Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
JCI Insight ; 2(2): e88848, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-28138555

RESUMEN

The neonatal mouse kidney retains nephron progenitor cells in a nephrogenic zone for 3 days after birth. We evaluated whether de novo nephrogenesis can be induced postnatally beyond 3 days. Given the long-term implications of nephron number for kidney health, it would be useful to enhance nephrogenesis in the neonate. We induced nephron reduction by cryoinjury with or without contralateral nephrectomy during the neonatal period or after 1 week of age. There was no detectable compensatory de novo nephrogenesis, as determined by glomerular counting and lineage tracing. Contralateral nephrectomy resulted in additional adaptive healing, with little or no fibrosis, but did not also stimulate de novo nephrogenesis. In contrast, injury initiated at 1 week of age led to healing with fibrosis. Thus, despite the presence of progenitor cells and ongoing nephron maturation in the newborn mouse kidney, de novo nephrogenesis is not inducible by acute nephron reduction. This indicates that additional nephron progenitors cannot be recruited after birth despite partial renal ablation providing a reparative stimulus and suggests that nephron number in the mouse is predetermined at birth.


Asunto(s)
Criocirugía , Nefrectomía , Nefronas/crecimiento & desarrollo , Células Madre , Animales , Animales Recién Nacidos , Linaje de la Célula , Fibrosis , Proteínas de Homeodominio/metabolismo , Riñón/patología , Glomérulos Renales/crecimiento & desarrollo , Glomérulos Renales/patología , Proteínas con Homeodominio LIM/metabolismo , Ratones , Nefronas/patología , Organogénesis , Factor de Transcripción PAX2/metabolismo , Factores de Transcripción/metabolismo , Transcriptoma
2.
J Am Soc Nephrol ; 27(11): 3394-3404, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26984884

RESUMEN

Erythropoietin (EPO) may be a beneficial tissue-protective cytokine. However, high doses of EPO are associate with adverse effects, including thrombosis, tumor growth, and hypertension. Carbamylated erythropoietin (CEPO) lacks both erythropoietic and vasoconstrictive actions. In this study, we compared the renoprotective, hemodynamic, and hematologic activities and survival effects of identical EPO and CEPO doses in rat models of clinically relevant AKI presentations, including ischemia-reperfusion-induced AKI superimposed on CKD (5000 U/kg EPO or CEPO; three subcutaneous injections) and ischemia-reperfusion-induced AKI in old versus young animals and male versus female animals (1000 U/kg EPO or CEPO; three subcutaneous injections). Compared with EPO therapy, CEPO therapy induced greater improvements in renal function and body weight in AKI on CKD animals, with smaller increases in hematocrit levels and similarly improved survival. Compared with EPO therapy in the other AKI groups, CEPO therapy induced greater improvements in protection and recovery of renal function and survival, with smaller increases in systolic BP and hematocrit levels. Overall, old or male animals had more severe loss in kidney function and higher mortality rates than young or female animals, respectively. Notably, mRNA and protein expression analyses confirmed the renal expression of the heterodimeric EPO receptor/CD131 complex, which is required for the tissue-protective effects of CEPO signaling. In conclusion, CEPO improves renal function, body and kidney weight, and survival in AKI models without raising hematocrit levels and BP as substantially as EPO. Thus, CEPO therapy may be superior to EPO in improving outcomes in common forms of clinical AKI.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Eritropoyetina/análogos & derivados , Eritropoyetina/uso terapéutico , Insuficiencia Renal Crónica/tratamiento farmacológico , Factores de Edad , Animales , Femenino , Masculino , Ratas , Ratas Endogámicas F344 , Factores Sexuales
3.
F1000Prime Rep ; 6: 83, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25343040

RESUMEN

Acute kidney injury (AKI) is a common clinical entity associated with high morbidity and mortality and clinical costs. The pathophysiology is multifaceted and involves inflammation, tubular injury, and vascular damage. Recently identified components include necroptosis, a special form of cell death, and autophagy. Most of the pathophysiological knowledge is obtained from animal models but these do not directly reflect the reality of the clinical situation. Tubular cells have a remarkable capacity to regenerate, and the role of stem/progenitor cells is discussed. Acute kidney injury is frequently associated with chronic kidney disease, and the implications are widespread.

5.
Am J Kidney Dis ; 60(6): 1012-22, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23036928

RESUMEN

Acute kidney injury (AKI) is a common clinical entity with high morbidity and mortality rates and ever increasing medical costs. A large number of patients who are hospitalized with morbidities such as diabetes, vascular disease, or chronic kidney disease are at high risk to develop AKI due to ischemic and nephrotoxic insults. The pathophysiology of ischemic and toxic forms of AKI is complex and includes tubular and vascular cell damage and inflammation. Given the seriousness of this essentially therapy-resistant complication, treatment beyond supportive measures and renal replacement therapy is urgently needed. Recent stem cell research has shown promising results, and cell therapy-based interventions are advancing into clinical trials. An example is our phase 1 clinical trial (NCT00733876) in which cardiac surgery patients at high risk of postoperative AKI were treated safely with allogeneic mesenchymal stem cells. Together with the introduction of biomarkers for an earlier and specific AKI diagnosis, currently tested stem cell-based therapies are expected to provide an entirely new class of diagnostic and therapeutic tools.


Asunto(s)
Lesión Renal Aguda/fisiopatología , Lesión Renal Aguda/terapia , Riñón/fisiología , Trasplante de Células Madre Mesenquimatosas/métodos , Regeneración/fisiología , Lesión Renal Aguda/diagnóstico , Anciano , Ensayos Clínicos Fase I como Asunto/tendencias , Humanos , Masculino , Diálisis Renal/métodos
6.
Organogenesis ; 7(2): 96-100, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21521944

RESUMEN

An extensive body of preclinical and clinical data has shown that administration of adult multipotent marrow stromal cells (MSCs) effectively ameliorates experimental and clinical conditions of many different organ systems. Differentiation into organ parenchymal cells, however, is very rare, and the main mechanism for organ protection and regeneration from different types of injury is the exertion of paracrine effects and stimulation of tissue repair. A large number of clinical trials have been conducted and are ongoing to investigate the safety and efficacy of MSCs in different organs after various types of organ injury. This article intends to give a brief overview about current applications of MSCs and mechanisms involved in organ protection and regeneration.


Asunto(s)
Células de la Médula Ósea/citología , Regeneración Tisular Dirigida/métodos , Células Madre Multipotentes/citología , Humanos , Especificidad de Órganos , Trasplante de Células Madre , Células del Estroma/citología
7.
Kidney Int Suppl (2011) ; 1(3): 87-89, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25018907

RESUMEN

Both the homing of hematopoietic stem cells (HSCs) to the bone marrow and their engraftment in recipients of bone marrow transplants are primarily mediated by the chemokine stromal-derived factor-1 (SDF-1) or CXCL12, which activates CXCR4, its cognate receptor on HSCs. We showed that the recruitment and temporary attachment of CXCR4-expressing cells, such as HSCs and a fraction of mesenchymal stem cells (MSCs), to the kidney, following ischemia/reperfusion acute kidney injury, are similarly mediated by robustly upregulated SDF-1 in the kidney, indicating that such organ injury appears to lead to the transient expression of a facultative stem cell niche. This SDF-1 response of the injured kidney facilitates both the mobilization from the bone marrow and homing of precursor cells, and other CXCR4-expressing cells such as administered MSCs, to the kidney, where they aid in its protection and repair. Similar responses have been observed subsequent to the injury of other solid organs such as the heart, liver, and brain.

8.
Kidney Int Suppl (2011) ; 1(3): 103-106, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25018910

RESUMEN

Current therapies for acute kidney injury remain primarily supportive and have failed to reduce morbidity, mortality (>50%), and associated costs. This prompted our studies in which rats with bilateral ischemia/reperfusion-induced acute kidney injury were treated with bone marrow-derived, culture-expanded allogeneic mesenchymal stem cells. Their administration into the suprarenal aorta after reflow significantly protected renal function and hastened repair, mediated by complex antiapoptotic, mitogenic, anti-inflammatory, and immune modulating actions that were not elicited by isogeneic fibroblasts. Infused mesenchymal stem cells, recruited to renal sites of injury, did not significantly differentiate into target cells but rather disappeared from kidneys and other organs within 72 h. Furthermore, at 3 months, compared with vehicle-treated controls, renal function was well preserved and interstitial fibrosis was absent. These preclinical data served as the scientific basis for a recently completed Phase I Clinical Trial (http://www.clinicaltrials.gov; # NCT00733876), in which patients at high risk for cardiac surgery-associated AKI were treated with allogeneic mesenchymal stem cells. Until now, MSC therapy in the study subjects has been safe, and none of the patients has developed postoperative AKI or subsequent loss of renal function, suggesting that this novel form of therapy may have promise in this group of high-risk patients, which will be further investigated in a Phase II Trial.

9.
Nat Rev Nephrol ; 6(3): 179-83, 2010 03.
Artículo en Inglés | MEDLINE | ID: mdl-20186233

RESUMEN

Acute kidney injury (AKI) is a common clinical complication, associated with poor outcomes and the development of chronic kidney disease. Despite major advances in the understanding of its pathophysiology, available therapies for AKI are only supportive; therefore, adequate functional recovery from AKI must predominantly rely on the kidney's own reparative ability. An extensive body of preclinical data from our own and from other laboratories has shown that administration of adult multipotent marrow stromal cells (commonly referred to as mesenchymal stem cells [MSCs]), effectively ameliorates experimental AKI by exerting paracrine renoprotective effects and by stimulating tissue repair. Based on these findings, a clinical trial has been conducted to investigate the safety and efficacy of MSCs administered to open-heart surgery patients who are at high risk of postoperative AKI. In this Perspectives article, we discuss some of the early data from this trial and describe potential applications for stem cell therapies in other fields of nephrology.


Asunto(s)
Lesión Renal Aguda/terapia , Puente de Arteria Coronaria/efectos adversos , Fallo Renal Crónico/prevención & control , Trasplante de Células Madre Mesenquimatosas/métodos , Lesión Renal Aguda/etiología , Lesión Renal Aguda/mortalidad , Animales , Ensayos Clínicos Fase I como Asunto , Puente de Arteria Coronaria/métodos , Enfermedad Coronaria/diagnóstico , Enfermedad Coronaria/terapia , Modelos Animales de Enfermedad , Femenino , Estudios de Seguimiento , Rechazo de Injerto , Supervivencia de Injerto , Humanos , Fallo Renal Crónico/etiología , Fallo Renal Crónico/fisiopatología , Masculino , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Ratones , Ratas , Medición de Riesgo , Tasa de Supervivencia , Trasplante Autólogo , Resultado del Tratamiento
10.
J Cell Mol Med ; 13(8B): 2109-2114, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19397783

RESUMEN

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.


Asunto(s)
Riñón/fisiopatología , Células Madre Multipotentes/citología , Células del Estroma/citología , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Secuencia de Bases , Medios de Cultivo Condicionados , Cartilla de ADN , Modelos Animales de Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley , Factor A de Crecimiento Endotelial Vascular/genética
11.
Stem Cells Dev ; 18(3): 475-85, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18564903

RESUMEN

Acute kidney injury (AKI) is a major clinical problem associated with high morbidity and mortality. Likely due to its complex pathophysiology, therapies with a single pharmacological agent have generally failed to improve outcomes. In contrast, stem cell-based interventions utilize these cells' ability to simultaneously target multiple pathophysiological components of AKI and thus represent a promising new tool for the treatment of AKI. The aims of the this study were to investigate the long-term outcome and safety of treatment with autologous and allogeneic mesenchymal stem cells (MSCs) after AKI and the role of vascular endothelial growth factor (VEGF) as one of the principal paracrine mediators of renoprotection of MSCs. MSC administration after AKI was not associated with adverse events and proved to be renoprotective in animals with severe renal failure. Identical doses of autologous MSC were more effective than allogeneic. At 3 months, MSCs were not engrafted in any tissues except in the bone marrow in 50% of animals given the highest allogeneic cell dose. There was no long-term fibrotic response in the kidneys attributable to MSC therapy, and animals with severe AKI were protected from development of fibrotic lesions after AKI. Furthermore, this study establishes VEGF as a critical factor mediating renal recovery. VEGF knockdown by small-interfering RNA reduced effectiveness of MSCs significantly and decreased survival. In summary, our results show that both autologous and allogeneic MSC are safe and effective in AKI, and importantly, reduce late renal fibrosis and loss of renal function in surviving animals and that VEGF is a critical factor in renoprotection by MSCs. Together, we posit that these data provide further justification for the conduct of clinical trails in which AKI is treated with MSC.


Asunto(s)
Lesión Renal Aguda/terapia , Células de la Médula Ósea/fisiología , Riñón/patología , Trasplante de Células Madre , Células del Estroma/fisiología , Trasplante Autólogo , Trasplante Homólogo , Lesión Renal Aguda/patología , Animales , Células de la Médula Ósea/citología , Expresión Génica , Humanos , Riñón/citología , Masculino , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley , Ratas Transgénicas , Células del Estroma/citología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
Am J Physiol Renal Physiol ; 295(1): F315-21, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18480180

RESUMEN

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.


Asunto(s)
Lesión Renal Aguda/patología , Mediciones Luminiscentes/métodos , Trasplante de Células Madre Mesenquimatosas/métodos , Lesión Renal Aguda/terapia , Animales , Femenino , Luciferina de Luciérnaga , Masculino , Ratones , Ratones Endogámicos C57BL
13.
Dev Dyn ; 236(12): 3321-31, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17685479

RESUMEN

Stem cells have been recognized as a potential tool for the development of innovative therapeutic strategies. There are in general two types of stem cells, embryonic and adult stem cells. While embryonic stem cell therapy has been riddled with problems of allogeneic rejection and ethical concerns, adult stem cells have long been used in the treatment of hematological malignancies. With the recognition of additional, potentially therapeutic characteristics, bone marrow-derived stem cells have become a tool in regenerative medicine. The bone marrow is an ideal source of stem cells because it is easily accessible and harbors two types of stem cells. Hematopoietic stem cells give rise to all blood cell types and have been shown to exhibit plasticity, while multipotent marrow stromal cells are the source of osteocytes, chondrocytes, and fat cells and have been shown to support and generate a large number of different cell types. This review describes the general characteristics of these stem cell populations and their current and potential future applications in regenerative medicine.


Asunto(s)
Células Madre Adultas/trasplante , Trasplante de Células Madre Hematopoyéticas , Regeneración , Ensayos Clínicos como Asunto , Movilización de Célula Madre Hematopoyética , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Trasplante de Células Madre Mesenquimatosas , Células Madre Multipotentes/trasplante , Infarto del Miocardio/terapia
14.
J Magn Reson Imaging ; 25(6): 1179-91, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17520738

RESUMEN

PURPOSE: To evaluate MRI for a qualitative and quantitative in vivo tracking of intraaortal injected iron oxide-labeled mesenchymal stem cells (MSC) into rats with acute kidney injury (AKI). MATERIALS AND METHODS: In vitro MRI and R2* measurement of nonlabeled and superparamagnetic iron oxide (SPIO)-labeled MSC (MSC(SPIO)) was performed in correlation to cellular iron content and cytological examination (Prussian blue, electron microscopy). In vivo MRI and R2* evaluation were performed before and after ischemic/reperfusion AKI (N = 14) and intraaortal injection of 1.5 x 10(6) MSC(SPIO) (N = 7), fetal calf serum (FCS) (medium, N = 6), and SPIO alone (N = 1) up to 14 days using a clinical 3T scanner. Signal to noise ratios (SNR), R2* of kidneys, liver, spleen, and bone marrow, renal function (creatinine [CREA], blood urea nitrogen [BUN]), and kidney volume were measured and tested for statistical significance (Student's t-test, P < 0.05) in comparison histology (hematoxylin and eosin [H&E], Prussian blue, periodic acid-Schiff [PAS], CD68). RESULTS: In vitro, MSC(SPIO) showed a reduction of SNR and T2* with R2* approximately number of MSC(SPIO) (R2 = 0.98). In vivo MSC(SPIO) administration resulted in a SNR decrease (35 +/- 15%) and R2* increase (101 +/- 18.3%) in renal cortex caused by MSC(SPIO) accumulation in contrast to control animals (P < 0.01). Liver, spleen, and bone marrow (MSC(SPIO)) showed a delayed SNR decline/R2* increase (P < 0.05) resulting from MSC(SPIO) migration. The increase of kidney volume and the decrease in renal function (P < 0.05) was reduced in MSC-treated animals. CONCLUSION: Qualitative and quantitative in vivo cell-tracking and monitoring of organ distribution of intraaortal injected MSC(SPIO) in AKI is feasible in MRI at 3T.


Asunto(s)
Isquemia/diagnóstico , Isquemia/terapia , Riñón/citología , Imagen por Resonancia Magnética/métodos , Trasplante de Células Madre Mesenquimatosas , Animales , Medios de Contraste , Dextranos , Estudios de Factibilidad , Óxido Ferrosoférrico , Procesamiento de Imagen Asistido por Computador , Inyecciones Intraarteriales , Hierro , Isquemia/fisiopatología , Nanopartículas de Magnetita , Óxidos , Ratas , Ratas Sprague-Dawley , Coloración y Etiquetado
15.
Am J Physiol Renal Physiol ; 292(5): F1626-35, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17213465

RESUMEN

Acute kidney injury (AKI) is a major clinical problem in which a critical vascular, pathophysiological component is recognized. We demonstrated previously that mesenchymal stem cells (MSC), unlike fibroblasts, are significantly renoprotective after ischemia-reperfusion injury and concluded that this renoprotection is mediated primarily by paracrine mechanisms. In this study, we investigated whether MSC possess vasculoprotective activity that may contribute, at least in part, to an improved outcome after ischemia-reperfusion AKI. MSC-conditioned medium contains VEGF, HGF, and IGF-1 and augments aortic endothelial cell (EC) growth and survival, a response not observed with fibroblast-conditioned medium. MSC and EC share vasculotropic gene expression profiles, as both form capillary tubes in vitro on Matrigel alone or in cooperation without fusion. MSC undergo differentiation into an endothelial-like cell phenotype in culture and develop into vascular structures in vivo. Infused MSC were readily detected in the kidney early after reflow but were only rarely engrafted at 1 wk post-AKI. MSC attached in the renal microvascular circulation significantly decreased apoptosis of adjacent cells. Infusion of MSC immediately after reflow in severe ischemia-reperfusion AKI did not improve renal blood flow, renovascular resistance, or outer cortical blood flow. These data demonstrate that the unique vasculotropic, paracrine actions elicited by MSC play a significant renoprotective role after AKI, further demonstrating that cell therapy has promise as a novel intervention in AKI.


Asunto(s)
Riñón/irrigación sanguínea , Riñón/fisiopatología , Trasplante de Células Madre Mesenquimatosas , Comunicación Paracrina , Daño por Reperfusión/fisiopatología , Enfermedad Aguda , Animales , Aorta/patología , Aorta/fisiopatología , Apoptosis , Vasos Sanguíneos/fisiopatología , Adhesión Celular , Comunicación Celular , Supervivencia Celular , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Células Endoteliales , Perfilación de la Expresión Génica , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Riñón/cirugía , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Endogámicos C57BL , Microcirculación , Ratas , Recuperación de la Función , Circulación Renal , Daño por Reperfusión/cirugía
16.
Nephron Exp Nephrol ; 105(1): e33-40, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17108708

RESUMEN

BACKGROUND: Acute kidney injury (AKI) secondary to ischemia continues to be a major clinical problem due to its high morbidity and mortality, and limited treatment options. Animal models are critical to both the study of the pathophysiology of AKI and the development of new interventions. Although histological changes at the glomerulotubular junction have been described in AKI, we examined here whether the extent of glomerular tubularization correlates with the degree of renal insufficiency in this condition. METHODS: Groups of mice with ischemia/reperfusion AKI were utilized in which the severity of renal insufficiency was defined. The resulting level of glomerular tubularization was analyzed, and the involved cell type was identified by immunohistochemistry and electron microscopy. RESULTS: The extent of glomerular tubularization increased significantly with the degree of renal insufficiency. Low level glomerular tubularization was present in normal mouse kidneys, while it was more common and increasingly circumferential in mice with more severe loss of kidney function. The parietal monolayer of cuboidal cells in glomeruli was contiguous with proximal tubular cells, showing a well-developed luminal brush border and positive staining for proliferating cell nuclear antigen and Lotus tetragonolobus, a proximal tubular cell-specific lectin. CONCLUSION: Increased levels of glomerular tubularization represent a poorly understood response to ischemic AKI in mice. As such, this glomerular 'tubularization score' may be useful to complement standard injury scores in experimental and, if detected, in clinical AKI.


Asunto(s)
Isquemia/patología , Glomérulos Renales/patología , Riñón/irrigación sanguínea , Enfermedad Aguda , Animales , Masculino , Ratones , Antígeno Nuclear de Célula en Proliferación/análisis
17.
Proc Natl Acad Sci U S A ; 103(15): 5965-70, 2006 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-16585502

RESUMEN

Recombinant human erythropoietin (rhEPO) is receiving increasing attention as a potential therapy for prevention of injury and restoration of function in nonhematopoietic tissues. However, the minimum effective dose required to mimic and augment these normal paracrine functions of erythropoietin (EPO) in some organs (e.g., the brain) is higher than for treatment of anemia. Notably, a dose-dependent risk of adverse effects has been associated with rhEPO administration, especially in high-risk groups, including polycythemia-hyperviscosity syndrome, hypertension, and vascular thrombosis. Of note, several clinical trials employing relatively high dosages of rhEPO in oncology patients were recently halted after an increase in mortality and morbidity, primarily because of thrombotic events. We recently identified a heteromeric EPO receptor complex that mediates tissue protection and is distinct from the homodimeric receptor responsible for the support of erythropoiesis. Moreover, we developed receptor-selective ligands that provide tools to assess which receptor isoform mediates which biological consequence of rhEPO therapy. Here, we demonstrate that rhEPO administration in the rat increases systemic blood pressure, reduces regional renal blood flow, and increases platelet counts and procoagulant activities. In contrast, carbamylated rhEPO, a heteromeric receptor-specific ligand that is fully tissue protective, increases renal blood flow, promotes sodium excretion, reduces injury-induced elevation in procoagulant activity, and does not effect platelet production. These preclinical findings suggest that nonerythropoietic tissue-protective ligands, which appear to elicit fewer adverse effects, may be especially useful in clinical settings for tissue protection.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Coagulantes/farmacología , Eritropoyetina/análogos & derivados , Eritropoyetina/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/fisiología , Línea Celular , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Eritropoyetina/uso terapéutico , Hematopoyesis/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Humanos , Ratones , Circulación Renal/efectos de los fármacos , Circulación Renal/fisiología , Células Madre/efectos de los fármacos , Células Madre/fisiología , Venas Umbilicales
18.
Kidney Int ; 68(4): 1613-7, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16164638

RESUMEN

BACKGROUND: Adult stem cells are promising for the development of novel therapies in regenerative medicine. Acute renal failure (ARF) remains a frequent clinical complication, associated with an unacceptably high mortality rate, in large part due to the ineffectiveness of currently available therapies. The aim of this study was, therefore, to evaluate the therapeutic effectiveness of bone marrow-derived mesenchymal stem cells in a rat model of ischemia/reperfusion (I/R) ARF. METHODS: We used a common I/R model in rats to induce ARF by clamping both renal pedicles for 40 minutes. Mesenchymal stem cells were iron-dextran-labeled for in vivo tracking studies by magnetic resonance imaging (MRI) and kidneys were imaged for mesenchymal stem cells immediately after infusion and at day 3 after ARF. Renal injury was scored on day 3 and cells were additionally tracked by Prussian blue staining. RESULTS: We show in I/R-induced ARF in rats, modeling the most common form of clinical ARF, that infusion of mesenchymal stem cells enhances recovery of renal function. Mesenchymal stem cells were found to be located in the kidney cortex after injection, as demonstrated by MRI. Mesenchymal stem cells-treated animals had both significantly better renal function on days 2 and 3 and better injury scores at day 3 after ARF. Histologically, mesenchymal stem cells were predominantly located in glomerular capillaries, while tubules showed no iron labeling, indicating absent tubular transdifferentiation. CONCLUSION: We conclude that the highly renoprotective capacity of mesenchymal stem cells opens the possibility for a cell-based paradigm shift in the treatment of I/R ARF.


Asunto(s)
Lesión Renal Aguda/terapia , Daño por Reperfusión/terapia , Trasplante de Células Madre , Lesión Renal Aguda/patología , Animales , Colorantes , Ferrocianuros , Supervivencia de Injerto , Complejo Hierro-Dextran , Riñón/patología , Imagen por Resonancia Magnética , Masculino , Mesodermo/citología , Ratas , Ratas Sprague-Dawley , Recuperación de la Función , Daño por Reperfusión/patología
19.
Kidney Int ; 67(5): 1772-84, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15840024

RESUMEN

BACKGROUND: Stem cell and leukocyte migration during homeostasis and inflammation is regulated by a number of chemokines. Stromal cell-derived factor-1 (SDF-1) and its receptor CXCR4 are important mediators of leukocyte homeostasis. The postischemic kidney has been shown to recruit different leukocyte populations, including bone marrow-derived stem cells. Therefore, we investigated the SDF-1/CXCR4 system in the kidney after ischemic acute renal failure (ARF). METHODS: We used immunohistochemistry, in situ hybridization, enzyme-linked immunosorbent assay (ELISA) and real-time reverse transcription-polymerase chain reaction (RT-PCR) to detect SDF-1 and CXCR4 in the normal kidney and the kidney after ischemia/reperfusion (I/R) ARF. Mobilization was assessed by flow cytometry for CD34 and colony assays. RESULTS: We show that SDF-1 is expressed in the normal mouse kidney and tubular cells express CXCR4. SDF-1 expression in the kidney increases after I/R induced ARF and decreases in the bone marrow, thereby reversing the normal gradient between bone marrow and the periphery. This causes mobilization of CD34-positive cells into the circulation and their subsequent homing to the kidney with ARF. In vitro and in vivo chemotaxis of bone marrow cells toward damaged kidney epithelium is reversibly inhibited by anti-CXCR4 antibodies. CONCLUSION: Our data show that renal SDF-1 is a currently unrecognized mediator of homing to and migration of CXCR4 expressing cells in the injured kidney. Because certain cells that express CXCR4 may have renoprotective effects, our results suggest that SDF-1 may be a major signal involved in kidney repair.


Asunto(s)
Quimiocinas CXC/metabolismo , Riñón/lesiones , Riñón/metabolismo , Receptores CXCR4/metabolismo , Daño por Reperfusión/metabolismo , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Antígenos CD34/metabolismo , Secuencia de Bases , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Movimiento Celular , Quimiocina CXCL12 , Quimiocinas CXC/genética , Femenino , Hibridación in Situ , Riñón/patología , Masculino , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores CXCR4/genética , Daño por Reperfusión/genética , Daño por Reperfusión/patología , Transducción de Señal , Regulación hacia Arriba
20.
Am J Physiol Renal Physiol ; 289(1): F31-42, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15713913

RESUMEN

Severe acute renal failure (ARF) remains a common, largely treatment-resistant clinical problem with disturbingly high mortality rates. Therefore, we tested whether administration of multipotent mesenchymal stem cells (MSC) to anesthetized rats with ischemia-reperfusion-induced ARF (40-min bilateral renal pedicle clamping) could improve the outcome through amelioration of inflammatory, vascular, and apoptotic/necrotic manifestations of ischemic kidney injury. Accordingly, intracarotid administration of MSC (approximately 10(6)/animal) either immediately or 24 h after renal ischemia resulted in significantly improved renal function, higher proliferative and lower apoptotic indexes, as well as lower renal injury and unchanged leukocyte infiltration scores. Such renoprotection was not obtained with syngeneic fibroblasts. Using in vivo two-photon laser confocal microscopy, fluorescence-labeled MSC were detected early after injection in glomeruli, and low numbers attached at microvasculature sites. However, within 3 days of administration, none of the administered MSC had differentiated into a tubular or endothelial cell phenotype. At 24 h after injury, expression of proinflammatory cytokines IL-1beta, TNF-alpha, IFN-gamma, and inducible nitric oxide synthase was significantly reduced and that of anti-inflammatory IL-10 and bFGF, TGF-alpha, and Bcl-2 was highly upregulated in treated kidneys. We conclude that the early, highly significant renoprotection obtained with MSC is of considerable therapeutic promise for the cell-based management of clinical ARF. The beneficial effects of MSC are primarily mediated via complex paracrine actions and not by their differentiation into target cells, which, as such, appears to be a more protracted response that may become important in late-stage organ repair.


Asunto(s)
Lesión Renal Aguda/fisiopatología , Lesión Renal Aguda/terapia , Diferenciación Celular/fisiología , Trasplante de Células Madre Mesenquimatosas , Daño por Reperfusión/fisiopatología , Animales , Células de la Médula Ósea/fisiología , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Riñón/metabolismo , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA