Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Renal Physiol ; 326(1): F120-F134, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37855038

RESUMEN

As life expectancy continues to rise, age-related diseases are becoming more prevalent. For example, proteinuric glomerular diseases typified by podocyte injury have worse outcomes in the elderly compared with young patients. However, the reasons are not well understood. We hypothesized that injury to nonaged podocytes induces senescence, which in turn augments their aging processes. In primary cultured human podocytes, injury induced by a cytopathic antipodocyte antibody, adriamycin, or puromycin aminonucleoside increased the senescence-related genes CDKN2A (p16INK4a/p14ARF), CDKN2D (p19INK4d), and CDKN1A (p21). Podocyte injury in human kidney organoids was accompanied by increased expression of CDKN2A, CDKN2D, and CDKN1A. In young mice, experimental focal segmental glomerulosclerosis (FSGS) induced by adriamycin and antipodocyte antibody increased the glomerular expression of p16, p21, and senescence-associated ß-galactosidase (SA-ß-gal). To assess the long-term effects of early podocyte injury-induced senescence, we temporally followed young mice with experimental FSGS through adulthood (12 m of age) and middle age (18 m of age). p16 and Sudan black staining were higher at middle age in mice with earlier FSGS compared with age-matched mice that did not get FSGS when young. This was accompanied by lower podocyte density, reduced canonical podocyte protein expression, and increased glomerular scarring. These results are consistent with injury-induced senescence in young podocytes, leading to increased senescence of podocytes by middle age accompanied by lower podocyte lifespan and health span.NEW & NOTEWORTHY Glomerular function is decreased by aging. However, little is known about the molecular mechanisms involved in age-related glomerular changes and which factors could contribute to a worse glomerular aging process. Here, we reported that podocyte injury in young mice and culture podocytes induced senescence, a marker of aging, and accelerates glomerular aging when compared with healthy aging mice.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria , Enfermedades Renales , Podocitos , Persona de Mediana Edad , Humanos , Ratones , Animales , Anciano , Podocitos/metabolismo , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Glomérulos Renales/metabolismo , Enfermedades Renales/metabolismo , Envejecimiento , Doxorrubicina/toxicidad , Doxorrubicina/metabolismo
2.
Kidney Int ; 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38901605

RESUMEN

Vascularization plays a critical role in organ maturation and cell-type development. Drug discovery, organ mimicry, and ultimately transplantation hinge on achieving robust vascularization of in vitro engineered organs. Here, focusing on human kidney organoids, we overcame this hurdle by combining a human induced pluripotent stem cell (iPSC) line containing an inducible ETS translocation variant 2 (ETV2) (a transcription factor playing a role in endothelial cell development) that directs endothelial differentiation in vitro, with a non-transgenic iPSC line in suspension organoid culture. The resulting human kidney organoids show extensive endothelialization with a cellular identity most closely related to human kidney endothelia. Endothelialized kidney organoids also show increased maturation of nephron structures, an associated fenestrated endothelium with de novo formation of glomerular and venous subtypes, and the emergence of drug-responsive renin expressing cells. The creation of an engineered vascular niche capable of improving kidney organoid maturation and cell type complexity is a significant step forward in the path to clinical translation. Thus, incorporation of an engineered endothelial niche into a previously published kidney organoid protocol allowed the orthogonal differentiation of endothelial and parenchymal cell types, demonstrating the potential for applicability to other basic and translational organoid studies.

3.
Am J Physiol Renal Physiol ; 323(4): F479-F491, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35979965

RESUMEN

Kidney organoids derived from human or rodent pluripotent stem cells have glomerular structures and differentiated/polarized nephron segments. Although there is an increasing understanding of the patterns of expression of transcripts and proteins within kidney organoids, there is a paucity of data regarding functional protein expression, in particular on transporters that mediate the vectorial transport of solutes. Using cells derived from kidney organoids, we examined the functional expression of key ion channels that are expressed in distal nephron segments: the large-conductance Ca2+-activated K+ (BKCa) channel, the renal outer medullary K+ (ROMK, Kir1.1) channel, and the epithelial Na+ channel (ENaC). RNA-sequencing analyses showed that genes encoding the pore-forming subunits of these transporters, and for BKCa channels, key accessory subunits, are expressed in kidney organoids. Expression and localization of selected ion channels was confirmed by immunofluorescence microscopy and immunoblot analysis. Electrophysiological analysis showed that BKCa and ROMK channels are expressed in different cell populations. These two cell populations also expressed other unidentified Ba2+-sensitive K+ channels. BKCa expression was confirmed at a single channel level, based on its high conductance and voltage dependence of activation. We also found a population of cells expressing amiloride-sensitive ENaC currents. In summary, our results show that human kidney organoids functionally produce key distal nephron K+ and Na+ channels.NEW & NOTEWORTHY Our results show that human kidney organoids express key K+ and Na+ channels that are expressed on the apical membranes of cells in the aldosterone-sensitive distal nephron, including the large-conductance Ca2+-activated K+ channel, renal outer medullary K+ channel, and epithelial Na+ channel.


Asunto(s)
Células Madre Pluripotentes Inducidas , Canales de Potasio de Rectificación Interna , Aldosterona/metabolismo , Amilorida/farmacología , Canales Epiteliales de Sodio/genética , Canales Epiteliales de Sodio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Riñón/metabolismo , Organoides/metabolismo , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo , ARN/metabolismo , Sodio/metabolismo
4.
Kidney Int ; 101(5): 845-853, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35276204

RESUMEN

Acute kidney injury impacts âˆ¼13.3 million individuals and causes âˆ¼1.7 million deaths per year globally. Numerous injury pathways contribute to acute kidney injury, including cell cycle arrest, senescence, inflammation, mitochondrial dysfunction, and endothelial injury and dysfunction, and can lead to chronic inflammation and fibrosis. However, factors enabling productive repair versus nonproductive, persistent injury states remain less understood. The (Re)Building a Kidney (RBK) consortium is a National Institute of Diabetes and Digestive and Kidney Diseases consortium focused on both endogenous kidney repair mechanisms and the generation of new kidney tissue. This short review provides an update on RBK studies of endogenous nephron repair, addressing the following questions: (i) What is productive nephron repair? (ii) What are the cellular sources and drivers of repair? and (iii) How do RBK studies promote development of therapeutics? Also, we provide a guide to RBK's open access data hub for accessing, downloading, and further analyzing data sets.


Asunto(s)
Lesión Renal Aguda , Riñón , Lesión Renal Aguda/patología , Femenino , Fibrosis , Humanos , Inflamación/patología , Riñón/patología , Masculino , National Institute of Diabetes and Digestive and Kidney Diseases (U.S.) , Regeneración , Estados Unidos
5.
Development ; 146(8)2019 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-31036548

RESUMEN

Zebrafish kidneys use resident kidney stem cells to replace damaged tubules with new nephrons: the filtration units of the kidney. What stimulates kidney progenitor cells to form new nephrons is not known. Here, we show that wnt9a and wnt9b are induced in the injured kidney at sites where frizzled9b- and lef1-expressing progenitor cells form new nephrons. New nephron aggregates are patterned by Wnt signaling, with high canonical Wnt-signaling cells forming a single cell thick rosette that demarcates: domains of cell proliferation in the elongating nephron; and tubule fusion where the new nephron plumbs into the distal tubule and establishes blood filtrate drainage. Pharmacological blockade of canonical Wnt signaling inhibited new nephron formation after injury by inhibiting cell proliferation, and resulted in loss of polarized rosette structures in the aggregates. Mutation in frizzled9b reduced total kidney nephron number, caused defects in tubule morphology and reduced regeneration of new nephrons after injury. Our results demonstrate an essential role for Wnt/frizzled signaling in adult zebrafish kidney development and regeneration, highlighting conserved mechanisms underlying both mammalian kidney development and kidney stem cell-directed neonephrogenesis in zebrafish.


Asunto(s)
Riñón/citología , Riñón/metabolismo , Nefronas/citología , Nefronas/metabolismo , Vía de Señalización Wnt/fisiología , Animales , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Regeneración/fisiología , Vía de Señalización Wnt/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
6.
J Am Soc Nephrol ; 31(1): 67-83, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31676724

RESUMEN

BACKGROUND: Lowe syndrome (LS) is an X-linked recessive disorder caused by mutations in OCRL, which encodes the enzyme OCRL. Symptoms of LS include proximal tubule (PT) dysfunction typically characterized by low molecular weight proteinuria, renal tubular acidosis (RTA), aminoaciduria, and hypercalciuria. How mutant OCRL causes these symptoms isn't clear. METHODS: We examined the effect of deleting OCRL on endocytic traffic and cell division in newly created human PT CRISPR/Cas9 OCRL knockout cells, multiple PT cell lines treated with OCRL-targeting siRNA, and in orcl-mutant zebrafish. RESULTS: OCRL-depleted human cells proliferated more slowly and about 10% of them were multinucleated compared with fewer than 2% of matched control cells. Heterologous expression of wild-type, but not phosphatase-deficient, OCRL prevented the accumulation of multinucleated cells after acute knockdown of OCRL but could not rescue the phenotype in stably edited knockout cell lines. Mathematic modeling confirmed that reduced PT length can account for the urinary excretion profile in LS. Both ocrl mutant zebrafish and zebrafish injected with ocrl morpholino showed truncated expression of megalin along the pronephric kidney, consistent with a shortened S1 segment. CONCLUSIONS: Our data suggest a unifying model to explain how loss of OCRL results in tubular proteinuria as well as the other commonly observed renal manifestations of LS. We hypothesize that defective cell division during kidney development and/or repair compromises PT length and impairs kidney function in LS patients.


Asunto(s)
Túbulos Renales Proximales/fisiología , Síndrome Oculocerebrorrenal/metabolismo , Proteínas/metabolismo , Línea Celular , Humanos , Modelos Biológicos , Mutación , Síndrome Oculocerebrorrenal/genética , Monoéster Fosfórico Hidrolasas/genética
7.
J Biol Chem ; 294(28): 10773-10788, 2019 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-31152064

RESUMEN

Nephrin is an immunoglobulin-type cell-adhesion molecule with a key role in the glomerular interpodocyte slit diaphragm. Mutations in the nephrin gene are associated with defects in the slit diaphragm, leading to early-onset nephrotic syndrome, typically resistant to treatment. Although the endocytic trafficking of nephrin is essential for the assembly of the slit diaphragm, nephrin's specific endocytic motifs remain unknown. To search for endocytic motifs, here we performed a multisequence alignment of nephrin and identified a canonical YXXØ-type motif, Y1139RSL, in the nephrin cytoplasmic tail, expressed only in primates. Using site-directed mutagenesis, various biochemical methods, single-plane illumination microscopy, a human podocyte line, and a human nephrin-expressing zebrafish model, we found that Y1139RSL is a novel endocytic motif and a structural element for clathrin-mediated nephrin endocytosis that functions as a phosphorylation-sensitive signal. We observed that Y1139RSL motif-mediated endocytosis helps to localize nephrin to specialized plasma membrane domains in podocytes and is essential for normal foot process organization into a functional slit diaphragm between neighboring foot processes in zebrafish. The importance of nephrin Y1139RSL for healthy podocyte development was supported by population-level analyses of genetic variations at this motif, revealing that such variations are very rare, suggesting that mutations in this motif have autosomal-recessive negative effects on kidney health. These findings expand our understanding of the mechanism underlying nephrin endocytosis and may lead to improved diagnostic tools or therapeutic strategies for managing early-onset, treatment-resistant nephrotic syndrome.


Asunto(s)
Glomérulos Renales/metabolismo , Proteínas de la Membrana/metabolismo , Secuencias de Aminoácidos , Animales , Línea Celular , Membrana Celular/metabolismo , Movimiento Celular , Clatrina/metabolismo , Embrión no Mamífero/metabolismo , Endocitosis , Humanos , Glomérulos Renales/ultraestructura , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Morfolinos/metabolismo , Mutagénesis Sitio-Dirigida , Fosforilación , Podocitos/citología , Podocitos/metabolismo , Pez Cebra/crecimiento & desarrollo
8.
Pediatr Nephrol ; 34(4): 561-569, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-29383444

RESUMEN

Acute kidney injury (AKI) is defined by a rapid decline in renal function. Regardless of the initial cause of injury, the influx of immune cells is a common theme during AKI. While an inflammatory response is critical for the initial control of injury, a prolonged response can negatively affect tissue repair. In this review, we focus on the role of macrophages, from early inflammation to resolution, during AKI. These cells serve as the innate defense system by phagocytosing cellular debris and pathogenic molecules and bridge communication with the adaptive immune system by acting as antigen-presenting cells and secreting cytokines. While many immune cells function to initiate inflammation, macrophages play a complex role throughout AKI. This complexity is driven by their functional plasticity: the ability to polarize from a "pro-inflammatory" phenotype to a "pro-reparative" phenotype. Importantly, experimental and translational studies indicate that macrophage polarization opens the possibility to generate novel therapeutics to promote repair during AKI. A thorough understanding of the biological roles these phagocytes play during both injury and repair is necessary to understand the limitations while furthering the therapeutic application.


Asunto(s)
Lesión Renal Aguda/patología , Plasticidad de la Célula , Riñón/patología , Activación de Macrófagos , Macrófagos/patología , Regeneración , Lesión Renal Aguda/inmunología , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/fisiopatología , Inmunidad Adaptativa , Animales , Citocinas/metabolismo , Humanos , Inmunidad Innata , Mediadores de Inflamación/metabolismo , Riñón/inmunología , Riñón/metabolismo , Riñón/fisiopatología , Macrófagos/inmunología , Macrófagos/metabolismo , Fenotipo , Transducción de Señal
9.
Dev Biol ; 425(2): 130-141, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28359809

RESUMEN

During zebrafish embryogenesis the pronephric kidney arises from a small population of posterior mesoderm cells that then undergo expansion during early stages of renal organogenesis. While wnt8 is required for posterior mesoderm formation during gastrulation, it is also transiently expressed in the post-gastrula embryo in the intermediate mesoderm, the precursor to the pronephros and some blood/vascular lineages. Here, we show that knockdown of wnt8a, using a low dose of morpholino that does not disrupt early mesoderm patterning, reduces the number of kidney and blood cells. For the kidney, wnt8a deficiency decreases renal progenitor growth during early somitogenesis, as detected by EdU incorporation, but has no effect on apoptosis. The depletion of the renal progenitor pool in wnt8a knockdown embryos leads to cellular deficits in the pronephros at 24 hpf that are characterised by a shortened distal-most segment and stretched proximal tubule cells. A pulse of the canonical Wnt pathway agonist BIO during early somitogenesis is sufficient to rescue the size of the renal progenitor pool while longer treatment expands the number of kidney cells. Taken together, these observations indicate that Wnt8, in addition to its well-established role in posterior mesoderm patterning, also plays a later role as a factor that expands the renal progenitor pool prior to kidney morphogenesis.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Embrión no Mamífero/citología , Riñón/citología , Riñón/embriología , Células Madre/citología , Proteínas Wnt/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Animales , Apoptosis/efectos de los fármacos , Células Sanguíneas/citología , Células Sanguíneas/efectos de los fármacos , Tipificación del Cuerpo/efectos de los fármacos , Recuento de Células , Proliferación Celular/efectos de los fármacos , Embrión no Mamífero/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Indoles/farmacología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/patología , Mesodermo/efectos de los fármacos , Mesodermo/embriología , Mesodermo/metabolismo , Morfolinos/farmacología , Oximas/farmacología , Pronefro/citología , Pronefro/embriología , Células Madre/efectos de los fármacos , Células Madre/metabolismo
10.
Am J Physiol Renal Physiol ; 315(2): F291-F299, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29537312

RESUMEN

Sepsis-associated acute kidney injury (S-AKI) independently predicts mortality among critically ill patients. The role of innate immunity in this process is unclear, and there is an unmet need for S-AKI models to delineate the pathophysiological response. Mammals and zebrafish ( Danio rerio) share a conserved nephron structure and homologous innate immune systems, making the latter suitable for S-AKI research. We introduced Edwardsiella tarda to the zebrafish. Systemic E. tarda bacteremia resulted in sustained bacterial infection and dose-dependent mortality. A systemic immune reaction was characterized by increased mRNA expressions of il1b, tnfa, tgfb1a, and cxcl8-l1 ( P < 0.0001, P < 0.001, P < 0.001, and P < 0.01, respectively). Increase of host stress response genes ccnd1 and tp53 was observed at 24 h postinjection ( P < 0.0001 and P < 0.05, respectively). Moderate E. tarda infection induced zebrafish mortality of over 50% in larvae and 20% in adults, accompanied by pericardial edema in larvae and renal dysfunction in both larval and adult zebrafish. Expression of AKI markers insulin-like growth factor-binding protein-7 (IGFBP7), tissue inhibitor of metalloproteinases 2 (TIMP-2), and kidney injury molecule-1 (KIM-1) was found to be significantly increased in the septic animals at the transcription level ( P < 0.01, P < 0.05, and P < 0.05) and in nephric tubules compared with noninfected animals. In conclusion, we established a zebrafish model of S-AKI induced by E. tarda injection, with both larval and adult zebrafish showing nephron injury in the setting of infection.


Asunto(s)
Lesión Renal Aguda/microbiología , Edwardsiella tarda/patogenicidad , Infecciones por Enterobacteriaceae/microbiología , Nefronas/microbiología , Sepsis/microbiología , Pez Cebra , Lesión Renal Aguda/inmunología , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Edwardsiella tarda/inmunología , Infecciones por Enterobacteriaceae/inmunología , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Inmunidad Innata , Larva , Nefronas/inmunología , Nefronas/metabolismo , Nefronas/patología , Sepsis/inmunología , Sepsis/metabolismo , Sepsis/patología , Transducción de Señal , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
11.
FASEB J ; 31(8): 3540-3554, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28446592

RESUMEN

Low nephron endowment at birth has been associated with an increased risk for developing hypertension and chronic kidney disease. We demonstrated in an earlier study that conditional deletion of the microRNA (miRNA)-processing enzyme Dicer from nephron progenitors results in premature depletion of the progenitors and increased expression of the proapoptotic protein Bim (also known as Bcl-2L11). In this study, we generated a compound mouse model with conditional deletion of both Dicer and Bim, to determine the biologic significance of increased Bim expression in Dicer-deficient nephron progenitors. The loss of Bim partially restored the number of nephron progenitors and improved nephron formation. The number of progenitors undergoing apoptosis was significantly reduced in kidneys with loss of a single allele, or both alleles, of Bim compared to mutant kidneys. Furthermore, 2 miRNAs expressed in nephron progenitors (miR-17 and miR-106b) regulated Bim levels in vitro and in vivo Together, these data suggest that miRNA-mediated regulation of Bim controls nephron progenitor survival during nephrogenesis, as one potential means of regulating nephron endowment.-Cerqueira, D. M., Bodnar, A. J., Phua, Y. L., Freer, R., Hemker, S. L., Walensky, L. D., Hukriede, N. A., Ho, J. Bim gene dosage is critical in modulating nephron progenitor survival in the absence of microRNAs during kidney development.


Asunto(s)
Proteína 11 Similar a Bcl2/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Riñón/embriología , MicroARNs/metabolismo , Animales , Apoptosis/fisiología , Proteína 11 Similar a Bcl2/metabolismo , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Embrión no Mamífero , Eliminación de Gen , Dosificación de Gen/fisiología , Células HEK293 , Humanos , Ratones , Ratones Transgénicos , MicroARNs/genética , Nefronas/citología , Ribonucleasa III/genética , Ribonucleasa III/metabolismo , Células Madre/citología , Xenopus laevis/embriología
12.
Nature ; 470(7332): 95-100, 2011 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-21270795

RESUMEN

Loss of kidney function underlies many renal diseases. Mammals can partly repair their nephrons (the functional units of the kidney), but cannot form new ones. By contrast, fish add nephrons throughout their lifespan and regenerate nephrons de novo after injury, providing a model for understanding how mammalian renal regeneration may be therapeutically activated. Here we trace the source of new nephrons in the adult zebrafish to small cellular aggregates containing nephron progenitors. Transplantation of single aggregates comprising 10-30 cells is sufficient to engraft adults and generate multiple nephrons. Serial transplantation experiments to test self-renewal revealed that nephron progenitors are long-lived and possess significant replicative potential, consistent with stem-cell activity. Transplantation of mixed nephron progenitors tagged with either green or red fluorescent proteins yielded some mosaic nephrons, indicating that multiple nephron progenitors contribute to a single nephron. Consistent with this, live imaging of nephron formation in transparent larvae showed that nephrogenic aggregates form by the coalescence of multiple cells and then differentiate into nephrons. Taken together, these data demonstrate that the zebrafish kidney probably contains self-renewing nephron stem/progenitor cells. The identification of these cells paves the way to isolating or engineering the equivalent cells in mammals and developing novel renal regenerative therapies.


Asunto(s)
Riñón/citología , Riñón/crecimiento & desarrollo , Nefronas/citología , Regeneración/fisiología , Células Madre/citología , Pez Cebra/crecimiento & desarrollo , Envejecimiento/fisiología , Animales , Animales Modificados Genéticamente , Proliferación Celular , Riñón/lesiones , Riñón/metabolismo , Larva , Modelos Animales , Nefronas/crecimiento & desarrollo , Organogénesis , Trasplante de Células Madre
13.
J Am Soc Nephrol ; 27(2): 495-508, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26109319

RESUMEN

Retinoic acid (RA) has been used therapeutically to reduce injury and fibrosis in models of AKI, but little is known about the regulation of this pathway and what role it has in regulating injury and repair after AKI. In these studies, we show that RA signaling is activated in mouse and zebrafish models of AKI, and that these responses limit the extent of injury and promote normal repair. These effects were mediated through a novel mechanism by which RA signaling coordinated the dynamic equilibrium of inflammatory M1 spectrum versus alternatively activated M2 spectrum macrophages. Our data suggest that locally synthesized RA represses proinflammatory macrophages, thereby reducing macrophage-dependent injury post-AKI, and activates RA signaling in injured tubular epithelium, which in turn promotes alternatively activated M2 spectrum macrophages. Because RA signaling has an essential role in kidney development but is repressed in the adult, these findings provide evidence of an embryonic signaling pathway that is reactivated after AKI and involved in reducing injury and enhancing repair.


Asunto(s)
Lesión Renal Aguda/etiología , Macrófagos/fisiología , Transducción de Señal , Tretinoina/fisiología , Animales , Masculino , Ratones , Ratones Endogámicos BALB C
14.
J Biol Chem ; 290(40): 24278-93, 2015 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-26269593

RESUMEN

DNA polymerase ν (POLN) is one of 16 DNA polymerases encoded in vertebrate genomes. It is important to determine its gene expression patterns, biological roles, and biochemical activities. By quantitative analysis of mRNA expression, we found that POLN from the zebrafish Danio rerio is expressed predominantly in testis. POLN is not detectably expressed in zebrafish embryos or in mouse embryonic stem cells. Consistent with this, injection of POLN-specific morpholino antisense oligonucleotides did not interfere with zebrafish embryonic development. Analysis of transcripts revealed that vertebrate POLN has an unusual gene expression arrangement, sharing a first exon with HAUS3, the gene encoding augmin-like complex subunit 3. HAUS3 is broadly expressed in embryonic and adult tissues, in contrast to POLN. Differential expression of POLN and HAUS3 appears to arise by alternate splicing of transcripts in mammalian cells and zebrafish. When POLN was ectopically overexpressed in human cells, it specifically coimmunoprecipitated with the homologous recombination factors BRCA1 and FANCJ, but not with previously suggested interaction partners (HELQ and members of the Fanconi anemia core complex). Purified zebrafish POLN protein is capable of thymine glycol bypass and strand displacement, with activity dependent on a basic amino acid residue known to stabilize the primer-template. These properties are conserved with the human enzyme. Although the physiological function of pol ν remains to be clarified, this study uncovers distinctive aspects of its expression control and evolutionarily conserved properties of this DNA polymerase.


Asunto(s)
ADN Polimerasa Dirigida por ADN/metabolismo , Regulación de la Expresión Génica , Testículo/metabolismo , Proteínas de Pez Cebra/metabolismo , Empalme Alternativo , Animales , Proteína BRCA1/metabolismo , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , ADN/química , Daño del ADN , Exones , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Orden Génico , Genes Sobrepuestos , Células HEK293 , Células HeLa , Humanos , Masculino , Ratones , Recombinación Genética , Transgenes , Pez Cebra
15.
Am J Physiol Renal Physiol ; 310(8): F705-F716, 2016 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-26661656

RESUMEN

No therapies have been shown to accelerate recovery or prevent fibrosis after acute kidney injury (AKI). In part, this is because most therapeutic candidates have to be given at the time of injury and the diagnosis of AKI is usually made too late for drugs to be efficacious. Strategies to enhance post-AKI repair represent an attractive approach to address this. Using a phenotypic screen in zebrafish, we identified 4-(phenylthio)butanoic acid (PTBA), which promotes proliferation of embryonic kidney progenitor cells (EKPCs), and the PTBA methyl ester UPHD25, which also increases postinjury repair in ischemia-reperfusion and aristolochic acid-induced AKI in mice. In these studies, a new panel of PTBA analogs was evaluated. Initial screening was performed in zebrafish EKPC assays followed by survival assays in a gentamicin-induced AKI larvae zebrafish model. Using this approach, we identified UPHD186, which in contrast to UPHD25, accelerates recovery and reduces fibrosis when administered several days after ischemia-reperfusion AKI and reduces fibrosis after unilateral ureteric obstruction in mice. UPHD25 and 186 are efficiently metabolized to the active analog PTBA in liver and kidney microsome assays, indicating both compounds may act as PTBA prodrugs in vivo. UPHD186 persists longer in the circulation than UPHD25, suggesting that sustained levels of UPHD186 may increase efficacy by acting as a reservoir for renal metabolism to PTBA. These findings validate use of zebrafish EKPC and AKI assays as a drug discovery strategy for molecules that reduce fibrosis in multiple AKI models and can be administered days after initiation of injury.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Butiratos/uso terapéutico , Riñón/efectos de los fármacos , Sulfuros/uso terapéutico , Lesión Renal Aguda/patología , Animales , Butiratos/farmacología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Fibrosis/tratamiento farmacológico , Fibrosis/patología , Riñón/patología , Masculino , Ratones , Sulfuros/farmacología , Pez Cebra
16.
PLoS Biol ; 11(11): e1001720, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24302887

RESUMEN

Planar cell polarity (PCP) regulates cell alignment required for collective cell movement during embryonic development. This requires PCP/PCP effector proteins, some of which also play essential roles in ciliogenesis, highlighting the long-standing question of the role of the cilium in PCP. Wdpcp, a PCP effector, was recently shown to regulate both ciliogenesis and collective cell movement, but the underlying mechanism is unknown. Here we show Wdpcp can regulate PCP by direct modulation of the actin cytoskeleton. These studies were made possible by recovery of a Wdpcp mutant mouse model. Wdpcp-deficient mice exhibit phenotypes reminiscent of Bardet-Biedl/Meckel-Gruber ciliopathy syndromes, including cardiac outflow tract and cochlea defects associated with PCP perturbation. We observed Wdpcp is localized to the transition zone, and in Wdpcp-deficient cells, Sept2, Nphp1, and Mks1 were lost from the transition zone, indicating Wdpcp is required for recruitment of proteins essential for ciliogenesis. Wdpcp is also found in the cytoplasm, where it is localized in the actin cytoskeleton and in focal adhesions. Wdpcp interacts with Sept2 and is colocalized with Sept2 in actin filaments, but in Wdpcp-deficient cells, Sept2 was lost from the actin cytoskeleton, suggesting Wdpcp is required for Sept2 recruitment to actin filaments. Significantly, organization of the actin filaments and focal contacts were markedly changed in Wdpcp-deficient cells. This was associated with decreased membrane ruffling, failure to establish cell polarity, and loss of directional cell migration. These results suggest the PCP defects in Wdpcp mutants are not caused by loss of cilia, but by direct disruption of the actin cytoskeleton. Consistent with this, Wdpcp mutant cochlea has normal kinocilia and yet exhibits PCP defects. Together, these findings provide the first evidence, to our knowledge, that a PCP component required for ciliogenesis can directly modulate the actin cytoskeleton to regulate cell polarity and directional cell migration.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Movimiento Celular , Cilios/fisiología , Proteínas del Citoesqueleto/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Polaridad Celular , Células Cultivadas , Análisis Mutacional de ADN , Adhesiones Focales/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple , Transporte de Proteínas , Septinas/metabolismo , Imagen de Lapso de Tiempo , Vía de Señalización Wnt , Pez Cebra
17.
J Am Soc Nephrol ; 25(11): 2539-45, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24722440

RESUMEN

Odd-skipped related 1 (Osr1) encodes a zinc finger transcription factor required for kidney development. Osr1 deficiency in mice results in metanephric kidney agenesis, whereas knockdown or mutation studies in zebrafish revealed that pronephric nephrons require osr1 for proximal tubule and podocyte development. osr1-deficient pronephric podocyte progenitors express the Wilms' tumor suppressor wt1a but do not undergo glomerular morphogenesis or express the foot process junctional markers nephrin and podocin. The function of osr1 in podocyte differentiation remains unclear, however. Here, we found by double fluorescence in situ hybridization that podocyte progenitors coexpress osr1 and wt1a. Knockdown of wt1a disrupted podocyte differentiation and prevented expression of osr1. Blocking retinoic acid signaling, which regulates wt1a, also prevented osr1 expression in podocyte progenitors. Furthermore, unlike the osr1-deficient proximal tubule phenotype, which can be rescued by manipulation of endoderm development, podocyte differentiation was not affected by altered endoderm development, as assessed by nephrin and podocin expression in double osr1/sox32-deficient embryos. These results suggest a different, possibly cell- autonomous requirement for osr1 in podocyte differentiation downstream of wt1a. Indeed, osr1-deficient embryos did not exhibit podocyte progenitor expression of the transcription factor lhx1a, and forced expression of activated forms of the lhx1a gene product rescued nephrin expression in osr1-deficient podocytes. Our results place osr1 in a framework of transcriptional regulators that control the expression of podocin and nephrin and thereby mediate podocyte differentiation.


Asunto(s)
Podocitos/fisiología , Factores de Transcripción/fisiología , Proteínas WT1/fisiología , Proteínas de Pez Cebra/fisiología , Animales , Diferenciación Celular/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Túbulos Renales/citología , Túbulos Renales/embriología , Túbulos Renales/fisiología , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/fisiología , Masculino , Podocitos/citología , Células Madre/citología , Células Madre/fisiología , Factores de Transcripción/genética , Transcripción Genética/fisiología , Proteínas WT1/genética , Pez Cebra , Proteínas de Pez Cebra/genética
18.
Traffic ; 13(9): 1295-305, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22680056

RESUMEN

Lowe syndrome is an X-linked disorder characterized by cataracts at birth, mental retardation and progressive renal malfunction that results from loss of function of the OCRL1 (oculocerebrorenal syndrome of Lowe) protein. OCRL1 is a lipid phosphatase that converts phosphatidylinositol 4,5-bisphosphate to phosphatidylinositol 4-phosphate. The renal pathogenesis of Lowe syndrome patients has been suggested to result from alterations in membrane trafficking, but this cannot fully explain the disease progression. We found that knockdown of OCRL1 in zebrafish caused developmental defects consistent with disruption of ciliary function, including body axis curvature, pericardial edema, hydrocephaly and impaired renal clearance. In addition, cilia in the proximal tubule of the zebrafish pronephric kidney were longer in ocrl morphant embryos. We also found that knockdown of OCRL1 in polarized renal epithelial cells caused elongation of the primary cilium and disrupted formation of cysts in three-dimensional cultures. Calcium release in response to ATP was blunted in OCRL1 knockdown cells, suggesting changes in signaling that could lead to altered cell function. Our results suggest a new role for OCRL1 in renal epithelial cell function that could contribute to the pathogenesis of Lowe syndrome.


Asunto(s)
Túbulos Renales Proximales/ultraestructura , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas de Pez Cebra/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Tipificación del Cuerpo , Señalización del Calcio , Línea Celular , Cilios/metabolismo , Cilios/ultraestructura , Perros , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Túbulos Renales Proximales/citología , Organogénesis , Monoéster Fosfórico Hidrolasas/genética , ARN Interferente Pequeño , Pez Cebra/embriología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
19.
Am J Physiol Renal Physiol ; 306(5): F496-504, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24370591

RESUMEN

Phenylthiobutanoic acids (PTBAs) are a new class of histone deacetylase (HDAC) inhibitors that accelerate recovery and reduce postinjury fibrosis after ischemia-reperfusion-induced acute kidney injury. However, unlike the more common scenario in which patients present with protracted and less clearly defined onset of renal injury, this model of acute kidney injury gives rise to a clearly defined injury that begins to resolve over a short period of time. In these studies, we show for the first time that treatment with the PTBA analog methyl-4-(phenylthio)butanoate (M4PTB) accelerates recovery and reduces postinjury fibrosis in a progressive model of acute kidney injury and renal fibrosis that occurs after aristolochic acid injection in mice. These effects are apparent when M4PTB treatment is delayed 4 days after the initiating injury and are associated with increased proliferation and decreased G2/M arrest of regenerating renal tubular epithelial cells. In addition, there is reduced peritubular macrophage infiltration and decreased expression of the macrophage chemokines CX3Cl1 and CCL2. Since macrophage infiltration plays a role in promoting kidney injury, and since renal tubular epithelial cells show defective repair and a marked increase in maladaptive G2/M arrest after aristolochic acid injury, these findings suggest M4PTB may be particularly beneficial in reducing injury and enhancing intrinsic cellular repair even when administered days after aristolochic acid ingestion.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Butiratos/farmacología , Sulfuros/farmacología , Lesión Renal Aguda/inducido químicamente , Animales , Ácidos Aristolóquicos/farmacología , Butiratos/análisis , Modelos Animales de Enfermedad , Fibrosis/tratamiento farmacológico , Fibrosis/prevención & control , Inhibidores de Histona Desacetilasas/farmacología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/metabolismo , Ratones , Ratones Biozzi , Daño por Reperfusión/inducido químicamente , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Sulfuros/análisis
20.
J Cell Sci ; 125(Pt 22): 5546-54, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22976307

RESUMEN

Kidney function requires the appropriate distribution of membrane proteins between the apical and basolateral surfaces along the kidney tubule. Further, the absolute amount of a protein at the cell surface versus intracellular compartments must be attuned to specific physiological needs. Endolyn (CD164) is a transmembrane protein that is expressed at the brush border and in apical endosomes of the proximal convoluted tubule and in lysosomes of more distal segments of the kidney. Endolyn has been shown to regulate CXCR4 signaling in hematopoietic precursor cells and myoblasts; however, little is known about endolyn function in the adult or developing kidney. Here we identify endolyn as a gene important for zebrafish pronephric kidney function. Zebrafish endolyn lacks the N-terminal mucin-like domain of the mammalian protein, but is otherwise highly conserved. Using in situ hybridization we show that endolyn is expressed early during development in zebrafish brain, eye, gut and pronephric kidney. Embryos injected with a translation-inhibiting morpholino oligonucleotide targeted against endolyn developed pericardial edema, hydrocephaly and body curvature. The pronephric kidney appeared normal morphologically, but clearance of fluorescent dextran injected into the common cardinal vein was delayed, consistent with a defect in the regulation of water balance in morphant embryos. Heterologous expression of rat endolyn rescued the morphant phenotypes. Interestingly, rescue experiments using mutant rat endolyn constructs revealed that both apical sorting and endocytic/lysosomal targeting motifs are required for normal pronephric kidney function. This suggests that both polarized targeting and postendocytic trafficking of endolyn are essential for the protein's proper function in mammalian kidney.


Asunto(s)
Polaridad Celular , Endocitosis , Endolina/metabolismo , Riñón/embriología , Riñón/metabolismo , Pronefro/embriología , Pez Cebra/embriología , Envejecimiento/metabolismo , Animales , Polaridad Celular/efectos de los fármacos , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Endocitosis/efectos de los fármacos , Endolina/química , Técnicas de Silenciamiento del Gen , Riñón/anatomía & histología , Riñón/citología , Células de Riñón Canino Madin Darby , Mamíferos/embriología , Mamíferos/metabolismo , Morfolinos/farmacología , Especificidad de Órganos , Pronefro/metabolismo , Estructura Terciaria de Proteína , Ratas , Relación Estructura-Actividad , Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA