Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
2.
JCI Insight ; 9(6)2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38516889

RESUMEN

Here, we used digital spatial profiling (DSP) to describe the glomerular transcriptomic signatures that may characterize the complex molecular mechanisms underlying progressive kidney disease in Alport syndrome, focal segmental glomerulosclerosis, and membranous nephropathy. Our results revealed significant transcriptional heterogeneity among diseased glomeruli, and this analysis showed that histologically similar glomeruli manifested different transcriptional profiles. Using glomerular pathology scores to establish an axis of progression, we identified molecular pathways with progressively decreased expression in response to increasing pathology scores, including signal recognition particle-dependent cotranslational protein targeting to membrane and selenocysteine synthesis pathways. We also identified a distinct signature of upregulated and downregulated genes common to all the diseases investigated when compared with nondiseased tissue from nephrectomies. These analyses using DSP at the single-glomerulus level could help to increase insight into the pathophysiology of kidney disease and possibly the identification of biomarkers of disease progression in glomerulopathies.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria , Nefritis Hereditaria , Insuficiencia Renal Crónica , Humanos , Transcriptoma , Glomérulos Renales/patología , Glomeruloesclerosis Focal y Segmentaria/patología , Nefritis Hereditaria/patología , Insuficiencia Renal Crónica/metabolismo
3.
JCI Insight ; 9(4)2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38227377

RESUMEN

The deposition of antipodocyte autoantibodies in the glomerular subepithelial space induces primary membranous nephropathy (MN), the leading cause of nephrotic syndrome worldwide. Taking advantage of the glomerulus-on-a-chip system, we modeled human primary MN induced by anti-PLA2R antibodies. Here we show that exposure of primary human podocytes expressing PLA2R to MN serum results in IgG deposition and complement activation on their surface, leading to loss of the chip permselectivity to albumin. C3a receptor (C3aR) antagonists as well as C3AR gene silencing in podocytes reduced oxidative stress induced by MN serum and prevented albumin leakage. In contrast, inhibition of the formation of the membrane-attack-complex (MAC), previously thought to play a major role in MN pathogenesis, did not affect permselectivity to albumin. In addition, treatment with a C3aR antagonist effectively prevented proteinuria in a mouse model of MN, substantiating the chip findings. In conclusion, using a combination of pathophysiologically relevant in vitro and in vivo models, we established that C3a/C3aR signaling plays a critical role in complement-mediated MN pathogenesis, indicating an alternative therapeutic target for MN.


Asunto(s)
Glomerulonefritis Membranosa , Síndrome Nefrótico , Podocitos , Animales , Humanos , Ratones , Albúminas , Glomerulonefritis Membranosa/genética , Glomérulos Renales/patología , Síndrome Nefrótico/patología , Podocitos/patología
4.
Pediatr Nephrol ; 2023 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-37775581

RESUMEN

Extracellular vesicles (EVs) are membranous cargo particles that mediate intercellular communication. They are heterogeneous in size and mechanism of release, and found in all biological fluids. Since EV content is in relation to the originating cell type and to its physiopathological conditions, EVs are under study to understand organ physiology and pathology. In addition, EV surface cargo, or corona, can be influenced by the microenvironment, leading to the concept that EV-associated molecules can represent useful biomarkers for diseases. Recent studies also focus on the use of natural, engineered, or synthetic EVs for therapeutic purposes. This review highlights the role of EVs in kidney development, pediatric kidney diseases, including inherited disorders, and kidney transplantation. Although few studies exist, they have promising results and may guide researchers in this field. Main limitations, including the influence of age on EV analyses, are also discussed.

5.
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
6.
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
7.
Kidney Int ; 101(1): 131-136, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34555393

RESUMEN

Analysis of the transcriptional profile of graft biopsies represents a promising strategy to study T cell-mediated-rejection (TCMR), also known as acute cellular rejection. However, bulk RNA sequencing of graft biopsies may not capture the focal nature of acute rejection. Herein, we used the whole exome GeoMX Digital Space Profiling platform to study five tubular and three glomerular regions of interest in the kidney graft biopsy from a patient with a chronic-active TCMR episode and in analogous areas from two different normal kidney control biopsies. All kidney sections were from paraffin blocks. Overall, inflammatory genes were significantly upregulated in the tubular areas of the TCMR biopsy and showed an enrichment for gene-ontology terms associated with T-cell activation, differentiation, and proliferation. Enrichment analysis of the 100 genes with the highest coefficient of variation across the TCMR tubular regions of interest revealed that these highly variable genes are involved in kidney development and injury and interestingly do not associate with the 2019 Banff classification pathology scores within the individual regions of interest. Spatial transcriptomics allowed us to unravel a previously unappreciated variability across different areas of the TCMR biopsy related to the graft response to the alloimmune attack, rather than to the immune cells. Thus, our approach has the potential to decipher clinically relevant, new pathogenic mechanisms, and therapeutic targets in acute cellular rejection and other kidney diseases with a focal nature.


Asunto(s)
Trasplante de Riñón , Linfocitos T , Aloinjertos/patología , Biopsia , Rechazo de Injerto , Humanos , Riñón/patología , Trasplante de Riñón/efectos adversos
8.
JCI Insight ; 7(1)2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-34793332

RESUMEN

Alport syndrome (AS) is a genetic disorder caused by mutations in type IV collagen that lead to defective glomerular basement membrane, glomerular filtration barrier (GFB) damage, and progressive chronic kidney disease. While the genetic basis of AS is well known, the molecular and cellular mechanistic details of disease pathogenesis have been elusive, hindering the development of mechanism-based therapies. Here, we performed intravital multiphoton imaging of the local kidney tissue microenvironment in a X-linked AS mouse model to directly visualize the major drivers of AS pathology. Severely distended glomerular capillaries and aneurysms were found accompanied by numerous microthrombi, increased glomerular endothelial surface layer (glycocalyx) and immune cell homing, GFB albumin leakage, glomerulosclerosis, and interstitial fibrosis by 5 months of age, with an intermediate phenotype at 2 months. Renal histology in mouse or patient tissues largely failed to detect capillary aberrations. Treatment of AS mice with hyaluronidase or the ACE inhibitor enalapril reduced the excess glomerular endothelial glycocalyx and blocked immune cell homing and GFB albumin leakage. This study identified central roles of glomerular mechanical forces and endothelial and immune cell activation early in AS, which could be therapeutically targeted to reduce mechanical strain and local tissue inflammation and improve kidney function.


Asunto(s)
Capilares , Microscopía Intravital , Glomérulos Renales , Nefritis Hereditaria , Animales , Capilares/diagnóstico por imagen , Capilares/inmunología , Capilares/patología , Microambiente Celular/fisiología , Modelos Animales de Enfermedad , Humanos , Glomérulos Renales/irrigación sanguínea , Glomérulos Renales/diagnóstico por imagen , Glomérulos Renales/inmunología , Glomérulos Renales/patología , Masculino , Ratones , Nefritis Hereditaria/diagnóstico por imagen , Nefritis Hereditaria/patología
9.
Front Med (Lausanne) ; 8: 772883, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34901088

RESUMEN

Kidney disease is characterized by loss of glomerular function with clinical manifestation of proteinuria. Identifying the cellular and molecular changes that lead to loss of protein in the urine is challenging due to the complexity of the filtration barrier, constituted by podocytes, glomerular endothelial cells, and glomerular basement membrane. In this review, we will discuss how technologies like single cell RNA sequencing and bioinformatics-based spatial transcriptomics, as well as in vitro systems like kidney organoids and the glomerulus-on-a-chip, have contributed to our understanding of glomerular pathophysiology. Knowledge gained from these studies will contribute toward the development of personalized therapeutic approaches for patients affected by proteinuric diseases.

10.
Sci Rep ; 10(1): 11414, 2020 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-32651395

RESUMEN

Glomerular endothelial cells (GEC) are a crucial component of the glomerular physiology and their damage contributes to the progression of chronic kidney diseases. How GEC affect the pathology of Alport syndrome (AS) however, is unclear. We characterized GEC from wild type (WT) and col4α5 knockout AS mice, a hereditary disorder characterized by progressive renal failure. We used endothelial-specific Tek-tdTomato reporter mice to isolate GEC by FACS and performed transcriptome analysis on them from WT and AS mice, followed by in vitro functional assays and confocal and intravital imaging studies. Biopsies from patients with chronic kidney disease, including AS were compared with our findings in mice. We identified two subpopulations of GEC (dimtdT and brighttdT) based on the fluorescence intensity of the TektdT signal. In AS mice, the brighttdT cell number increased and presented differential expression of endothelial markers compared to WT. RNA-seq analysis revealed differences in the immune and metabolic signaling pathways. In AS mice, dimtdT and brighttdT cells had different expression profiles of matrix-associated genes (Svep1, Itgß6), metabolic activity (Apom, Pgc1α) and immune modulation (Apelin, Icam1) compared to WT mice. We confirmed a new pro-inflammatory role of Apelin in AS mice and in cultured human GEC. Gene modulations were identified comparable to the biopsies from patients with AS and focal segmental glomerulosclerosis, possibly indicating that the same mechanisms apply to humans. We report the presence of two GEC subpopulations that differ between AS and healthy mice or humans. This finding paves the way to a better understanding of the pathogenic role of GEC in AS progression and could lead to novel therapeutic targets.


Asunto(s)
Células Endoteliales/citología , Glomérulos Renales/citología , Nefritis Hereditaria/patología , Adolescente , Adulto , Animales , Apelina/metabolismo , Biopsia , Separación Celular , Progresión de la Enfermedad , Citometría de Flujo , Perfilación de la Expresión Génica , Genes Reporteros , Glomeruloesclerosis Focal y Segmentaria/patología , Humanos , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Proteinuria/orina , Insuficiencia Renal Crónica/patología , Transducción de Señal , Transcriptoma , Adulto Joven
11.
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
12.
Thyroid ; 27(10): 1316-1322, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28847239

RESUMEN

BACKGROUND: Myopathic changes are commonly described in hypothyroid and hyperthyroid patients, including muscular atrophy and weakness. Satellite cells (SCs) play a major role in skeletal muscle maintenance and regeneration after injury. A mouse model of resistance to thyroid hormone-TRα1PV demonstrated impaired skeletal muscle regeneration after injury with significant reduction of SCs, suggesting that exhaustion of the SC pool contributes to the impaired regeneration. To test this hypothesis, SC activation and proliferation were analyzed in vivo in response to skeletal muscle injury and during aging. METHODS: SCs of TRα1PV male mice were analyzed four days after cardiotoxin-induced muscle injury, and they were compared to wild-type (WT) male animals. TRα-knockdown C2C12 myoblasts were injected into injured skeletal muscle, and four days after transplantation, the in vivo behavior was compared to control C2C12 myoblasts. Skeletal muscle regeneration was compared in younger and older TRα1PV and WT animals. RESULTS: The total number of SCs in skeletal muscle of TRα1PV mice was significantly lower than control, both before and shortly after muscle injury, with significant impairment of SC activation, consistent with SC pool exhaustion. TRα-knockdown myoblasts showed impaired in vivo proliferation and migration. TRα1PV mice had skeletal muscle loss and significant impairment in skeletal muscle regeneration with aging. This translated to a significant reduction of the SC pool with aging compared to WT mice. CONCLUSION: TRα plays an important role in the maintenance of the SC pool. Impaired skeletal muscle regeneration in TRα1PV mice is associated with insufficient SC activation and proliferation, as well as the progressive loss of the SC pool with aging. Regulation of the SC pool and SC proliferation provides a therapeutic target to enhance skeletal muscle regeneration and possibly slow age-associated sarcopenia.


Asunto(s)
Envejecimiento/metabolismo , Músculo Esquelético/metabolismo , Sarcopenia/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Receptores alfa de Hormona Tiroidea/metabolismo , Envejecimiento/genética , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Masculino , Ratones , Músculo Esquelético/lesiones , Sarcopenia/genética , Receptores alfa de Hormona Tiroidea/genética
13.
Stem Cells Transl Med ; 6(2): 419-433, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28191781

RESUMEN

Mature nephrons originate from a small population of uninduced nephrogenic progenitor cells (NPs) within the cap mesenchyme. These cells are characterized by the coexpression of SIX2 and CITED1. Many studies on mouse models as well as on human pluripotent stem cells have advanced our knowledge of NPs, but very little is known about this population in humans, since it is exhausted before birth and strategies for its direct isolation are still limited. Here we report an efficient protocol for direct isolation of human NPs without genetic manipulation or stepwise induction procedures. With the use of RNA-labeling probes, we isolated SIX2+ CITED1+ cells from human fetal kidney for the first time. We confirmed their nephrogenic state by gene profiling and evaluated their nephrogenic capabilities in giving rise to mature renal cells. We also evaluated the ability to culture these cells without complete loss of SIX2 and CITED1 expression over time. In addition to defining the gene profile of human NPs, this in vitro system facilitates studies of human renal development and provides a novel tool for renal regeneration and bioengineering purposes. Stem Cells Translational Medicine 2017;6:419-433.


Asunto(s)
Separación Celular/métodos , Nefronas/embriología , Células Madre/fisiología , Animales , Proteínas Reguladoras de la Apoptosis , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Morfogénesis , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fenotipo , Células Madre/metabolismo , Factores de Tiempo , Transactivadores , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma
14.
Endocrinology ; 157(1): 4-15, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26451739

RESUMEN

Thyroid hormone plays an essential role in myogenesis, the process required for skeletal muscle development and repair, although the mechanisms have not been established. Skeletal muscle develops from the fusion of precursor myoblasts into myofibers. We have used the C2C12 skeletal muscle myoblast cell line, primary myoblasts, and mouse models of resistance to thyroid hormone (RTH) α and ß, to determine the role of thyroid hormone in the regulation of myoblast differentiation. T3, which activates thyroid hormone receptor (TR) α and ß, increased myoblast differentiation whereas GC1, a selective TRß agonist, was minimally effective. Genetic approaches confirmed that TRα plays an important role in normal myoblast proliferation and differentiation and acts through the Wnt/ß-catenin signaling pathway. Myoblasts with TRα knockdown, or derived from RTH-TRα PV (a frame-shift mutation) mice, displayed reduced proliferation and myogenic differentiation. Moreover, skeletal muscle from the TRα1PV mutant mouse had impaired in vivo regeneration after injury. RTH-TRß PV mutant mouse model skeletal muscle and derived primary myoblasts did not have altered proliferation, myogenic differentiation, or response to injury when compared with control. In conclusion, TRα plays an essential role in myoblast homeostasis and provides a potential therapeutic target to enhance skeletal muscle regeneration.


Asunto(s)
Desarrollo de Músculos , Músculo Esquelético/fisiología , Mioblastos Esqueléticos/citología , Regeneración , Receptores alfa de Hormona Tiroidea/agonistas , Triyodotironina/metabolismo , Acetatos/farmacología , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Resistencia a Medicamentos , Mutación del Sistema de Lectura , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Desarrollo de Músculos/efectos de los fármacos , Músculo Esquelético/citología , Músculo Esquelético/lesiones , Mioblastos Esqueléticos/efectos de los fármacos , Mioblastos Esqueléticos/metabolismo , Fenoles/farmacología , Interferencia de ARN , Receptores alfa de Hormona Tiroidea/antagonistas & inhibidores , Receptores alfa de Hormona Tiroidea/genética , Receptores alfa de Hormona Tiroidea/metabolismo , Receptores beta de Hormona Tiroidea/agonistas , Receptores beta de Hormona Tiroidea/genética , Receptores beta de Hormona Tiroidea/metabolismo , Triyodotironina/análogos & derivados , Triyodotironina/farmacología , Vía de Señalización Wnt/efectos de los fármacos
15.
Tissue Eng Part B Rev ; 22(3): 183-92, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26653996

RESUMEN

Over the past years, extracellular matrix (ECM) obtained from whole organ decellularization has been investigated as a platform for organ engineering. The ECM is composed of fibrous and nonfibrous molecules providing structural and biochemical support to the surrounding cells. Multiple decellularization techniques, including ours, have been optimized to maintain the composition, microstructure, and biomechanical properties of the native renal ECM that are difficult to obtain during the generation of synthetic substrates. There are evidences suggesting that in vivo implanted renal ECM has the capacity to induce formation of vasculature-like structures, but long-term in vivo transplantation and filtration activity by these tissue-engineered constructs have not been investigated or reported. Therefore, even if the process of renal decellularization is possible, the repopulation of the renal matrix with functional renal cell types is still very challenging. This review aims to summarize the current reports on kidney tissue engineering with the use of decellularized matrices and addresses the challenges in creating functional kidney units. Finally, this review discusses how future studies investigating cell-matrix interaction may aid the generation of a functional renal unit that would be transplantable into patients one day.


Asunto(s)
Riñón , Matriz Extracelular , Humanos , Medicina Regenerativa , Ingeniería de Tejidos , Andamios del Tejido
16.
Cell Metab ; 22(1): 86-99, 2015 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-26094889

RESUMEN

Prolonged fasting (PF) promotes stress resistance, but its effects on longevity are poorly understood. We show that alternating PF and nutrient-rich medium extended yeast lifespan independently of established pro-longevity genes. In mice, 4 days of a diet that mimics fasting (FMD), developed to minimize the burden of PF, decreased the size of multiple organs/systems, an effect followed upon re-feeding by an elevated number of progenitor and stem cells and regeneration. Bi-monthly FMD cycles started at middle age extended longevity, lowered visceral fat, reduced cancer incidence and skin lesions, rejuvenated the immune system, and retarded bone mineral density loss. In old mice, FMD cycles promoted hippocampal neurogenesis, lowered IGF-1 levels and PKA activity, elevated NeuroD1, and improved cognitive performance. In a pilot clinical trial, three FMD cycles decreased risk factors/biomarkers for aging, diabetes, cardiovascular disease, and cancer without major adverse effects, providing support for the use of FMDs to promote healthspan.


Asunto(s)
Cognición , Ayuno , Longevidad , Grasa Abdominal/metabolismo , Adulto , Anciano , Envejecimiento , Animales , Peso Corporal , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/dietoterapia , Enfermedades Cardiovasculares/metabolismo , Dieta , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Neoplasias/dietoterapia , Neoplasias/metabolismo , Neoplasias/patología , Neurogénesis , Proyectos Piloto , Desempeño Psicomotor , Regeneración , Saccharomyces cerevisiae/citología , Adulto Joven
17.
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
18.
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
19.
PLoS One ; 8(8): e71679, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23967234

RESUMEN

The potential for amniotic fluid stem cell (AFSC) treatment to inhibit the progression of fibrotic lung injury has not been described. We have previously demonstrated that AFSC can attenuate both acute and chronic-fibrotic kidney injury through modification of the cytokine environment. Fibrotic lung injury, such as in Idiopathic Pulmonary Fibrosis (IPF), is mediated through pro-fibrotic and pro-inflammatory cytokine activity. Thus, we hypothesized that AFSC treatment might inhibit the progression of bleomycin-induced pulmonary fibrosis through cytokine modulation. In particular, we aimed to investigate the effect of AFSC treatment on the modulation of the pro-fibrotic cytokine CCL2, which is increased in human IPF patients and is correlated with poor prognoses, advanced disease states and worse fibrotic outcomes. The impacts of intravenous murine AFSC given at acute (day 0) or chronic (day 14) intervention time-points after bleomycin injury were analyzed at either day 3 or day 28 post-injury. Murine AFSC treatment at either day 0 or day 14 post-bleomycin injury significantly inhibited collagen deposition and preserved pulmonary function. CCL2 expression increased in bleomycin-injured bronchoalveolar lavage (BAL), but significantly decreased following AFSC treatment at either day 0 or at day 14. AFSC were observed to localize within fibrotic lesions in the lung, showing preferential targeting of AFSC to the area of fibrosis. We also observed that MMP-2 was transiently increased in BAL following AFSC treatment. Increased MMP-2 activity was further associated with cleavage of CCL2, rendering it a putative antagonist for CCL2/CCR2 signaling, which we surmise is a potential mechanism for CCL2 reduction in BAL following AFSC treatment. Based on this data, we concluded that AFSC have the potential to inhibit the development or progression of fibrosis in a bleomycin injury model during both acute and chronic remodeling events.


Asunto(s)
Líquido Amniótico/citología , Lavado Broncoalveolar , Quimiocina CCL2/metabolismo , Fibrosis Pulmonar/metabolismo , Células Madre/metabolismo , Células Epiteliales Alveolares/metabolismo , Animales , Bleomicina/efectos adversos , Quimiotaxis/inmunología , Técnicas de Cocultivo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Feto , Humanos , Mediadores de Inflamación/metabolismo , Pulmón/metabolismo , Pulmón/patología , Pulmón/fisiopatología , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Modelos Biológicos , Embarazo , Proteolisis , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/terapia , Trasplante de Células Madre , Células Madre/inmunología , Factores de Tiempo
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA