Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Ann Surg ; 278(6): e1313-e1326, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37450698

RESUMEN

OBJECTIVES: To test whether mitochondrial transplantation (MITO) mitigates damage in 2 models of acute kidney injury (AKI). BACKGROUND: MITO is a process where exogenous isolated mitochondria are taken up by cells. As virtually any morbid clinical condition is characterized by mitochondrial distress, MITO may find a role as a treatment modality in numerous clinical scenarios including AKI. METHODS: For the in vitro experiments, human proximal tubular cells were damaged and then treated with mitochondria or placebo. For the ex vivo experiments, we developed a non-survival ex vivo porcine model mimicking the donation after cardiac death renal transplantation scenario. One kidney was treated with mitochondria, although the mate organ received placebo, before being perfused at room temperature for 24 hours. Perfusate samples were collected at different time points and analyzed with Raman spectroscopy. Biopsies taken at baseline and 24 hours were analyzed with standard pathology, immunohistochemistry, and RNA sequencing analysis. RESULTS: In vitro, cells treated with MITO showed higher proliferative capacity and adenosine 5'-triphosphate production, preservation of physiological polarization of the organelles and lower toxicity and reactive oxygen species production. Ex vivo, kidneys treated with MITO shed fewer molecular species, indicating stability. In these kidneys, pathology showed less damage whereas RNAseq analysis showed modulation of genes and pathways most consistent with mitochondrial biogenesis and energy metabolism and downregulation of genes involved in neutrophil recruitment, including IL1A, CXCL8, and PIK3R1. CONCLUSIONS: MITO mitigates AKI both in vitro and ex vivo.


Asunto(s)
Lesión Renal Aguda , Trasplante de Riñón , Daño por Reperfusión , Humanos , Porcinos , Animales , Riñón/metabolismo , Mitocondrias/metabolismo , Mitocondrias/patología , Lesión Renal Aguda/prevención & control , Lesión Renal Aguda/metabolismo
2.
Adv Sci (Weinh) ; 10(20): e2206787, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37114795

RESUMEN

A nephrogenic progenitor cell (NP) with cancer stem cell characteristics driving Wilms tumor (WT) using spatial transcriptomics, bulk and single cell RNA sequencing, and complementary in vitro and transplantation experiments is identified and characterized. NP from WT samples with NP from the developing human kidney is compared. Cells expressing SIX2 and CITED1 fulfill cancer stem cell criteria by reliably recapitulating WT in transplantation studies. It is shown that self-renewal versus differentiation in SIX2+CITED1+ cells is regulated by the interplay between integrins ITGß1 and ITGß4. The spatial transcriptomic analysis defines gene expression maps of SIX2+CITED1+ cells in WT samples and identifies the interactive gene networks involved in WT development. These studies define SIX2+CITED1+ cells as the nephrogenic-like cancer stem cells of WT and points to the renal developmental transcriptome changes as a possible driver in regulating WT formation and progression.


Asunto(s)
Neoplasias Renales , Tumor de Wilms , Humanos , Factores de Transcripción/genética , Tumor de Wilms/genética , Tumor de Wilms/metabolismo , Tumor de Wilms/patología , Riñón , Células Madre Neoplásicas/metabolismo , Neoplasias Renales/genética
3.
Transplant Direct ; 5(9): e481, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31579809

RESUMEN

Development of anti-human leukocyte antigen donor-specific antibodies (DSAs) is associated with antibody-mediated rejection (AMR) and reduced allograft survival in kidney transplant recipients. Whether changes in circulating lymphocytes anticipate DSA or AMR development is unclear. METHODS: We used time-of-flight mass cytometry to analyze prospectively collected peripheral blood mononuclear cells (PBMC) from pediatric kidney transplant recipients who developed DSA (DSA-positive recipients [DSAPOS], n = 10). PBMC were obtained at 2 months posttransplant, 3 months before DSA development, and at DSA detection. PBMC collected at the same time points posttransplant from recipients who did not develop DSA (DSA-negative recipients [DSANEG], n = 11) were used as controls. RESULTS: DSAPOS and DSANEG recipients had similar baseline characteristics and comparable frequencies of total B and T cells. Within DSAPOS recipients, there was no difference in DSA levels (mean fluorescence intensity [MFI]: 13 687 ± 4159 vs 11 375 ± 1894 in DSAPOSAMR-positive recipients (AMRPOS) vs DSAPOSAMR-negative recipients (AMRNEG), respectively; P = 0.630), C1q binding (5 DSAPOSAMRPOS [100%] vs 4 DSAPOSAMRNEG [80%]; P = 1.000), or C3d binding (3 DSAPOSAMRPOS [60%] vs 1 DSAPOSAMRNEG [20%]; P = 0.520) between patients who developed AMR and those who did not. However, DSAPOS patients who developed AMR (n = 5; 18.0 ± 3.6 mo post-DSA detection) had increased B cells with antibody-secreting (IgD-CD27+CD38+; P = 0.002) and memory (IgD-CD27+CD38-; P = 0.003) phenotypes compared with DSANEG and DSAPOSAMRNEG recipients at DSA detection. CONCLUSIONS: Despite the small sample size, our comprehensive phenotypic analyses show that circulating B cells with memory and antibody-secreting phenotypes are present at DSA onset, >1 year before biopsy-proven AMR in pediatric kidney transplant recipients.

4.
Pediatr Nephrol ; 33(6): 935-945, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-28620747

RESUMEN

Amniotic fluid (AF) contains a heterogeneous population of cells that have been identified to possess pluripotent and progenitor-like characteristics. These cells have been applied in various regenerative medicine applications ranging from in vitro cell differentiation to tissue engineering to cellular therapies for different organs including the heart, the liver, the lung, and the kidneys. In this review, we examine the different methodologies used for the derivation of amniotic fluid stem cells and renal progenitors, and their application in renal repair and regeneration. Moreover, we discuss the recent achievements and newly emerging challenges in our understanding of their biology, their immunoregulatory characteristics, and their paracrine-mediated therapeutic potential for the treatment of acute and chronic kidney diseases.


Asunto(s)
Líquido Amniótico/citología , Enfermedades Renales/terapia , Trasplante de Células Madre/métodos , Animales , Humanos , Riñón/fisiopatología , Medicina Regenerativa/métodos
5.
Matrix Biol ; 57-58: 334-346, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27575985

RESUMEN

The outcome of tissue engineered organ transplants depends on the capacity of the biomaterial to promote a pro-healing response once implanted in vivo. Multiple studies, including ours, have demonstrated the possibility of using the extracellular matrix (ECM) of animal organs as platform for tissue engineering and more recently, discarded human organs have also been proposed as scaffold source. In contrast to artificial biomaterials, natural ECM has the advantage of undergoing continuous remodeling which allows adaptation to diverse conditions. It is known that natural matrices present diverse immune properties when compared to artificial biomaterials. However, how these properties compare between diseased and healthy ECM and artificial scaffolds has not yet been defined. To answer this question, we used decellularized renal ECM derived from WT mice and from mice affected by Alport Syndrome at different time-points of disease progression as a model of renal failure with extensive fibrosis. We characterized the morphology and composition of these ECMs and compared their in vitro effects on macrophage activation with that of synthetic scaffolds commonly used in the clinic (collagen type I and poly-L-(lactic) acid, PLLA). We showed that ECM derived from Alport kidneys differed in fibrous protein deposition and cytokine content when compared to ECM derived from WT kidneys. Yet, both WT and Alport renal ECM induced macrophage differentiation mainly towards a reparative (M2) phenotype, while artificial biomaterials towards an inflammatory (M1) phenotype. Anti-inflammatory properties of natural ECMs were lost when homogenized, hence three-dimensional structure of ECM seems crucial for generating an anti-inflammatory response. Together, these data support the notion that natural ECM, even if derived from diseased kidneys promote a M2 protolerogenic macrophage polarization, thus providing novel insights on the applicability of ECM obtained from discarded organs as ideal scaffold for tissue engineering.


Asunto(s)
Matriz Extracelular/química , Riñón/química , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Nefritis Hereditaria/inmunología , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacología , Colágeno Tipo I/química , Colágeno Tipo I/farmacología , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Matriz Extracelular/inmunología , Matriz Extracelular/ultraestructura , Humanos , Inmunohistoquímica , Inmunofenotipificación , Riñón/inmunología , Macrófagos/clasificación , Macrófagos/citología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Nefritis Hereditaria/metabolismo , Nefritis Hereditaria/patología , Fenotipo , Poliésteres/química , Poliésteres/farmacología , Cultivo Primario de Células , Ingeniería de Tejidos/métodos , Andamios del Tejido
6.
Transplantation ; 99(9): 1807-16, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26018349

RESUMEN

BACKGROUND: Extracellular matrix (ECM) scaffolds, obtained through detergent-based decellularization of native kidneys, represent the most promising platform for investigations aiming at manufacturing kidneys for transplant purposes. We previously showed that decellularization of the human kidney yields renal ECM scaffolds (hrECMs) that maintain their basic molecular components, are cytocompatible, stimulate angiogenesis, and show an intact innate vasculature. However, evidence that the decellularization preserves glomerular morphometric characteristics, physiological parameters (pressures and resistances of the vasculature bed), and biological properties of the renal ECM, including retention of important growth factors (GFs), is still missing. METHODS: To address these issues, we studied the morphometry and resilience of hrECMs' native vasculature with resin casting at electronic microscopy and pulse-wave measurements, respectively. Moreover, we determined the fate of 40 critical GFs post decellularization with a glass chip-based multiplex enzyme-linked immunosorbent assay array and in vitro immunofluorescence. RESULTS: Our method preserves the 3-dimensional conformation of the native glomerulus. Resin casting and pulse-wave measurements, showed that hrECMs preserves the microvascular morphology and morphometry, and physiological function. Moreover, GFs including vascular endothelial growth factor and its receptors are retained within the matrices. CONCLUSIONS: Our results indicate that discarded human kidneys are a suitable source of renal scaffolds because they maintain a well-preserved structure and function of the vasculature, as well as GFs that are fundamental to achieve a satisfying recellularization of the scaffold in vivo due to their angiogenic properties.


Asunto(s)
Matriz Extracelular , Hemodinámica , Péptidos y Proteínas de Señalización Intercelular/análisis , Glomérulos Renales , Microvasos , Andamios del Tejido , Molde por Corrosión , Matriz Extracelular/química , Matriz Extracelular/ultraestructura , Humanos , Glomérulos Renales/irrigación sanguínea , Glomérulos Renales/química , Glomérulos Renales/citología , Glomérulos Renales/ultraestructura , Microscopía Electrónica de Rastreo , Microvasos/química , Microvasos/fisiología , Microvasos/ultraestructura , Perfusión , Análisis por Matrices de Proteínas , Análisis de la Onda del Pulso , Receptores de Factores de Crecimiento Endotelial Vascular/análisis , Factor A de Crecimiento Endotelial Vascular/análisis
7.
Cell Stem Cell ; 14(6): 810-23, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-24905167

RESUMEN

Immune system defects are at the center of aging and a range of diseases. Here, we show that prolonged fasting reduces circulating IGF-1 levels and PKA activity in various cell populations, leading to signal transduction changes in long-term hematopoietic stem cells (LT-HSCs) and niche cells that promote stress resistance, self-renewal, and lineage-balanced regeneration. Multiple cycles of fasting abated the immunosuppression and mortality caused by chemotherapy and reversed age-dependent myeloid-bias in mice, in agreement with preliminary data on the protection of lymphocytes from chemotoxicity in fasting patients. The proregenerative effects of fasting on stem cells were recapitulated by deficiencies in either IGF-1 or PKA and blunted by exogenous IGF-1. These findings link the reduced levels of IGF-1 caused by fasting to PKA signaling and establish their crucial role in regulating hematopoietic stem cell protection, self-renewal, and regeneration.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ayuno/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Terapia de Inmunosupresión , Factor I del Crecimiento Similar a la Insulina/metabolismo , Regeneración , Animales , Células Madre Hematopoyéticas/enzimología , Ratones , Ratones Endogámicos C57BL
8.
Cytotherapy ; 16(1): 41-55, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24210784

RESUMEN

BACKGROUND AIMS: The contribution of amniotic fluid stem cells (AFSC) to tissue protection and regeneration in models of acute and chronic kidney injuries and lung failure has been shown in recent years. In the present study, we used a chemically induced mouse model of type 1 diabetes to determine whether AFSC could play a role in modulating ß-cell injury and restoring ß-cell function. METHODS: Streptozotocin-induced diabetic mice were given intracardial injection of AFSC; morphological and physiological parameters and gene expression profile for the insulin pathway were evaluated after cell transplantation. RESULTS: AFSC injection resulted in protection from ß-cell damage and increased ß-cell regeneration in a subset of mice as indicated by glucose and insulin levels, increased islet mass and preservation of islet structure. Moreover, ß-cell preservation/regeneration correlated with activation of the insulin receptor/Pi3K/Akt signaling pathway and vascular endothelial growth factor-A expression involved in maintaining ß-cell mass and function. CONCLUSIONS: Our results suggest a therapeutic role for AFSC in preserving and promoting endogenous ß-cell functionality and proliferation. The protective role of AFSC is evident when stem cell transplantation is performed before severe hyperglycemia occurs, which suggests the importance of early intervention. The present study demonstrates the possible benefits of the application of a non-genetically engineered stem cell population derived from amniotic fluid for the treatment of type 1 diabetes mellitus and gives new insight on the mechanism by which the beneficial effect is achieved.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Líquido Amniótico/química , Diabetes Mellitus Experimental/tratamiento farmacológico , Células Madre/química , Lesión Renal Aguda/complicaciones , Lesión Renal Aguda/patología , Líquido Amniótico/citología , Animales , Tratamiento Basado en Trasplante de Células y Tejidos , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/patología , Modelos Animales de Enfermedad , Humanos , Inyecciones , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Pulmón/patología , Ratones , Regeneración , Trasplante de Células Madre , Células Madre/citología
9.
PLoS One ; 8(12): e81812, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24349133

RESUMEN

Amniotic fluid is in continuity with multiple developing organ systems, including the kidney. Committed, but still stem-like cells from these organs may thus appear in amniotic fluid. We report having established for the first time a stem-like cell population derived from human amniotic fluid and possessing characteristics of podocyte precursors. Using a method of triple positive selection we obtained a population of cells (hAKPC-P) that can be propagated in vitro for many passages without immortalization or genetic manipulation. Under specific culture conditions, these cells can be differentiated to mature podocytes. In this work we compared these cells with conditionally immortalized podocytes, the current gold standard for in vitro studies. After in vitro differentiation, both cell lines have similar expression of the major podocyte proteins, such as nephrin and type IV collagen, that are characteristic of mature functional podocytes. In addition, differentiated hAKPC-P respond to angiotensin II and the podocyte toxin, puromycin aminonucleoside, in a way typical of podocytes. In contrast to immortalized cells, hAKPC-P have a more nearly normal cell cycle regulation and a pronounced developmental pattern of specific protein expression, suggesting their suitability for studies of podocyte development for the first time in vitro. These novel progenitor cells appear to have several distinct advantages for studies of podocyte cell biology and potentially for translational therapies.


Asunto(s)
Líquido Amniótico/citología , Ciclo Celular/genética , Podocitos/citología , Líquido Amniótico/metabolismo , Angiotensina II/farmacología , Antimetabolitos Antineoplásicos/farmacología , Biomarcadores/metabolismo , Ciclo Celular/efectos de los fármacos , Diferenciación Celular , Proliferación Celular , Separación Celular , Células Cultivadas , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Podocitos/efectos de los fármacos , Podocitos/metabolismo , Puromicina Aminonucleósido/farmacología
10.
PLoS One ; 7(8): e42177, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22879915

RESUMEN

Bone marrow mesenchymal stem cells (BMSCs) have been shown to ameliorate diabetes in animal models. The mechanism, however, remains largely unknown. An unanswered question is whether BMSCs are able to differentiate into ß-cells in vivo, or whether BMSCs are able to mediate recovery and/or regeneration of endogenous ß-cells. Here we examined these questions by testing the ability of hBMSCs genetically modified to transiently express vascular endothelial growth factor (VEGF) or pancreatic-duodenal homeobox 1 (PDX1) to reverse diabetes and whether these cells were differentiated into ß-cells or mediated recovery through alternative mechanisms. Human BMSCs expressing VEGF and PDX1 reversed hyperglycemia in more than half of the diabetic mice and induced overall improved survival and weight maintenance in all mice. Recovery was sustained only in the mice treated with hBMSCs-VEGF. However, de novo ß-cell differentiation from human cells was observed in mice in both cases, treated with either hBMSCs-VEGF or hBMSCs- PDX1, confirmed by detectable level of serum human insulin. Sustained reversion of diabetes mediated by hBMSCs-VEGF was secondary to endogenous ß-cell regeneration and correlated with activation of the insulin/IGF receptor signaling pathway involved in maintaining ß-cell mass and function. Our study demonstrated the possible benefit of hBMSCs for the treatment of insulin-dependent diabetes and gives new insight into the mechanism of ß-cell recovery after injury mediated by hBMSC therapy.


Asunto(s)
Células de la Médula Ósea/citología , Células Secretoras de Insulina/fisiología , Células Madre Mesenquimatosas/citología , Regeneración/fisiología , Adulto , Animales , Células de la Médula Ósea/metabolismo , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/terapia , Proteínas de Homeodominio/metabolismo , Humanos , Hiperglucemia/complicaciones , Hiperglucemia/metabolismo , Hiperglucemia/patología , Insulina/sangre , Células Secretoras de Insulina/enzimología , Células Secretoras de Insulina/patología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor IGF Tipo 1/metabolismo , Transducción de Señal/genética , Estreptozocina , Transactivadores/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
J Am Soc Nephrol ; 23(4): 661-73, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22302195

RESUMEN

Injection of amniotic fluid stem cells ameliorates the acute phase of acute tubular necrosis in animals by promoting proliferation of injured tubular cells and decreasing apoptosis, but whether these stem cells could be of benefit in CKD is unknown. Here, we used a mouse model of Alport syndrome, Col4a5(-/-) mice, to determine whether amniotic fluid stem cells could modify the course of progressive renal fibrosis. Intracardiac administration of amniotic fluid stem cells before the onset of proteinuria delayed interstitial fibrosis and progression of glomerular sclerosis, prolonged animal survival, and ameliorated the decline in kidney function. Treated animals exhibited decreased recruitment and activation of M1-type macrophages and a higher proportion of M2-type macrophages, which promote tissue remodeling. Amniotic fluid stem cells did not differentiate into podocyte-like cells and did not stimulate production of the collagen IVa5 needed for normal formation and function of the glomerular basement membrane. Instead, the mechanism of renal protection was probably the paracrine/endocrine modulation of both profibrotic cytokine expression and recruitment of macrophages to the interstitial space. Furthermore, injected mice retained a normal number of podocytes and had better integrity of the glomerular basement membrane compared with untreated Col4a5(-/-) mice. Inhibition of the renin-angiotensin system by amniotic fluid stem cells may contribute to these beneficial effects. In conclusion, treatment with amniotic fluid stem cells may be beneficial in kidney diseases characterized by progressive renal fibrosis.


Asunto(s)
Riñón/patología , Nefritis Hereditaria/terapia , Sistema Renina-Angiotensina/fisiología , Trasplante de Células Madre/métodos , Líquido Amniótico/citología , Análisis de Varianza , Animales , Western Blotting , Células Cultivadas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fibrosis/patología , Fibrosis/terapia , Inmunohistoquímica , Riñón/fisiopatología , Pruebas de Función Renal , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nefritis Hereditaria/patología , Podocitos/metabolismo , Distribución Aleatoria , Reacción en Cadena en Tiempo Real de la Polimerasa , Estadísticas no Paramétricas
12.
Adv Drug Deliv Rev ; 63(4-5): 379-87, 2011 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-21145933

RESUMEN

End stage renal disease is a major health problem in this country and worldwide. Although dialysis and kidney transplantation are currently used to treat this condition, kidney regeneration resulting in complete healing would be a desirable alternative. In this review we focus our attention on current therapeutic approaches used clinically to delay the onset of kidney failure. In addition we describe novel approaches, like Tissue Engineering, Stem cell Applications, Gene Therapy, and Renal Replacement Therapy that may one day be possible alternative therapies for patients with the hope of delaying kidney failure or even stopping the progression of renal disease.


Asunto(s)
Enfermedades Renales/terapia , Regeneración , Medicina Regenerativa/métodos , Animales , Terapia Genética/métodos , Humanos , Riñón/fisiología , Enfermedades Renales/fisiopatología , Fallo Renal Crónico/prevención & control , Terapia de Reemplazo Renal/métodos , Trasplante de Células Madre/métodos , Ingeniería de Tejidos/métodos
13.
Curr Opin Organ Transplant ; 16(1): 101-5, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21157345

RESUMEN

PURPOSE OF REVIEW: Amniotic fluid, due to its contact to the fetus during development, is considered an important diagnostic tool to evaluate the health status of the fetus during pregnancy. However, amniotic fluid also contains a heterogeneous cellular population that can be safely collected by amniocentesis and easily cultured. Many different cell types have been found within amniotic fluid and currently some of them are being tested for their possible use for cellular therapy. RECENT FINDINGS: Potential of pluripotent and multipotent cells isolated from the amniotic fluid has been tested and in-vitro differentiations toward various cell types have been successfully performed. Furthermore, in-vivo studies are highlighting the benefits and mechanisms of amniotic fluid cells for therapy, with particular focus on kidney and lung diseases. SUMMARY: Amniotic fluid may represent a precious source for easily and safely retrievable cell types that may be used for regenerative medicine purposes.


Asunto(s)
Líquido Amniótico/citología , Diferenciación Celular/fisiología , Células Madre Multipotentes/citología , Regeneración/fisiología , Medicina Regenerativa/métodos , Animales , Femenino , Humanos , Embarazo
14.
PLoS One ; 5(2): e9357, 2010 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-20195358

RESUMEN

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-kit(pos) 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.


Asunto(s)
Modelos Animales de Enfermedad , Células Madre Embrionarias/trasplante , Necrosis Tubular Aguda/cirugía , Trasplante de Células Madre/métodos , Líquido Amniótico/citología , Animales , Apoptosis/inmunología , Nitrógeno de la Urea Sanguínea , Proliferación Celular , Creatinina/sangre , Citocinas/metabolismo , Células Madre Embrionarias/inmunología , Células Madre Embrionarias/metabolismo , Expresión Génica , Glicerol , Humanos , Cariotipificación , Riñón/metabolismo , Riñón/patología , Riñón/cirugía , Necrosis Tubular Aguda/inducido químicamente , Necrosis Tubular Aguda/inmunología , Luciferasas/genética , Luciferasas/metabolismo , Mediciones Luminiscentes , Proteínas de la Membrana/genética , Ratones , Ratones Desnudos , Factor de Transcripción PAX2/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rabdomiólisis/inducido químicamente , Rabdomiólisis/inmunología , Rabdomiólisis/cirugía , Trasplante Heterólogo
15.
J Urol ; 183(3): 1193-200, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20096867

RESUMEN

PURPOSE: Human amniotic fluid contains multiple cell types, including pluripotent and committed progenitor cells, and fully differentiated cells. We characterized various cell populations in amniotic fluid. MATERIALS AND METHODS: Optimum culture techniques for multiple cell line passages with minimal morphological change were established. Cell line analysis and characterization were done with reverse transcriptase and real-time polymerase chain reaction. Immunoseparation was done to distinguish native progenitor cell lines and their various subpopulations. RESULTS: Endodermal and mesodermal marker expression was greatest in samples of early gestational age while ectodermal markers showed a constant rate across all samples. Pluripotent and mesenchymal cells were always present but hematopoietic cell markers were expressed only in older samples. Specific markers for lung, kidney, liver and heart progenitor cells were increasingly expressed after 18 weeks of gestation. We specifically focused on a CD24+OB-cadherin+ population that could identify uninduced metanephric mesenchyma-like cells, which in vivo are nephron precursors. The CD24+OB-cadherin+ cell line was isolated and subjected to further immunoseparation to select 5 distinct amniotic fluid kidney progenitor cell subpopulations based on E-cadherin, podocalyxin, nephrin, TRKA and PDGFRA expression, respectively. CONCLUSIONS: These subpopulations may represent different precursor cell lineages committed to specific renal cell fates. Committed progenitor cells in amniotic fluid may provide an important and novel resource of useful cells for regenerative medicine purposes.


Asunto(s)
Líquido Amniótico/citología , Medicina Regenerativa/métodos , Células Madre , Células Cultivadas , Predicción , Humanos , Riñón/fisiología , Regeneración , Medicina Regenerativa/tendencias
16.
Pediatr Res ; 63(5): 467-71, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18427289

RESUMEN

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.


Asunto(s)
Riñón , Medicina Regenerativa/métodos , Células Madre , Enfermedad Aguda , Enfermedad Crónica , Humanos , Enfermedades Renales/patología , Enfermedades Renales/terapia , Ingeniería de Tejidos/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA