Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Mol Gastroenterol Hepatol ; 16(3): 325-339, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37270061

RESUMO

BACKGROUND & AIMS: Acute and chronic gastric injury induces alterations in differentiation within the corpus of the stomach called pyloric metaplasia. Pyloric metaplasia is characterized by the death of parietal cells and reprogramming of mitotically quiescent zymogenic chief cells into proliferative, mucin-rich spasmolytic polypeptide-expressing metaplasia (SPEM) cells. Overall, pyloric metaplastic units show increased proliferation and specific expansion of mucous lineages, both by proliferation of normal mucous neck cells and recruitment of SPEM cells. Here, we identify Sox9 as a potential gene of interest in the regulation of mucous neck and SPEM cell identity in the stomach. METHODS: We used immunostaining and electron microscopy to characterize the expression pattern of SRY-box transcription factor 9 (SOX9) during murine gastric development, homeostasis, and injury in homeostasis, after genetic deletion of Sox9 and after targeted genetic misexpression of Sox9 in the gastric epithelium and chief cells. RESULTS: SOX9 is expressed in all early gastric progenitors and strongly expressed in mature mucous neck cells with minor expression in the other principal gastric lineages during adult homeostasis. After injury, strong SOX9 expression was induced in the neck and base of corpus units in SPEM cells. Adult corpus units derived from Sox9-deficient gastric progenitors lacked normal mucous neck cells. Misexpression of Sox9 during postnatal development and adult homeostasis expanded mucous gene expression throughout corpus units including within the chief cell zone in the base. Sox9 deletion specifically in chief cells blunts their reprogramming into SPEM. CONCLUSIONS: Sox9 is a master regulator of mucous neck cell differentiation during gastric development. Sox9 also is required for chief cells to fully reprogram into SPEM after injury.


Assuntos
Celulas Principais Gástricas , Animais , Camundongos , Celulas Principais Gástricas/metabolismo , Mucosa Gástrica/metabolismo , Metaplasia/metabolismo , Células Parietais Gástricas/metabolismo , Estômago
2.
Dev Cell ; 52(5): 647-658.e6, 2020 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-32155439

RESUMO

During development, intestinal epithelia undergo dramatic morphogenesis mediated by mesenchymal signaling to form villi, which are required for efficient nutrient absorption and host defense. Although both smooth-muscle-induced physical forces and mesenchymal cell clustering beneath emerging villi are implicated in epithelial folding, the underlying cellular mechanisms are unclear. Hedgehog (Hh) signaling can mediate both processes. We therefore analyzed its direct targetome and revealed GLI2 transcriptional activation of atypical cadherin and planar cell polarity (PCP) genes. By examining Fat4 and Dchs1 knockout mice, we demonstrate their critical roles in villus formation. Analyses of PCP-mutant mice and genetic interaction studies show that the Fat4-Dchs1 axis acts in parallel to the core-Vangl2 PCP axis to control mesenchymal cell clustering. Moreover, live light-sheet fluorescence microscopy and cultured PDGFRα+ cells reveal a requirement for PCP in their oriented cell migration guided by WNT5A. Therefore, mesenchymal PCP induced by Hh signaling drives cell clustering and subsequent epithelial remodeling.


Assuntos
Caderinas/metabolismo , Polaridade Celular , Proteínas Hedgehog/metabolismo , Mucosa Intestinal/crescimento & desenvolvimento , Células-Tronco Mesenquimais/metabolismo , Microvilosidades/metabolismo , Animais , Caderinas/genética , Diferenciação Celular , Movimento Celular , Células Cultivadas , Feminino , Proteínas Hedgehog/genética , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Morfogênese , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Transdução de Sinais , Proteína Wnt-5a/genética , Proteína Wnt-5a/metabolismo , Proteína Gli2 com Dedos de Zinco/genética , Proteína Gli2 com Dedos de Zinco/metabolismo
3.
J Pathol ; 249(1): 114-125, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31038742

RESUMO

Autosomal Dominant Polycystic Kidney Disease is characterised by the development of fluid-filled cysts in the kidneys which lead to end-stage renal disease (ESRD). In the majority of cases, the disease is caused by a mutation in the Pkd1 gene. In a previous study, we demonstrated that renal injury can accelerate cyst formation in Pkd1 knock-out (KO) mice. In that study, we found that after injury four-jointed (Fjx1), an upstream regulator of planar cell polarity and the Hippo pathway, was aberrantly expressed in Pkd1 KO mice compared to WT. Therefore, we hypothesised a role for Fjx1 in injury/repair and cyst formation. We generated single and double deletion mice for Pkd1 and Fjx1, and we induced toxic renal injury using the nephrotoxic compound 1,2-dichlorovinyl-cysteine. We confirmed that nephrotoxic injury can accelerate cyst formation in Pkd1 mutant mice. This caused Pkd1 KO mice to reach ESRD significantly faster; unexpectedly, double KO mice survived significantly longer. Cyst formation was comparable in both models, but we found significantly less fibrosis and macrophage infiltration in double KO mice. Taken together, these data suggest that Fjx1 disruption protects the cystic kidneys against kidney failure by reducing inflammation and fibrosis. Moreover, we describe, for the first time, an interesting (yet unidentified) mechanism that partially discriminates cyst growth from fibrogenesis. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Assuntos
Injúria Renal Aguda/complicações , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Falência Renal Crônica/etiologia , Rim/metabolismo , Rim Policístico Autossômico Dominante/complicações , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/genética , Injúria Renal Aguda/metabolismo , Animais , Cisteína/análogos & derivados , Modelos Animais de Doenças , Progressão da Doença , Fibrose , Mediadores da Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Rim/patologia , Falência Renal Crônica/genética , Falência Renal Crônica/metabolismo , Masculino , Camundongos Knockout , Mutação , Rim Policístico Autossômico Dominante/genética , Rim Policístico Autossômico Dominante/metabolismo , Canais de Cátion TRPP/genética , Fatores de Tempo , Via de Sinalização Wnt
4.
Dev Cell ; 48(6): 780-792.e4, 2019 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-30853441

RESUMO

FAT4 mutations lead to several human diseases that disrupt the normal development of the kidney. However, the underlying mechanism remains elusive. In studying the duplex kidney phenotypes observed upon deletion of Fat4 in mice, we have uncovered an interaction between the atypical cadherin FAT4 and RET, a tyrosine kinase receptor essential for kidney development. Analysis of kidney development in Fat4-/- kidneys revealed abnormal ureteric budding and excessive RET signaling. Removal of one copy of the RET ligand Gdnf rescues Fat4-/- kidney development, supporting the proposal that loss of Fat4 hyperactivates RET signaling. Conditional knockout analyses revealed a non-autonomous role for Fat4 in regulating RET signaling. Mechanistically, we found that FAT4 interacts with RET through extracellular cadherin repeats. Importantly, expression of FAT4 perturbs the assembly of the RET-GFRA1-GDNF complex, reducing RET signaling. Thus, FAT4 interacts with RET to fine-tune RET signaling, establishing a juxtacrine mechanism controlling kidney development.


Assuntos
Caderinas/metabolismo , Rim/embriologia , Rim/metabolismo , Proteínas Proto-Oncogênicas c-ret/metabolismo , Transdução de Sinais , Animais , Caderinas/química , Caderinas/deficiência , Deleção de Genes , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Rim/anormalidades , Camundongos , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/metabolismo , Ligação Proteica , Regulação para Cima
5.
J Am Soc Nephrol ; 29(3): 775-783, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29326158

RESUMO

The epithelial Wolffian duct (WD) inserts into the cloaca (primitive bladder) before metanephric kidney development, thereby establishing the initial plumbing for eventual joining of the ureters and bladder. Defects in this process cause common anomalies in the spectrum of congenital anomalies of the kidney and urinary tract (CAKUT). However, developmental, cellular, and molecular mechanisms of WD-cloaca fusion are poorly understood. Through systematic analysis of early WD tip development in mice, we discovered that a novel process of spatiotemporally regulated apoptosis in WD and cloaca was necessary for WD-cloaca fusion. Aberrant RET tyrosine kinase signaling through tyrosine (Y) 1062, to which PI3K- or ERK-activating proteins dock, or Y1015, to which PLCγ docks, has been shown to cause CAKUT-like defects. Cloacal apoptosis did not occur in RetY1062F mutants, in which WDs did not reach the cloaca, or in RetY1015F mutants, in which WD tips reached the cloaca but did not fuse. Moreover, inhibition of ERK or apoptosis prevented WD-cloaca fusion in cultures, and WD-specific genetic deletion of YAP attenuated cloacal apoptosis and WD-cloacal fusion in vivo Thus, cloacal apoptosis requires direct contact and signals from the WD tip and is necessary for WD-cloacal fusion. These findings may explain the mechanisms of many CAKUT.


Assuntos
Apoptose/genética , Cloaca/embriologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas Proto-Oncogênicas c-ret/genética , Anormalidades Urogenitais/genética , Ductos Mesonéfricos/embriologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas de Ciclo Celular , Cloaca/anormalidades , Cloaca/metabolismo , Rim/embriologia , Sistema de Sinalização das MAP Quinases , Camundongos , Mutação , Fosfoproteínas/genética , Proteínas Proto-Oncogênicas c-ret/metabolismo , Ureter/embriologia , Ductos Mesonéfricos/anormalidades , Ductos Mesonéfricos/metabolismo , Proteínas de Sinalização YAP
6.
J Am Soc Nephrol ; 28(3): 852-861, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27647853

RESUMO

In the kidney, formation of the functional filtration units, the nephrons, is essential for postnatal life. During development, mesenchymal progenitors tightly regulate the balance between self-renewal and differentiation to give rise to all nephron epithelia. Here, we investigated the functions of the Hippo pathway serine/threonine-protein kinases Lats1 and Lats2, which phosphorylate and inhibit the transcriptional coactivators Yap and Taz, in nephron progenitor cells. Genetic deletion of Lats1 and Lats2 in nephron progenitors of mice led to disruption of nephrogenesis, with an accumulation of spindle-shaped cells in both cortical and medullary regions of the kidney. Lineage-tracing experiments revealed that the cells that accumulated in the interstitium derived from nephron progenitor cells and expressed E-cadherin as well as vimentin, a myofibroblastic marker not usually detected after mesenchymal-to-epithelial transition. The accumulation of these interstitial cells associated with collagen deposition and ectopic expression of the myofibroblastic markers vimentin and α-smooth-muscle actin in developing kidneys. Although these myofibroblastic cells had high Yap and Taz accumulation in the nucleus concomitant with a loss of phosphorylated Yap, reduction of Yap and/or Taz expression levels completely rescued the Lats1/2 phenotype. Taken together, our results demonstrate that Lats1/2 kinases restrict Yap/Taz activities to promote nephron progenitor cell differentiation in the mammalian kidney. Notably, our data also show that myofibroblastic cells can differentiate from nephron progenitors.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Miofibroblastos/fisiologia , Néfrons/citologia , Fosfoproteínas/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Células-Tronco/fisiologia , Fatores de Transcrição/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Aciltransferases , Animais , Proteínas de Ciclo Celular , Diferenciação Celular , Epitélio , Camundongos , Fosforilação , Proteínas de Sinalização YAP
7.
Nat Commun ; 7: 12309, 2016 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-27480037

RESUMO

Branching morphogenesis is a complex biological process common to the development of most epithelial organs. Here we demonstrate that NF2, LATS1/2 and YAP play a critical role in branching morphogenesis in the mouse kidney. Removal of Nf2 or Lats1/2 from the ureteric bud (UB) lineage causes loss of branching morphogenesis that is rescued by loss of one copy of Yap and Taz, and phenocopied by YAP overexpression. Mosaic analysis demonstrates that cells with high YAP expression have reduced contribution to UB tips, similar to Ret(-/-) cells, and that YAP suppresses RET signalling and tip identity. Conversely, Yap/Taz UB-deletion leads to cyst-like branching and expansion of UB tip markers, suggesting a shift towards tip cell identity. Based on these data we propose that NF2 and the Hippo pathway locally repress YAP/TAZ activity in the UB to promote subsequent splitting of the tip to allow branching morphogenesis.


Assuntos
Rim/metabolismo , Morfogênese/genética , Neurofibromina 2/genética , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Ciclo Celular , Via de Sinalização Hippo , Rim/embriologia , Camundongos Knockout , Camundongos Transgênicos , Neurofibromina 2/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Ureter/embriologia , Ureter/metabolismo , Proteínas de Sinalização YAP
8.
Sci Rep ; 6: 22879, 2016 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-26971715

RESUMO

In addition to their canonical roles in the cell cycle, RB family proteins regulate numerous developmental pathways, although the mechanisms remain obscure. We found that Drosophila Rbf1 associates with genes encoding components of the highly conserved apical-basal and planar cell polarity pathways, suggesting a possible regulatory role. Here, we show that depletion of Rbf1 in Drosophila tissues is indeed associated with polarity defects in the wing and eye. Key polarity genes aPKC, par6, vang, pk, and fmi are upregulated, and an aPKC mutation suppresses the Rbf1-induced phenotypes. RB control of cell polarity may be an evolutionarily conserved function, with important implications in cancer metastasis.


Assuntos
Polaridade Celular/genética , Proteínas de Drosophila/genética , Olho/metabolismo , Interferência de RNA , Fatores de Transcrição/genética , Asas de Animais/metabolismo , Animais , Animais Geneticamente Modificados , Caderinas/genética , Caderinas/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Epistasia Genética , Olho/citologia , Olho/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Discos Imaginais/crescimento & desenvolvimento , Discos Imaginais/metabolismo , Proteínas com Domínio LIM/genética , Proteínas com Domínio LIM/metabolismo , Larva/citologia , Larva/genética , Larva/crescimento & desenvolvimento , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Ligação Proteica , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Proteína do Retinoblastoma , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/metabolismo , Asas de Animais/citologia , Asas de Animais/crescimento & desenvolvimento
9.
PLoS One ; 11(3): e0152259, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27015411

RESUMO

The insulin/IGF-1 signaling pathway mediates various physiological processes associated with human health. Components of this pathway are highly conserved throughout eukaryotic evolution. In Drosophila, the PTEN ortholog and its mammalian counterpart downregulate insulin/IGF signaling by antagonizing the PI3-kinase function. From a dominant loss-of-function genetic screen, we discovered that mutations of a Dbl-family member, the guanine nucleotide exchange factor DRhoGEF2 (DRhoGEF22(l)04291), suppressed the PTEN-overexpression eye phenotype. dAkt/dPKB phosphorylation, a measure of PI3K signaling pathway activation, increased in the eye discs from the heterozygous DRhoGEF2 wandering third instar larvae. Overexpression of DRhoGEF2, and it's functional mammalian ortholog PDZ-RhoGEF (ArhGEF11), at various stages of eye development, resulted in both dPKB/Akt-dependent and -independent phenotypes, reflecting the complexity in the crosstalk between PI3K and Rho signaling in Drosophila.


Assuntos
Proteínas de Drosophila/genética , Drosophila/genética , Olho/crescimento & desenvolvimento , Fosfatidilinositol 3-Quinases/genética , Proteínas rho de Ligação ao GTP/genética , Animais , Proteínas de Ciclo Celular , Drosophila/crescimento & desenvolvimento , Proteínas de Drosophila/biossíntese , Olho/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Insulina/genética , Fator de Crescimento Insulin-Like I/genética , Mutação , Proteínas Proto-Oncogênicas c-akt/genética , Transdução de Sinais , Proteínas rho de Ligação ao GTP/biossíntese
10.
Nat Commun ; 7: 10822, 2016 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-26905694

RESUMO

Steroid-resistant nephrotic syndrome (SRNS) causes 15% of chronic kidney disease (CKD). Here we show that recessive mutations in FAT1 cause a distinct renal disease entity in four families with a combination of SRNS, tubular ectasia, haematuria and facultative neurological involvement. Loss of FAT1 results in decreased cell adhesion and migration in fibroblasts and podocytes and the decreased migration is partially reversed by a RAC1/CDC42 activator. Podocyte-specific deletion of Fat1 in mice induces abnormal glomerular filtration barrier development, leading to podocyte foot process effacement. Knockdown of Fat1 in renal tubular cells reduces migration, decreases active RAC1 and CDC42, and induces defects in lumen formation. Knockdown of fat1 in zebrafish causes pronephric cysts, which is partially rescued by RAC1/CDC42 activators, confirming a role of the two small GTPases in the pathogenesis. These findings provide new insights into the pathogenesis of SRNS and tubulopathy, linking FAT1 and RAC1/CDC42 to podocyte and tubular cell function.


Assuntos
Caderinas/genética , Adesão Celular/genética , Movimento Celular/genética , Fibroblastos/metabolismo , Síndrome Nefrótica/congênito , Podócitos/metabolismo , Proteínas de Peixe-Zebra/genética , Animais , Dilatação Patológica/genética , Técnicas de Silenciamento de Genes , Hematúria/genética , Humanos , Túbulos Renais/citologia , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Lisencefalia/genética , Camundongos , Mutação , Síndrome Nefrótica/genética , Síndrome , Peixe-Zebra , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
11.
Dev Cell ; 36(1): 50-62, 2016 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-26766442

RESUMO

Control of cell-division orientation is integral to epithelial morphogenesis and asymmetric cell division. Proper spatiotemporal localization of the evolutionarily conserved Gαi-LGN-NuMA protein complex is critical for mitotic spindle orientation, but how this is achieved remains unclear. Here we identify Suppressor APC domain containing 2 (SAPCD2) as a previously unreported LGN-interacting protein. We show that SAPCD2 is essential to instruct planar mitotic spindle orientation in both epithelial cell cultures and mouse retinal progenitor cells in vivo. Loss of SAPCD2 randomizes spindle orientation, which in turn disrupts cyst morphogenesis in three-dimensional cultures, and triples the number of terminal asymmetric cell divisions in the developing retina. Mechanistically, we show that SAPCD2 negatively regulates the localization of LGN at the cell cortex, likely by competing with NuMA for its binding. These results uncover SAPCD2 as a key regulator of the ternary complex controlling spindle orientation during morphogenesis and asymmetric cell divisions.


Assuntos
Antígenos Nucleares/metabolismo , Polaridade Celular/fisiologia , Mitose/fisiologia , Proteínas Associadas à Matriz Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Fuso Acromático/metabolismo , Animais , Ciclo Celular/genética , Proteínas de Ciclo Celular , Polaridade Celular/genética , Humanos , Camundongos , Morfogênese/fisiologia , Proteínas Nucleares/genética , Ligação Proteica
12.
Genes Dev ; 30(24): 2696-2709, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-28087714

RESUMO

Disruption of apical-basal polarity is implicated in developmental disorders and cancer; however, the mechanisms connecting cell polarity proteins with intracellular signaling pathways are largely unknown. We determined previously that membrane-associated guanylate kinase (MAGUK) protein discs large homolog 5 (DLG5) functions in cell polarity and regulates cellular proliferation and differentiation via undefined mechanisms. We report here that DLG5 functions as an evolutionarily conserved scaffold and negative regulator of Hippo signaling, which controls organ size through the modulation of cell proliferation and differentiation. Affinity purification/mass spectrometry revealed a critical role of DLG5 in the formation of protein assemblies containing core Hippo kinases mammalian ste20 homologs 1/2 (MST1/2) and Par-1 polarity proteins microtubule affinity-regulating kinases 1/2/3 (MARK1/2/3). Consistent with this finding, Hippo signaling is markedly hyperactive in mammalian Dlg5-/- tissues and cells in vivo and ex vivo and in Drosophila upon dlg5 knockdown. Conditional deletion of Mst1/2 fully rescued the phenotypes of brain-specific Dlg5 knockout mice. Dlg5 also interacts genetically with Hippo effectors Yap1/Taz Mechanistically, we show that DLG5 inhibits the association between MST1/2 and large tumor suppressor homologs 1/2 (LATS1/2), uses its scaffolding function to link MST1/2 with MARK3, and inhibits MST1/2 kinase activity. These data reveal a direct connection between cell polarity proteins and Hippo, which is essential for proper development of multicellular organisms.


Assuntos
Polaridade Celular/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Membrana/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/genética , Proteínas Supressoras de Tumor/metabolismo , Animais , Células Cultivadas , Drosophila/embriologia , Drosophila/enzimologia , Drosophila/genética , Deleção de Genes , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Ligação Proteica , Domínios Proteicos , Proteínas Serina-Treonina Quinases/genética , Proteômica , Interferência de RNA , Proteínas Supressoras de Tumor/genética
13.
J Am Soc Nephrol ; 27(1): 216-26, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26015453

RESUMO

FSGS is the most common primary glomerular disease underlying ESRD in the United States and is increasing in incidence globally. FSGS results from podocyte injury, yet the mechanistic details of disease pathogenesis remain unclear. This has resulted in an unmet clinical need for cell-specific therapy in the treatment of FSGS and other proteinuric kidney diseases. We previously identified Yes-associated protein (YAP) as a prosurvival signaling molecule, the in vitro silencing of which increases podocyte susceptibility to apoptotic stimulus. YAP is a potent oncogene that is a prominent target for chemotherapeutic drug development. In this study, we tested the hypothesis that podocyte-specific deletion of Yap leads to proteinuric kidney disease through increased podocyte apoptosis. Yap was selectively silenced in podocytes using Cre-mediated recombination controlled by the podocin promoter. Yap silencing in podocytes resulted in podocyte apoptosis, podocyte depletion, proteinuria, and an increase in serum creatinine. Histologically, features characteristic of FSGS, including mesangial sclerosis, podocyte foot process effacement, tubular atrophy, interstitial fibrosis, and casts, were observed. In human primary FSGS, we noted reduced glomerular expression of YAP. Taken together, these results suggest a role for YAP as a physiologic antagonist of podocyte apoptosis, the signaling of which is essential for maintaining the integrity of the glomerular filtration barrier. These data suggest potential nephrotoxicity with strategies directed toward inhibition of YAP function. Further studies should evaluate the role of YAP in proteinuric glomerular disease pathogenesis and its potential utility as a therapeutic target.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Deleção de Genes , Glomerulosclerose Segmentar e Focal/genética , Fosfoproteínas/genética , Podócitos/fisiologia , Insuficiência Renal/genética , Animais , Proteínas de Ciclo Celular , Progressão da Doença , Humanos , Camundongos , Camundongos Knockout , Proteínas de Sinalização YAP
14.
Development ; 142(15): 2696-703, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26243870

RESUMO

Despite the high occurrence of congenital abnormalities of the lower urinary tract in humans, the molecular, cellular and morphological aspects of their development are still poorly understood. Here, we use a conditional knockout approach to inactivate within the nephric duct (ND) lineage the two effectors of the Hippo pathway, Yap and Taz. Deletion of Yap leads to hydronephrotic kidneys with blind-ending megaureters at birth. In Yap mutants, the ND successfully migrates towards, and contacts, the cloaca. However, close analysis reveals that the tip of the Yap(-/-) ND forms an aberrant connection with the cloaca and does not properly insert into the cloaca, leading to later detachment of the ND from the cloaca. Taz deletion from the ND does not cause any defect, but analysis of Yap(-/-);Taz(-/-) NDs indicates that both genes play partially redundant roles in ureterovesical junction formation. Aspects of the Yap(-/-) phenotype resemble hypersensitivity to RET signaling, including excess budding of the ND, increased phospho-ERK and increased expression of Crlf1, Sprouty1, Etv4 and Etv5. Importantly, the Yap(ND) (-/-) ND phenotype can be largely rescued by reducing Ret gene dosage. Taken together, these results suggest that disrupting Yap/Taz activities enhances Ret pathway activity and contributes to pathogenesis of lower urinary tract defects in human infants.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Morfogênese/fisiologia , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-ret/metabolismo , Transdução de Sinais/fisiologia , Sistema Urinário/embriologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Bromodesoxiuridina , Proteínas de Ciclo Celular , Imunofluorescência , Galactosídeos , Técnicas de Inativação de Genes , Humanos , Imuno-Histoquímica , Hibridização In Situ , Marcação In Situ das Extremidades Cortadas , Indóis , Camundongos , Fosfoproteínas/genética , Transativadores , Proteínas de Sinalização YAP
15.
Invest Ophthalmol Vis Sci ; 56(6): 4099-107, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26114487

RESUMO

PURPOSE: The Fat family of atypical cadherins, originally identified in Drosophila, play diverse roles during embryogenesis and adult tissue maintenance. Among four mammalian members, Fat1 is essential for kidney and muscle organization, and is also essential for eye development; Fat1 knockout causes partial penetrant microphthalmia or anophthalmia. To account for the partial penetrance of the Fat1 phenotype, involvement of Fat4 in eye development was assessed. Lens phenotypes in Fat1 and 4 knockouts were also examined. METHODS: Fat1 and Fat4 mRNA expression was examined by in situ hybridization. Knockout phenotypes of Fat1 and Fat4 were analyzed by hematoxylin and eosin (H&E) and immunofluorescent staining. RESULTS: We found Fat4 knockout did not affect eye induction or enhance severity of Fat1 eye defects. Although Fat1 and Fat4 mRNAs are similarly expressed in the lens epithelial cells, only Fat1 knockout caused a fully penetrant lens epithelial cell defect, which was apparent at embryonic day 14.5 (E14.5). The columnar structure of the lens epithelial cells was disrupted and in some regions cell aggregates were formed. In these multilayered regions, apical cell junctions were fragmented and the apical-basal polarity was lost. EdU incorporation assay also showed enhanced proliferation in the lens epithelial cells. Interestingly, these defects were found mainly in the central zone of the epithelial layer. The lens epithelial cells of the germinative zone maintained their normal morphology and fiber differentiation occurred normally at the equator. CONCLUSIONS: These observations indicate that Fat1 is essential for lens epithelial cell polarity and proliferation but not for terminal differentiation.


Assuntos
Caderinas/metabolismo , Polaridade Celular/fisiologia , Proliferação de Células/fisiologia , Células Epiteliais/fisiologia , Cristalino/metabolismo , Animais , Caderinas/genética , Diferenciação Celular/fisiologia , Modelos Animais de Doenças , Junções Intercelulares/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/metabolismo
16.
Cell ; 158(6): 1293-1308, 2014 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-25215488

RESUMO

Fat (Ft) cadherins are enormous cell adhesion molecules that function at the cell surface to regulate the tumor-suppressive Hippo signaling pathway and planar cell polarity (PCP) tissue organization. Mutations in Ft cadherins are found in a variety of tumors, and it is presumed that this is due to defects in either Hippo signaling or PCP. Here, we show Drosophila Ft functions in mitochondria to directly regulate mitochondrial electron transport chain integrity and promote oxidative phosphorylation. Proteolytic cleavage releases a soluble 68 kDa fragment (Ft(mito)) that is imported into mitochondria. Ft(mito) binds directly to NADH dehydrogenase ubiquinone flavoprotein 2 (Ndufv2), a core component of complex I, stabilizing the holoenzyme. Loss of Ft leads to loss of complex I activity, increases in reactive oxygen species, and a switch to aerobic glycolysis. Defects in mitochondrial activity in ft mutants are independent of Hippo and PCP signaling and are reminiscent of the Warburg effect.


Assuntos
Caderinas/metabolismo , Moléculas de Adesão Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Mitocôndrias/metabolismo , Sequência de Aminoácidos , Animais , Moléculas de Adesão Celular/química , Polaridade Celular , Proteínas de Drosophila/química , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Complexo I de Transporte de Elétrons/metabolismo , Olho/crescimento & desenvolvimento , Genes Supressores de Tumor , Humanos , MAP Quinase Quinase 4/metabolismo , Dados de Sequência Molecular , Transporte Proteico , Espécies Reativas de Oxigênio/metabolismo , Asas de Animais/crescimento & desenvolvimento
17.
Sci Signal ; 6(296): re7, 2013 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-24106343

RESUMO

The Hippo pathway is a kinase cascade, formed by Hippo, Salvador, Warts, and Mats, that regulates the subcellular distribution and transcriptional activity of Yorkie. Yorkie is a transcriptional coactivator that promotes the expression of genes that inhibit apoptosis and drive cell proliferation. We review recent studies indicating that activity of the Hippo pathway is controlled by cell-cell junctions, cell adhesion molecules, scaffolding proteins, and cytoskeletal proteins, as well as by regulators of apical-basal polarity and extracellular tension.


Assuntos
Proteínas de Drosophila/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/fisiologia , Animais , Apoptose/fisiologia , Moléculas de Adesão Celular/biossíntese , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Polaridade Celular/fisiologia , Proliferação de Células , Proteínas de Drosophila/genética , Drosophila melanogaster , Regulação da Expressão Gênica/fisiologia , Junções Intercelulares/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
18.
Nat Genet ; 45(11): 1300-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24056717

RESUMO

The regulated proliferation and differentiation of neural stem cells before the generation and migration of neurons in the cerebral cortex are central aspects of mammalian development. Periventricular neuronal heterotopia, a specific form of mislocalization of cortical neurons, can arise from neuronal progenitors that fail to negotiate aspects of these developmental processes. Here we show that mutations in genes encoding the receptor-ligand cadherin pair DCHS1 and FAT4 lead to a recessive syndrome in humans that includes periventricular neuronal heterotopia. Reducing the expression of Dchs1 or Fat4 within mouse embryonic neuroepithelium increased progenitor cell numbers and reduced their differentiation into neurons, resulting in the heterotopic accumulation of cells below the neuronal layers in the neocortex, reminiscent of the human phenotype. These effects were countered by concurrent knockdown of Yap, a transcriptional effector of the Hippo signaling pathway. These findings implicate Dchs1 and Fat4 upstream of Yap as key regulators of mammalian neurogenesis.


Assuntos
Caderinas/genética , Córtex Cerebral/embriologia , Células-Tronco Neurais/metabolismo , Neurogênese/genética , Proteínas Supressoras de Tumor/genética , Anormalidades Múltiplas/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Sequência de Bases , Proteínas Relacionadas a Caderinas , Proteínas de Ciclo Celular , Diferenciação Celular , Proliferação de Células , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Anormalidades Craniofaciais/genética , Deformidades Congênitas do Pé/genética , Técnicas de Silenciamento de Genes , Deformidades Congênitas da Mão/genética , Humanos , Deficiência Intelectual/genética , Instabilidade Articular/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Heterotopia Nodular Periventricular/genética , Fosfoproteínas/genética , Análise de Sequência de DNA , Transdução de Sinais/genética , Proteínas de Sinalização YAP
19.
Development ; 139(10): 1806-20, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22510986

RESUMO

The atypical cadherin fat (ft) was originally discovered as a tumor suppressor in Drosophila and later shown to regulate a form of tissue patterning known as planar polarity. In mammals, four ft homologs have been identified (Fat1-4). Recently, we demonstrated that Fat4 plays a role in vertebrate planar polarity. Fat4 has the highest homology to ft, whereas other Fat family members are homologous to the second ft-like gene, ft2. Genetic studies in flies and mice imply significant functional differences between the two groups of Fat cadherins. Here, we demonstrate that Fat family proteins act both synergistically and antagonistically to influence multiple aspects of tissue morphogenesis. We find that Fat1 and Fat4 cooperate during mouse development to control renal tubular elongation, cochlear extension, cranial neural tube formation and patterning of outer hair cells in the cochlea. Similarly, Fat3 and Fat4 synergize to drive vertebral arch fusion at the dorsal midline during caudal vertebra morphogenesis. We provide evidence that these effects depend on conserved interactions with planar polarity signaling components. In flies, the transcriptional co-repressor Atrophin (Atro) physically interacts with Ft and acts as a component of Fat signaling for planar polarity. We find that the mammalian orthologs of atro, Atn1 and Atn2l, modulate Fat4 activity during vertebral arch fusion and renal tubular elongation, respectively. Moreover, Fat4 morphogenetic defects are enhanced by mutations in Vangl2, a 'core' planar cell polarity gene. These studies highlight the wide range and complexity of Fat activities and suggest that a Fat-Atrophin interaction is a conserved element of planar polarity signaling.


Assuntos
Caderinas/metabolismo , Polaridade Celular/fisiologia , Morfogênese/fisiologia , Animais , Caderinas/genética , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Polaridade Celular/genética , Cóclea/embriologia , Cóclea/metabolismo , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Camundongos , Morfogênese/genética , Ligação Proteica/genética , Ligação Proteica/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
20.
Mol Cell Biol ; 32(1): 173-85, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22037766

RESUMO

ß-Catenin-independent Wnt signaling pathways have been implicated in the regulation of planar cell polarity (PCP) and convergent extension (CE) cell movements. Prickle, one of the core proteins of these pathways, is known to asymmetrically localize proximally at the adherens junction of Drosophila melanogaster wing cells and to locally accumulate within plasma membrane subdomains in cells undergoing CE movements during vertebrate development. Using mass spectrometry, we have identified the Ste20 kinase Mink1 as a Prickle-associated protein and found that they genetically interact during the establishment of PCP in the Drosophila eye and CE in Xenopus laevis embryos. We show that Mink1 phosphorylates Prickle on a conserved threonine residue and regulates its Rab5-dependent endosomal trafficking, a process required for the localized plasma membrane accumulation and function of Prickle. Mink1 also was found to be important for the clustering of Vangl within plasma membrane puncta. Our results provide a link between Mink and the Vangl-Prickle complex and highlight the importance of Prickle phosphorylation and endosomal trafficking for its function during Wnt-PCP signaling.


Assuntos
Proteínas com Domínio LIM/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Endossomos/metabolismo , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/análise , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas com Domínio LIM/análise , Proteínas de Membrana/análise , Proteínas de Membrana/metabolismo , Dados de Sequência Molecular , Fosforilação , Proteínas Serina-Treonina Quinases/análise , Transporte Proteico , Proteínas Supressoras de Tumor/análise , Xenopus/embriologia , Xenopus/metabolismo , Proteínas de Xenopus/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA