Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Curr Top Dev Biol ; 153: 195-227, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36967195

RESUMO

Deciphering non-canonical WNT signaling has proven to be both fascinating and challenging. Discovered almost 30 years ago, non-canonical WNT ligands signal independently of the transcriptional co-activator ß-catenin to regulate a wide range of morphogenetic processes during development. The molecular and cellular mechanisms that underlie non-canonical WNT function, however, remain nebulous. Recent results from various model systems have converged to define a core non-canonical WNT pathway consisting of the prototypic non-canonical WNT ligand, WNT5A, the receptor tyrosine kinase ROR, the seven transmembrane receptor Frizzled and the cytoplasmic scaffold protein Dishevelled. Importantly, mutations in each of these signaling components cause Robinow syndrome, a congenital disorder characterized by profound tissue morphogenetic abnormalities. Moreover, dysregulation of the pathway has also been linked to cancer metastasis. As new knowledge concerning the WNT5A-ROR pathway continues to grow, modeling these mutations will likely provide crucial insights into both the physiological regulation of the pathway and the etiology of WNT5A-ROR-driven diseases.


Assuntos
Receptores Órfãos Semelhantes a Receptor Tirosina Quinase , Proteínas Wnt , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Proteína Wnt-5a/genética , Proteína Wnt-5a/metabolismo , Transdução de Sinais/fisiologia , Morfogênese , Via de Sinalização Wnt
2.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34135125

RESUMO

Wnt5a-Ror signaling is a conserved pathway that regulates morphogenetic processes during vertebrate development [R. T. Moon et al, Development 119, 97-111 (1993); I. Oishi et al, Genes Cells 8, 645-654 (2003)], but its downstream signaling events remain poorly understood. Through a large-scale proteomic screen in mouse embryonic fibroblasts, we identified the E3 ubiquitin ligase Pdzrn3 as a regulatory target of the Wnt5a-Ror pathway. Upon pathway activation, Pdzrn3 is degraded in a ß-catenin-independent, ubiquitin-proteasome system-dependent manner. We developed a flow cytometry-based reporter to monitor Pdzrn3 abundance and delineated a signaling cascade involving Frizzled, Dishevelled, Casein kinase 1, and Glycogen synthase kinase 3 that regulates Pdzrn3 stability. Epistatically, Pdzrn3 is regulated independently of Kif26b, another Wnt5a-Ror effector. Wnt5a-dependent degradation of Pdzrn3 requires phosphorylation of three conserved amino acids within its C-terminal LNX3H domain [M. Flynn, O. Saha, P. Young, BMC Evol. Biol. 11, 235 (2011)], which acts as a bona fide Wnt5a-responsive element. Importantly, this phospho-dependent degradation is essential for Wnt5a-Ror modulation of cell migration. Collectively, this work establishes a Wnt5a-Ror cell morphogenetic cascade involving Pdzrn3 phosphorylation and degradation.


Assuntos
Proteômica , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Via de Sinalização Wnt , Proteína Wnt-5a/metabolismo , Animais , Movimento Celular , Camundongos , Fosforilação , Complexo de Endopeptidases do Proteassoma/metabolismo , Domínios Proteicos , Proteólise , Reprodutibilidade dos Testes , Ubiquitina/metabolismo
3.
Cells ; 9(2)2020 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-32046118

RESUMO

WNT5a is a mainly "non-canonical" WNT ligand whose dysregulation is observed in lung diseases such as idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD) and asthma. Germline deletion of Wnt5a disrupts embryonic lung development. However, the temporal-specific function of WNT5a remains unknown. In this study, we generated a conditional loss-of-function mouse model (Wnt5aCAG) and examined the specific role of Wnt5a during the saccular and alveolar phases of lung development. The lack of Wnt5a in the saccular phase blocked distal airway expansion and attenuated differentiation of endothelial and alveolar epithelial type I (AT1) cells and myofibroblasts. Postnatal Wnt5a inactivation disrupted alveologenesis, producing a phenotype resembling human bronchopulmonary dysplasia (BPD). Mutant lungs showed hypoalveolization, but endothelial and epithelial differentiation was unaffected. The major impact of Wnt5a inactivation on alveologenesis was on myofibroblast differentiation and migration, with reduced expression of key regulatory genes. These findings were validated in vitro using isolated lung fibroblasts. Conditional inactivation of the WNT5a receptors Ror1 and Ror2 in alveolar myofibroblasts recapitulated the Wnt5aCAG phenotype, demonstrating that myofibroblast defects are the major cause of arrested alveologenesis in Wnt5aCAG lungs. Finally, we show that WNT5a is reduced in human BPD lung samples, indicating the clinical relevance and potential role for WNT5a in pathogenesis of BPD.


Assuntos
Organogênese , Alvéolos Pulmonares/embriologia , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Transdução de Sinais , Proteína Wnt-5a/metabolismo , Animais , Animais Recém-Nascidos , Diferenciação Celular , Movimento Celular , Células Cultivadas , Células Endoteliais/citologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Recém-Nascido , Camundongos , Modelos Biológicos , Miofibroblastos/citologia
4.
Elife ; 82019 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-31268420

RESUMO

Adult neurogenesis in the hippocampus may represent a form of plasticity in brain functions including mood, learning and memory. However, mechanisms underlying neural stem/progenitor cells (NSPCs) proliferation are not well understood. We found that Agrin, a factor critical for neuromuscular junction formation, is elevated in the hippocampus of mice that are stimulated by enriched environment (EE). Genetic deletion of the Agrn gene in excitatory neurons decreases NSPCs proliferation and increases depressive-like behavior. Low-density lipoprotein receptor-related protein 4 (Lrp4), a receptor for Agrin, is expressed in hippocampal NSPCs and its mutation blocked basal as well as EE-induced NSPCs proliferation and maturation of newborn neurons. Finally, we show that Lrp4 interacts with and activates receptor tyrosine kinase-like orphan receptor 2 (Ror2); and Ror2 mutation impairs NSPCs proliferation. Together, these observations identify a role of Agrin-Lrp4-Ror2 signaling for adult neurogenesis, uncovering previously unexpected functions of Agrin and Lrp4 in the brain.


Assuntos
Agrina/metabolismo , Hipocampo/crescimento & desenvolvimento , Proteínas Relacionadas a Receptor de LDL/metabolismo , Células-Tronco Neurais/fisiologia , Neurogênese , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Transdução de Sinais , Agrina/deficiência , Animais , Proliferação de Células , Técnicas de Inativação de Genes , Proteínas Relacionadas a Receptor de LDL/deficiência , Camundongos , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/deficiência
5.
Nat Commun ; 10(1): 1703, 2019 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-30979871

RESUMO

Multiple vertebrate embryonic structures such as organ primordia are composed of confluent cells. Although mechanisms that shape tissue sheets are increasingly understood, those which shape a volume of cells remain obscure. Here we show that 3D mesenchymal cell intercalations are essential to shape the mandibular arch of the mouse embryo. Using a genetically encoded vinculin tension sensor that we knock-in to the mouse genome, we show that cortical force oscillations promote these intercalations. Genetic loss- and gain-of-function approaches show that Wnt5a functions as a spatial cue to coordinate cell polarity and cytoskeletal oscillation. These processes diminish tissue rigidity and help cells to overcome the energy barrier to intercalation. YAP/TAZ and PIEZO1 serve as downstream effectors of Wnt5a-mediated actomyosin polarity and cytosolic calcium transients that orient and drive mesenchymal cell intercalations. These findings advance our understanding of how developmental pathways regulate biophysical properties and forces to shape a solid organ primordium.


Assuntos
Polaridade Celular , Citoesqueleto/fisiologia , Mandíbula/embriologia , Mandíbula/fisiologia , Proteína Wnt-5a/fisiologia , Citoesqueleto de Actina , Actomiosina/metabolismo , Animais , Cálcio/metabolismo , Ciclo Celular , Citosol/metabolismo , Elasticidade , Células Epiteliais/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Mutação , Oscilometria , Transdução de Sinais , Estresse Mecânico , Vinculina/metabolismo , Viscosidade
6.
Cancer Res ; 79(8): 1719-1729, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30952630

RESUMO

Our understanding of the cellular mechanisms governing carcinoma invasiveness and metastasis has evolved dramatically over the last several years. The previous emphasis on the epithelial-mesenchymal transition as a driver of the migratory properties of single cells has expanded with the observation that carcinoma cells often invade and migrate collectively as adherent groups. Moreover, recent analyses suggest that circulating tumor cells within the vasculature often exist as multicellular clusters and that clusters more efficiently seed metastatic lesions than single circulating tumor cells. While these observations point to a key role for collective cell migration in carcinoma metastasis, the molecular mechanisms driving collective tumor cell migration remain to be discerned. Wnt/PCP (planar cell polarity) signaling, one of the noncanonical Wnt signaling pathways, mediates collective migratory events such as convergent extension during developmental processes. Wnt/PCP signaling components are frequently dysregulated in solid tumors, and aberrant pathway activation contributes to tumor cell migratory properties. Here we summarize key studies that address the mechanisms by which Wnt/PCP signaling mediate collective cell migration in developmental and tumor contexts. We emphasize Wnt/PCP component localization within migrating cells and discuss how component asymmetry may govern the spatiotemporal control of downstream cytoskeletal effectors to promote collective cell motility.


Assuntos
Carcinoma/patologia , Movimento Celular , Polaridade Celular , Transição Epitelial-Mesenquimal , Proteínas Wnt/metabolismo , Animais , Carcinoma/metabolismo , Humanos , Metástase Neoplásica
7.
Genes Cells ; 24(4): 307-317, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30801848

RESUMO

Mutations in the human receptor tyrosine kinase ROR2 are associated with Robinow syndrome (RRS) and brachydactyly type B1. Amongst others, the shortened limb phenotype associated with RRS is recapitulated in Ror2-/- mutant mice. In contrast, Ror1-/- mutant mice are viable and show no limb phenotype. Ror1-/- ;Ror2-/- double mutants are embryonic lethal, whereas double mutants containing a hypomorphic Ror1 allele (Ror1hyp ) survive up to birth and display a more severe shortened limb phenotype. Both orphan receptors have been shown to act as possible Wnt coreceptors and to mediate the Wnt5a signal. Here, we analyzed genetic interactions between the Wnt ligand, Wnt9a, and Ror2 or Ror1, as Wnt9a has also been implicated in skeletal development. Wnt9a-/- single mutants display a mild shortening of the long bones, whereas these are severely shortened in Ror2-/- mutants. Ror2-/- ;Wnt9a-/- double mutants displayed even more severely shortened long bones, and intermediate phenotypes were observed in compound Ror2;Wnt9a mutants. Long bones were also shorter in Ror1hyp/hyp ;Wnt9a-/- double mutants. In addition, Ror1hyp/hyp ;Wnt9a-/- double mutants displayed a secondary palate cleft phenotype, which was not present in the respective single mutants. Interestingly, 50% of compound mutant pups heterozygous for Ror2 and homozygous mutant for Ror1 also developed a secondary palate cleft phenotype.


Assuntos
Fissura Palatina/genética , Epistasia Genética , Deformidades Congênitas dos Membros/genética , Mutação , Osteogênese/genética , Proteínas Wnt/genética , Animais , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase
8.
PLoS Genet ; 14(12): e1007850, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30521570

RESUMO

Domestic dog breeds exhibit remarkable morphological variations that result from centuries of artificial selection and breeding. Identifying the genetic changes that contribute to these variations could provide critical insights into the molecular basis of tissue and organismal morphogenesis. Bulldogs, French Bulldogs and Boston Terriers share many morphological and disease-predisposition traits, including brachycephalic skull morphology, widely set eyes and short stature. Unlike other brachycephalic dogs, these breeds also exhibit vertebral malformations that result in a truncated, kinked tail (screw tail). Whole genome sequencing of 100 dogs from 21 breeds identified 12.4 million bi-allelic variants that met inclusion criteria. Whole Genome Association of these variants with the breed defining phenotype of screw tail was performed using 10 cases and 84 controls and identified a frameshift mutation in the WNT pathway gene DISHEVELLED 2 (DVL2) (Chr5: 32195043_32195044del, p = 4.37 X 10-37) as the most strongly associated variant in the canine genome. This DVL2 variant was fixed in Bulldogs and French Bulldogs and had a high allele frequency (0.94) in Boston Terriers. The DVL2 variant segregated with thoracic and caudal vertebral column malformations in a recessive manner with incomplete and variable penetrance for thoracic vertebral malformations between different breeds. Importantly, analogous frameshift mutations in the human DVL1 and DVL3 genes cause Robinow syndrome, a congenital disorder characterized by similar craniofacial, limb and vertebral malformations. Analysis of the canine DVL2 variant protein showed that its ability to undergo WNT-induced phosphorylation is reduced, suggesting that altered WNT signaling may contribute to the Robinow-like syndrome in the screwtail breeds.


Assuntos
Anormalidades Craniofaciais/veterinária , Proteínas Desgrenhadas/genética , Doenças do Cão/genética , Cães/genética , Nanismo/veterinária , Deformidades Congênitas dos Membros/veterinária , Anormalidades Urogenitais/veterinária , Sequência de Aminoácidos , Animais , Anormalidades Craniofaciais/genética , Anormalidades Craniofaciais/metabolismo , Proteínas Desgrenhadas/metabolismo , Doenças do Cão/metabolismo , Cães/anatomia & histologia , Cães/classificação , Nanismo/genética , Nanismo/metabolismo , Feminino , Mutação da Fase de Leitura , Variação Genética , Estudo de Associação Genômica Ampla , Humanos , Deformidades Congênitas dos Membros/genética , Deformidades Congênitas dos Membros/metabolismo , Masculino , Compostos de Organossilício , Homologia de Sequência de Aminoácidos , Especificidade da Espécie , Cauda/anatomia & histologia , Anormalidades Urogenitais/genética , Anormalidades Urogenitais/metabolismo , Via de Sinalização Wnt/genética
9.
Am J Med Genet A ; 176(12): 2623-2629, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30151950

RESUMO

KIF26B is a member of the kinesin superfamily with evolutionarily conserved functions in controlling aspects of embryogenesis, including the development of the nervous system, though its function is incompletely understood. We describe an infant with progressive microcephaly, pontocerebellar hypoplasia, and arthrogryposis secondary to the involvement of anterior horn cells and ventral (motor) nerves. We performed whole exome sequencing on the trio and identified a de novo KIF26B missense variant, p.Gly546Ser, in the proband. This variant alters a highly conserved amino acid residue that is part of the phosphate-binding loop motif and motor-like domain and is deemed pathogenic by several in silico methods. Functional analysis of the variant protein in cultured cells revealed a reduction in the KIF26B protein's ability to promote cell adhesion, a defect that potentially contributes to its pathogenicity. Overall, KIF26B may play a critical role in the brain development and, when mutated, cause pontocerebellar hypoplasia with arthrogryposis.


Assuntos
Cinesinas/genética , Atrofias Olivopontocerebelares/genética , Atrofias Musculares Espinais da Infância/diagnóstico , Atrofias Musculares Espinais da Infância/genética , Sequência de Aminoácidos , Animais , Encéfalo/anormalidades , Encéfalo/diagnóstico por imagem , Adesão Celular , Modelos Animais de Doenças , Expressão Gênica , Humanos , Cinesinas/química , Imageamento por Ressonância Magnética/métodos , Camundongos , Modelos Moleculares , Conformação Proteica , Sequenciamento do Exoma
10.
Bio Protoc ; 8(6)2018 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-29770348

RESUMO

Noncanonical Wnt signaling functions independently of the ß-catenin pathway to control diverse developmental processes, and dysfunction of the pathway contributes to a number of human pathological conditions, including birth defects and metastatic cancer. Progress in the field, however, has been hampered by the scarcity of functional assays for measuring noncanonical Wnt signaling activity. We recently described the Wnt5a-Ror-Kif26b (WRK) reporter assay, which directly monitors a post-transcriptional regulatory event in noncanonical Wnt signaling. In this protocol, we describe the generation of the stable GFP-Kif26b reporter cell line and a quantitative reporter assay for detecting and measuring Wnt5a signaling activities in live cells via flow cytometry.

11.
Genes (Basel) ; 9(4)2018 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-29621187

RESUMO

Noncanonical WNT pathways function independently of the ß-catenin transcriptional co-activator to regulate diverse morphogenetic and pathogenic processes. Recent studies showed that noncanonical WNTs, such as WNT5A, can signal the degradation of several downstream effectors, thereby modulating these effectors' cellular activities. The protein domain(s) that mediates the WNT5A-dependent degradation response, however, has not been identified. By coupling protein mutagenesis experiments with a flow cytometry-based degradation reporter assay, we have defined a protein domain in the kinesin superfamily protein KIF26B that is essential for WNT5A-dependent degradation. We found that a human disease-causing KIF26B mutation located at a conserved amino acid within this domain compromises the ability of WNT5A to induce KIF26B degradation. Using pharmacological perturbation, we further uncovered a role of glycogen synthase kinase 3 (GSK3) in WNT5A regulation of KIF26B degradation. Lastly, based on the identification of the WNT5A-responsive domain, we developed a new reporter system that allows for efficient profiling of WNT5A-KIF26B signaling activity in both somatic and stem cells. In conclusion, our study identifies a new protein domain that mediates WNT5A-dependent degradation of KIF26B and provides a new tool for functional characterization of noncanonical WNT5A signaling in cells.

12.
Elife ; 62017 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-28885975

RESUMO

Wnt5a-Ror signaling constitutes a developmental pathway crucial for embryonic tissue morphogenesis, reproduction and adult tissue regeneration, yet the molecular mechanisms by which the Wnt5a-Ror pathway mediates these processes are largely unknown. Using a proteomic screen, we identify the kinesin superfamily protein Kif26b as a downstream target of the Wnt5a-Ror pathway. Wnt5a-Ror, through a process independent of the canonical Wnt/ß-catenin-dependent pathway, regulates the cellular stability of Kif26b by inducing its degradation via the ubiquitin-proteasome system. Through this mechanism, Kif26b modulates the migratory behavior of cultured mesenchymal cells in a Wnt5a-dependent manner. Genetic perturbation of Kif26b function in vivo caused embryonic axis malformations and depletion of primordial germ cells in the developing gonad, two phenotypes characteristic of disrupted Wnt5a-Ror signaling. These findings indicate that Kif26b links Wnt5a-Ror signaling to the control of morphogenetic cell and tissue behaviors in vertebrates and reveal a new role for regulated proteolysis in noncanonical Wnt5a-Ror signal transduction.


Assuntos
Cinesinas/metabolismo , Transdução de Sinais , Proteína Wnt-5a/metabolismo , Animais , Linhagem Celular , Desenvolvimento Embrionário/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Humanos , Cinesinas/genética , Camundongos , Camundongos Endogâmicos C57BL , Morfogênese/efeitos dos fármacos , Proteômica , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Via de Sinalização Wnt , Proteína Wnt-5a/farmacologia , beta Catenina/metabolismo
13.
J Biol Chem ; 292(38): 15939-15951, 2017 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-28790171

RESUMO

The Ror family receptor tyrosine kinases, Ror1 and Ror2, play important roles in regulating developmental morphogenesis and tissue- and organogenesis, but their roles in tissue regeneration in adult animals remain largely unknown. In this study, we examined the expression and function of Ror1 and Ror2 during skeletal muscle regeneration. Using an in vivo skeletal muscle injury model, we show that expression of Ror1 and Ror2 in skeletal muscles is induced transiently by the inflammatory cytokines, TNF-α and IL-1ß, after injury and that inhibition of TNF-α and IL-1ß by neutralizing antibodies suppresses expression of Ror1 and Ror2 in injured muscles. Importantly, expression of Ror1, but not Ror2, was induced primarily in Pax7-positive satellite cells (SCs) after muscle injury, and administration of neutralizing antibodies decreased the proportion of Pax7-positive proliferative SCs after muscle injury. We also found that stimulation of a mouse myogenic cell line, C2C12 cells, with TNF-α or IL-1ß induced expression of Ror1 via NF-κB activation and that suppressed expression of Ror1 inhibited their proliferative responses in SCs. Intriguingly, SC-specific depletion of Ror1 decreased the number of Pax7-positive SCs after muscle injury. Collectively, these findings indicate for the first time that Ror1 has a critical role in regulating SC proliferation during skeletal muscle regeneration. We conclude that Ror1 might be a suitable target in the development of diagnostic and therapeutic approaches to manage muscular disorders.


Assuntos
Músculo Esquelético/lesões , Músculo Esquelético/fisiologia , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Regeneração , Células Satélites de Músculo Esquelético/citologia , Animais , Linhagem Celular , Proliferação de Células , Regulação Enzimológica da Expressão Gênica , Interleucina-1beta/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Desenvolvimento Muscular , NF-kappa B/metabolismo , Fator de Transcrição PAX7/metabolismo , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Células Satélites de Músculo Esquelético/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
14.
Proc Natl Acad Sci U S A ; 113(50): E8079-E8088, 2016 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-27911818

RESUMO

Blastocyst implantation is a complex process requiring coordination of a dynamic sequence of embryo-uterine interactions. Blood vessels enter the uterus from the mesometrium, demarcating the uterus into mesometrial (M) and antimesometrial (AM) domains. Implantation occurs along the uterine longitudinal axis within specialized implantation chambers (crypts) that originate within the evaginations directed from the primary lumen toward the AM domain. The morphological orientation of crypts in rodent uteri was recognized more than a century ago, but the mechanism remained unknown. Here we provide evidence that planar cell polarity (PCP) signaling orchestrates directed epithelial evaginations to form crypts for implantation in mice. Uterine deletion of Vang-like protein 2, but not Vang-like protein 1, conferred aberrant PCP signaling, misdirected epithelial evaginations, defective crypt formation, and blastocyst attachment, leading to severely compromised pregnancy outcomes. The study reveals a previously unrecognized role for PCP in executing spatial cues for crypt formation and implantation. Because PCP is an evolutionarily conserved phenomenon, our study is likely to inspire implantation studies of this signaling pathway in humans and other species.


Assuntos
Polaridade Celular/fisiologia , Implantação do Embrião/fisiologia , Útero/fisiologia , Animais , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/fisiologia , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Comunicação Celular/fisiologia , Proteínas Desgrenhadas/fisiologia , Epitélio/anatomia & histologia , Epitélio/fisiologia , Feminino , Masculino , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Gravidez , Resultado da Gravidez , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/deficiência , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/fisiologia , Transdução de Sinais/fisiologia , Útero/anatomia & histologia , Proteína Wnt-5a/deficiência , Proteína Wnt-5a/genética , Proteína Wnt-5a/fisiologia
15.
J Cell Biol ; 215(2): 217-229, 2016 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-27810913

RESUMO

Cell segregation is the process by which cells self-organize to establish developmental boundaries, an essential step in tissue formation. Cell segregation is a common outcome of Eph/ephrin signaling, but the mechanisms remain unclear. In craniofrontonasal syndrome, X-linked mosaicism for ephrin-B1 expression has been hypothesized to lead to aberrant Eph/ephrin-mediated cell segregation. Here, we use mouse genetics to exploit mosaicism to study cell segregation in the mammalian embryo and integrate live-cell imaging to examine the underlying cellular and molecular mechanisms. Our data demonstrate that dramatic ephrin-B1-mediated cell segregation occurs in the early neuroepithelium. In contrast to the paradigm that repulsive bidirectional signaling drives cell segregation, unidirectional EphB kinase signaling leads to cell sorting by the Rho kinase-dependent generation of a cortical actin differential between ephrin-B1- and EphB-expressing cells. These results define mechanisms of Eph/ephrin-mediated cell segregation, implicating unidirectional regulation of cortical actomyosin contractility as a key effector of this fundamental process.


Assuntos
Actomiosina/metabolismo , Diferenciação Celular , Efrina-B1/metabolismo , Células Neuroepiteliais/citologia , Receptores da Família Eph/metabolismo , Transdução de Sinais , Actinas/metabolismo , Animais , Contagem de Células , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Camundongos , Modelos Biológicos , Células Neuroepiteliais/metabolismo , Quinases Associadas a rho/metabolismo
16.
J Cell Sci ; 129(13): 2493-9, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27199373

RESUMO

Mouse ovarian germ cells enter meiosis in a wave that propagates from anterior to posterior, but little is known about contribution of germ cells to initiation or propagation of meiosis. In a Ror2 mutant with diminished germ cell number and migration, we find that overall timing of meiotic initiation is delayed at the population level. We use chemotherapeutic depletion to exclude a profoundly reduced number of germ cells as a cause for meiotic delay. We rule out sex reversal or failure to specify somatic support cells as contributors to the meiotic phenotype. Instead, we find that anomalies in the distribution of germ cells as well as gonad shape in mutants contribute to aberrant initiation of meiosis. Our analysis supports a model of meiotic initiation via diffusible signal(s), excludes a role for germ cells in commencing the meiotic wave and furnishes the first phenotypic demonstration of the wave of meiotic entry. Finally, our studies underscore the importance of considering germ cell migration defects while studying meiosis to discern secondary effects resulting from positioning versus primary meiotic entry phenotypes.


Assuntos
Células Germinativas/metabolismo , Gônadas/patologia , Meiose/genética , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Animais , Contagem de Células , Movimento Celular/genética , Forma Celular/genética , Feminino , Células Germinativas/crescimento & desenvolvimento , Células Germinativas/patologia , Gônadas/crescimento & desenvolvimento , Camundongos , Mutação , Ovário/crescimento & desenvolvimento , Ovário/metabolismo , Ovário/patologia , Transdução de Sinais/genética
17.
Dev Neurobiol ; 76(4): 405-20, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26148571

RESUMO

The two cortical hemispheres of the mammalian forebrain are interconnected by major white matter tracts, including the corpus callosum (CC) and the posterior branch of the anterior commissure (ACp), that bridge the telencephalic midline. We show here that the intracellular signaling domains of the EphB1 and EphB2 receptors are critical for formation of both the ACp and CC. We observe partial and complete agenesis of the corpus callosum, as well as highly penetrant ACp misprojection phenotypes in truncated EphB1/2 mice that lack intracellular signaling domains. Consistent with the roles for these receptors in formation of the CC and ACp, we detect expression of these receptors in multiple brain regions associated with the formation of these forebrain structures. Taken together, our findings suggest that a combination of forward and reverse EphB1/2 receptor-mediated signaling contribute to ACp and CC axon guidance.


Assuntos
Comissura Anterior/embriologia , Comissura Anterior/metabolismo , Corpo Caloso/embriologia , Corpo Caloso/metabolismo , Receptor EphB1/metabolismo , Receptor EphB2/metabolismo , Animais , Comissura Anterior/citologia , Axônios/metabolismo , Movimento Celular/fisiologia , Corpo Caloso/citologia , Técnicas de Introdução de Genes , Técnicas de Inativação de Genes , Imuno-Histoquímica , Espaço Intracelular , Camundongos Transgênicos , Técnicas de Rastreamento Neuroanatômico , Domínios Proteicos , Receptor EphB1/genética , Receptor EphB2/genética , Transdução de Sinais
18.
Cell Rep ; 8(2): 382-92, 2014 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-25043182

RESUMO

Embryo homing and implantation occur within a crypt (implantation chamber) at the antimesometrial (AM) pole along the uterus. The mechanism by which this is achieved is not known. Here, we show that villi-like epithelial projections from the main uterine lumen toward the AM pole at regularly spaced intervals that form crypts for embryo implantation were disrupted in mice with uterine loss or gain of function of Wnt5a, or loss of function of both Ror1 and Ror2. This disruption of Wnt5a-ROR signaling resulted in disorderly epithelial projections, crypt formation, embryo spacing, and impaired implantation. These early disturbances under abnormal Wnt5a-ROR signaling were reflected in adverse late pregnancy events, including defective decidualization and placentation, ultimately leading to compromised pregnancy outcomes. This study presents deeper insight regarding the formation of organized epithelial projections for crypt formation and embryo implantation for pregnancy success.


Assuntos
Decídua/metabolismo , Implantação do Embrião , Células Epiteliais/citologia , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , Animais , Decídua/citologia , Decídua/fisiologia , Células Epiteliais/metabolismo , Feminino , Camundongos , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Proteínas Wnt/genética , Proteína Wnt-5a
19.
Endocr Relat Cancer ; 21(5): R345-56, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24981109

RESUMO

Cancers commonly reactivate embryonic developmental pathways to promote the aggressive behavior of their cells, resulting in metastasis and poor patient outcome. While developmental pathways such as canonical Wnt signaling and epithelial-to-mesenchymal transition have received much attention, our understanding of the role of the planar cell polarity (PCP) pathway in tumor progression remains rudimentary. Protein components of PCP, including a subset that overlaps with the canonical Wnt pathway, partition in polarized epithelial cells along the planar axis and are required for the establishment and maintenance of lateral epithelial polarity. Significant insight into PCP regulation of developmental and cellular processes has come from analysis of the functions of the core PCP scaffolding proteins Vangl1 and Vangl2. In particular, studies on zebrafish and with Looptail (Lp) mice, which harbor point mutations in Vangl2 that alter its trafficking and localization, point to roles for the PCP pathway in maintaining cell polarization along both the apical-basal and planar axes as well as in collective cell motility and invasiveness. Recent findings have suggested that the Vangls can promote similar processes in tumor cells. Initial data-mining efforts suggest that VANGL1 and VANGL2 are dysregulated in human cancers, and estrogen receptor (ER)-positive breast cancer patients whose tumors exhibit elevated VANGL1 expression suffer from shortened overall survival. Overall, evidence is beginning to accumulate that the heightened cellular motility and invasiveness associated with PCP reactivation may contribute to the malignancy of some cancer subtypes.


Assuntos
Proteínas de Transporte/metabolismo , Polaridade Celular , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Neoplasias/metabolismo , Animais , Proteínas de Transporte/química , Células Epiteliais/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Proteínas de Membrana/química , Neoplasias/patologia
20.
Proc Natl Acad Sci U S A ; 111(6): 2188-93, 2014 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-24453220

RESUMO

In early brain development, ascending thalamocortical axons (TCAs) navigate through the ventral telencephalon (VTel) to reach their target regions in the young cerebral cortex. Descending, deep-layer cortical axons subsequently target appropriate thalamic and subcortical target regions. However, precisely how and when corticothalamic axons (CTAs) identify their appropriate, reciprocal thalamic targets remains unclear. We show here that EphB1 and EphB2 receptors control proper navigation of a subset of TCA and CTA projections through the VTel. We show in vivo that EphB receptor forward signaling and the ephrinB1 ligand are required during the early navigation of L1-CAM(+) thalamic fibers in the VTel, and that the misguided thalamic fibers in EphB1/2 KO mice appear to interact with cortical subregion-specific axon populations during reciprocal cortical axon guidance. As such, our findings suggest that descending cortical axons identify specific TCA subpopulations in the dorsal VTel to coordinate reciprocal cortical-thalamic connectivity in the early developing brain.


Assuntos
Axônios , Córtex Cerebral/metabolismo , Receptores da Família Eph/metabolismo , Transdução de Sinais , Tálamo/metabolismo , Animais , Camundongos , Camundongos Knockout , Receptores da Família Eph/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...