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.
J Cell Sci ; 134(24)2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34841431

RESUMO

The main laminin-binding integrins α3ß1, α6ß1 and α6ß4 are co-expressed in the developing kidney collecting duct system. We previously showed that deleting the integrin α3 or α6 subunit in the ureteric bud, which gives rise to the kidney collecting system, caused either a mild or no branching morphogenesis phenotype, respectively. To determine whether these two integrin subunits cooperate in kidney collecting duct development, we deleted α3 and α6 in the developing ureteric bud. The collecting system of the double knockout phenocopied the α3 integrin conditional knockout. However, with age, the mice developed severe inflammation and fibrosis around the collecting ducts, resulting in kidney failure. Integrin α3α6-null collecting duct epithelial cells showed increased secretion of pro-inflammatory cytokines and displayed mesenchymal characteristics, causing loss of barrier function. These features resulted from increased nuclear factor kappa-B (NF-κB) activity, which regulated the Snail and Slug (also known as Snai1 and Snai2, respectively) transcription factors and their downstream targets. These data suggest that laminin-binding integrins play a key role in the maintenance of kidney tubule epithelial cell polarity and decrease pro-inflammatory cytokine secretion by regulating NF-κB-dependent signaling.


Assuntos
Integrinas , Túbulos Renais Coletores , Animais , Células Epiteliais , Inflamação/genética , Integrina alfa3beta1 , Integrinas/genética , Laminina/genética , Camundongos , NF-kappa B/genética
2.
J Cell Biol ; 220(11)2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34647970

RESUMO

A polarized collecting duct (CD), formed from the branching ureteric bud (UB), is a prerequisite for an intact kidney. The small Rho GTPase Rac1 is critical for actin cytoskeletal regulation. We investigated the role of Rac1 in the kidney collecting system by selectively deleting it in mice at the initiation of UB development. The mice exhibited only a mild developmental phenotype; however, with aging, the CD developed a disruption of epithelial integrity and function. Despite intact integrin signaling, Rac1-null CD cells had profound adhesion and polarity abnormalities that were independent of the major downstream Rac1 effector, Pak1. These cells did however have a defect in the WAVE2-Arp2/3 actin nucleation and polymerization apparatus, resulting in actomyosin hyperactivity. The epithelial defects were reversible with direct myosin II inhibition. Furthermore, Rac1 controlled lateral membrane height and overall epithelial morphology by maintaining lateral F-actin and restricting actomyosin. Thus, Rac1 promotes CD epithelial integrity and morphology by restricting actomyosin via Arp2/3-dependent cytoskeletal branching.


Assuntos
Actomiosina/metabolismo , Túbulos Renais Coletores/metabolismo , Neuropeptídeos/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animais , Adesão Celular/fisiologia , Polaridade Celular/fisiologia , Células Cultivadas , Citoesqueleto/metabolismo , Células Epiteliais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Miosina Tipo II/metabolismo , Transdução de Sinais/fisiologia
5.
Nat Commun ; 9(1): 814, 2018 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-29483507

RESUMO

Polycystic kidney disease (PKD) is a common genetic disorder characterized by the growth of fluid-filled cysts in the kidneys. Several studies reported that the serine-threonine kinase Lkb1 is dysregulated in PKD. Here we show that genetic ablation of Lkb1 in the embryonic ureteric bud has no effects on tubule formation, maintenance, or growth. However, co-ablation of Lkb1 and Tsc1, an mTOR repressor, results in an early developing, aggressive form of PKD. We find that both loss of Lkb1 and loss of Pkd1 render cells dependent on glutamine for growth. Metabolomics analysis suggests that Lkb1 mutant kidneys require glutamine for non-essential amino acid and glutathione metabolism. Inhibition of glutamine metabolism in both Lkb1/Tsc1 and Pkd1 mutant mice significantly reduces cyst progression. Thus, we identify a role for Lkb1 in glutamine metabolism within the kidney epithelia and suggest that drugs targeting glutamine metabolism may help reduce cyst number and/or size in PKD.


Assuntos
Glutamina/metabolismo , Doenças Renais Policísticas/enzimologia , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Quinases Ativadas por AMP , Animais , Progressão da Doença , Feminino , Humanos , Rim/embriologia , Rim/metabolismo , Rim/patologia , Masculino , Camundongos , Camundongos Knockout , Doenças Renais Policísticas/embriologia , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/metabolismo , Proteínas Serina-Treonina Quinases/genética
6.
Development ; 144(22): 4148-4158, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-28993400

RESUMO

Kidney collecting system development requires integrin-dependent cell-extracellular matrix interactions. Integrins are heterodimeric transmembrane receptors consisting of α and ß subunits; crucial integrins in the kidney collecting system express the ß1 subunit. The ß1 cytoplasmic tail has two NPxY motifs that mediate functions by binding to cytoplasmic signaling and scaffolding molecules. Talins, scaffolding proteins that bind to the membrane proximal NPxY motif, are proposed to activate integrins and to link them to the actin cytoskeleton. We have defined the role of talin binding to the ß1 proximal NPxY motif in the developing kidney collecting system in mice that selectively express a Y-to-A mutation in this motif. The mice developed a hypoplastic dysplastic collecting system. Collecting duct cells expressing this mutation had moderate abnormalities in cell adhesion, migration, proliferation and growth factor-dependent signaling. In contrast, mice lacking talins in the developing ureteric bud developed kidney agenesis and collecting duct cells had severe cytoskeletal, adhesion and polarity defects. Thus, talins are essential for kidney collecting duct development through mechanisms that extend beyond those requiring binding to the ß1 integrin subunit NPxY motif.


Assuntos
Integrina beta1/metabolismo , Morfogênese , Talina/metabolismo , Ureter/citologia , Ureter/embriologia , Junções Aderentes/metabolismo , Motivos de Aminoácidos , Animais , Sítios de Ligação , Adesão Celular , Membrana Celular/metabolismo , Polaridade Celular , Regulação da Expressão Gênica no Desenvolvimento , Integrina beta1/química , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/embriologia , Camundongos Endogâmicos C57BL , Mutação/genética , Proteínas de Junções Íntimas/genética , Proteínas de Junções Íntimas/metabolismo , Ureter/metabolismo
7.
Matrix Biol ; 57-58: 244-257, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28043890

RESUMO

Laminins are a major constituent of the basement membranes of the kidney collecting system. Integrins, transmembrane receptors formed by non-covalently bound α and ß subunits, serve as laminin receptors, but their role in development and homeostasis of the kidney collecting system is poorly defined. Integrin α3ß1, one of the major laminin receptors, plays a minor role in kidney collecting system development, while the role of α6 containing integrins (α6ß1 and α6ß4), the other major laminin receptors, is unknown. Patients with mutations in α6 containing integrins not only develop epidermolysis bullosa, but also have abnormalities in the kidney collecting system. In this study, we show that selectively deleting the α6 or ß4 integrin subunits at the initiation of ureteric bud development in mice does not affect morphogenesis. However, the collecting system becomes dilated and dysmorphic as the mice age. The collecting system in both null genotypes was also highly susceptible to unilateral ureteric obstruction injury with evidence of excessive tubule dilatation and epithelial cell apoptosis. Mechanistically, integrin α6-null collecting duct cells are unable to withstand high mechanical force when adhered to laminin. Thus, we conclude that α6 integrins are important for maintaining the integrity of the kidney collecting system by enhancing tight adhesion of the epithelial cells to the basement membrane. These data give a mechanistic explanation for the association between kidney collecting system abnormalities in patients and epidermolysis bullosa.


Assuntos
Membrana Basal/metabolismo , Integrina alfa6beta1/genética , Integrina alfa6beta4/genética , Túbulos Renais Coletores/metabolismo , Laminina/genética , Obstrução Ureteral/metabolismo , Animais , Apoptose , Membrana Basal/patologia , Adesão Celular , Movimento Celular , Proliferação de Células , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Fibrose , Regulação da Expressão Gênica , Humanos , Integrina alfa6beta1/deficiência , Integrina alfa6beta4/deficiência , Túbulos Renais Coletores/patologia , Laminina/metabolismo , Camundongos , Camundongos Knockout , Ligação Proteica , Transdução de Sinais , Ureter/cirurgia , Obstrução Ureteral/patologia , Obstrução Ureteral/cirurgia
8.
J Cell Sci ; 128(23): 4293-305, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26490995

RESUMO

The Rho GTPase Cdc42 regulates key signaling pathways required for multiple cell functions, including maintenance of shape, polarity, proliferation, migration, differentiation and morphogenesis. Although previous studies have shown that Cdc42 is required for proper epithelial development and maintenance, its exact molecular function in kidney development is not well understood. In this study, we define the specific role of Cdc42 during murine kidney epithelial tubulogenesis by deleting it selectively at the initiation of ureteric bud or metanephric mesenchyme development. Deletion in either lineage results in abnormal tubulogenesis, with profound defects in polarity, lumen formation and the actin cytoskeleton. Ultimately, these defects lead to renal failure. Additionally, in vitro analysis of Cdc42-null collecting duct cells shows that Cdc42 controls these processes by regulating the polarity Par complex (Par3-Par6-aPKC-Cdc42) and the cytoskeletal proteins N-Wasp and ezrin. Thus, we conclude that the principal role of Cdc42 in ureteric bud and metanephric mesenchyme development is to regulate epithelial cell polarity and the actin cytoskeleton.


Assuntos
Polaridade Celular/fisiologia , Citoesqueleto/metabolismo , Células Epiteliais/metabolismo , Túbulos Renais/embriologia , Proteína cdc42 de Ligação ao GTP/metabolismo , Animais , Citoesqueleto/genética , Células Epiteliais/citologia , Camundongos , Proteína cdc42 de Ligação ao GTP/genética
9.
Mol Biol Cell ; 26(10): 1857-74, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25808491

RESUMO

The collecting system of the kidney develops from the ureteric bud (UB), which undergoes branching morphogenesis, a process regulated by multiple factors, including integrin-extracellular matrix interactions. The laminin (LM)-binding integrin α3ß1 is crucial for this developmental program; however, the LM types and LM/integrin α3ß1-dependent signaling pathways are poorly defined. We show that α3 chain-containing LMs promote normal UB branching morphogenesis and that LM-332 is a better substrate than LM-511 for stimulating integrin α3ß1-dependent collecting duct cell functions. We demonstrate that integrin α3ß1-mediated cell adhesion to LM-332 modulates Akt activation in the developing collecting system and that Akt activation is PI3K independent but requires decreased PTEN activity and K63-linked polyubiquitination. We identified the ubiquitin-modifying enzyme TRAF6 as an interactor with the integrin ß1 subunit and regulator of integrin α3ß1-dependent Akt activation. Finally, we established that the developmental defects of TRAF6- and integrin α3-null mouse kidneys are similar. Thus K63-linked polyubiquitination plays a previously unrecognized role in integrin α3ß1-dependent cell signaling required for UB development and may represent a novel mechanism whereby integrins regulate signaling pathways.


Assuntos
Integrina alfa3beta1/metabolismo , Túbulos Renais Coletores/embriologia , Morfogênese , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo , Animais , Túbulos Renais Coletores/metabolismo , Camundongos , Camundongos Knockout , Transdução de Sinais , Fator 6 Associado a Receptor de TNF/genética , Ubiquitinação
10.
J Clin Invest ; 124(8): 3295-310, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24983314

RESUMO

Tubulointerstitial fibrosis underlies all forms of end-stage kidney disease. TGF-ß mediates both the development and the progression of kidney fibrosis through binding and activation of the serine/threonine kinase type II TGF-ß receptor (TßRII), which in turn promotes a TßRI-mediated SMAD-dependent fibrotic signaling cascade. Autophosphorylation of serine residues within TßRII is considered the principal regulatory mechanism of TßRII-induced signaling; however, there are 5 tyrosine residues within the cytoplasmic tail that could potentially mediate TßRII-dependent SMAD activation. Here, we determined that phosphorylation of tyrosines within the TßRII tail was essential for SMAD-dependent fibrotic signaling within cells of the kidney collecting duct. Conversely, the T cell protein tyrosine phosphatase (TCPTP) dephosphorylated TßRII tail tyrosine residues, resulting in inhibition of TßR-dependent fibrotic signaling. The collagen-binding receptor integrin α1ß1 was required for recruitment of TCPTP to the TßRII tail, as mice lacking this integrin exhibited impaired TCPTP-mediated tyrosine dephosphorylation of TßRII that led to severe fibrosis in a unilateral ureteral obstruction model of renal fibrosis. Together, these findings uncover a crosstalk between integrin α1ß1 and TßRII that is essential for TßRII-mediated SMAD activation and fibrotic signaling pathways.


Assuntos
Integrina alfa1beta1/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteínas Smad/metabolismo , Animais , Colágeno/biossíntese , Transição Epitelial-Mesenquimal , Fibrose , Integrina alfa1/genética , Integrina alfa1/metabolismo , Rim/metabolismo , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Proteínas Serina-Treonina Quinases/química , Proteína Tirosina Fosfatase não Receptora Tipo 2/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/química , Transdução de Sinais , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Tirosina/química , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia
11.
J Biol Chem ; 289(12): 8532-44, 2014 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-24509849

RESUMO

Epithelial cells lining the gastrointestinal tract and kidney have different abilities to facilitate paracellular and transcellular transport of water and solutes. In the kidney, the proximal tubule allows both transcellular and paracellular transport, while the collecting duct primarily facilitates transcellular transport. The claudins and E-cadherin are major structural and functional components regulating paracellular transport. In this study we present the novel finding that the transmembrane matrix receptors, integrins, play a role in regulating paracellular transport of renal proximal tubule cells. Deleting the integrin ß1 subunit in these cells converts them from a "loose" epithelium, characterized by low expression of E-cadherin and claudin-7 and high expression of claudin-2, to a "tight" epithelium with increased E-cadherin and claudin-7 expression and decreased claudin-2 expression. This effect is mediated by the integrin ß1 cytoplasmic tail and does not entail ß1 heterodimerization with an α-subunit or its localization to the cell surface. In addition, we demonstrate that deleting the ß1 subunit in the proximal tubule of the kidney results in a major urine-concentrating defect. Thus, the integrin ß1 tail plays a key role in regulating the composition and function of tight and adherens junctions that define paracellular transport properties of terminally differentiated renal proximal tubule epithelial cells.


Assuntos
Deleção de Genes , Integrina beta1/genética , Integrina beta1/metabolismo , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/metabolismo , Animais , Caderinas/genética , Caderinas/metabolismo , Permeabilidade da Membrana Celular , Células Cultivadas , Claudina-2/genética , Claudina-2/metabolismo , Regulação para Baixo , Células Epiteliais/metabolismo , Integrina beta1/análise , Camundongos , Permeabilidade , Regulação para Cima , Urina/química
12.
Mol Cell Biol ; 32(20): 4080-91, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22869523

RESUMO

Loss of ß1 integrin expression inhibits renal collecting-system development. Two highly conserved NPXY motifs in the distal ß1 tail regulate integrin function by associating with phosphtyrosine binding (PTB) proteins, such as talin and kindlin. Here, we define the roles of these two tyrosines in collecting-system development and delineate the structural determinants of the distal ß1 tail using nuclear magnetic resonance (NMR). Mice carrying alanine mutations have moderate renal collecting-system developmental abnormalities relative to ß1-null mice. Phenylalanine mutations did not affect renal collecting-system development but increased susceptibility to renal injury. NMR spectra in bicelles showed the distal ß1 tail is disordered and does not interact with the model membrane surface. Alanine or phenylalanine mutations did not alter ß1 structure or interactions between α and ß1 subunit transmembrane/cytoplasmic domains; however, they did decrease talin and kindlin binding. Thus, these studies highlight the fact that the functional roles of the NPXY motifs are organ dependent. Moreover, the ß1 cytoplasmic tail, in the context of the adjacent transmembrane domain in bicelles, is significantly different from the more ordered, membrane-associated ß3 integrin tail. Finally, tyrosine mutations of ß1 NPXY motifs induce phenotypes by disrupting their interactions with critical integrin binding proteins like talins and kindlins.


Assuntos
Integrina beta1/metabolismo , Túbulos Renais Coletores/crescimento & desenvolvimento , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Linhagem Celular , Citosol/metabolismo , Humanos , Integrina beta1/genética , Integrina beta3/química , Integrina beta3/metabolismo , Proteínas de Membrana/química , Camundongos , Dados de Sequência Molecular , Mutação , Proteínas de Neoplasias/química , Ligação Proteica , Conformação Proteica , Talina/química , Tirosina/química , Tirosina/genética , Tirosina/metabolismo
13.
Development ; 138(20): 4535-44, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21903675

RESUMO

In order to understand the functions of laminins in the renal collecting system, the Lamc1 gene was inactivated in the developing mouse ureteric bud (UB). Embryos bearing null alleles exhibited laminin deficiency prior to mesenchymal tubular induction and either failed to develop a UB with involution of the mesenchyme, or developed small kidneys with decreased proliferation and branching, delayed renal vesicle formation and postnatal emergence of a water transport deficit. Embryonic day 12.5 kidneys revealed an almost complete absence of basement membrane proteins and reduced levels of α6 integrin and FGF2. mRNA levels for fibroblast growth factor 2 (FGF2) and mediators of the GDNF/RET and WNT11 signaling pathway were also decreased. Furthermore, collecting duct cells derived from laminin-deficient kidneys and grown in collagen gels were found to proliferate and branch slowly. The laminin-deficient cells exhibited decreased activation of growth factor- and integrin-dependent pathways, whereas heparin lyase-treated and ß1 integrin-null cells exhibited more selective decreases. Collectively, these data support a requirement of γ1 laminins for assembly of the collecting duct system basement membrane, in which immobilized ligands act as solid-phase agonists to promote branching morphogenesis, growth and water transport functions.


Assuntos
Túbulos Renais Coletores/embriologia , Túbulos Renais Coletores/metabolismo , Laminina/metabolismo , Animais , Membrana Basal/embriologia , Membrana Basal/metabolismo , Diabetes Insípido/embriologia , Diabetes Insípido/genética , Diabetes Insípido/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Substâncias de Crescimento/metabolismo , Heparitina Sulfato/metabolismo , Hidronefrose/embriologia , Hidronefrose/genética , Hidronefrose/metabolismo , Integrina beta1/metabolismo , Túbulos Renais Coletores/anormalidades , Laminina/deficiência , Laminina/genética , Masculino , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfogênese , Gravidez , Transdução de Sinais
14.
Development ; 137(19): 3233-43, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20823064

RESUMO

The integrin-linked kinase (ILK), pinch and parvin ternary complex connects the cytoplasmic tails of beta1 integrins to the actin cytoskeleton. We recently showed that constitutive expression of ILK and alpha parvin in both the ureteric bud and the metanephric mesenchyme of the kidney is required for kidney development. In this study, we define the selective role of ILK in the ureteric bud of the mouse kidney in renal development by deleting it in the ureteric cell lineage before the onset of branching morphogenesis (E10.5). Although deleting ILK resulted in only a moderate decrease in branching, the mice died at 8 weeks of age from obstruction due to the unprecedented finding of intraluminal collecting duct cellular proliferation. ILK deletion in the ureteric bud resulted in the inability of collecting duct cells to undergo contact inhibition and to activate p38 mitogen-activated protein kinase (MAPK) in vivo and in vitro. p38 MAPK activation was not dependent on the kinase activity of ILK. Thus, we conclude that ILK plays a crucial role in activating p38 MAPK, which regulates cell cycle arrest of epithelial cells in renal tubulogenesis.


Assuntos
Ciclo Celular , Proteínas Serina-Treonina Quinases/metabolismo , Ureter/embriologia , Ureter/enzimologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Adesão Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais , Ureter/citologia
15.
Exp Cell Res ; 316(17): 2993-3005, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20727881

RESUMO

Extracellular matrix (ECM) remodeling regulates multiple cellular functions required for normal development and tissue repair. Matrix metalloproteinases (MMPs) are key mediators of this process and membrane targeted MMPs (MT-MMPs) in particular have been shown to be important in normal development of specific organs. In this study we investigated the role of MT1-MMP in kidney development. We demonstrate that loss of MT1-MMP leads to a renal phenotype characterized by a moderate decrease in ureteric bud branching morphogenesis and a severe proliferation defect. The kidneys of MT1-MMP-null mice have increased deposition of collagen IV, laminins, perlecan, and nidogen and the phenotype is independent of the MT-1MMP target, MMP-2. Utilizing in vitro systems we demonstrated that MTI-MMP proteolytic activity is required for renal tubule cells to proliferate in three dimensional matrices and to migrate on collagen IV and laminins. Together these data suggest an important role for MT1-MMP in kidney development, which is mediated by its ability to regulate cell proliferation and migration by proteolytically cleaving kidney basement membrane components.


Assuntos
Membrana Basal/metabolismo , Rim/crescimento & desenvolvimento , Metaloproteinase 14 da Matriz/fisiologia , Animais , Membrana Basal/citologia , Movimento Celular , Proliferação de Células , Matriz Extracelular , Rim/citologia , Metaloproteinase 14 da Matriz/deficiência , Camundongos , Camundongos Knockout , Fenótipo
16.
J Am Soc Nephrol ; 21(8): 1334-43, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20576806

RESUMO

TGF-beta plays a key role in upregulating matrix production in injury-induced renal fibrosis, but how TGF-beta signaling in distinct compartments of the kidney, such as specific segments of the nephron, affects the response to injury is unknown. In this study, we determined the role of TGF-beta signaling both in development of the renal collecting system and in response to injury by selectively deleting the TGF-beta type II receptor in mice at the initiation of ureteric bud development. These mice developed normally but demonstrated a paradoxic increase in fibrosis associated with enhanced levels of active TGF-beta after unilateral ureteral obstruction. Consistent with this observation, TGF-beta type II receptor deletion in cultured collecting duct cells resulted in excessive integrin alphavbeta6-dependent TGF-beta activation that increased collagen synthesis in co-cultured renal interstitial fibroblasts. These results suggest that inhibiting TGF-beta receptor-mediated function in collecting ducts may exacerbate renal fibrosis by enhancing paracrine TGF-beta signaling between epithelial and interstitial cells.


Assuntos
Túbulos Renais Coletores , Rim/patologia , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Animais , Células Cultivadas , Fibrose/etiologia , Camundongos , Obstrução Ureteral
17.
Development ; 136(19): 3357-66, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19710172

RESUMO

The kidney collecting system develops from branching morphogenesis of the ureteric bud (UB). This process requires signaling by growth factors such as glial cell line derived neurotrophic factor (GDNF) and fibroblast growth factors (FGFs) as well as cell extracellular matrix interactions mediated by integrins. The importance of integrin signaling in UB development was investigated by deleting integrin beta1 at initiation (E10.5) and late (E18.5) stages of development. Deletion at E10.5 resulted in a severe branching morphogenesis phenotype. Deletion at E18.5 did not alter renal development but predisposed the collecting system to severe injury following ureteric obstruction. beta1 integrin was required for renal tubular epithelial cells to mediate GDNF- and FGF-dependent signaling despite normal receptor localization and activation in vitro. Aberrations in the same signaling molecules were present in the beta1-null UBs in vivo. Thus beta1 integrins can regulate organ branching morphogenesis during development by mediating growth-factor-dependent signaling in addition to their well-defined role as adhesion receptors.


Assuntos
Integrina beta1/metabolismo , Túbulos Renais Coletores/embriologia , Túbulos Renais Coletores/metabolismo , Ureter/embriologia , Ureter/metabolismo , Animais , Adesão Celular , Linhagem Celular , Movimento Celular , Proliferação de Células , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Substâncias de Crescimento/metabolismo , Integrina beta1/genética , Túbulos Renais Coletores/citologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Morfogênese , Técnicas de Cultura de Órgãos , Transdução de Sinais
18.
Cancer Res ; 68(15): 6127-35, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18676835

RESUMO

The collagen IV binding receptor integrin alpha1beta1 has been shown to regulate lung cancer due to its proangiogenic properties; however, it is unclear whether this receptor also plays a direct role in promoting primary lung tumors. To investigate this possibility, integrin alpha1-null mice were crossed with KrasLA2 mice that carry an oncogenic mutation of the Kras gene (G12D) and develop spontaneous primary tumors with features of non-small cell lung cancer. We provide evidence that KrasLA2/alpha1-null mice have a decreased incidence of primary lung tumors and longer survival compared with KrasLA2/alpha1 wild-type controls. Tumors from KrasLA2/alpha1-null mice were also smaller, less vascularized, and exhibited reduced cell proliferation and increased apoptosis, as determined by proliferating cell nuclear antigen and terminal deoxynucleotidyl-transferase-mediated dUTP nick-end staining, respectively. Moreover, tumors from the KrasLA2/alpha1-null mice showed diminished extracellular signal-regulated kinase (ERK) but enhanced p38 mitogen-activated protein kinase activation. Primary lung tumor epithelial cells isolated from KrasLA2/alpha1-null mice showed a significant decrease in anchorage-independent colony formation, collagen-mediated cell proliferation, ERK activation, and, most importantly, tumorigenicity when injected into nude mice compared with KrasLA2/alpha1 wild-type tumor cells. These results indicate that loss of the integrin alpha1 subunit decreases the incidence and growth of lung epithelial tumors initiated by oncogenic Kras, suggesting that both Kras and integrin alpha1beta1 cooperate to drive the growth of non-small cell lung cancer in vivo.


Assuntos
Genes ras , Integrina alfa1beta1/fisiologia , Neoplasias Pulmonares/genética , Animais , Adesão Celular , Proliferação de Células , Colágeno Tipo IV/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Imuno-Histoquímica , Integrina alfa1beta1/genética , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Knockout , Análise de Sobrevida
19.
Mol Biol Cell ; 17(4): 2046-56, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16467383

RESUMO

The collecting system of the kidney, derived from the ureteric bud (UB), undergoes repetitive bifid branching events during early development followed by a phase of tubular growth and elongation. Although members of the Ras GTPase family control cell growth, differentiation, proliferation, and migration, their role in development of the collecting system of the kidney is unexplored. In this study, we demonstrate that members of the R-Ras family of proteins, R-Ras and TC21, are expressed in the murine collecting system at E13.5, whereas H-Ras is only detected at day E17.5. Using murine UB cells expressing activated H-Ras, R-Ras, and TC21, we demonstrate that R-Ras-expressing cells show increased branching morphogenesis and cell growth, TC21-expressing cells branch excessively but lose their ability to migrate, whereas H-Ras-expressing cells migrated the most and formed long unbranched tubules. These differences in branching morphogenesis are mediated by differential regulation/activation of the Rho family of GTPases and mitogen-activated protein kinases. Because most branching of the UB occurs early in development, it is conceivable that R-Ras and TC-21 play a role in facilitating branching and growth in early UB development, whereas H-Ras might favor cell migration and elongation of tubules, events that occur later in development.


Assuntos
Túbulos Renais Coletores/embriologia , Proteínas de Membrana/fisiologia , Proteínas Monoméricas de Ligação ao GTP/fisiologia , Morfogênese , Ureter/embriologia , Proteínas ras/fisiologia , Animais , Movimento Celular , Proliferação de Células , Células Cultivadas , Ativação Enzimática , Epitélio/embriologia , Epitélio/enzimologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Túbulos Renais Coletores/química , Túbulos Renais Coletores/enzimologia , Proteínas de Membrana/análise , Proteínas de Membrana/genética , Mesoderma/enzimologia , Camundongos , Proteínas Monoméricas de Ligação ao GTP/análise , Proteínas Monoméricas de Ligação ao GTP/genética , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Transdução de Sinais , Ureter/química , Ureter/enzimologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas ras/análise , Proteínas ras/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...