Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Renal Physiol ; 326(3): F326-F337, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38205542

RESUMO

Acute kidney injury activates both proliferative and antiproliferative pathways, the consequences of which are not fully elucidated. If an initial proliferation of the renal epithelium is necessary for the successful repair, the persistence of proliferation markers is associated with the occurrence of chronic kidney disease. We hypothesized that proliferation in stress conditions impacts cell viability and renal outcomes. We found that proliferation is associated with cell death after various stresses in kidney cells. In vitro, the ATP/ADP ratio oscillates reproducibly throughout the cell cycle, and cell proliferation is associated with a decreased intracellular ATP/ADP ratio. In vivo, transcriptomic data from transplanted kidneys revealed that proliferation was strongly associated with a decrease in the expression of the mitochondria-encoded genes of the oxidative phosphorylation pathway, but not of the nucleus-encoded ones. These observations suggest that mitochondrial function is a limiting factor for energy production in proliferative kidney cells after injury. The association of increased proliferation and decreased mitochondrial function was indeed associated with poor renal outcomes. In summary, proliferation is an energy-demanding process impairing the cellular ability to cope with an injury, highlighting proliferative repair and metabolic recovery as indispensable and interdependent features for successful kidney repair.NEW & NOTEWORTHY ATP depletion is a hallmark of acute kidney injury. Proliferation is instrumental to kidney repair. We show that ATP levels vary during the cell cycle and that proliferation sensitizes renal epithelial cells to superimposed injuries in vitro. More proliferation and less energy production by the mitochondria are associated with adverse outcomes in injured kidney allografts. This suggests that controlling the timing of kidney repair might be beneficial to mitigate the extent of acute kidney injury.


Assuntos
Injúria Renal Aguda , Traumatismo por Reperfusão , Humanos , Rim/metabolismo , Injúria Renal Aguda/genética , Injúria Renal Aguda/metabolismo , Células Epiteliais/metabolismo , Proliferação de Células , Trifosfato de Adenosina/metabolismo , Traumatismo por Reperfusão/metabolismo
2.
Sci Adv ; 8(38): eabq0866, 2022 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-36129975

RESUMO

Organoids serve as a novel tool for disease modeling in three-dimensional multicellular contexts. Static organoids, however, lack the requisite biophysical microenvironment such as fluid flow, limiting their ability to faithfully recapitulate disease pathology. Here, we unite organoids with organ-on-a-chip technology to unravel disease pathology and develop therapies for autosomal recessive polycystic kidney disease. PKHD1-mutant organoids-on-a-chip are subjected to flow that induces clinically relevant phenotypes of distal nephron dilatation. Transcriptomics discover 229 signal pathways that are not identified by static models. Mechanosensing molecules, RAC1 and FOS, are identified as potential therapeutic targets and validated by patient kidney samples. On the basis of this insight, we tested two U.S. Food and Drug Administration-approved and one investigational new drugs that target RAC1 and FOS in our organoid-on-a-chip model, which suppressed cyst formation. Our observations highlight the vast potential of organoid-on-a-chip models to elucidate complex disease mechanisms for therapeutic testing and discovery.


Assuntos
Rim Policístico Autossômico Recessivo , Descoberta de Drogas , Drogas em Investigação , Humanos , Dispositivos Lab-On-A-Chip , Organoides/metabolismo , Rim Policístico Autossômico Recessivo/genética , Rim Policístico Autossômico Recessivo/metabolismo , Rim Policístico Autossômico Recessivo/patologia
3.
Nat Rev Nephrol ; 16(11): 686-696, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32939051

RESUMO

An important need exists to better understand and stratify kidney disease according to its underlying pathophysiology in order to develop more precise and effective therapeutic agents. National collaborative efforts such as the Kidney Precision Medicine Project are working towards this goal through the collection and integration of large, disparate clinical, biological and imaging data from patients with kidney disease. Ontologies are powerful tools that facilitate these efforts by enabling researchers to organize and make sense of different data elements and the relationships between them. Ontologies are critical to support the types of big data analysis necessary for kidney precision medicine, where heterogeneous clinical, imaging and biopsy data from diverse sources must be combined to define a patient's phenotype. The development of two new ontologies - the Kidney Tissue Atlas Ontology and the Ontology of Precision Medicine and Investigation - will support the creation of the Kidney Tissue Atlas, which aims to provide a comprehensive molecular, cellular and anatomical map of the kidney. These ontologies will improve the annotation of kidney-relevant data, and eventually lead to new definitions of kidney disease in support of precision medicine.


Assuntos
Atlas como Assunto , Ontologias Biológicas , Nefropatias/classificação , Medicina de Precisão , Big Data , Humanos , Fenótipo
4.
Dev Biol ; 440(1): 13-21, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29705331

RESUMO

Formation of a functional kidney depends on the balance between renewal and differentiation of nephron progenitors. Failure to sustain this balance can lead to kidney failure or stem cell tumors. For nearly 60 years, we have known that signals from an epithelial structure known as the ureteric bud were essential for maintaining this balance. More recently it was discovered that one molecule, Wnt9b, was necessary for both renewal and differentiation of the nephron progenitor cells. How one ligand signaling through one transcription factor promoted two seemingly contradictory cellular processes was unclear. In this study, we show that Wnt9b/beta-catenin signaling alone is sufficient to promote both renewal and differentiation. Moreover, we show that discrete levels of beta-catenin can promote these two disparate fates, with low levels fostering progenitor renewal and high levels driving differentiation. These results provide insight into how Wnt9b regulates distinct target genes that balance nephron progenitor renewal and differentiation.


Assuntos
Néfrons/fisiologia , beta Catenina/metabolismo , beta Catenina/fisiologia , Animais , Diferenciação Celular/genética , Regulação da Expressão Gênica/genética , Rim/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Néfrons/embriologia , Transdução de Sinais/fisiologia , Células-Tronco/metabolismo , Células-Tronco/fisiologia , Fatores de Transcrição/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt/fisiologia
5.
JCI Insight ; 2(2): e88848, 2017 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-28138555

RESUMO

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.


Assuntos
Criocirurgia , Nefrectomia , Néfrons/crescimento & desenvolvimento , Células-Tronco , Animais , Animais Recém-Nascidos , Linhagem da Célula , Fibrose , Proteínas de Homeodomínio/metabolismo , Rim/patologia , Glomérulos Renais/crescimento & desenvolvimento , Glomérulos Renais/patologia , Proteínas com Homeodomínio LIM/metabolismo , Camundongos , Néfrons/patologia , Organogênese , Fator de Transcrição PAX2/metabolismo , Fatores de Transcrição/metabolismo , Transcriptoma
6.
Nat Commun ; 6: 8715, 2015 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-26493500

RESUMO

Human-pluripotent-stem-cell-derived kidney cells (hPSC-KCs) have important potential for disease modelling and regeneration. Whether the hPSC-KCs can reconstitute tissue-specific phenotypes is currently unknown. Here we show that hPSC-KCs self-organize into kidney organoids that functionally recapitulate tissue-specific epithelial physiology, including disease phenotypes after genome editing. In three-dimensional cultures, epiblast-stage hPSCs form spheroids surrounding hollow, amniotic-like cavities. GSK3ß inhibition differentiates spheroids into segmented, nephron-like kidney organoids containing cell populations with characteristics of proximal tubules, podocytes and endothelium. Tubules accumulate dextran and methotrexate transport cargoes, and express kidney injury molecule-1 after nephrotoxic chemical injury. CRISPR/Cas9 knockout of podocalyxin causes junctional organization defects in podocyte-like cells. Knockout of the polycystic kidney disease genes PKD1 or PKD2 induces cyst formation from kidney tubules. All of these functional phenotypes are distinct from effects in epiblast spheroids, indicating that they are tissue specific. Our findings establish a reproducible, versatile three-dimensional framework for human epithelial disease modelling and regenerative medicine applications.


Assuntos
Células-Tronco Embrionárias/citologia , Camadas Germinativas/citologia , Nefropatias/genética , Rim/citologia , Organoides/citologia , Células-Tronco Pluripotentes/citologia , Diferenciação Celular , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Células-Tronco Embrionárias/metabolismo , Técnicas de Inativação de Genes , Camadas Germinativas/metabolismo , Humanos , Rim/metabolismo , Nefropatias/metabolismo , Nefropatias/fisiopatologia , Modelos Biológicos , Organoides/metabolismo , Células-Tronco Pluripotentes/metabolismo , Sialoglicoproteínas/genética , Sialoglicoproteínas/metabolismo
7.
J Clin Invest ; 125(8): 3198-214, 2015 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-26121749

RESUMO

Macrophages (Mø) are integral in ischemia/reperfusion injury-incited (I/R-incited) acute kidney injury (AKI) that leads to fibrosis and chronic kidney disease (CKD). IL-34 and CSF-1 share a receptor (c-FMS), and both cytokines mediate Mø survival and proliferation but also have distinct features. CSF-1 is central to kidney repair and destruction. We tested the hypothesis that IL-34-dependent, Mø-mediated mechanisms promote persistent ischemia-incited AKI that worsens subsequent CKD. In renal I/R, the time-related magnitude of Mø-mediated AKI and subsequent CKD were markedly reduced in IL-34-deficient mice compared with controls. IL-34, c-FMS, and a second IL-34 receptor, protein-tyrosine phosphatase ζ (PTP-ζ) were upregulated in the kidney after I/R. IL-34 was generated by tubular epithelial cells (TECs) and promoted Mø-mediated TEC destruction during AKI that worsened subsequent CKD via 2 distinct mechanisms: enhanced intrarenal Mø proliferation and elevated BM myeloid cell proliferation, which increases circulating monocytes that are drawn into the kidney by chemokines. CSF-1 expression in TECs did not compensate for IL-34 deficiency. In patients, kidney transplants subject to I/R expressed IL-34, c-FMS, and PTP-ζ in TECs during AKI that increased with advancing injury. Moreover, IL-34 expression increased, along with more enduring ischemia in donor kidneys. In conclusion, IL-34-dependent, Mø-mediated, CSF-1 nonredundant mechanisms promote persistent ischemia-incited AKI that worsens subsequent CKD.


Assuntos
Injúria Renal Aguda/metabolismo , Interleucinas/metabolismo , Transplante de Rim , Macrófagos/metabolismo , Disfunção Primária do Enxerto/metabolismo , Insuficiência Renal Crônica/metabolismo , Injúria Renal Aguda/genética , Injúria Renal Aguda/patologia , Animais , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Interleucinas/genética , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Macrófagos/patologia , Camundongos , Camundongos Transgênicos , Disfunção Primária do Enxerto/genética , Disfunção Primária do Enxerto/patologia , Receptor de Fator Estimulador de Colônias de Macrófagos/genética , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/patologia
8.
J Am Soc Nephrol ; 25(6): 1211-25, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24357672

RESUMO

Human pluripotent stem cells (hPSCs) can generate a diversity of cell types, but few methods have been developed to derive cells of the kidney lineage. Here, we report a highly efficient system for differentiating human embryonic stem cells and induced pluripotent stem cells (referred to collectively as hPSCs) into cells expressing markers of the intermediate mesoderm (IM) that subsequently form tubule-like structures. Treatment of hPSCs with the glycogen synthase kinase-3ß inhibitor CHIR99021 induced BRACHYURY(+)MIXL1(+) mesendoderm differentiation with nearly 100% efficiency. In the absence of additional exogenous factors, CHIR99021-induced mesendodermal cells preferentially differentiated into cells expressing markers of lateral plate mesoderm with minimal IM differentiation. However, the sequential treatment of hPSCs with CHIR99021 followed by fibroblast growth factor-2 and retinoic acid generated PAX2(+)LHX1(+) cells with 70%-80% efficiency after 3 days of differentiation. Upon growth factor withdrawal, these PAX2(+)LHX1(+) cells gave rise to apically ciliated tubular structures that coexpressed the proximal tubule markers Lotus tetragonolobus lectin, N-cadherin, and kidney-specific protein and partially integrated into embryonic kidney explant cultures. With the addition of FGF9 and activin, PAX2(+)LHX1(+) cells specifically differentiated into cells expressing SIX2, SALL1, and WT1, markers of cap mesenchyme nephron progenitor cells. Our findings demonstrate the effective role of fibroblast growth factor signaling in inducing IM differentiation in hPSCs and establish the most rapid and efficient system whereby hPSCs can be differentiated into cells with features characteristic of kidney lineage cells.


Assuntos
Diferenciação Celular/fisiologia , Túbulos Renais Proximais/citologia , Mesoderma/citologia , Células-Tronco Pluripotentes/citologia , Animais , Antineoplásicos/farmacologia , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Feminino , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fibroblastos/citologia , Prepúcio do Pênis/citologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Humanos , Túbulos Renais Proximais/embriologia , Túbulos Renais Proximais/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Masculino , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos , Fator de Transcrição PAX2/metabolismo , Células-Tronco Pluripotentes/efeitos dos fármacos , Gravidez , Piridinas/farmacologia , Pirimidinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Tretinoína/farmacologia
9.
J Am Soc Nephrol ; 23(4): 674-86, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22343121

RESUMO

Nur77 and its family members Nurr1 and Nor-1 are inducible orphan nuclear receptors that orchestrate cellular responses to diverse extracellular signals. In epithelia, Nur77 can act as a potent proapoptotic molecule in response to cellular stress, suggesting a possible role for this nuclear receptor in the tissue response to injury. Here, we found that Nur77 promotes epithelial cell apoptosis after AKI. Injury of proximal tubular epithelial cells rapidly and strongly induced Nur77, Nor-1, and Nurr1 both in vitro and in vivo. After renal ischemia-reperfusion, Nurr77-deficient mice exhibited less apoptosis of tubular epithelial cells and better renal function than wild-type mice. Nur77-mediated renal injury involved a conformational change of Bcl2 and an increase in the protein levels of proapoptotic Bcl-xS. Ligand-activated retinoic acid receptors repressed Nur77 induction and function. Pretreatment of wild-type mice with retinoic acid before renal ischemia-reperfusion blunted the induction of Nur77, conferred protection of renal function, attenuated renal histologic injury, and reduced the expression of epithelial-derived proinflammatory cytokines. Retinoic acid also inhibited hypoxia-mediated induction of proinflammatory cytokines in cultured renal epithelial cells. Results obtained from proximal tubule cultures derived from Nur77-deficient mice suggested that the inhibition of Nur77 expression mediated the renoprotective effects of retinoic acid. In summary, Nur77 promotes epithelial apoptosis after ischemia-reperfusion injury, and retinoic acid-mediated inhibition of Nur77 expression is a promising therapeutic strategy for the prevention of AKI.


Assuntos
Injúria Renal Aguda/genética , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Traumatismo por Reperfusão/prevenção & controle , Tretinoína/farmacologia , Injúria Renal Aguda/patologia , Animais , Apoptose/genética , Apoptose/fisiologia , Células Cultivadas , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/metabolismo , Dimetil Sulfóxido/farmacologia , Modelos Animais de Doenças , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/fisiologia , Hibridização In Situ , Testes de Função Renal , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Distribuição Aleatória , Valores de Referência , Traumatismo por Reperfusão/patologia , Índice de Gravidade de Doença
11.
Development ; 138(19): 4245-54, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21852398

RESUMO

The primary excretory organ in vertebrates is the kidney, which is responsible for blood filtration, solute homeostasis and pH balance. These functions are carried out by specialized epithelial cells organized into tubules called nephrons. Each of these cell types arise during embryonic development from a mesenchymal stem cell pool through a process of mesenchymal-to-epithelial transition (MET) that requires sequential action of specific Wnt signals. Induction by Wnt9b directs cells to exit the stem cell niche and express Wnt4, which is both necessary and sufficient for the formation of epithelia. Without either factor, MET fails, nephrons do not form and newborn mice die owing to kidney failure. Ectopic Notch activation in stem cells induces mass differentiation and exhaustion of the stem cell pool. To investigate whether this reflected an interaction between Notch and Wnt, we employed a novel gene manipulation strategy in cultured embryonic kidneys. We show that Notch activation is capable of inducing MET in the absence of both Wnt4 and Wnt9b. Following MET, the presence of Notch directs cells primarily to the proximal tubule fate. Only nephron stem cells have the ability to undergo MET in response to Wnt or Notch, as activation in the closely related stromal mesenchyme has no inductive effect. These data demonstrate that stem cells for renal epithelia are uniquely poised to undergo MET, and that Notch activation can replace key inductive Wnt signals in this process. After MET, Notch provides an instructive signal directing cells towards the proximal tubule lineage at the expense of other renal epithelial fates.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Néfrons/embriologia , Receptores Notch/metabolismo , Células-Tronco/citologia , Proteínas Wnt/metabolismo , Proteína Wnt4/metabolismo , Animais , Desdiferenciação Celular , Linhagem da Célula , Células Epiteliais/citologia , Mesoderma/metabolismo , Camundongos , Modelos Biológicos , Podócitos/citologia
12.
Kidney Int ; 79(3): 317-30, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20944551

RESUMO

MicroRNAs (miRNAs) are a large and growing class of small, non-coding, regulatory RNAs that control gene expression predominantly at the post-transcriptional level. The production of most functional miRNAs depends on the enzymatic activity of Dicer, an RNase III class enzyme. To address the potential action of Dicer-dependent miRNAs in mammalian kidney development, we conditionally ablated Dicer function within cells of nephron lineage and the ureteric bud-derived collecting duct system. Six2Cre-mediated removal of Dicer activity from the progenitors of the nephron epithelium led to elevated apoptosis and premature termination of nephrogenesis. Thus, Dicer action is important for maintaining the viability of this critical self-renewing progenitor pool and, consequently, development of a normal nephron complement. HoxB7Cre-mediated removal of Dicer function from the ureteric bud epithelium led to the development of renal cysts. This was preceded by excessive cell proliferation and apoptosis, and accompanied by disrupted ciliogenesis within the ureteric bud epithelium. Dicer removal also disrupted branching morphogenesis with the phenotype correlating with downregulation of Wnt11 and c-Ret expression at ureteric tips. Thus Dicer, and by inference Dicer-dependent miRNA activity, have distinct regulatory roles within different components of the developing mouse kidney. Furthermore, an understanding of miRNA action may provide new insights into the etiology and pathogenesis of renal cyst-based kidney disease.


Assuntos
RNA Helicases DEAD-box/metabolismo , Endorribonucleases/metabolismo , Células Epiteliais/metabolismo , Néfrons/metabolismo , Ureter/metabolismo , Animais , Animais Recém-Nascidos , Apoptose , Proliferação de Células , Sobrevivência Celular , RNA Helicases DEAD-box/deficiência , RNA Helicases DEAD-box/genética , Endorribonucleases/deficiência , Endorribonucleases/genética , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Idade Gestacional , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Doenças Renais Císticas/embriologia , Doenças Renais Císticas/metabolismo , Camundongos , Camundongos Knockout , MicroRNAs/metabolismo , Morfogênese , Mutação , Néfrons/embriologia , Fenótipo , Proteínas Proto-Oncogênicas c-ret/genética , RNA Mensageiro/metabolismo , Ribonuclease III , Células-Tronco/metabolismo , Ureter/embriologia , Proteínas Wnt
13.
Am J Pathol ; 176(1): 85-97, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20008127

RESUMO

Understanding the origin of myofibroblasts in kidney is of great interest because these cells are responsible for scar formation in fibrotic kidney disease. Recent studies suggest epithelial cells are an important source of myofibroblasts through a process described as the epithelial-to-mesenchymal transition; however, confirmatory studies in vivo are lacking. To quantitatively assess the contribution of renal epithelial cells to myofibroblasts, we used Cre/Lox techniques to genetically label and fate map renal epithelia in models of kidney fibrosis. Genetically labeled primary proximal epithelial cells cultured in vitro from these mice readily induce markers of myofibroblasts after transforming growth factor beta(1) treatment. However, using either red fluorescent protein or beta-galactosidase as fate markers, we found no evidence that epithelial cells migrate outside of the tubular basement membrane and differentiate into interstitial myofibroblasts in vivo. Thus, although renal epithelial cells can acquire mesenchymal markers in vitro, they do not directly contribute to interstitial myofibroblast cells in vivo. Lineage analysis shows that during nephrogenesis, FoxD1-positive((+)) mesenchymal cells give rise to adult CD73(+), platelet derived growth factor receptor beta(+), smooth muscle actin-negative interstitial pericytes, and these FoxD1-derivative interstitial cells expand and differentiate into smooth muscle actin(+) myofibroblasts during fibrosis, accounting for a large majority of myofibroblasts. These data indicate that therapeutic strategies directly targeting pericyte differentiation in vivo may productively impact fibrotic kidney disease.


Assuntos
Linhagem da Célula , Células Epiteliais/patologia , Fibroblastos/patologia , Rim/patologia , Pericitos/patologia , Actinas/metabolismo , Animais , Movimento Celular , Células Cultivadas , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Fibroblastos/metabolismo , Fibrose , Fatores de Transcrição Forkhead/metabolismo , Proteínas de Fluorescência Verde , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Integrases/metabolismo , Rim/metabolismo , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Mesoderma/metabolismo , Mesoderma/patologia , Camundongos , Pericitos/metabolismo , Regiões Promotoras Genéticas/genética , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/metabolismo , Fatores de Transcrição/genética , Regulação para Cima
14.
Cell Stem Cell ; 3(2): 169-81, 2008 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-18682239

RESUMO

Nephrons, the basic functional units of the kidney, are generated repetitively during kidney organogenesis from a mesenchymal progenitor population. Which cells within this pool give rise to nephrons and how multiple nephron lineages form during this protracted developmental process are unclear. We demonstrate that the Six2-expressing cap mesenchyme represents a multipotent nephron progenitor population. Six2-expressing cells give rise to all cell types of the main body of the nephron during all stages of nephrogenesis. Pulse labeling of Six2-expressing nephron progenitors at the onset of kidney development suggests that the Six2-expressing population is maintained by self-renewal. Clonal analysis indicates that at least some Six2-expressing cells are multipotent, contributing to multiple domains of the nephron. Furthermore, Six2 functions cell autonomously to maintain a progenitor cell status, as cap mesenchyme cells lacking Six2 activity contribute to ectopic nephron tubules, a mechanism dependent on a Wnt9b inductive signal. Taken together, our observations suggest that Six2 activity cell-autonomously regulates a multipotent nephron progenitor population.


Assuntos
Proteínas de Homeodomínio/genética , Rim/citologia , Néfrons/citologia , Organogênese/genética , Fatores de Transcrição/genética , Animais , Diferenciação Celular/genética , Quimera/genética , Quimera/metabolismo , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Rim/embriologia , Rim/fisiologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , Néfrons/embriologia , Néfrons/fisiologia , Fatores de Transcrição/metabolismo , Ativação Transcricional , Proteínas Wnt/genética
15.
Cell Stem Cell ; 2(3): 284-91, 2008 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-18371453

RESUMO

Understanding the mechanisms of nephron repair is critical for the design of new therapeutic approaches to treat kidney disease. The kidney can repair after even a severe insult, but whether adult stem or progenitor cells contribute to epithelial renewal after injury and the cellular origin of regenerating cells remain controversial. Using genetic fate-mapping techniques, we generated transgenic mice in which 94%-95% of tubular epithelial cells, but no interstitial cells, were labeled with either beta-galactosidase (lacZ) or red fluorescent protein (RFP). Two days after ischemia-reperfusion injury (IRI), 50.5% of outer medullary epithelial cells coexpress Ki67 and RFP, indicating that differentiated epithelial cells that survived injury undergo proliferative expansion. After repair was complete, 66.9% of epithelial cells had incorporated BrdU, compared to only 3.5% of cells in the uninjured kidney. Despite this extensive cell proliferation, no dilution of either cell-fate marker was observed after repair. These results indicate that regeneration by surviving tubular epithelial cells is the predominant mechanism of repair after ischemic tubular injury in the adult mammalian kidney.


Assuntos
Células-Tronco Adultas/metabolismo , Proliferação de Células , Células Epiteliais/metabolismo , Nefropatias/metabolismo , Túbulos Renais/metabolismo , Regeneração , Traumatismo por Reperfusão/metabolismo , Células-Tronco Adultas/citologia , Animais , Células Epiteliais/patologia , Nefropatias/patologia , Túbulos Renais/patologia , Camundongos , Camundongos Transgênicos , Traumatismo por Reperfusão/patologia , Fatores de Tempo
16.
Development ; 134(13): 2533-9, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17537789

RESUMO

Mammalian nephrons form as a result of a complex morphogenesis and patterning of a simple epithelial precursor, the renal vesicle. Renal vesicles are established from a mesenchymal progenitor population in response to inductive signals. Several lines of evidence support the sequential roles of two Wnt family members, Wnt9b and Wnt4, in renal vesicle induction. Using genetic approaches to specifically manipulate the activity of beta-catenin within the mesenchymal progenitor pool in mice, we investigated the potential role of the canonical Wnt pathway in these inductive events. Progenitor-cell-specific removal of beta-catenin activity completely blocked both the formation of renal vesicles and the expected molecular signature of an earlier inductive response. By contrast, activation of stabilized beta-catenin in the same cell population causes ectopic expression of mesenchymal induction markers in vitro and functionally replaces the requirement for Wnt9b and Wnt4 in their inductive roles in vivo. Thus, canonical Wnt signaling is both necessary and sufficient for initiating and maintaining inductive pathways mediated by Wnt9b and Wnt4. However, the failure of induced mesenchyme with high levels of beta-catenin activity to form epithelial structures suggests that modulating canonical signaling may be crucial for the cellular transition to the renal vesicle.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Rim/embriologia , Rim/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animais , Biomarcadores , Diferenciação Celular , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Rim/citologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas Wnt/genética , beta Catenina/genética
17.
Proc Natl Acad Sci U S A ; 99(12): 8090-5, 2002 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-12060755

RESUMO

Mutation of the functionally redundant Hoxa 11/Hoxd 11 genes gives absent or rudimentary kidneys resulting from a dramatic reduction of the growth and branching of the ureteric bud. To understand better the molecular mechanisms of Hoxa 11/Hoxd 11 function in kidney development, it is necessary to identify the downstream target genes regulated by their encoded transcription factors. To this end, we conducted a screen for Hoxa 11-responsive genes in two kidney cell lines. HEK293 cells, which usually do not express Hoxa 11, were modified to allow inducible Hoxa 11 expression. The mK10 cells, derived specifically for this study from Hoxa 11/Hoxd 11 double-mutant mice, were also modified to give cell populations with and without Hoxa 11 expression. Differential display, Gene Discovery Arrays, and Affymetrix genechip probe arrays were used to screen for genes up- or down-regulated by Hoxa 11. Nine genes, PDGF A, Cathepsin L, annexin A1, Mm.112139, Est2 repressor factor, NrCAM, ZNF192, integrin-associated protein, and GCM1, showed reproducible 3-fold or smaller changes in gene expression in response to Hoxa 11. One gene, the Integrin alpha8, was up-regulated approximately 20-fold after Hoxa 11 expression. The Integrin alpha8 gene is expressed together with Hoxa 11 in metanephric mesenchyme cells, and mutation of Integrin alpha8 gives a bud-branching morphogenesis defect very similar to that observed in Hoxa 11/Hoxd 11 mutant mice. In situ hybridizations showed a dramatic regional reduction in Integrin alpha8 expression in the developing kidneys of Hoxa 11/Hoxd 11 mutant mice. This work suggests that the Integrin alpha8 gene may be a major effector of Hoxa 11/Hoxd 11 function in the developing kidney.


Assuntos
Proteínas de Homeodomínio/genética , Cadeias alfa de Integrinas , Integrinas/genética , Rim/embriologia , Proteínas Oncogênicas/genética , Animais , Sequência de Bases , Linhagem Celular , Primers do DNA , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Mutagênese , Reação em Cadeia da Polimerase , Proteínas Proto-Oncogênicas , Proteínas Recombinantes/metabolismo , Transfecção
18.
Mech Dev ; 112(1-2): 219-32, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11850199

RESUMO

Clonal cell lines representing different developmental stages of the metanephric mesenchyme were made from transgenic mice with the Simian Virus 40 T-antigen (SV40 Tag) gene driven by the Hoxa 11 promoter. The resulting mK3 cell line represented early metanephric mesenchyme, prior to induction by the ureteric bud. These cells showed a spindle-shaped, fibroblast morphology. They expressed genes characteristic of early mesenchyme, including Hoxa 11, Hoxd 11, collagen I, and vimentin. Moreover, the mK3 cells displayed early metanephric mesenchyme biological function. In organ co-culture experiments they were able to induce growth and branching of the ureteric bud. Another cell line, mK4, represented later, induced metanephric mesenchyme undergoing epithelial conversion. These cells were more polygonal, or epithelial in shape, and expressed genes diagnostic of late mesenchyme, including Pax-2, Pax-8, Wnt-4, Cadherin-6, Collagen IV, and LFB3. To better define the gene expression patterns of kidney metanephric mesenchyme cells at these two stages of development, RNAs from the mK3 and mK4 cells were hybridized to Affymetrix GeneChip probe arrays. Over 4000 expressed genes were identified and thereby implicated in kidney formation. Comparison of the mK3 and mK4 gene expression profiles revealed 121 genes showing greater than a ten-fold difference in expression level. Several are known to be expressed during metanephric mesenchyme differentiation, but most had not been previously associated with this process. In situ hybridizations were used to confirm that selected novel genes were expressed in the developing kidney.


Assuntos
Rim/embriologia , Animais , Antígenos Transformantes de Poliomavirus/genética , Northern Blotting , Linhagem Celular , Perfilação da Expressão Gênica , Proteínas de Homeodomínio/genética , Imuno-Histoquímica , Hibridização In Situ , Mesoderma/citologia , Camundongos , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ureter/embriologia
19.
Mech Dev ; 110(1-2): 151-64, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11744376

RESUMO

Clonal cell lines representing different developmental stages of the metanephric mesenchyme were made from transgenic mice with the Simian Virus 40 T-antigen (SV40 Tag) gene driven by the Hoxa 11 promoter. The resulting mK3 cell line represented early metanephric mesenchyme, prior to induction by the ureteric bud. These cells showed a spindle-shaped, fibroblast morphology. They expressed genes characteristic of early mesenchyme, including Hoxa 11, Hoxd 11, collagen I, and vimentin. Moreover, the mK3 cells displayed early metanephric mesenchyme biological function. In organ co-culture experiments they were able to induce growth and branching of the ureteric bud. Another cell line, mK4, represented later, induced metanephric mesenchyme undergoing epithelial conversion. These cells were more polygonal, or epithelial in shape, and expressed genes diagnostic of late mesenchyme, including Pax-2, Pax-8, Wnt-4, Cadherin-6, Collagen IV, and LFB3. To better define the gene expression patterns of kidney metanephric mesenchyme cells at these two stages of development, RNAs from the mK3 and mK4 cells were hybridized to Affymetrix GeneChip probe arrays. Over 4000 expressed genes were identified and thereby implicated in kidney formation. Comparison of the mK3 and mK4 gene expression profiles revealed 121 genes showing greater than a ten-fold difference in expression level. Several are known to be expressed during metanephric mesenchyme differentiation, but most had not been previously associated with this process. In situ hybridizations were used to confirm that selected novel genes were expressed in the developing kidney.


Assuntos
Rim/embriologia , Animais , Antígenos Transformantes de Poliomavirus/genética , Northern Blotting , Linhagem Celular , Perfilação da Expressão Gênica , Proteínas de Homeodomínio/genética , Imuno-Histoquímica , Hibridização In Situ , Mesoderma/citologia , Camundongos , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ureter/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA