can result in acute kidney injury (AKI), a condition associated with high mortality and an increased risk to develop end-stage renal disease. Nevertheless, kidney function is often quickly recovered after tubular injury. How this happens has only partially been unveiled. Indeed, although it has clearly been demonstrated that regenerated epithelial cells arise from survived intratubular cells, the true entity, as well as the cellular source of this regenerative process, remains mostly unknown. Is whichever proximal tubular epithelial cell able to dedifferentiate and divide to replace neighboring lost tubular cells, thus suggesting an extreme regenerative ability of residual tubular epithelium, or is the regenerative potential of tubular epithelium limited, and mostly related to a preexisting population of intratubular scattered progenitor cells which are more resistant to death? Gaining insights on how this process takes place is essential for developing new therapeutic strategies to prevent AKI, as well as AKI-related chronic kidney disease. The aim of this review is to discuss why the answers to these questions are still open, and how further investigations are needed to understand which is the true regenerative potential of the tubule and who are the players that allow functional recovery after AKI.
Collapse
Affiliation(s)
- Duccio Lombardi
- Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies (DENOTHE), University of Florence, Florence, Italy
| | - Francesca Becherucci
- Nephrology and Dialysis Unit, Meyer Children's University Hospital, Florence, Italy
| | - Paola Romagnani
- Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies (DENOTHE), University of Florence, Florence, Italy Nephrology and Dialysis Unit, Meyer Children's University Hospital, Florence, Italy Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy
| |
Collapse
|
11
|
Reyes EE, Gillard M, Duggan R, Wroblewski K, Kregel S, Isikbay M, Kach J, Brechka H, Weele DJV, Szmulewitz RZ, Griend DJV. Molecular analysis of CD133-positive circulating tumor cells from patients with metastatic castration-resistant prostate cancer. JOURNAL OF TRANSLATIONAL SCIENCE 2015; 1:http://oatext.com/Molecular-analysis-of-CD133-positive-circulating-tumor-cells-from-patients-with-metastatic-castration-resistant-prostate-cancer.php. [PMID: 26753099 PMCID: PMC4704802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The function and clinical utility of stem cell markers in metastatic castration-resistant prostate cancer (mCRPC) remains unresolved, and their expression may confer important therapeutic opportunities for staging and therapy. In the adult human prostate, CD133 (PROM1) expression identifies infrequent prostate epithelial progenitor cells and putative cancer stem cells. Previous work demonstrated an association with CD133 and cancer cell proliferation using in vitro model systems. The primary objective here was to investigate the expression of CD133 in circulating tumor cells (CTCs) from patients with mCRPC and to test the hypothesis that patients with mCRPC had CD133-positive CTCs associated with increased cell proliferation, changes in the androgen receptor (AR) protein expression, or AR nuclear co-localization. We utilized ImageStreamX technology, which combines flow cytometry and fluorescence microscopy, to capture and analyze CD45-negative/EpCAM-positive CTCs for CD133, Ki-67, and AR. All patient samples (20/20) contained CD133-positive populations of CTCs, and on average 50.9 ± 28.2% (range of 18.2% to 100%) of CTCs were CD133-positive. CD133-positive CTCs have increased Ki-67 protein expression compared to CD133-negative CTCs, implying that CD133-positive CTCs may have greater proliferative potential when compared to their CD133-negative counterparts. CD133-positive and CD133-negative CTCs have similar levels of AR protein expression and cellular co-localization with nuclear markers, implying that CD133 expression is independent of AR pathway activity and an AR-independent marker of mCRPC proliferation. These studies demonstrate the presence of CD133-positive populations in CTCs from mCRPC with increased proliferative potential.
Collapse
Affiliation(s)
- Edwin E Reyes
- Committee on Immunology, The University of Chicago, Chicago, IL, USA
| | - Marc Gillard
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL, USA
| | - Ryan Duggan
- Flow Cytometry Facility, The University of Chicago, Chicago, IL, USA
| | - Kristen Wroblewski
- Department of Public Health Sciences, The University of Chicago, Chicago, IL, USA
| | - Steven Kregel
- Committee on Cancer Biology, The University of Chicago, Chicago, IL, USA
| | - Masis Isikbay
- Department of Surgery, Section of Urology, The University of Chicago, Chicago, IL, USA
| | - Jacob Kach
- Department of Surgery, Section of Urology, The University of Chicago, Chicago, IL, USA
| | - Hannah Brechka
- Committee on Cancer Biology, The University of Chicago, Chicago, IL, USA
| | - David J Vander Weele
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL, USA
| | - Russell Z Szmulewitz
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL, USA
| | - Donald J Vander Griend
- Committee on Cancer Biology, The University of Chicago, Chicago, IL, USA
- Department of Surgery, Section of Urology, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
12
|
Wang HL, Liu NM, Li R. Role of adult resident renal progenitor cells in tubular repair after acute kidney injury. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2015; 12:469-75. [PMID: 25412664 DOI: 10.1016/s2095-4964(14)60053-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Acute kidney injury is a serious global health problem and determinant of morbidity and mortality. Recent advancements in the field of stem cell research raise hopes for stem cell-based regenerative approaches to treat acute kidney diseases. In this review, the authors summarized the latest research advances of the adult resident renal progenitor cells (ARPCs) on kidney repair, the role of ARPCs on tubular regeneration after acute kidney injury, the current understanding of the mechanisms related to ARPC activation and modulation, as well as the challenges that remain to be faced.
Collapse
Affiliation(s)
- Hui-ling Wang
- Department of Nephrology, the 455th Hospital; Institute of Nephrology of Nanjing Military Command, Shanghai 200052, China; E-mail:
| | - Nan-mei Liu
- Department of Nephrology, the 455th Hospital; Institute of Nephrology of Nanjing Military Command, Shanghai 200052, China
| | - Rui Li
- Department of Nephrology, the 455th Hospital; Institute of Nephrology of Nanjing Military Command, Shanghai 200052, China
| |
Collapse
|
13
|
Dimuccio V, Ranghino A, Praticò Barbato L, Fop F, Biancone L, Camussi G, Bussolati B. Urinary CD133+ extracellular vesicles are decreased in kidney transplanted patients with slow graft function and vascular damage. PLoS One 2014; 9:e104490. [PMID: 25100147 PMCID: PMC4123993 DOI: 10.1371/journal.pone.0104490] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 07/10/2014] [Indexed: 01/17/2023] Open
Abstract
Extracellular vesicles (EVs) present in the urine are mainly released from cells of the nephron and can therefore provide information on kidney function. We here evaluated the presence of vesicles expressing the progenitor marker CD133 in the urine of normal subjects and of patients undergoing renal transplant. We found that EV expressing CD133 were present in the urine of normal subjects, but not of patients with end stage renal disease. The first day after transplant, urinary CD133+ EVs were present at low levels, to increase thereafter (at day 7). Urinary CD133+ EVs significantly increased in patients with slow graft function in respect to those with early graft function. In patients with a severe pre-transplant vascular damage of the graft, CD133+ EVs did not increase at day 7. At variance, the levels of EVs expressing the renal exosomal marker CD24 did not vary in the urine of patients with end stage renal disease or in transplanted patients in respect to controls. Sorted CD133+ EVs were found to express glomerular and proximal tubular markers. These data indicate that urinary CD133+ EVs are continuously released during the homeostatic turnover of the nephron and may provide information on its function or regenerative potential.
Collapse
Affiliation(s)
- Veronica Dimuccio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Andrea Ranghino
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Loredana Praticò Barbato
- Immunogenetics and Transplant Biology Service, Città della Salute e della Scienza Hospital, Torino, Italy
| | - Fabrizio Fop
- Division of Nephrology, Dialysis and Transplantation, Città della Salute e della Scienza Hospital, Torino, Italy
| | - Luigi Biancone
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Giovanni Camussi
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- * E-mail:
| |
Collapse
|
14
|
Qin D, Long T, Deng J, Zhang Y. Urine-derived stem cells for potential use in bladder repair. Stem Cell Res Ther 2014; 5:69. [PMID: 25157812 PMCID: PMC4055102 DOI: 10.1186/scrt458] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Engineered bladder tissues, created with autologous bladder cells seeded on biodegradable scaffolds, are being developed for use in patients who need cystoplasty. However, in individuals with organ damage from congenital disorders, infection, irradiation, or cancer, abnormal cells obtained by biopsy from the compromised tissue could potentially contaminate the engineered tissue. Thus, an alternative cell source for construction of the neo-organ would be useful. Although other types of stem cells have been investigated, autologous mesenchymal stem cells (MSCs) are most suitable to use in bladder regeneration. These cells are often used as a cell source for bladder repair in three ways - secreting paracrine factors, recruiting resident cells, and trans-differentiation, inducing MSCs to differentiate into bladder smooth muscle cells and urothelial cells. Adult stem cell populations have been demonstrated in bone marrow, fat, muscle, hair follicles, and amniotic fluid. These cells remain an area of intense study, as their potential for therapy may be applicable to bladder disorders. Recently, we have found stem cells in the urine and the cells are highly expandable, and have self-renewal capacity and paracrine properties. As a novel cell source, urine-derived stem cells (USCs) provide advantages for cell therapy and tissue engineering applications in bladder tissue repair because they originate from the urinary tract system. Importantly, USCs can be obtained via a noninvasive, simple, and low-cost approach and induced with high efficiency to differentiate into bladder cells.
Collapse
|
15
|
Chen D, Chen Z, Zhang Y, Park C, Al-Omari A, Moeckel GW. Role of medullary progenitor cells in epithelial cell migration and proliferation. Am J Physiol Renal Physiol 2014; 307:F64-74. [PMID: 24808539 DOI: 10.1152/ajprenal.00547.2013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
This study is aimed at characterizing medullary interstitial progenitor cells and to examine their capacity to induce tubular epithelial cell migration and proliferation. We have isolated a progenitor cell side population from a primary medullary interstitial cell line. We show that the medullary progenitor cells (MPCs) express CD24, CD44, CXCR7, CXCR4, nestin, and PAX7. MPCs are CD34 negative, which indicates that they are not bone marrow-derived stem cells. MPCs survive >50 passages, and when grown in epithelial differentiation medium develop phenotypic characteristics of epithelial cells. Inner medulla collecting duct (IMCD3) cells treated with conditioned medium from MPCs show significantly accelerated cell proliferation and migration. Conditioned medium from PGE2-treated MPCs induce tubule formation in IMCD3 cells grown in 3D Matrigel. Moreover, most of the MPCs express the pericyte marker PDGFR-b. Our study shows that the medullary interstitium harbors a side population of progenitor cells that can differentiate to epithelial cells and can stimulate tubular epithelial cell migration and proliferation. The findings of this study suggest that medullary pericyte/progenitor cells may play a critical role in collecting duct cell injury repair.
Collapse
Affiliation(s)
- Dong Chen
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut; and
| | - Zhiyong Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
| | - Yuning Zhang
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut; and
| | - Chanyoung Park
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut; and
| | - Ahmed Al-Omari
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut; and
| | - Gilbert W Moeckel
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut; and
| |
Collapse
|
16
|
Evidence for a morphologically distinct and functionally robust cell type in the proximal tubules of human kidney. Hum Pathol 2014; 45:382-93. [DOI: 10.1016/j.humpath.2013.10.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 09/23/2013] [Accepted: 10/02/2013] [Indexed: 12/13/2022]
|
17
|
Epithelial–mesenchymal transition of renal tubules: Divergent processes of repairing in acute or chronic injury? Med Hypotheses 2013; 81:73-5. [DOI: 10.1016/j.mehy.2013.03.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 03/09/2013] [Indexed: 11/18/2022]
|
18
|
Romagnani P. Of mice and men: the riddle of tubular regeneration. J Pathol 2013; 229:641-4. [PMID: 23299489 DOI: 10.1002/path.4162] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2012] [Revised: 12/22/2012] [Accepted: 12/24/2012] [Indexed: 12/29/2022]
Abstract
Regeneration can occur through multiple distinct mechanisms, such as pluripotent stem cells, lineage-committed progenitors or dedifferentiation. The respective contribution of each of these regenerative strategies in every organ or tissue may be different. Recent results indicate that dedifferentiation contributes less than previously thought, and that stem or progenitor cells seem to be the main drivers of regenerative processes. Our views of regeneration in the kidney are undergoing the same process of revision. Indeed, studies in humans have established the existence of a scattered population of tubular progenitors in the adult kidney. Renal progenitors have been discovered also in other animal classes such as fish and insects. In contrast, in rodents a tubular progenitor phenotype seems to be induced only after tubular injury, suggesting some differences may exist. Is this difference really related to a distinct regenerative strategy or is it simply a matter of the type and modality of cellular markers expressed? It may also be possible that progenitor cells, as well as tubular cell dedifferentiation, act in concert to allow regeneration of a complex organ like the adult mammalian kidney, as recently proposed also for the liver. Further studies are needed to resolve the riddle of tubular regeneration. However, beyond the controversial results obtained from humans and rodents, identification of tubular progenitors in humans can move the field forward and provide a novel perspective for understanding tubular regeneration.
Collapse
Affiliation(s)
- Paola Romagnani
- Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies (DENOTHE), University of Florence, Florence, Italy.
| |
Collapse
|
19
|
Romagnani P, Lasagni L, Remuzzi G. Renal progenitors: an evolutionary conserved strategy for kidney regeneration. Nat Rev Nephrol 2013; 9:137-46. [PMID: 23338209 DOI: 10.1038/nrneph.2012.290] [Citation(s) in RCA: 147] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Following kidney injury, repair can result in functional tissue becoming a patch of cells and disorganized extracellular matrix--a scar--or it can recapitulate the original tissue architecture through the process of regeneration. Regeneration can potentially occur in all animal species and humans. Indeed, the repair of portions of the existing nephron after tubular damage, a response that has been designated classically as cellular regeneration, is conserved in all animal species from the ancestral phases of evolution. By contrast, another type of regenerative response--nephron neogenesis--has been described in lower branches of the animal kingdom, but does not occur in adult mammals. Converging evidence suggests that a renal progenitor system is present in the adult kidney across different stages of evolution, with renal progenitors having been identified as the main drivers of kidney regenerative responses in fish, insects, rodents and humans. In this Review, we describe similarities and differences between the renal progenitor systems through evolution, and propose explanations for how progressive kidney adaptation to environmental changes both required and permitted neonephrogenesis to be given up and for cellular regeneration to be retained as the main regenerative strategy. Understanding the mechanisms that drive renal progenitor growth and differentiation represent the key step for modulating this potential for therapeutic purposes.
Collapse
Affiliation(s)
- Paola Romagnani
- Pediatric Nephrology Unit, Meyer Children's Hospital, University of Florence, Viale Pieraccini 24, 50139 Florence, Italy.
| | | | | |
Collapse
|
20
|
Mozaffari MS, Abdelsayed R, Liu JY, Zakhary I, Baban B. Renal distal tubule proliferation and increased aquaporin 2 level but decreased urine osmolality in db/db mouse: treatment with chromium picolinate. Exp Mol Pathol 2011; 92:54-8. [PMID: 21983138 DOI: 10.1016/j.yexmp.2011.09.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 09/20/2011] [Indexed: 10/17/2022]
Abstract
Hallmark features of type 2 diabetes mellitus include glucosuria and polyuria. Further, renal aquaporin 2 is pivotal to regulation of fluid excretion and urine osmolality. Accordingly, we tested the hypothesis that the db/db mouse displays increased glucosuria and fluid excretion but reduced urine osmolality in association with decreased renal aquaporin 2 level. In addition, we examined the effect of chromium picolinate (Cr(pic)3) which is purported to improve glycemic control. The db/db mice excreted more urine in association with marked glucose excretion but lower urine osmolality than db/m control group. Light microscopic examination of renal tissue revealed proliferation of tubular structures in db/db compared to the db/m mice, a feature validated with Ki67 immunostaining. Further, these tubules showed generally similar immunostaining intensity and pattern for aquaporin 2 indicating that proliferated tubules are of distal origin. On the other hand, renal aquaporin 2 protein level was significantly higher in the db/db than db/m group. Treatment of db/db mice with Cr(pic)3 reduced plasma glucose and hemoglobin A1c (~15-17%, p<0.05) and Ki67 positive cells but other parameters were similar to their untreated counterparts. Collectively, these findings suggest that proliferation of renal distal tubules and increased aquaporin 2 level likely represent an adaptive mechanism to regulate fluid excretion to prevent dehydration in the setting of marked glucosuria in the db/db mouse, features not affected by Cr(pic)3 treatment. These observations are of relevance to increasing interest in developing therapeutic agents that facilitate renal glucose elimination.
Collapse
Affiliation(s)
- Mahmood S Mozaffari
- Department of Oral Biology, College of Dental Medicine, Georgia Health Sciences University, Augusta, GA 30912, USA.
| | | | | | | | | |
Collapse
|
21
|
Otto WR, Wright NA. Mesenchymal stem cells: from experiment to clinic. FIBROGENESIS & TISSUE REPAIR 2011; 4:20. [PMID: 21902837 PMCID: PMC3182886 DOI: 10.1186/1755-1536-4-20] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 09/08/2011] [Indexed: 02/07/2023]
Abstract
There is currently much interest in adult mesenchymal stem cells (MSCs) and their ability to differentiate into other cell types, and to partake in the anatomy and physiology of remote organs. It is now clear these cells may be purified from several organs in the body besides bone marrow. MSCs take part in wound healing by contributing to myofibroblast and possibly fibroblast populations, and may be involved in epithelial tissue regeneration in certain organs, although this remains more controversial. In this review, we examine the ability of MSCs to modulate liver, kidney, heart and intestinal repair, and we update their opposing qualities of being less immunogenic and therefore tolerated in a transplant situation, yet being able to contribute to xenograft models of human tumour formation in other contexts. However, such observations have not been replicated in the clinic. Recent studies showing the clinical safety of MSC in several pathologies are discussed. The possible opposing powers of MSC need careful understanding and control if their clinical potential is to be realised with long-term safety for patients.
Collapse
Affiliation(s)
- William R Otto
- Histopathology Laboratory, Cancer Research UK, London Research Institute, 44, Lincoln's Inn Fields, London WC2A 3LY, UK.
| | | |
Collapse
|