Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Development ; 149(3)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35112129

RESUMO

The tracheal epithelium is a primary target for pulmonary diseases as it provides a conduit for air flow between the environment and the lung lobes. The cellular and molecular mechanisms underlying airway epithelial cell proliferation and differentiation remain poorly understood. Hedgehog (HH) signaling orchestrates communication between epithelial and mesenchymal cells in the lung, where it modulates stromal cell proliferation, differentiation and signaling back to the epithelium. Here, we reveal a previously unreported autocrine function of HH signaling in airway epithelial cells. Epithelial cell depletion of the ligand sonic hedgehog (SHH) or its effector smoothened (SMO) causes defects in both epithelial cell proliferation and differentiation. In cultured primary human airway epithelial cells, HH signaling inhibition also hampers cell proliferation and differentiation. Epithelial HH function is mediated, at least in part, through transcriptional activation, as HH signaling inhibition leads to downregulation of cell type-specific transcription factor genes in both the mouse trachea and human airway epithelial cells. These results provide new insights into the role of HH signaling in epithelial cell proliferation and differentiation during airway development.


Assuntos
Comunicação Autócrina/fisiologia , Diferenciação Celular , Proliferação de Células , Proteínas Hedgehog/metabolismo , Transdução de Sinais/genética , Animais , Células Cultivadas , Regulação para Baixo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Proteínas Hedgehog/deficiência , Proteínas Hedgehog/genética , Humanos , Pulmão/crescimento & desenvolvimento , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Knockout , Receptor Smoothened/deficiência , Receptor Smoothened/genética , Receptor Smoothened/metabolismo , Traqueia/citologia , Traqueia/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
2.
Mol Genet Genomics ; 296(1): 33-40, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32944789

RESUMO

Joubert syndrome (JBTS), a rare genetic disorder resulted from primary cilium defects or basal-body dysfunction, is characterized by agenesis of cerebellar vermis and abnormal brain stem. Both genotypes and phenotypes of JBTS are highly heterogeneous. The identification of pathogenic gene variation is essential for making a definite diagnosis on JBTS. Here, we found that hypoplasia of cerebellar vermis occurred in three male members in a Chinese family. Then, we performed whole exome sequencing to identify a novel missense mutation c.599T > C (p. L200P) in the OFD1 gene which is the candidate gene of X-linked JBTS (JBST10). The following analysis showed that the variant was absent in the 1000 Genomes, ExAC and the 200 female controls; the position 200 Leucine residue was highly conserved across species; the missense variant was predicted to be deleterious using PolyPhen-2, PROVEAN, SIFT and Mutation Taster. The OFD1 expression was heavily lower in the proband and an induced male fetus compared with a healthy male with a wild-type OFD1 gene. The in vitro expression analysis of transiently transfecting c.599T or c.599C plasmids into HEK-293T cells confirmed that the missense mutation caused OFD1 reduction at the protein level. And further the mutated OFD1 decreased the level of Gli1 protein, a read-out of Sonic hedgehog (SHH) signaling essential for development of central neural system. A known pathogenic variant c.515T > C (p. L172P) showed the similar results. All of these observations suggested that the missense mutation causes the loss function of OFD1, resulting in SHH signaling impairs and brain development abnormality. In addition, the three patients have Dandy-Walker malformation, macrogyria and tetralogy of Fallot, respectively, the latter two of which are firstly found in JBTS10 patients. In conclusion, our findings expand the context of genotype and phenotype in the JBTS10 patients.


Assuntos
Anormalidades Múltiplas/genética , Cerebelo/anormalidades , Síndrome de Dandy-Walker/genética , Anormalidades do Olho/genética , Doenças Renais Císticas/genética , Lisencefalia/genética , Mutação de Sentido Incorreto , Proteínas/genética , Retina/anormalidades , Tetralogia de Fallot/genética , Anormalidades Múltiplas/diagnóstico por imagem , Anormalidades Múltiplas/metabolismo , Anormalidades Múltiplas/patologia , Sequência de Aminoácidos , Tronco Encefálico/anormalidades , Tronco Encefálico/diagnóstico por imagem , Tronco Encefálico/metabolismo , Vermis Cerebelar/anormalidades , Vermis Cerebelar/diagnóstico por imagem , Vermis Cerebelar/metabolismo , Cerebelo/diagnóstico por imagem , Cerebelo/metabolismo , Cerebelo/patologia , Pré-Escolar , Síndrome de Dandy-Walker/diagnóstico por imagem , Síndrome de Dandy-Walker/metabolismo , Síndrome de Dandy-Walker/patologia , Anormalidades do Olho/diagnóstico por imagem , Anormalidades do Olho/metabolismo , Anormalidades do Olho/patologia , Família , Feminino , Expressão Gênica , Genótipo , Células HEK293 , Proteínas Hedgehog/deficiência , Proteínas Hedgehog/genética , Humanos , Doenças Renais Císticas/diagnóstico por imagem , Doenças Renais Císticas/metabolismo , Doenças Renais Císticas/patologia , Lisencefalia/diagnóstico por imagem , Lisencefalia/metabolismo , Lisencefalia/patologia , Masculino , Linhagem , Fenótipo , Proteínas/metabolismo , Retina/diagnóstico por imagem , Retina/metabolismo , Retina/patologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Fatores Sexuais , Transdução de Sinais , Tetralogia de Fallot/diagnóstico por imagem , Tetralogia de Fallot/metabolismo , Tetralogia de Fallot/patologia , Proteína GLI1 em Dedos de Zinco/deficiência , Proteína GLI1 em Dedos de Zinco/genética
3.
Commun Biol ; 3(1): 144, 2020 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-32214226

RESUMO

The Creeper (Cp) chicken is characterized by chondrodystrophy in Cp/+ heterozygotes and embryonic lethality in Cp/Cp homozygotes. However, the genes underlying the phenotypes have not been fully known. Here, we show that a 25 kb deletion on chromosome 7, which contains the Indian hedgehog (IHH) and non-homologous end-joining factor 1 (NHEJ1) genes, is responsible for the Cp trait in Japanese bantam chickens. IHH is essential for chondrocyte maturation and is downregulated in the Cp/+ embryos and completely lost in the Cp/Cp embryos. This indicates that chondrodystrophy is caused by the loss of IHH and that chondrocyte maturation is delayed in Cp/+ heterozygotes. The Cp/Cp homozygotes exhibit impaired DNA double-strand break (DSB) repair due to the loss of NHEJ1, resulting in DSB accumulation in the vascular and nervous systems, which leads to apoptosis and early embryonic death.


Assuntos
Doenças do Desenvolvimento Ósseo/veterinária , Osso e Ossos/embriologia , Enzimas Reparadoras do DNA/genética , Proteínas de Ligação a DNA/genética , Deleção de Genes , Proteínas Hedgehog/genética , Doenças das Aves Domésticas/genética , Animais , Apoptose , Doenças do Desenvolvimento Ósseo/embriologia , Doenças do Desenvolvimento Ósseo/genética , Doenças do Desenvolvimento Ósseo/metabolismo , Osso e Ossos/metabolismo , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Embrião de Galinha , Enzimas Reparadoras do DNA/deficiência , Proteínas de Ligação a DNA/deficiência , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Predisposição Genética para Doença , Proteínas Hedgehog/deficiência , Heterozigoto , Homozigoto , Fenótipo , Doenças das Aves Domésticas/embriologia , Doenças das Aves Domésticas/metabolismo
4.
Circ Res ; 123(9): 1053-1065, 2018 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-30355159

RESUMO

RATIONALE: Klf (kruppel-like factor) 2 is critical to establish and maintain endothelial integrity. OBJECTIVE: Therefore, determining upstream and downstream mediators of Klf2 would lead to alternative therapeutic targets in cardiovascular disease management. METHODS AND RESULTS: Here we identify Dhh (desert hedgehog) as a downstream effector of Klf2, whose expression in endothelial cells (ECs) is upregulated by shear stress and decreased by inflammatory cytokines. Consequently, we show that Dhh knockdown in ECs promotes endothelial permeability and EC activation and that Dhh agonist prevents TNF-α (tumor necrosis factor alpha) or glucose-induced EC dysfunction. Moreover, we demonstrate that human critical limb ischemia, a pathological condition linked to diabetes mellitus and inflammation, is associated to major EC dysfunction. By recreating a complex model of critical limb ischemia in diabetic mice, we found that Dhh-signaling agonist significantly improved EC function without promoting angiogenesis, which subsequently improved muscle perfusion. CONCLUSION: Restoring EC function leads to significant critical limb ischemia recovery. Dhh appears to be a promising target, downstream of Klf2, to prevent the endothelial dysfunction involved in ischemic vascular diseases.


Assuntos
Células Endoteliais/metabolismo , Proteínas Hedgehog/metabolismo , Isquemia/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Músculo Esquelético/irrigação sanguínea , Neovascularização Fisiológica , Animais , Comunicação Autócrina , Permeabilidade Capilar , Células Cultivadas , Estado Terminal , Cicloexilaminas/farmacologia , Citocinas/metabolismo , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Regulação da Expressão Gênica , Proteínas Hedgehog/deficiência , Proteínas Hedgehog/genética , Membro Posterior , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Isquemia/tratamento farmacológico , Isquemia/genética , Isquemia/fisiopatologia , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Fisiológica/efeitos dos fármacos , Fluxo Sanguíneo Regional , Transdução de Sinais , Estresse Mecânico , Tiofenos/farmacologia
5.
FASEB J ; 32(10): 5703-5715, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29768039

RESUMO

Sonic hedgehog ( Shh) is crucial for organogenesis in the foregut. This study investigated the function of Shh at the late-gestational stage; during which, the esophagus continues to differentiate. We established cytokeratin 14 ( CK14)-Cre;Shhfl/fl mice in which the down-regulation of Shh in the epithelium occurred at approximately the same time as esophageal muscle conversion. Hematoxylin and eosin and immunohistochemical staining, with antibodies against keratin 14, Shh, patched 1 (Ptch1), Gli1, proliferating cell nuclear antigen (PCNA), α-smooth muscle actin (αSMA), high-molecular-weight caldesmon (hCD), myogenin, paired box 7 (Pax7), ß3-tubulin, and protein gene product 9.5 (PGP9.5), was performed to detect specific tissue dysplasia. Organ culture was conducted in vitro, and total mRNA was extracted to determine the transcriptional dysregulation. The esophagus of CK14-Cre;Shhfl/fl mice developed into an independent tube with an obvious dilatation at postnatal d 0.5. The number of cell layers and the expression of PCNA were decreased in mutant mice, compared with those in wild-type mice. The expression of hCD declined progressively in the middle, distal, and lower esophageal sphincter levels of the mutant esophagus from embryonic d 17.5, compared with the expression in wild-type littermates. Pax7 accumulation and myogenin reduction in mutant mice indicated that esophageal skeletal-myoblast progression was blocked. RNA sequencing analysis revealed a significant down-regulation of genes involved in proliferation and muscular motivation in CK14-Cre;Shhfl/fl mice. Thus, loss of Shh at the late-gestational stage leads to megaesophagus with reduced proliferation and a muscle development disorder in mice.-Jia, X., Min, L., Zhu, S., Zhang, S., Huang, X. Loss of sonic hedgehog gene leads to muscle development disorder and megaesophagus in mice.


Assuntos
Acalasia Esofágica , Proteínas Hedgehog/deficiência , Proteínas Musculares , Músculo Esquelético , Doenças Musculares , Mutação , Animais , Animais Geneticamente Modificados , Acalasia Esofágica/embriologia , Acalasia Esofágica/genética , Acalasia Esofágica/metabolismo , Acalasia Esofágica/patologia , Camundongos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/embriologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Doenças Musculares/epidemiologia , Doenças Musculares/genética , Doenças Musculares/metabolismo , Doenças Musculares/patologia
6.
Cardiovasc Res ; 114(5): 759-770, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29365079

RESUMO

Aims: Hedgehog (Hh) signalling has been shown to be re-activated in ischaemic tissues and participate in ischaemia-induced angiogenesis. Sonic Hedgehog (Shh) is upregulated by more than 80-fold in the ischaemic skeletal muscle, however its specific role in ischaemia-induced angiogenesis has not yet been fully investigated. The purpose of the present study was to investigate the role of endogenous Shh in ischaemia-induced angiogenesis. Methods and results: To this aim, we used inducible Shh knock-out (KO) mice and unexpectedly found that capillary density was significantly increased in re-generating muscle of Shh deficient mice 5 days after hind limb ischaemia was induced, demonstrating that endogenous Shh does not promote angiogenesis but more likely limits it. Myosin and MyoD expression were equivalent in Shh deficient mice and control mice, indicating that endogenous Shh is not required for ischaemia-induced myogenesis. Additionally, we observed a significant increase in macrophage infiltration in the ischaemic muscle of Shh deficient mice. Our data indicate that this was due to an increase in chemokine expression by myoblasts in the setting of impaired Hh signalling, using tissue specific Smoothened conditional KO mice. The increased macrophage infiltration in mice deficient for Hh signalling in myocytes was associated with increased VEGFA expression and a transiently increased angiogenesis, demonstrating that Shh limits inflammation and angiogenesis indirectly by signalling to myocytes. Conclusion: Although ectopic administration of Shh has previously been shown to promote ischaemia-induced angiogenesis, the present study reveals that endogenous Shh does not promote ischaemia-induced angiogenesis. On the contrary, the absence of Shh leads to aberrant ischaemic tissue inflammation and a transiently increased angiogenesis.


Assuntos
Proteínas Hedgehog/metabolismo , Inflamação/metabolismo , Isquemia/metabolismo , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/metabolismo , Neovascularização Fisiológica , Animais , Velocidade do Fluxo Sanguíneo , Quimiocinas/metabolismo , Quimiotaxia , Modelos Animais de Doenças , Proteínas Hedgehog/deficiência , Proteínas Hedgehog/genética , Membro Posterior , Inflamação/genética , Inflamação/fisiopatologia , Inflamação/prevenção & controle , Isquemia/genética , Isquemia/fisiopatologia , Macrófagos/metabolismo , Camundongos Knockout , Mioblastos Esqueléticos/metabolismo , Fluxo Sanguíneo Regional , Transdução de Sinais , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Nat Genet ; 49(10): 1539-1545, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28846100

RESUMO

Copy number variations (CNVs) often include noncoding sequences and putative enhancers, but how these rearrangements induce disease is poorly understood. Here we investigate CNVs involving the regulatory landscape of IHH (encoding Indian hedgehog), which cause multiple, highly localized phenotypes including craniosynostosis and synpolydactyly. We show through transgenic reporter and genome-editing studies in mice that Ihh is regulated by a constellation of at least nine enhancers with individual tissue specificities in the digit anlagen, growth plates, skull sutures and fingertips. Consecutive deletions, resulting in growth defects of the skull and long bones, showed that these enhancers function in an additive manner. Duplications, in contrast, caused not only dose-dependent upregulation but also misexpression of Ihh, leading to abnormal phalanges, fusion of sutures and syndactyly. Thus, precise spatiotemporal control of developmental gene expression is achieved by complex multipartite enhancer ensembles. Alterations in the composition of such clusters can result in gene misexpression and disease.


Assuntos
Doenças do Desenvolvimento Ósseo/genética , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas Hedgehog/fisiologia , Osteogênese/genética , Animais , Sequência de Bases , Variações do Número de Cópias de DNA , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Deformidades Congênitas do Pé/genética , Deleção de Genes , Dosagem de Genes , Duplicação Gênica , Técnicas de Inativação de Genes , Genes Reporter , Proteínas Hedgehog/deficiência , Proteínas Hedgehog/genética , Camundongos , Camundongos Endogâmicos C57BL , Polidactilia/genética , Sequências Reguladoras de Ácido Nucleico , Análise de Sequência de DNA , Crânio/anormalidades , Transcrição Gênica
8.
Cell Rep ; 19(4): 809-821, 2017 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-28445731

RESUMO

During development and regeneration, matrix progenitors undergo terminal differentiation to form the concentric layers of the hair follicle. These differentiation events are thought to require signals from the mesenchymal dermal papilla (DP); however, it remains unclear how DP-progenitor cell interactions govern specific cell fate decisions. Here, we show that the hair follicle differentiated layers are specified asynchronously, with early matrix progenitors initiating differentiation before surrounding the DP. Furthermore, these early matrix cells can undergo terminal differentiation in the absence of Shh, BMP signaling, and DP maturation. Whereas early matrix progenitors form the hair follicle companion layer, later matrix populations progressively form the inner root sheath and hair shaft. Altogether, our findings characterize some of the earliest terminal differentiation events in the hair follicle and reveal that the matrix progenitor pool can be divided into early and late phases based on distinct temporal, molecular, and functional characteristics.


Assuntos
Diferenciação Celular/fisiologia , Folículo Piloso/citologia , Células-Tronco Mesenquimais/citologia , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Derme/citologia , Fator de Transcrição GATA3/metabolismo , Folículo Piloso/metabolismo , Proteínas Hedgehog/deficiência , Proteínas Hedgehog/genética , Queratina-6/metabolismo , Queratinas/genética , Queratinas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais
9.
PLoS One ; 11(2): e0150263, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26918743

RESUMO

The developing long bone is a model of endochondral ossification that displays the morphological layers of chondrocytes toward the ossification center of the diaphysis. Indian hedgehog (Ihh), a member of the hedgehog family of secreted molecules, regulates chondrocyte proliferation and differentiation, as well as osteoblast differentiation, through the process of endochondral ossification. Here, we report that the basic helix-loop-helix transcription factor Hand1, which is expressed in the cartilage primordia, is involved in proper osteogenesis of the bone collar via its control of Ihh production. Genetic overexpression of Hand1 in the osteochondral progenitors resulted in prenatal hypoplastic or aplastic ossification in the diaphyses, mimicking an Ihh loss-of-function phenotype. Ihh expression was downregulated in femur epiphyses of Hand1-overexpressing mice. We also confirmed that Hand1 downregulated Ihh gene expression in vitro by inhibiting Runx2 transactivation of the Ihh proximal promoter. These results demonstrate that Hand1 in chondrocytes regulates endochondral ossification, at least in part through the Runx2-Ihh axis.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Subunidade alfa 1 de Fator de Ligação ao Core/antagonistas & inibidores , Proteínas Hedgehog/fisiologia , Osteogênese/fisiologia , Ativação Transcricional , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Osso e Ossos/embriologia , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Linhagem Celular , Subunidade alfa 1 de Fator de Ligação ao Core/fisiologia , Diáfises , Regulação para Baixo , Feminino , Genes Reporter , Lâmina de Crescimento/metabolismo , Proteínas Hedgehog/biossíntese , Proteínas Hedgehog/deficiência , Proteínas Hedgehog/genética , Deformidades Congênitas dos Membros/genética , Masculino , Camundongos , Camundongos Transgênicos , Osteogênese/genética , Osteopontina/biossíntese , Osteopontina/genética , Fenótipo , Regiões Promotoras Genéticas/genética , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras/genética , Transdução de Sinais/fisiologia , Transfecção , Proteína 1 Relacionada a Twist/genética
10.
Nature ; 526(7574): 578-82, 2015 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-26436454

RESUMO

Postnatal tissue quiescence is thought to be a default state in the absence of a proliferative stimulus such as injury. Although previous studies have demonstrated that certain embryonic developmental programs are reactivated aberrantly in adult organs to drive repair and regeneration, it is not well understood how quiescence is maintained in organs such as the lung, which displays a remarkably low level of cellular turnover. Here we demonstrate that quiescence in the adult lung is an actively maintained state and is regulated by hedgehog signalling. Epithelial-specific deletion of sonic hedgehog (Shh) during postnatal homeostasis in the murine lung results in a proliferative expansion of the adjacent lung mesenchyme. Hedgehog signalling is initially downregulated during the acute phase of epithelial injury as the mesenchyme proliferates in response, but returns to baseline during injury resolution as quiescence is restored. Activation of hedgehog during acute epithelial injury attenuates the proliferative expansion of the lung mesenchyme, whereas inactivation of hedgehog signalling prevents the restoration of quiescence during injury resolution. Finally, we show that hedgehog also regulates epithelial quiescence and regeneration in response to injury via a mesenchymal feedback mechanism. These results demonstrate that epithelial-mesenchymal interactions coordinated by hedgehog actively maintain postnatal tissue homeostasis, and deregulation of hedgehog during injury leads to aberrant repair and regeneration in the lung.


Assuntos
Proteínas Hedgehog/metabolismo , Lesão Pulmonar/metabolismo , Lesão Pulmonar/patologia , Pulmão/citologia , Pulmão/metabolismo , Regeneração , Cicatrização , Animais , Proliferação de Células , Regulação para Baixo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Retroalimentação Fisiológica , Proteínas Hedgehog/deficiência , Proteínas Hedgehog/genética , Homeostase , Pulmão/patologia , Masculino , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Comunicação Parácrina
11.
Exp Neurol ; 271: 84-94, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25936873

RESUMO

The secreted protein desert hedgehog (dhh) controls the formation of the nerve perineurium during development and is a key component of Schwann cells that ensures peripheral nerve survival. We postulated that dhh may play a critical role in maintaining myelination and investigated its role in demyelination-induced compression neuropathies by using a post-natal model of a chronic nerve injury in wildtype and dhh(-/-) mice. We evaluated demyelination using electrophysiological, morphological, and molecular approaches. dhh transcripts and protein are down-regulated early after injury in wild-type mice, suggesting an intimate relationship between the hedgehog pathway and demyelination. In dhh(-/-) mice, nerve injury induced more prominent and severe demyelination relative to their wild-type counterparts, suggesting a protective role of dhh. Alterations in nerve fiber characteristics included significant decreases in nerve conduction velocity, increased myelin debris, and substantial decreases in internodal length. Furthermore, in vitro studies showed that dhh blockade via either adenovirus-mediated (shRNA) or pharmacological inhibition both resulted in severe demyelination, which could be rescued by exogenous Dhh. Exogenous Dhh was protective against this demyelination and maintained myelination at baseline levels in a custom in vitro bioreactor to applied biophysical forces to myelinated DRG/Schwann cell co-cultures. Together, these results demonstrate a pivotal role for dhh in maintaining myelination. Furthermore, dhh signaling reveals a potential target for therapeutic intervention to prevent and treat demyelination of peripheral nerves in compression neuropathies.


Assuntos
Artrogripose/complicações , Doenças Desmielinizantes/etiologia , Doenças Desmielinizantes/metabolismo , Regulação da Expressão Gênica/fisiologia , Proteínas Hedgehog/deficiência , Proteínas Hedgehog/metabolismo , Neuropatia Hereditária Motora e Sensorial/complicações , Animais , Animais Recém-Nascidos , Axônios/patologia , Técnicas de Cultura de Células , Modelos Animais de Doenças , Embrião de Mamíferos , Lateralidade Funcional/genética , Gânglios Espinais/citologia , Proteínas Hedgehog/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Bainha de Mielina/metabolismo , Bainha de Mielina/patologia , Condução Nervosa/genética , Neurônios/fisiologia , Nervos Periféricos/patologia , Nervos Periféricos/ultraestrutura , Ratos , Ratos Sprague-Dawley , Células de Schwann/fisiologia
12.
PLoS Biol ; 13(3): e1002119, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25826604

RESUMO

During nervous system development, gradients of Sonic Hedgehog (Shh) and Netrin-1 attract growth cones of commissural axons toward the floor plate of the embryonic spinal cord. Mice defective for either Shh or Netrin-1 signaling have commissural axon guidance defects, suggesting that both Shh and Netrin-1 are required for correct axon guidance. However, how Shh and Netrin-1 collaborate to guide axons is not known. We first quantified the steepness of the Shh gradient in the spinal cord and found that it is mostly very shallow. We then developed an in vitro microfluidic guidance assay to simulate these shallow gradients. We found that axons of dissociated commissural neurons respond to steep but not shallow gradients of Shh or Netrin-1. However, when we presented axons with combined Shh and Netrin-1 gradients, they had heightened sensitivity to the guidance cues, turning in response to shallower gradients that were unable to guide axons when only one cue was present. Furthermore, these shallow gradients polarized growth cone Src-family kinase (SFK) activity only when Shh and Netrin-1 were combined, indicating that SFKs can integrate the two guidance cues. Together, our results indicate that Shh and Netrin-1 synergize to enable growth cones to sense shallow gradients in regions of the spinal cord where the steepness of a single guidance cue is insufficient to guide axons, and we identify a novel type of synergy that occurs when the steepness (and not the concentration) of a guidance cue is limiting.


Assuntos
Cones de Crescimento/efeitos dos fármacos , Proteínas Hedgehog/farmacologia , Fatores de Crescimento Neural/farmacologia , Medula Espinal/efeitos dos fármacos , Proteínas Supressoras de Tumor/farmacologia , Quinases da Família src/genética , Animais , Quimiotaxia/fisiologia , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento , Cones de Crescimento/metabolismo , Cones de Crescimento/ultraestrutura , Proteínas Hedgehog/deficiência , Proteínas Hedgehog/genética , Dispositivos Lab-On-A-Chip , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Imagem Molecular , Fatores de Crescimento Neural/deficiência , Fatores de Crescimento Neural/genética , Netrina-1 , Cultura Primária de Células , Transdução de Sinais , Medula Espinal/crescimento & desenvolvimento , Medula Espinal/metabolismo , Medula Espinal/ultraestrutura , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética , Quinases da Família src/metabolismo
13.
Med Sci (Paris) ; 30(11): 1024-33, 2014 Nov.
Artigo em Francês | MEDLINE | ID: mdl-25388585

RESUMO

Advances in genomics, bioinformatics and the creation of model organisms have identified many genes associated with polycystic kidney diseases. Historically, these genes were not necessarily associated with ciliopathies, but it appeared that many connections can be made between the cystic kidney disease and function of the primary cilium. Indeed, the proteins encoded by these genes are localized to the cilium itself, to the basal body or are known to regulate the expression and localization of ciliary proteins. The goal of this article is to describe the multiple cellular processes that may lead to the development of renal cysts if they are deregulated. These include changes in proliferation rate, cell polarity or signaling pathways involved in embryonic kidney development. To highlight the role of the primary cilium in cystogenesis, I will discuss several studies investigating the function of ciliary genes and cilia in the kidneys of different model organisms.


Assuntos
Cílios/fisiologia , Transtornos da Motilidade Ciliar/patologia , Doenças Renais Císticas/patologia , Animais , Divisão Celular , Movimento Celular , Polaridade Celular , Transtornos da Motilidade Ciliar/genética , Modelos Animais de Doenças , Células Epiteliais/ultraestrutura , Proteínas Hedgehog/deficiência , Proteínas Hedgehog/genética , Proteínas Hedgehog/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Rim/embriologia , Rim/ultraestrutura , Doenças Renais Císticas/classificação , Doenças Renais Císticas/genética , Camundongos , Mitose , Modelos Biológicos , Morfogênese , Ratos , Transdução de Sinais/fisiologia , Proteínas Wnt/deficiência , Proteínas Wnt/genética , Proteínas Wnt/fisiologia
14.
Gastroenterology ; 147(3): 655-666.e9, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24859162

RESUMO

BACKGROUND & AIMS: Loss of expression of Sonic Hedgehog (Shh) from parietal cells results in hypergastrinemia in mice, accompanied by increased expression of Indian Hedgehog (Ihh) and hyperproliferation of surface mucous cells. We investigated whether hypergastrinemia induces gastric epithelial proliferation by activating Ihh signaling in mice. METHODS: We studied mice with parietal cell-specific deletion of Shh (PC-Shh(KO)) and hypergastrinemia, crossed with gastrin-deficient (GKO) mice (PC-Shh(KO)/GKO). When mice were 3-4 months old, gastric tissues were collected and analyzed by histology, for incorporation of bromodeoxyuridine, and for expression of the surface mucous cell marker Ulex europaeus. PC-Shh(KO)/GKO mice were given gastrin infusions for 7 days; gastric surface epithelium was collected and expression of Ihh was quantified by laser capture microdissection followed by quantitative reverse transcriptase polymerase chain reaction. Mouse stomach-derived organoids were incubated with or without inhibitors of WNT (DKK1) or Smoothened (vismodegib) and then cocultured with immortalized stomach mesenchymal cells, to assess proliferative responses to gastrin. RESULTS: Gastric tissues from PC-Shh(KO)/GKO mice with hypergastrinemia had an expanded surface pit epithelium, indicated by a significant increase in numbers of bromodeoxyuridine- and Ulex europaeus-positive cells, but there was no evidence for hyperproliferation. Gastrin infusion of PC PC-Shh(KO)/GKO mice increased expression of Ihh and proliferation within the surface epithelium compared with mice given infusions of saline. In gastric organoids cocultured with immortalized stomach mesenchymal cells, antagonists of WNT and Smoothened inhibited gastrin-induced proliferation and WNT activity. Activity of WNT in media collected from immortalized stomach mesenchymal cells correlated with increased expression of glioma-associated oncogene homolog 1, and was inhibited by DKK1 or vismodegib. CONCLUSIONS: Ihh signaling mediates gastrin-induced proliferation of epithelial cells in stomachs of adult mice.


Assuntos
Proliferação de Células , Células Epiteliais/metabolismo , Mucosa Gástrica/metabolismo , Gastrinas/metabolismo , Proteínas Hedgehog/metabolismo , Gastropatias/metabolismo , Animais , Linhagem Celular , Técnicas de Cocultura , Modelos Animais de Doenças , Células Epiteliais/patologia , Mucosa Gástrica/patologia , Gastrinas/administração & dosagem , Gastrinas/deficiência , Gastrinas/genética , Proteínas Hedgehog/deficiência , Proteínas Hedgehog/genética , Infusões Parenterais , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Organoides , Receptores Acoplados a Proteínas G/metabolismo , Receptor Smoothened , Gastropatias/genética , Gastropatias/patologia , Fatores de Tempo , Via de Sinalização Wnt , Proteína GLI1 em Dedos de Zinco
15.
Cancer Cell ; 25(6): 735-47, 2014 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-24856585

RESUMO

Sonic hedgehog (Shh), a soluble ligand overexpressed by neoplastic cells in pancreatic ductal adenocarcinoma (PDAC), drives formation of a fibroblast-rich desmoplastic stroma. To better understand its role in malignant progression, we deleted Shh in a well-defined mouse model of PDAC. As predicted, Shh-deficient tumors had reduced stromal content. Surprisingly, such tumors were more aggressive and exhibited undifferentiated histology, increased vascularity, and heightened proliferation--features that were fully recapitulated in control mice treated with a Smoothened inhibitor. Furthermore, administration of VEGFR blocking antibody selectively improved survival of Shh-deficient tumors, indicating that Hedgehog-driven stroma suppresses tumor growth in part by restraining tumor angiogenesis. Together, these data demonstrate that some components of the tumor stroma can act to restrain tumor growth.


Assuntos
Carcinoma Ductal Pancreático/patologia , Neoplasias Pancreáticas/patologia , Células Estromais/patologia , Animais , Anticorpos Monoclonais/farmacologia , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Progressão da Doença , Técnicas de Silenciamento de Genes , Proteínas Hedgehog/deficiência , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Imunoglobulina G/farmacologia , Camundongos , Camundongos Transgênicos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Distribuição Aleatória , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Transdução de Sinais , Células Estromais/metabolismo
16.
Nature ; 504(7479): 311-4, 2013 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-24336288

RESUMO

Primary cilia are solitary, non-motile extensions of the centriole found on nearly all nucleated eukaryotic cells between cell divisions. Only ∼200-300 nm in diameter and a few micrometres long, they are separated from the cytoplasm by the ciliary neck and basal body. Often called sensory cilia, they are thought to receive chemical and mechanical stimuli and initiate specific cellular signal transduction pathways. When activated by a ligand, hedgehog pathway proteins, such as GLI2 and smoothened (SMO), translocate from the cell into the cilium. Mutations in primary ciliary proteins are associated with severe developmental defects. The ionic conditions, permeability of the primary cilia membrane, and effectiveness of the diffusion barriers between the cilia and cell body are unknown. Here we show that cilia are a unique calcium compartment regulated by a heteromeric TRP channel, PKD1L1-PKD2L1, in mice and humans. In contrast to the hypothesis that polycystin (PKD) channels initiate changes in ciliary calcium that are conducted into the cytoplasm, we show that changes in ciliary calcium concentration occur without substantially altering global cytoplasmic calcium. PKD1L1-PKD2L1 acts as a ciliary calcium channel controlling ciliary calcium concentration and thereby modifying SMO-activated GLI2 translocation and GLI1 expression.


Assuntos
Canais de Cálcio/metabolismo , Sinalização do Cálcio , Cílios/metabolismo , Proteínas Hedgehog/metabolismo , Organelas/metabolismo , Animais , Cálcio/metabolismo , Canais de Cálcio/química , Células Cultivadas , Citoplasma/metabolismo , Feminino , Proteínas Hedgehog/deficiência , Proteínas Hedgehog/genética , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , Proteínas de Membrana/química , Proteínas de Membrana/deficiência , Proteínas de Membrana/metabolismo , Camundongos , Proteínas Nucleares/metabolismo , Receptores de Superfície Celular/química , Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptor Smoothened , Proteína GLI1 em Dedos de Zinco , Proteína Gli2 com Dedos de Zinco
17.
Circ Res ; 112(5): 762-70, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23343527

RESUMO

RATIONALE: Blood vessel growth and patterning have been shown to be regulated by nerve-derived signals. Desert hedgehog (Dhh), one of the Hedgehog family members, is expressed by Schwann cells of peripheral nerves. OBJECTIVE: The purpose of this study was to investigate the contribution of Dhh to angiogenesis in the setting of ischemia. METHODS AND RESULTS: We induced hindlimb ischemia in wild-type and Dhh(-/-) mice. First, we found that limb perfusion is significantly impaired in the absence of Dhh. This effect is associated with a significant decrease in capillary and artery density in Dhh(-/-). By using mice in which the Hedgehog signaling pathway effector Smoothened was specifically invalidated in endothelial cells, we demonstrated that Dhh does not promote angiogenesis by a direct activation of endothelial cells. On the contrary, we found that Dhh promotes peripheral nerve survival in the ischemic muscle and, by doing so, maintains the pool of nerve-derived proangiogenic factors. Consistently, we found that denervation of the leg, immediately after the onset of ischemia, severely impairs ischemia-induced angiogenesis and decreases expression of vascular endothelial growth factor A, angiopoietin 1, and neurotrophin 3 in the ischemic muscle. CONCLUSIONS: This study demonstrates the crucial roles of nerves and factors regulating nerve physiology in the setting of ischemia-induced angiogenesis.


Assuntos
Proteínas Hedgehog/fisiologia , Membro Posterior/irrigação sanguínea , Isquemia/fisiopatologia , Neovascularização Fisiológica/fisiologia , Nervos Periféricos/fisiologia , Angiopoietina-1/metabolismo , Animais , Sobrevivência Celular/fisiologia , Modelos Animais de Doenças , Proteínas Hedgehog/deficiência , Proteínas Hedgehog/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Denervação Muscular , Músculo Esquelético/inervação , Fatores de Crescimento Neural/metabolismo , Nervos Periféricos/citologia , Células de Schwann/citologia , Células de Schwann/fisiologia , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
18.
PLoS One ; 8(1): e53675, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23308271

RESUMO

In this study, we examined the expression of Sonic Hedgehog, Patched, Gli1, Gli2, Gli3 and Myocardin in the developing bladders of male and female normal and megabladder (mgb-/-) mutant mice at embryonic days 12 through 16 by in situ hybridization. This analysis indicated that each member of the Sonic Hedgehog signaling pathway as well as Myocardin displayed distinct temporal and spatial patterns of expression during normal bladder development. In contrast, mgb-/- bladders showed both temporal and spatial changes in the expression of Patched, Gli1 and Gli3 as well as a complete lack of Myocardin expression. These changes occurred primarily in the outer mesenchyme of developing mgb-/- bladders consistent with the development of an amuscular bladder phenotype in these animals. These results provide the first comprehensive analysis of the Sonic Hedgehog signaling pathway during normal bladder development and provide strong evidence that this key signaling cascade is critical in establishing radial patterning in the developing bladder. In addition, the lack of detrusor smooth muscle development observed in mgb-/- mice is associated with bladder-specific temporospatial changes in Sonic Hedgehog signaling coupled with a lack of Myocardin expression that appears to result in altered patterning of the outer mesenchyme and poor initiation and differentiation of smooth muscle cells within this region of the developing bladder.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/genética , Mesoderma/metabolismo , Músculo Liso/metabolismo , Transdução de Sinais/genética , Bexiga Urinária/metabolismo , Animais , Padronização Corporal/genética , Diferenciação Celular , Embrião de Mamíferos , Feminino , Proteínas Hedgehog/deficiência , Proteínas Hedgehog/metabolismo , Fatores de Transcrição Kruppel-Like/deficiência , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , Mesoderma/embriologia , Mesoderma/patologia , Camundongos , Camundongos Knockout , Músculo Liso/anormalidades , Músculo Liso/embriologia , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Receptores Patched , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Transativadores/deficiência , Transativadores/genética , Bexiga Urinária/anormalidades , Bexiga Urinária/embriologia , Proteína GLI1 em Dedos de Zinco , Proteína Gli2 com Dedos de Zinco , Proteína Gli3 com Dedos de Zinco
19.
Dev Cell ; 23(2): 280-91, 2012 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-22898775

RESUMO

The developmental mechanisms underlying human congenital heart disease (CHD) are poorly understood. Atrial septal defects (ASDs) can result from haploinsufficiency of cardiogenic transcription factors including TBX5. We demonstrated that Tbx5 is required in the second heart field (SHF) for atrial septation in mice. Conditional Tbx5 haploinsufficiency in the SHF but not the myocardium or endocardium caused ASDs. Tbx5 SHF knockout embryos lacked atrial septum progenitors. We found that Tbx5 mutant SHF progenitors demonstrated cell-cycle progression defects and that Tbx5 regulated cell-cycle progression genes including Cdk6. Activated hedgehog (Hh) signaling rescued ASDs in Tbx5 mutant embryos, placing Tbx5 upstream or parallel to Hh in cardiac progenitors. Tbx5 regulated SHF Gas1 and Osr1 expression, supporting both pathways. These results describe a SHF Tbx5-Hh network required for atrial septation. A paradigm defining molecular requirements in SHF cardiac progenitors for cardiac septum morphogenesis has implications for the ontogeny of CHD.


Assuntos
Septos Cardíacos/metabolismo , Transdução de Sinais , Proteínas com Domínio T/metabolismo , Artérias/metabolismo , Linhagem Celular , Proliferação de Células , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Septos Cardíacos/embriologia , Proteínas Hedgehog/deficiência , Proteínas Hedgehog/metabolismo , Mutação , Neovascularização Fisiológica , Células-Tronco/citologia , Células-Tronco/metabolismo , Proteínas com Domínio T/genética , Transcrição Gênica
20.
Dev Dyn ; 241(6): 1034-42, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22513894

RESUMO

BACKGROUND: The cellular mechanisms regulating branching and growth of the intersegmental vessels (ISVs) are not well understood. We have carried out studies demonstrating that Hedgehog (Hh) signaling is a major regulator of intersomitic vessel growth. RESULTS: Inhibition of Hh activity by cyclopamine completely blocks formation of intersomitic vessels in the avian embryo. Examination of gene expression patterns in Hh-deficient embryos shows that components of the VEGF and Notch signaling pathways are down-regulated. However, we find no evidence that Notch signaling plays a significant role in regulation of intersomitic vessel growth. Indeed, it appears that Hh modulation of Vascular Endothelial Growth Factor, VEGF, is the primary regulator of growth of intersomitic vessels in the avian embryo. CONCLUSIONS: Inhibition of the VEGF pathway results in absence of ISVs, whereas stimulation of VEGF expression leads to precocious branching of ISVs. These results demonstrate that Hh is an essential modulator of VEGF expression during developmental angiogenesis.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas Hedgehog/metabolismo , Neovascularização Fisiológica/fisiologia , Transdução de Sinais/fisiologia , Somitos/irrigação sanguínea , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Embrião de Galinha , Primers do DNA/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas Hedgehog/deficiência , Hibridização In Situ , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/genética , Alcaloides de Veratrum
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA