Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Development ; 149(14)2022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-35781558

RESUMO

Formation of highly unique and complex facial structures is controlled by genetic programs that are responsible for the precise coordination of three-dimensional tissue morphogenesis. However, the underlying mechanisms governing these processes remain poorly understood. We combined mouse genetic and genomic approaches to define the mechanisms underlying normal and defective midfacial morphogenesis. Conditional inactivation of the Wnt secretion protein Wls in Pax3-expressing lineage cells disrupted frontonasal primordial patterning, cell survival and directional outgrowth, resulting in altered facial structures, including midfacial hypoplasia and midline facial clefts. Single-cell RNA sequencing revealed unique transcriptomic atlases of mesenchymal subpopulations in the midfacial primordia, which are disrupted in the conditional Wls mutants. Differentially expressed genes and cis-regulatory sequence analyses uncovered that Wls modulates and integrates a core gene regulatory network, consisting of key midfacial regulatory transcription factors (including Msx1, Pax3 and Pax7) and their downstream targets (including Wnt, Shh, Tgfß and retinoic acid signaling components), in a mesenchymal subpopulation of the medial nasal prominences that is responsible for midline facial formation and fusion. These results reveal fundamental mechanisms underlying mammalian midfacial morphogenesis and related defects at single-cell resolution.


Assuntos
Redes Reguladoras de Genes , Transcriptoma , Animais , Face , Mamíferos/genética , Camundongos , Morfogênese/genética , Transcriptoma/genética , Proteínas Wnt/metabolismo
2.
Int J Mol Sci ; 23(2)2022 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-35055125

RESUMO

PTH induces phosphorylation of the transcriptional coregulator NACA on serine 99 through Gαs and PKA. This leads to nuclear translocation of NACA and expression of the target gene Lrp6, encoding a coreceptor of the PTH receptor (PTH1R) necessary for full anabolic response to intermittent PTH (iPTH) treatment. We hypothesized that maintaining enough functional PTH1R/LRP6 coreceptor complexes at the plasma membrane through NACA-dependent Lrp6 transcription is important to ensure maximal response to iPTH. To test this model, we generated compound heterozygous mice in which one allele each of Naca and Lrp6 is inactivated in osteoblasts and osteocytes, using a knock-in strain with a Naca99 Ser-to-Ala mutation and an Lrp6 floxed strain (test genotype: Naca99S/A; Lrp6+/fl;OCN-Cre). Four-month-old females were injected with vehicle or 100 µg/kg PTH(1-34) once daily, 5 days a week for 4 weeks. Control mice showed significant increases in vertebral trabecular bone mass and biomechanical properties that were abolished in compound heterozygotes. Lrp6 expression was reduced in compound heterozygotes vs. controls. The iPTH treatment increased Alpl and Col1a1 mRNA levels in the control but not in the test group. These results confirm that NACA and LRP6 form part of a common genetic pathway that is necessary for the full anabolic effect of iPTH.


Assuntos
Anabolizantes/administração & dosagem , Células-Tronco Embrionárias/citologia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Chaperonas Moleculares/genética , Hormônio Paratireóideo/administração & dosagem , Anabolizantes/farmacologia , Animais , Linhagem Celular , Membrana Celular/metabolismo , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Introdução de Genes , Camundongos , Chaperonas Moleculares/metabolismo , Mutagênese Sítio-Dirigida , Osteoblastos/metabolismo , Osteócitos/metabolismo , Hormônio Paratireóideo/farmacologia , Fosforilação , Transdução de Sinais/efeitos dos fármacos , Microtomografia por Raio-X
3.
Cell Mol Life Sci ; 77(18): 3597-3609, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31758234

RESUMO

The bHLH transcription factor Olig2 is required for sequential cell fate determination of both motor neurons and oligodendrocytes and for progenitor proliferation in the central nervous system. However, the role of Olig2 in peripheral sensory neurogenesis remains unknown. We report that Olig2 is transiently expressed in the newly differentiated olfactory sensory neurons (OSNs) and is down-regulated in the mature OSNs in mice from early gestation to adulthood. Genetic fate mapping demonstrates that Olig2-expressing cells solely give rise to OSNs in the peripheral olfactory system. Olig2 depletion does not affect the proliferation of peripheral olfactory progenitors and the fate determination of OSNs, sustentacular cells, and the olfactory ensheathing cells. However, the terminal differentiation and maturation of OSNs are compromised in either Olig2 single or Olig1/Olig2 double knockout mice, associated with significantly diminished expression of multiple OSN maturation and odorant signaling genes, including Omp, Gnal, Adcy3, and Olfr15. We further demonstrate that Olig2 binds to the E-box in the Omp promoter region to regulate its expression. Taken together, our results reveal a distinctly novel function of Olig2 in the periphery nervous system to regulate the terminal differentiation and maturation of olfactory sensory neurons.


Assuntos
Diferenciação Celular , Neurônios Receptores Olfatórios/metabolismo , Fator de Transcrição 2 de Oligodendrócitos/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Linhagem da Célula , Proliferação de Células , Proteína Duplacortina , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Camundongos , Camundongos Transgênicos , Proteína de Marcador Olfatório/genética , Mucosa Olfatória/citologia , Mucosa Olfatória/metabolismo , Fator de Transcrição 2 de Oligodendrócitos/deficiência , Fator de Transcrição 2 de Oligodendrócitos/genética , Regiões Promotoras Genéticas , Fatores de Transcrição SOXB1/deficiência , Fatores de Transcrição SOXB1/genética , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
4.
Biochem Biophys Res Commun ; 526(3): 647-653, 2020 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-32248972

RESUMO

The mechanisms underlying mammalian neural tube closure remain poorly understood. We report a unique cellular process involving multicellular rosette formation, convergent cellular protrusions, and F-actin cable network of the non-neural surface ectodermal cells encircling the closure site of the posterior neuropore, which are demonstrated by scanning electron microscopy and genetic fate mapping analyses during mouse spinal neurulation. These unique cellular structures are severely disrupted in the surface ectodermal transcription factor Grhl3 mutants that exhibit fully penetrant spina bifida. We propose a novel model of mammalian neural tube closure driven by surface ectodermal dynamics, which is computationally visualized.


Assuntos
Actinas/metabolismo , Ectoderma/embriologia , Defeitos do Tubo Neural/embriologia , Tubo Neural/embriologia , Neurulação , Actinas/análise , Animais , Proteínas de Ligação a DNA/genética , Ectoderma/anormalidades , Ectoderma/metabolismo , Ectoderma/ultraestrutura , Camundongos , Mutação , Tubo Neural/anormalidades , Tubo Neural/metabolismo , Tubo Neural/ultraestrutura , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo , Disrafismo Espinal/embriologia , Disrafismo Espinal/genética , Disrafismo Espinal/metabolismo , Coluna Vertebral/anormalidades , Coluna Vertebral/embriologia , Coluna Vertebral/metabolismo , Coluna Vertebral/ultraestrutura , Fatores de Transcrição/genética
5.
Biochem Biophys Res Commun ; 496(4): 1302-1307, 2018 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-29410176

RESUMO

After traumatic spinal cord injury (SCI), a scar may form with a fibrotic core (fibrotic scar) and surrounding reactive astrocytes (glial scar) at the lesion site. The scar tissue is considered a major obstacle preventing regeneration both as a physical barrier and as a source for secretion of inhibitors of axonal regeneration. Understanding the mechanism of scar formation and how to control it may lead to effective SCI therapies. Using a compression-SCI model on adult transgenic mice, we demonstrate that the canonical Wnt/ß-catenin signaling reporter TOPgal (TCF/Lef1-lacZ) positive cells appeared at the lesion site by 5 days, peaked on 7 days, and diminished by 14 days post injury. Using various representative cell lineage markers, we demonstrate that, these transiently TOPgal positive cells are a group of Fibronectin(+);GFAP(-) fibroblast-like cells in the core scar region. Some of them are proliferative. These results indicate that Wnt/ß-catenin signaling may play a key role in fibrotic scar formation after traumatic spinal cord injury.


Assuntos
Cicatriz/metabolismo , Cicatriz/patologia , Compressão da Medula Espinal/metabolismo , Compressão da Medula Espinal/patologia , Medula Espinal/patologia , Via de Sinalização Wnt , beta Catenina/metabolismo , Animais , Cicatriz/etiologia , Fibrose , Proteína Glial Fibrilar Ácida , Camundongos , Camundongos Transgênicos , Medula Espinal/metabolismo , Compressão da Medula Espinal/complicações
6.
Development ; 141(1): 148-57, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24284205

RESUMO

Non-canonical Wnt/planar cell polarity (PCP) signaling plays a primary role in the convergent extension that drives neural tube closure and body axis elongation. PCP signaling gene mutations cause severe neural tube defects (NTDs). However, the role of canonical Wnt/ß-catenin signaling in neural tube closure and NTDs remains poorly understood. This study shows that conditional gene targeting of ß-catenin in the dorsal neural folds of mouse embryos represses the expression of the homeobox-containing genes Pax3 and Cdx2 at the dorsal posterior neuropore (PNP), and subsequently diminishes the expression of the Wnt/ß-catenin signaling target genes T, Tbx6 and Fgf8 at the tail bud, leading to spina bifida aperta, caudal axis bending and tail truncation. We demonstrate that Pax3 and Cdx2 are novel downstream targets of Wnt/ß-catenin signaling. Transgenic activation of Pax3 cDNA can rescue the closure defect in the ß-catenin mutants, suggesting that Pax3 is a key downstream effector of ß-catenin signaling in the PNP closure process. Cdx2 is known to be crucial in posterior axis elongation and in neural tube closure. We found that Cdx2 expression is also repressed in the dorsal PNPs of Pax3-null embryos. However, the ectopically activated Pax3 in the ß-catenin mutants cannot restore Cdx2 mRNA in the dorsal PNP, suggesting that the presence of both ß-catenin and Pax3 is required for regional Cdx2 expression. Thus, ß-catenin signaling is required for caudal neural tube closure and elongation, acting through the transcriptional regulation of key target genes in the PNP.


Assuntos
Padronização Corporal/fisiologia , Proteínas de Homeodomínio/metabolismo , Tubo Neural/embriologia , Fatores de Transcrição Box Pareados/metabolismo , Fatores de Transcrição/metabolismo , beta Catenina/metabolismo , Animais , Padronização Corporal/genética , Fator de Transcrição CDX2 , Adesão Celular/genética , Polaridade Celular/fisiologia , Fator 8 de Crescimento de Fibroblasto/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Fator de Transcrição MSX1/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tubo Neural/crescimento & desenvolvimento , Tubo Neural/metabolismo , Defeitos do Tubo Neural/genética , Neurulação , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados/biossíntese , Fatores de Transcrição Box Pareados/genética , Disrafismo Espinal/genética , Proteínas com Domínio T , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Transcrição Gênica , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Via de Sinalização Wnt/genética , beta Catenina/genética
8.
J Am Soc Nephrol ; 27(2): 417-27, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26047795

RESUMO

Hypoplastic and/or cystic kidneys have been found in both LDL receptor-related protein 6 (Lrp6)- and ß-catenin-mutant mouse embryos, and these proteins are key molecules for Wnt signaling. However, the underlying mechanisms of Lrp6/ß-catenin signaling in renal development and cystic formation remain poorly understood. In this study, we found evidence that diminished cell proliferation and increased apoptosis occur before cystic dysplasia in the renal primordia of Lrp6-deficient mouse embryos. The expression of Ret proto-oncogene (Ret), a critical receptor for the growth factor glial cell line-derived neurotrophic factor (GDNF), which is required for early nephrogenesis, was dramatically diminished in the mutant renal primordia. The activities of other representative nephrogenic genes, including Lim1, Pax2, Pax8, GDNF, and Wnt11, were subsequently diminished in the mutant renal primordia. Molecular biology experiments demonstrated that Ret is a novel transcriptional target of Wnt/ß-catenin signaling. Wnt agonist lithium promoted Ret expression in vitro and in vivo. Furthermore, Lrp6-knockdown or lithium treatment in vitro led to downregulation or upregulation, respectively, of the phosphorylated mitogen-activated protein kinases 1 and 3, which act downstream of GDNF/Ret signaling. Mice with single and double mutations of Lrp6 and Ret were perinatal lethal and demonstrated gene dosage-dependent effects on the severity of renal hypoplasia during embryogenesis. Taken together, these results suggest that Lrp6-mediated Wnt/ß-catenin signaling modulates or interacts with a signaling network consisting of Ret cascades and related nephrogenic factors for renal development, and the disruption of these genes or signaling activities may cause a spectrum of hypoplastic and cystic kidney disorders.


Assuntos
Rim/crescimento & desenvolvimento , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Rim Displásico Multicístico/etiologia , Proteínas Proto-Oncogênicas c-ret/fisiologia , Transdução de Sinais , Animais , Camundongos , Camundongos Knockout , Rim Displásico Multicístico/genética
9.
Dev Biol ; 389(1): 39-49, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24315854

RESUMO

The neurogenic cranial placodes are a unique transient epithelial niche of neural progenitor cells that give rise to multiple derivatives of the peripheral nervous system, particularly, the sensory neurons. Placode neurogenesis occurs throughout an extended period of time with epithelial cells continually recruited as neural progenitor cells. Sensory neuron development in the trigeminal, epibranchial, otic, and olfactory placodes coincides with detachment of these neuroblasts from the encompassing epithelial sheet, leading to delamination and ingression into the mesenchyme where they continue to differentiate as neurons. Multiple signaling pathways are known to direct placodal development. This review defines the signaling pathways working at the finite spatiotemporal period when neuronal selection within the placodes occurs, and neuroblasts concomitantly delaminate from the epithelium. Examining neurogenesis and delamination after initial placodal patterning and specification has revealed a common trend throughout the neurogenic placodes, which suggests that both activated FGF and attenuated Notch signaling activities are required for neurogenesis and changes in epithelial cell adhesion leading to delamination. We also address the varying roles of other pathways such as the Wnt and BMP signaling families during sensory neurogenesis and neuroblast delamination in the differing placodes.


Assuntos
Ectoderma/inervação , Sistema Nervoso/embriologia , Neurogênese , Transdução de Sinais , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Ectoderma/citologia , Ectoderma/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Humanos , Modelos Neurológicos , Sistema Nervoso/citologia , Sistema Nervoso/metabolismo , Receptores Notch/metabolismo
10.
Stem Cells ; 32(1): 45-58, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24115331

RESUMO

The Wnt/ß-catenin pathway is a critical stem cell regulator and plays important roles in neuroepithelial cells during early gestation. However, the role of Wnt/ß-catenin signaling in radial glia, a major neural stem cell population expanded by midgestation, remains poorly understood. This study shows that genetic ablation of ß-catenin with hGFAP-Cre mice inhibits neocortical formation by disrupting radial glial development. Reduced radial glia and intermediate progenitors are found in the ß-catenin-deficient neocortex during late gestation. Increased apoptosis and divergent localization of radial glia in the subventricular zone are also observed in the mutant neocortex. In vivo and in vitro proliferation and neurogenesis as well as oligodendrogenesis by cortical radial glia or by dissociated neural stem cells are significantly defective in the mutants. Neocortical layer patterning is not apparently altered, while astrogliogenesis is ectopically increased in the mutants. At the molecular level, the expression of the transcription factor Pax6 is dramatically diminished in the cortical radial glia and the sphere-forming neural stem cells of ß-catenin-deficient mutants. Chromatin immunoprecipitation and luciferase assays demonstrate that ß-catenin/Tcf complex binds to Pax6 promoter and induces its transcriptional activities. The forced expression of Pax6 through lentiviral transduction partially rescues the defective proliferation and neurogenesis by ß-catenin-deficient neural stem cells. Thus, Pax6 is a novel downstream target of the Wnt/ß-catenin pathway, and ß-catenin/Pax6 signaling plays critical roles in self-renewal and neurogenesis of radial glia/neural stem cells during neocortical development.


Assuntos
Proteínas do Olho/metabolismo , Proteínas de Homeodomínio/metabolismo , Neocórtex/citologia , Células-Tronco Neurais/citologia , Neurogênese/fisiologia , Fatores de Transcrição Box Pareados/metabolismo , Proteínas Repressoras/metabolismo , beta Catenina/metabolismo , Animais , Diferenciação Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Camundongos , Camundongos Transgênicos , Neocórtex/metabolismo , Células-Tronco Neurais/metabolismo , Neuroglia/citologia , Neuroglia/metabolismo , Fator de Transcrição PAX6 , Transdução de Sinais , Transfecção , Via de Sinalização Wnt , beta Catenina/genética
11.
J Cell Sci ; 124(Pt 9): 1553-63, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21486944

RESUMO

The mammalian olfactory epithelium (OE) has a unique stem cell or progenitor niche, which is responsible for the constant peripheral neurogenesis throughout the lifespan of the animal. However, neither the signals that regulate the behavior of these cells nor the lineage properties of the OE stem cells are well understood. Multiple Wnt signaling components exhibit dynamic expression patterns in the developing OE. We generated Wnt signaling reporter TOPeGFP transgenic mice and found TOPeGFP activation predominantly in proliferating Sox2(+) OE basal cells during early postnatal development. FACS-isolated TOPeGFP(+) OE basal cells are required, but are not sufficient, for formation of spheres. Wnt3a significantly promotes the proliferation of the Sox2(+) OE sphere cells. Wnt-stimulated OE sphere cells maintain their multipotency and can differentiate into most types of neuronal and non-neuronal epithelial cells. Also, Wnt activators shift the production of differentiated cells toward olfactory sensory neurons. Moreover, TOPeGFP(+) cells are robustly increased in the adult OE after injury. In vivo administration of Wnt modulators significantly alters the regeneration potential. This study demonstrates the role of the canonical Wnt signaling pathway in the regulation of OE stem cells or progenitors during development and regeneration.


Assuntos
Diferenciação Celular/fisiologia , Neurogênese/fisiologia , Mucosa Olfatória/citologia , Células-Tronco/citologia , Animais , Apoptose/genética , Apoptose/fisiologia , Diferenciação Celular/genética , Proliferação de Células , Células Cultivadas , Citometria de Fluxo , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Transgênicos , Neurogênese/genética , Neurônios/citologia , Neurônios/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteína Wnt3 , Proteína Wnt3A
12.
Birth Defects Res ; 115(19): 1851-1865, 2023 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-37435868

RESUMO

BACKGROUND: In addition to genomic risk variants and environmental influences, increasing evidence suggests epigenetic modifications are important for orofacial development and their alterations can contribute to orofacial clefts. Ezh2 encodes a core catalytic component of the Polycomb repressive complex responsible for addition of methyl marks to Histone H3 as a mechanism of repressing target genes. The role of Ezh2 in orofacial clefts remains unknown. AIMS: To investigate the epithelial role of Ezh2-dependent methylation in secondary palatogenesis. METHODS: We used conditional gene-targeting methods to ablate Ezh2 in the surface ectoderm-derived oral epithelium of mouse embryos. We then performed single-cell RNA sequencing combined with immunofluorescence and RT-qPCR to investigate gene expression in conditional mutant palate. We also employed double knockout analyses of Ezh1 and Ezh2 to address if they have synergistic roles in palatogenesis. RESULTS: We found that conditional inactivation of Ezh2 in oral epithelia results in partially penetrant cleft palate. Double knockout analyses revealed that another family member Ezh1 is dispensable in orofacial development, and it does not have synergistic roles with Ezh2 in palatogenesis. Histochemistry and single-cell RNA-seq analyses revealed dysregulation of cell cycle regulators in the palatal epithelia of Ezh2 mutant mouse embryos disrupts palatogenesis. CONCLUSION: Ezh2-dependent histone H3K27 methylation represses expression of cell cycle regulator Cdkn1a and promotes proliferation in the epithelium of the developing palatal shelves. Loss of this regulation may perturb movement of the palatal shelves, causing a delay in palate elevation which may result in failure of the secondary palate to close altogether.


Assuntos
Fenda Labial , Fissura Palatina , Animais , Camundongos , Fissura Palatina/genética , Fissura Palatina/metabolismo , Histonas/genética , Histonas/metabolismo , Metilação , Proteínas do Grupo Polycomb
13.
Birth Defects Res ; 115(19): 1835-1850, 2023 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-37497595

RESUMO

Orofacial clefts (OFCs) are one of the most common types of structural birth defects. The etiologies are complicated, involving with genetic, epigenetic, and environmental factors. Studies have found that maternal diabetes and metabolic syndrome are associated with a higher risk of OFCs in offspring. Metabolic syndrome is a clustering of several disease risk factors, including hyperglycemia, dyslipidemia, obesity, and hypertension. Metabolic disease during pregnancy can increase risk of adverse outcomes and significantly influence fetal development, including orofacial formation and fusion. An altered metabolic state may contribute to developmental disorders or congenital defects including OFCs, potentially through epigenetic modulations, such as histone modification, DNA methylation, and noncoding RNA expression to alter activities of critical morphogenetic signaling or related developmental genes. This review summarizes the currently available evidence and underlying mechanisms of how the maternal metabolic syndrome is associated with OFCs in mostly human and some animal studies. It may provide a better understanding of the interactions between intrauterine metabolic status and fetal orofacial development which might be applied toward prevention and treatments of OFCs.


Assuntos
Fenda Labial , Fissura Palatina , Diabetes Gestacional , Síndrome Metabólica , Gravidez , Feminino , Animais , Humanos , Fenda Labial/complicações , Fenda Labial/genética , Fissura Palatina/complicações , Fissura Palatina/genética , Síndrome Metabólica/complicações , Síndrome Metabólica/genética , Epigênese Genética
14.
Front Physiol ; 14: 704406, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37250135

RESUMO

The secondary palate forms from two lateral primordia called the palatal shelves which form a contact in the midline, become adherent at the fusing interface (medial edge epithelia, MEE) and subsequently fuse. The gene encoding transforming growth factor-ß3 (Tgfb3) is strongly and specifically expressed in MEE cells. Our previous study suggested that Tgfb3 expression is controlled via upstream cis-regulatory elements in and around the neighboring Ift43 gene. Another study suggested that the canonical Wnt signaling via ß-Catenin is responsible for the MEE-specific Tgfb3 gene expression, since deletion of the Ctnnb1 gene by a commonly used Keratin 14-Cre (K14Cre) mouse line almost completely abolished Tgfb3 expression in the MEE resulting in cleft palate. Here, we wanted to analyze whether Tcf/Lef consensus binding sites located in the previously identified regions of the Ift43 gene are responsible for the spatiotemporal control of Tgfb3 expression during palatogenesis. We show that contrary to the previous report, deletion of the Ctnnb1 gene in basal MEE cells by the K14Cre driver (the same K14Cre mouse line was used as in the previous study referenced above) does not affect the MEE-specific Tgfb3 expression or TGFß3-dependent palatal epithelial fusion. All mutant embryos showed a lack of palatal rugae accompanied by other craniofacial defects, e.g., a narrow snout and a small upper lip, while only a small subset (<5%) of Ctnnb1 mutants displayed a cleft palate. Moreover, the K14Cre:Ctnnb1 embryos showed reduced levels and altered patterns of Shh expression. Our present data imply that epithelial ß-catenin may not be required for MEE-specific Tgfb3 expression or palatal epithelial fusion.

15.
Dev Biol ; 349(2): 250-60, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21070765

RESUMO

The canonical Wnt/ß-catenin signaling pathway has implications in early facial development; yet, its function and signaling mechanism remain poorly understood. We report here that the frontonasal and upper jaw primordia cannot be formed after conditional ablation of ß-catenin with Foxg1-Cre mice in the facial ectoderm and the adjacent telencephalic neuroepithelium. Gene expression of several cell-survival and patterning factors, including Fgf8, Fgf3, and Fgf17, is dramatically diminished in the anterior neural ridge (ANR, a rostral signaling center) and/or the adjacent frontonasal ectoderm of the ß-catenin conditional mutant mice. In addition, Shh expression is diminished in the ventral telencephalon of the mutants, while Tcfap2a expression is less affected in the facial primordia. Apoptosis occurs robustly in the rostral head tissues following inactivation of Fgf signaling in the conditional mutants. Consequently, the upper jaw, nasal, ocular and telencephalic structures are absent, but the tongue and mandible are relatively developed in the conditional mutants at birth. Using molecular biological approaches, we demonstrate that the Fgf8 gene is transcriptionally targeted by Wnt/ß-catenin signaling during early facial and forebrain development. Furthermore, we show that conditional gain-of-function of ß-catenin signaling causes drastic upregulation of Fgf8 mRNA in the ANR and the entire facial ectoderm, which also arrests facial and forebrain development. Taken together, our results suggest that canonical Wnt/ß-catenin signaling is required for early development of the mammalian face and related head structures, which mainly or partly acts through the initiation and modulation of balanced Fgf signaling activity.


Assuntos
Ossos Faciais/embriologia , Fatores de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Transdução de Sinais/fisiologia , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Técnicas de Ablação , Animais , Apoptose/fisiologia , Imunoprecipitação da Cromatina , Primers do DNA/genética , Ectoderma/metabolismo , Imunofluorescência , Fatores de Transcrição Forkhead/genética , Proteínas Hedgehog/metabolismo , Hibridização In Situ , Marcação In Situ das Extremidades Cortadas , Luciferases , Camundongos , Camundongos Mutantes , Proteínas do Tecido Nervoso/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Telencéfalo/citologia , Telencéfalo/metabolismo , beta Catenina/genética
16.
Development ; 136(18): 3161-71, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19700620

RESUMO

Neither the mechanisms that govern lip morphogenesis nor the cause of cleft lip are well understood. We report that genetic inactivation of Lrp6, a co-receptor of the Wnt/beta-catenin signaling pathway, leads to cleft lip with cleft palate. The activity of a Wnt signaling reporter is blocked in the orofacial primordia by Lrp6 deletion in mice. The morphological dynamic that is required for normal lip formation and fusion is disrupted in these mutants. The expression of the homeobox genes Msx1 and Msx2 is dramatically reduced in the mutants, which prevents the outgrowth of orofacial primordia, especially in the fusion site. We further demonstrate that Msx1 and Msx2 (but not their potential regulator Bmp4) are the downstream targets of the Wnt/beta-catenin signaling pathway during lip formation and fusion. By contrast, a ;fusion-resistant' gene, Raldh3 (also known as Aldh1a3), that encodes a retinoic acid-synthesizing enzyme is ectopically expressed in the upper lip primordia of Lrp6-deficient embryos, indicating a region-specific role of the Wnt/beta-catenin signaling pathway in repressing retinoic acid signaling. Thus, the Lrp6-mediated Wnt signaling pathway is required for lip development by orchestrating two distinctively different morphogenetic movements.


Assuntos
Proteínas Relacionadas a Receptor de LDL/metabolismo , Lábio/embriologia , Morfogênese/fisiologia , Transdução de Sinais/fisiologia , Proteínas Wnt/metabolismo , Aldeído Oxirredutases/genética , Aldeído Oxirredutases/metabolismo , Animais , Apoptose/fisiologia , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Proliferação de Células , Fenda Labial/metabolismo , Fenda Labial/patologia , Embrião de Mamíferos/anatomia & histologia , Embrião de Mamíferos/fisiologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Proteínas Relacionadas a Receptor de LDL/genética , Lábio/anatomia & histologia , Lábio/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Fator de Transcrição MSX1/genética , Fator de Transcrição MSX1/metabolismo , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Retinal Desidrogenase , Proteínas Wnt/genética
17.
Stem Cells ; 29(6): 907-12, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21557389

RESUMO

The adult mammalian central nervous system (CNS) lacks the capacity for regeneration, making it a highly sought-after topic for researchers. The identification of neural stem cells (NSCs) in the adult CNS wiped out a long-held dogma that the adult brain contains a set number of neurons and is incapable of replacing them. The discovery of adult NSCs (aNSCs) stoked the fire for researchers who dream of brain self-repair. Unfortunately, the quiescent nature and limited plasticity of aNSCs diminish their regenerative potential. Recent studies evaluating aNSC plasticity under pathological conditions indicate that a switch from quiescent to active aNSCs in neurogenic regions plays an important role in both repairing the damaged tissue and preserving progenitor pools. Here, we summarize the most recent findings and present questions about characterizing the active and quiescent aNSCs in major neurogenic regions, and factors for maintaining their active and quiescent states, hoping to outline an emerging view for promoting the endogenous aNSC-based regeneration.


Assuntos
Células-Tronco Adultas/fisiologia , Proliferação de Células , Senescência Celular , Células-Tronco Neurais/fisiologia , Animais , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular , Hipocampo/citologia , Hipocampo/fisiologia , Humanos , Ventrículos Laterais/citologia , Ventrículos Laterais/fisiologia , Mucosa Olfatória/citologia , Mucosa Olfatória/fisiologia , Nicho de Células-Tronco/metabolismo
18.
Dis Model Mech ; 15(6)2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35514236

RESUMO

Neural tube defects (NTDs) are among the common and severe birth defects with poorly understood etiology. Mutations in the Wnt co-receptor LRP6 are associated with NTDs in humans. Either gain-of-function (GOF) or loss-of-function (LOF) mutations of Lrp6 can cause NTDs in mice. NTDs in Lrp6-GOF mutants may be attributed to altered ß-catenin-independent noncanonical Wnt signaling. However, the mechanisms underlying NTDs in Lrp6-LOF mutants and the role of Lrp6-mediated canonical Wnt/ß-catenin signaling in neural tube closure remain unresolved. We previously demonstrated that ß-catenin signaling is required for posterior neuropore (PNP) closure. In the current study, conditional ablation of Lrp6 in dorsal PNP caused spinal NTDs with diminished activities of Wnt/ß-catenin signaling and its downstream target gene Pax3, which is required for PNP closure. ß-catenin-GOF rescued NTDs in Lrp6-LOF mutants. Moreover, maternal supplementation of a Wnt/ß-catenin signaling agonist reduced the frequency and severity of spinal NTDs in Lrp6-LOF mutants by restoring Pax3 expression. Together, these results demonstrate the essential role of Lrp6-mediated Wnt/ß-catenin signaling in PNP closure, which could also provide a therapeutic target for NTD intervention through manipulation of canonical Wnt/ß-catenin signaling activities.


Assuntos
Defeitos do Tubo Neural , Via de Sinalização Wnt , Animais , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Camundongos , Tubo Neural/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Via de Sinalização Wnt/genética , beta Catenina/metabolismo
19.
Front Physiol ; 13: 835198, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35350693

RESUMO

Circadian factors likely influence the occurrence, development, therapy, and prognosis of cardiovascular diseases (CVDs). To determine the association between the heart rate (HR) diurnal parameters and CVD risks, we designed an analytical strategy to detect diurnal rhythms of HR using longitudinal data collected by clinically used Holter monitors and wearable devices. By combining in-house developed algorithms with existing analytical tools, we obtained trough phase and nocturnal variation in HR for different purposes. The analytical strategy is robust and also sensitive enough to identify variations in HR rhythms influenced by multiple effectors such as jet lag, geological location and altitude, and age from total 211 volunteers. A total of 10,094 sets of 24-h Holter ECG data were analyzed by stepwise partial correlation to determine the critical points of HR trough phase and nocturnal variation. The following HR diurnal patterns correlate with high CVD risk: arrhythmic pattern, anti-phase pattern, rhythmic patterns with trough phase less than 0 (extremely advanced diurnal pattern) or more than 5 (extremely delayed diurnal pattern), and nocturnal variation less than 2.75 (extremely low) or more than 26 (extremely high). In addition, HR trough phases from wearable devices were nearly identical to those from 24-h Holter monitoring from 12 volunteers by linear correlation and Bland-Altman analysis. Our analytical system provides useful information to identify functional diurnal patterns and parameters by monitoring personalized, HR-based diurnal changes. These findings have important implications for understanding how a regular heart diurnal pattern benefits cardiac function and raising the possibility of non-pharmacological intervention against circadian related CVDs. With the rapid expansion of wearable devices, public cardiovascular health can be promoted if the analytical strategy is widely applied.

20.
Cell Death Discov ; 8(1): 345, 2022 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-35918318

RESUMO

Exploring the functions of human-specific genes (HSGs) is challenging due to the lack of a tractable genetic model system. Testosterone is essential for maintaining human spermatogenesis and fertility, but the underlying mechanism is unclear. Here, we identified Cancer/Testis Antigen gene family 47 (CT47) as an essential regulator of human-specific spermatogenesis by stabilizing arginine methyltransferase 5 (PRMT5). A humanized mouse model revealed that CT47 functions to arrest spermatogenesis by interacting with and regulating CT47/PRMT5 accumulation in the nucleus during the leptotene/zygotene-to-pachytene transition of meiosis. We demonstrate that testosterone induces nuclear depletion of CT47/PRMT5 and rescues leptotene-arrested spermatocyte progression in humanized testes. Loss of CT47 in human embryonic stem cells (hESCs) by CRISPR/Cas9 led to an increase in haploid cells but blocked the testosterone-induced increase in haploid cells when hESCs were differentiated into haploid spermatogenic cells. Moreover, CT47 levels were decreased in nonobstructive azoospermia. Together, these results established CT47 as a crucial regulator of human spermatogenesis by preventing meiosis initiation before the testosterone surge.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa