Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 154
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 543(7645): 424-427, 2017 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-28273069

RESUMO

The small intestinal epithelium self-renews every four or five days. Intestinal stem cells (Lgr5+ crypt base columnar cells (CBCs)) sustain this renewal and reside between terminally differentiated Paneth cells at the bottom of the intestinal crypt. Whereas the signalling requirements for maintaining stem cell function and crypt homeostasis have been well studied, little is known about how metabolism contributes to epithelial homeostasis. Here we show that freshly isolated Lgr5+ CBCs and Paneth cells from the mouse small intestine display different metabolic programs. Compared to Paneth cells, Lgr5+ CBCs display high mitochondrial activity. Inhibition of mitochondrial activity in Lgr5+ CBCs or inhibition of glycolysis in Paneth cells strongly affects stem cell function, as indicated by impaired organoid formation. In addition, Paneth cells support stem cell function by providing lactate to sustain the enhanced mitochondrial oxidative phosphorylation in the Lgr5+ CBCs. Mechanistically, we show that oxidative phosphorylation stimulates p38 MAPK activation by mitochondrial reactive oxygen species signalling, thereby establishing the mature crypt phenotype. Together, our results reveal a critical role for the metabolic identity of Lgr5+ CBCs and Paneth cells in supporting optimal stem cell function, and we identify mitochondria and reactive oxygen species signalling as a driving force of cellular differentiation.


Assuntos
Autorrenovação Celular , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Intestino Delgado/citologia , Intestino Delgado/metabolismo , Células-Tronco/citologia , Animais , Diferenciação Celular , Meios de Cultivo Condicionados/química , Meios de Cultivo Condicionados/farmacologia , Glicólise , Homeostase , Ácido Láctico/metabolismo , Camundongos , Mitocôndrias/metabolismo , Organoides/citologia , Organoides/efeitos dos fármacos , Organoides/metabolismo , Fosforilação Oxidativa , Celulas de Paneth/citologia , Celulas de Paneth/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Células-Tronco/fisiologia , Proteína Wnt3A/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
2.
J Biochem Mol Toxicol ; 36(12): e23213, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36111657

RESUMO

Stroke is a life-threatening disease with limited therapeutic options. Damage to the blood-brain barrier (BBB) is the key pathological feature of ischemic stroke. This study explored the role of the bradykinin (BK)/bradykinin 1 receptor (B1R) and its mechanism of action in the BBB. Human brain microvascular endothelial cells (BMECs) were used to test for cellular responses to BK by using the Cell Counting Kit-8 assay, 5-ethynyl-2'-deoxyuridine staining, enzyme-linked immunosorbent assay, flow cytometry, immunofluorescence, cellular permeability assays, and western blotting to evaluate cell viability, cytokine production, and reactive oxygen species (ROS) levels in vitro. A BBB induced by middle cerebral artery occlusion was used to evaluate BBB injuries, and the role played by BK/B1R in ischemic/reperfusion (I/R) was explored in a rat model. Results showed that BK reduced the viability of BMECs and increased the levels of proinflammatory cytokines (interleukin 6 [IL-6], IL-18, and monocyte chemoattractant protein-1) and ROS. Additionally, cellular permeability was increased by BK treatment, and the expression of tight junction proteins (claudin-5 and occludin) was decreased. Interestingly, Wnt3a expression was inhibited by BK and exogenous Wnt3a restored the effects of BK on BMECs. In an in vivo I/R rat model, knockdown of B1R significantly decreased infarct volume and inflammation in I/R rats. Our results suggest that BK might be a key inducer of BBB injury and B1R knockdown might provide a beneficial effect by upregulating Wnt3a.


Assuntos
Células Endoteliais , Receptores da Bradicinina , Animais , Ratos , Humanos , Células Endoteliais/metabolismo , Receptores da Bradicinina/metabolismo , Bradicinina/farmacologia , Bradicinina/metabolismo , Citocinas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Permeabilidade , Proteína Wnt3A/metabolismo , Proteína Wnt3A/farmacologia
3.
Med Mol Morphol ; 55(3): 199-209, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35578118

RESUMO

We investigated whether BMP4, FGF8, and/or WNT3a on neural crest-like cells (NCLC) derived from mouse induced pluripotent stem (miPS) cells will promote differentiation of odontoblasts-like cells. After the miPS cells matured into embryonic body (EB) cells, they were cultured in a neural induction medium to produce NCLC. As the differentiation of NCLC were confirmed by RT-qPCR, they were then disassociated and cultured with a medium containing, BMP4, FGF8, and/or WNT3a for 7 and 14 days. The effect of these stimuli on NCLC were assessed by RT-qPCR, ALP staining, and immunocytochemistry. The cultured EB cells presented a significant increase of Snai1, Slug, and Sox 10 substantiating the differentiation of NCLC. NCLC stimulated with more than two stimuli significantly increased the odontoblast markers Dmp-1, Dspp, Nestin, Alp, and Runx2 expression compared to control with no stimulus. The expression of Dmp-1 and Dspp upregulated more when FGF8 was combined with WNT3a. ALP staining was positive in groups containing BMP4 and fluorescence was observed in immunocytochemistry of the common significant groups between Dmp-1 and Dspp. After stimulation, the cell morphology demonstrated a spindle-shaped cells with long projections resembling odontoblasts. Simultaneous BMP4, FGF8, and WNT3a stimuli significantly differentiated NCLC into odontoblast-like cells.


Assuntos
Proteína Morfogenética Óssea 4 , Fator 8 de Crescimento de Fibroblasto , Células-Tronco Pluripotentes Induzidas , Odontoblastos , Animais , Proteína Morfogenética Óssea 4/farmacologia , Diferenciação Celular , Células Cultivadas , Fator 8 de Crescimento de Fibroblasto/farmacologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Camundongos , Crista Neural , Odontoblastos/metabolismo , Proteína Wnt3A/farmacologia
4.
Arterioscler Thromb Vasc Biol ; 40(9): 2244-2264, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32640900

RESUMO

OBJECTIVE: Nanog is expressed in adult endothelial cells (ECs) at a low-level, however, its functional significance is not known. The goal of our study was to elucidate the role of Nanog in adult ECs using a genetically engineered mouse model system. Approach and Results: Biochemical analyses showed that Nanog is expressed in both adult human and mouse tissues. Primary ECs isolated from adult mice showed detectable levels of Nanog, Tert (telomerase reverse transcriptase), and eNos (endothelial nitric oxide synthase). Wnt3a (Wnt family member 3A) increased the expression of Nanog and hTERT (human telomerase reverse transcriptase) in ECs and increased telomerase activity in these cells. In a chromatin immunoprecipitation experiment, Nanog directly bound to the hTERT and eNOS promoter/enhancer DNA elements, thereby regulating their transcription. Administration of low-dose tamoxifen to ROSAmT/mG::Nanogfl/+::Cdh5CreERT2 mice induced deletion of a single Nanog allele, simultaneously labeling ECs with green fluorescent protein and resulting in decreased Tert and eNos levels. Histological and morphometric analyses of heart tissue sections prepared from these mice revealed cell death, microvascular rarefaction, and increased fibrosis in cardiac vessels. Accordingly, EC-specific Nanog-haploinsufficiency resulted in impaired EC homeostasis and angiogenesis. Conversely, re-expression of cDNA encoding the hTERT in Nanog-depleted ECs, in part, restored the effect of loss of Nanog. CONCLUSIONS: We showed that low-level Nanog expression is required for normal EC homeostasis and angiogenesis in adulthood.


Assuntos
Proliferação de Células , Senescência Celular , Vasos Coronários/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Proteína Homeobox Nanog/metabolismo , Animais , Apoptose , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/patologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Feminino , Fibrose , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Homeobox Nanog/deficiência , Proteína Homeobox Nanog/genética , Neovascularização Fisiológica , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Telomerase/genética , Telomerase/metabolismo , Ativação Transcricional , Via de Sinalização Wnt , Proteína Wnt3A/farmacologia
5.
Mol Cell ; 49(3): 474-86, 2013 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-23273980

RESUMO

Nuclear accumulation of ß-catenin, a widely recognized marker of poor cancer prognosis, drives cancer cell proliferation and senescence bypass and regulates incretins, critical regulators of fat and glucose metabolism. Diabetes, characterized by elevated blood glucose levels, is associated with increased cancer risk, partly because of increased insulin growth factor 1 signaling, but whether elevated glucose directly impacts cancer-associated signal-transduction pathways is unknown. Here, we show that high glucose is essential for nuclear localization of ß-catenin in response to Wnt signaling. Glucose-dependent ß-catenin nuclear retention requires lysine 354 and is mediated by alteration of the balance between p300 and sirtuins that trigger ß-catenin acetylation. Consequently ß-catenin accumulates in the nucleus and activates target promoters under combined glucose and Wnt stimulation, but not with either stimulus alone. Our results reveal a mechanism by which high glucose enhances signaling through the cancer-associated Wnt/ß-catenin pathway and may explain the increased frequency of cancer associated with obesity and diabetes.


Assuntos
Glucose/farmacologia , Neoplasias/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , beta Catenina/metabolismo , Acetilação/efeitos dos fármacos , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Cromatina/metabolismo , Citosol/efeitos dos fármacos , Citosol/metabolismo , Proteína p300 Associada a E1A/metabolismo , Polipeptídeo Inibidor Gástrico/genética , Polipeptídeo Inibidor Gástrico/metabolismo , Humanos , Cloreto de Lítio/farmacologia , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Neoplasias/patologia , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , Sirtuínas/metabolismo , Fatores de Transcrição TCF/metabolismo , Transcrição Gênica/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/genética , Proteína Wnt3A/farmacologia
6.
J Biol Chem ; 294(31): 11677-11684, 2019 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-31235524

RESUMO

The network of Wingless/Int-1 (WNT)-induced signaling pathways includes ß-catenin-dependent and -independent pathways. ß-Catenin regulates T cell factor/lymphoid enhancer-binding factor (TCF/LEF)-mediated gene transcription, and in response to WNTs, ß-catenin signaling is initiated through engagement of a Frizzled (FZD)/LDL receptor-related protein 5/6 (LRP5/6) receptor complex. FZDs are G protein-coupled receptors, but the question of whether heterotrimeric G proteins are involved in WNT/ß-catenin signaling remains unanswered. Here, we investigate whether acute activation of WNT/ß-catenin signaling by purified WNT-3A requires functional signaling through heterotrimeric G proteins. Using genome editing, we ablated expression of Gs/Golf/Gq/G11/G12/G13/Gz in HEK293 (ΔG7) cells, leaving the expression of pertussis toxin (PTX)-sensitive Gi/o proteins unchanged, to assess whether WNT-3A activates WNT/ß-catenin signaling in WT and ΔG7 cells devoid of functional G protein signaling. We monitored WNT-3A-induced activation by detection of phosphorylation of LDL receptor-related protein 6 (LRP6), electrophoretic mobility shift of the phosphoprotein Dishevelled (DVL), ß-catenin stabilization and dephosphorylation, and TCF-dependent transcription. We found that purified, recombinant WNT-3A efficiently induces WNT/ß-catenin signaling in ΔG7 cells in both the absence and presence of Gi/o-blocking PTX. Furthermore, cells completely devoid of G protein expression, so called Gα-depleted HEK293 cells, maintain responsiveness to WNT-3A with regard to the hallmarks of WNT/ß-catenin signaling. These findings corroborate the concept that heterotrimeric G proteins are not required for this FZD- and DVL-mediated signaling branch. Our observations agree with previous results arguing for FZD conformation-dependent functional selectivity between DVL and heterotrimeric G proteins. In conclusion, WNT/ß-catenin signaling through FZDs does not require the involvement of heterotrimeric G proteins.


Assuntos
Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , Proteína Wnt3A/farmacologia , Proteínas Desgrenhadas , Edição de Genes , Células HEK293 , Proteínas Heterotriméricas de Ligação ao GTP/genética , Humanos , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Toxina Pertussis/farmacologia , Fosforilação , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/farmacologia , Fatores de Transcrição TCF/metabolismo , Proteína Wnt3A/genética , Proteína Wnt3A/metabolismo , beta Catenina/metabolismo
7.
Mol Cell Proteomics ; 17(5): 901-912, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29386236

RESUMO

Tumor: tumor microenvironment (TME) interactions are critical for tumor progression and the composition and structure of the local extracellular matrix (ECM) are key determinants of tumor metastasis. We recently reported that activation of Wnt/beta-catenin signaling in Ewing sarcoma cells induces widespread transcriptional changes that are associated with acquisition of a metastatic tumor phenotype. Significantly, ECM protein-encoding genes were found to be enriched among Wnt/beta-catenin induced transcripts, leading us to hypothesize that activation of canonical Wnt signaling might induce changes in the Ewing sarcoma secretome. To address this hypothesis, conditioned media from Ewing sarcoma cell lines cultured in the presence or absence of Wnt3a was collected for proteomic analysis. Label-free mass spectrometry was used to identify and quantify differentially secreted proteins. We then used in silico databases to identify only proteins annotated as secreted. Comparison of the secretomes of two Ewing sarcoma cell lines revealed numerous shared proteins, as well as a degree of heterogeneity, in both basal and Wnt-stimulated conditions. Gene set enrichment analysis of secreted proteins revealed that Wnt stimulation reproducibly resulted in increased secretion of proteins involved in ECM organization, ECM receptor interactions, and collagen formation. In particular, Wnt-stimulated Ewing sarcoma cells up-regulated secretion of structural collagens, as well as matricellular proteins, such as the metastasis-associated protein, tenascin C (TNC). Interrogation of published databases confirmed reproducible correlations between Wnt/beta-catenin activation and TNC and COL1A1 expression in patient tumors. In summary, this first study of the Ewing sarcoma secretome reveals that Wnt/beta-catenin activated tumor cells upregulate secretion of ECM proteins. Such Wnt/beta-catenin mediated changes are likely to impact on tumor: TME interactions that contribute to metastatic progression.


Assuntos
Proteoma/metabolismo , Sarcoma de Ewing/metabolismo , Via de Sinalização Wnt , Linhagem Celular Tumoral , Matriz Extracelular/metabolismo , Humanos , Espectrometria de Massas , Proteínas de Neoplasias/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Via de Sinalização Wnt/efeitos dos fármacos , Proteína Wnt3A/farmacologia
8.
J Neurosci ; 38(30): 6787-6801, 2018 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-29954850

RESUMO

After ischemic stroke, apoptosis of neurons is a primary factor in determining outcome. Wnt3a is a naturally occurring protein that has been shown to have protective effects in the brain for traumatic brain injury. Although wnt3a has been investigated in the phenomena of neurogenesis, anti-apoptosis, and anti-inflammation, it has never been investigated as a therapy for stroke. We hypothesized that the potential neuroprotective agent wnt3a would reduce infarction and improve behavior following ischemic stroke by attenuating neuronal apoptosis and promoting cell survival through the Frizzled-1/PIWI1a/FOXM1 pathway in middle cerebral artery occlusion (MCAO) rats. A total of 229 Sprague Dawley rats were assigned to male, female, and 9-month-old male MCAO or sham groups followed by reperfusion 2 h after MCAO. Animals assigned to MCAO were either given wnt3a or its control. To explore the downstream signaling of wnt3a, the following interventions were given: Frizzled-1 siRNA, PIWI1a siRNA, and PIWI1a-clustered regularly interspaced short palindromic repeats, along with the appropriate controls. Post-MCAO assessments included neurobehavioral tests, infarct volume, Western blot, and immunohistochemistry. Endogenous levels of wnt3a and Frizzled-1/PIWI1a/FOXM1 were lowered after MCAO. The administration of intranasal wnt3a, 1 h after MCAO, increased PIWIL1a and FOXM1 expression through Frizzled-1, reducing brain infarction and neurological deficits at 24 and 72 h. Frizzled-1 and PIWI1a siRNAs reversed the protective effects of wnt3a after MCAO. Restoration of PIWI1a after knockdown of Frizzled-1 increased FOXM1 survival protein and reduced cleaved caspase-3 levels. In summary, wnt3a decreases neuronal apoptosis and improves neurological deficits through Frizzled-1/PIWI1a/FOXM1 pathway after MCAO in rats. Therefore, wnt3a is a novel intranasal approach to decrease apoptosis after stroke.SIGNIFICANCE STATEMENT Only 5% of patients receive recombinant tissue plasminogen activator after stroke, and few qualify for mechanical thrombectomy. No neuroprotective agents have been successfully translated to promote neuronal survival in stroke. Thus, using a clinically relevant rat model of stroke, middle cerebral artery occlusion, we explored a novel intranasal administration of wnt3a. wnt3a naturally occurs in the body and crosses the blood-brain barrier, supporting the clinically translatable approach of intranasal administration. Significant neuronal apoptosis occurs during stroke, and wnt3a shows promise due to its antiapoptotic effects. We investigated whether wnt3a mediates its poststroke effects via Frizzled-1 and the impact on its downstream signaling molecules, PIWI1a and FOXM1, in apoptosis. Elucidating the mechanism of wnt3a will identify additional pharmacological targets and further understanding of stroke.


Assuntos
Apoptose/efeitos dos fármacos , Infarto da Artéria Cerebral Média/patologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Proteína Wnt3A/farmacologia , Administração Intranasal , Animais , Apoptose/fisiologia , Proteínas Argonautas/efeitos dos fármacos , Proteínas Argonautas/metabolismo , Feminino , Proteína Forkhead Box M1/efeitos dos fármacos , Proteína Forkhead Box M1/metabolismo , Receptores Frizzled/efeitos dos fármacos , Receptores Frizzled/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Masculino , Neurônios/metabolismo , Neurônios/patologia , Ratos , Ratos Sprague-Dawley , Receptores de Neurotransmissores/efeitos dos fármacos , Receptores de Neurotransmissores/metabolismo , Transdução de Sinais/efeitos dos fármacos
9.
Osteoporos Int ; 30(9): 1873-1885, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31338519

RESUMO

Our goal was to evaluate alveolar bone healing in OVX mice, and to assess the functional utility of a WNT-based treatment to accelerate healing in mice with an osteoporotic-like bony phenotype. INTRODUCTION: Is osteoporosis a risk factor for dental procedures? This relatively simple question is exceedingly difficult to answer in a clinical setting, for two reasons. First, as an age-related disease, osteoporosis is frequently accompanied by age-related co-morbidities that can contribute to slower tissue repair. Second, the intervals at which alveolar bone repair are assessed in a clinical study are often measured in months to years. This study aimed to evaluate alveolar bone repair in ovariectomized (OVX) mice and provide preclinical evidence to support a WNT-based treatment to accelerate alveolar bone formation. METHODS: OVX was performed in young mice to produce an osteoporotic-like bone phenotype. Thereafter, the rate of extraction socket healing and osteotomy repair was assessed. A liposomal WNT3A treatment was tested for its ability to promote alveolar bone formation in this OVX-induced model of bone loss. RESULTS: Bone loss was observed throughout the murine skeleton, including the maxilla, and mirrored the pattern of bone loss observed in aged mice. Injuries to the alveolar bone, including tooth extraction and osteotomy site preparation, both healed significantly slower than the same injuries produced in young controls. Given sufficient time, however, all injuries eventually healed. In OVX mice, osteotomies healed significantly faster if they were treated with L-WNT3A. CONCLUSIONS: Alveolar bone injuries heal slower in OVX mice that exhibit an osteoporotic-like phenotype. The rate of alveolar bone repair in OVX mice can be significantly promoted with local delivery of L-WNT3A.


Assuntos
Regeneração Óssea/efeitos dos fármacos , Osteoporose/fisiopatologia , Alvéolo Dental/efeitos dos fármacos , Proteína Wnt3A/farmacologia , Envelhecimento/fisiologia , Perda do Osso Alveolar/diagnóstico por imagem , Perda do Osso Alveolar/tratamento farmacológico , Perda do Osso Alveolar/etiologia , Perda do Osso Alveolar/fisiopatologia , Animais , Reabsorção Óssea/fisiopatologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos/métodos , Feminino , Maxila/efeitos dos fármacos , Maxila/fisiologia , Camundongos Endogâmicos BALB C , Dente Molar/cirurgia , Osteogênese/fisiologia , Osteoporose/complicações , Ovariectomia , Extração Dentária , Alvéolo Dental/diagnóstico por imagem , Alvéolo Dental/fisiologia , Cicatrização/efeitos dos fármacos , Microtomografia por Raio-X/métodos
10.
J Am Soc Nephrol ; 29(1): 182-193, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29021383

RESUMO

The Wnt/ß-catenin pathway is crucial in normal development and throughout life, but aberrant activation of this pathway has been linked to kidney fibrosis, although the mechanisms involved remain incompletely determined. Here, we investigated the role of Wnt/ß-catenin in regulating macrophage activation and the contribution thereof to kidney fibrosis. Treatment of macrophages with Wnt3a exacerbated IL-4- or TGFß1-induced macrophage alternative (M2) polarization and the phosphorylation and nuclear translocation of STAT3 in vitro Conversely, inhibition of Wnt/ß-catenin signaling prevented these IL-4- or TGFß1-induced processes. In a mouse model, induced deletion of ß-catenin in macrophages attenuated the fibrosis, macrophage accumulation, and M2 polarization observed in the kidneys of wild-type littermates after unilateral ureter obstruction. This study shows that activation of Wnt/ß-catenin signaling promotes kidney fibrosis by stimulating macrophage M2 polarization.


Assuntos
Rim/patologia , Ativação de Macrófagos , Macrófagos/fisiologia , Via de Sinalização Wnt , beta Catenina/genética , beta Catenina/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Linhagem Celular , Fibrose , Interleucina-4/metabolismo , Masculino , Camundongos , Camundongos Knockout , Fosforilação/efeitos dos fármacos , Fator de Transcrição STAT3/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Obstrução Ureteral/complicações , Proteína Wnt3A/farmacologia
11.
J Cell Biochem ; 119(1): 1041-1049, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28853173

RESUMO

mTORC1 signaling not only plays important physiological roles in the regulation of proliferation and osteogenic differentiation of BMSCs, but also mediates exogenous Wnt-induced protein anabolism and osteoblast differentiation. However, the downstream effectors of the mTORC1 signaling in the above processes are still poorly understood. In this study, we explored the specific role of S6K1, one of the major targets of the mTORC1 pathway, in BMSCs self-renewal and osteogenic differentiation. We first found that S6K1 was active in primary mouse bone marrow stromal cells, and further activated upon osteogenic induction. We then determined the effects of S6K1 inhibition by LY2584702 Tosylate, a selective inhibitor of S6K1 (hereafter S6KI), using both primary mouse bone marrow stromal cells and ST2 cells. Colony-Forming Unit-Fibroblast (CFU-F) assays showed that S6KI dramatically reduced the total number of colonies formed in primary BMSCs cultures. Under the basal osteogenic culture condition, S6KI significantly inhibited mRNA expression of osteoblast marker genes (Sp7, Bglap, Ibsp, and Col1a1), ALP activity and matrix mineralization. Upon Wnt3a treatments, S6KI inhibited Wnt3a-induced osteoblast differentiation and expression of protein anabolism genes in ST2 cells, but to a much lesser degree than rapamycin (a specific inhibitor of mTORC1 signaling). Collectively, our findings have demonstrated that pharmacological inhibition of S6K1 impaired self-renewal and osteogenic differentiation of BMSCs, but only partially suppressed exogenous Wnt3a-induced osteoblast differentiation and protein anabolism.


Assuntos
Células-Tronco Mesenquimais/citologia , Osteogênese/efeitos dos fármacos , Pirazóis/farmacologia , Pirimidinas/farmacologia , Proteínas Quinases S6 Ribossômicas 90-kDa/antagonistas & inibidores , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Transdução de Sinais/efeitos dos fármacos , Proteína Wnt3A/farmacologia
12.
J Cell Sci ; 129(20): 3892-3902, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27744318

RESUMO

Dishevelled (DVL) assembles Wnt signalosomes through dynamic head-to-tail polymerisation by means of its DIX domain. It thus transduces Wnt signals to cytoplasmic effectors including ß-catenin, to control cell fates during normal development, tissue homeostasis and also in cancer. To date, most functional studies of Dishevelled relied on its Wnt-independent signalling activity resulting from overexpression, which is sufficient to trigger polymerisation, bypassing the requirement for Wnt signals. Here, we generate a human cell line devoid of endogenous Dishevelled (DVL1- DVL3), which lacks Wnt signal transduction to ß-catenin. However, Wnt responses can be restored by DVL2 stably re-expressed at near-endogenous levels. Using this assay to test mutant DVL2, we show that its DEP domain is essential, whereas its PDZ domain is dispensable, for signalling to ß-catenin. Our results imply two mutually exclusive functions of the DEP domain in Wnt signal transduction - binding to Frizzled to recruit Dishevelled to the receptor complex, and dimerising to cross-link DIX domain polymers for signalosome assembly. Our assay avoids the caveats associated with overexpressing Dishevelled, and provides a powerful tool for rigorous functional tests of this pivotal human signalling protein.


Assuntos
Bioensaio/métodos , Proteínas Desgrenhadas/química , Proteínas Desgrenhadas/metabolismo , Proteína Wnt3A/farmacologia , Regulação para Baixo/efeitos dos fármacos , Receptores Frizzled/metabolismo , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Mutação/genética , Domínios PDZ , Peptídeos/metabolismo , Multimerização Proteica/efeitos dos fármacos , Relação Estrutura-Atividade , Via de Sinalização Wnt/efeitos dos fármacos , beta Catenina/metabolismo
13.
Gastroenterology ; 153(1): 154-165.e9, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28359679

RESUMO

BACKGROUND & AIMS: Neural stem and progenitor cells from the enteric nervous system (ENS) might serve as a source of cells for treatment of neurogastrointestinal disorders. Before we can use these cells, we must increase our understanding of the signaling mechanisms that regulate proliferation and differentiation. We systematically evaluated the effects of canonical Wnt signaling on proliferation and differentiation of cultured ENS progenitor cells from neonatal mice and humans. METHODS: We isolated ENS progenitors from tunica muscularis of the small intestine of newborn (postnatal day 0) wild-type C57BL/6 mice as well as from Wnt1-Cre2 reporter mice. We also obtained intestinal tissue samples from infants (2 and 7 months old) undergoing surgery for imperforate anus or focal intestinal perforation and isolated ENS cells. ENS cells were cultured under proliferation conditions leading to formation of 3-dimensional spheres, which we activated with Wnt3a and SB216763 in order to activate the ß-catenin-dependent canonical Wnt pathway. We used immunoblot and quantitative polymerase chain reaction to evaluate the molecular response to Wnt stimuli and immunohistochemistry, proliferation, and cell death assays to identify new neurons. RESULTS: In proliferating enterospheres derived from ENS progenitor cells, we verified the expression of Wnt receptors frizzled 1-10 and the co-receptors low-density lipoprotein receptor-related proteins 5 and 6. Pharmacologic stimulation with Wnt agonists led to intracellular accumulation of Wnt-dependent ß-catenin and up-regulated expression of known Wnt target genes axin2, lef1, and lgr5. Activation of the canonical Wnt pathway promoted growth of ENS cell spheres during cell expansion and increased the number of newborn neurons derived from mouse and human progenitor cells. CONCLUSIONS: In studies of human and mouse ENS progenitors, we found activation of the Wnt signaling pathway to promote neurogenesis of the ENS in vitro. The neurogenic effect of Wnt agonists on ENS progenitors supports their use in generation of cell pools for autologous cell replacement therapies.


Assuntos
Diferenciação Celular , Proliferação de Células , Sistema Nervoso Entérico/citologia , Neurônios , RNA Mensageiro/análise , Via de Sinalização Wnt , Animais , Animais Recém-Nascidos , Proteína Axina/genética , Contagem de Células , Morte Celular , Proliferação de Células/efeitos dos fármacos , Feminino , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Indóis/farmacologia , Lactente , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Fator 1 de Ligação ao Facilitador Linfoide/genética , Masculino , Maleimidas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores Acoplados a Proteínas G/genética , Esferoides Celulares/metabolismo , Células-Tronco , Regulação para Cima , Via de Sinalização Wnt/efeitos dos fármacos , Proteína Wnt3A/farmacologia , beta Catenina/metabolismo
14.
Biochem Biophys Res Commun ; 496(2): 346-351, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29331373

RESUMO

Dermal papilla (DP) regulates the growth and cycling of hair follicles. Cultured DP cells are useful for the study of their role in relation to hair growth and regeneration. However, cultivation of human DP cells is tedious and difficult. In addition, cultured DP cells possess a relatively short replicative life span, requiring immortalized human DP cell lines. We previously established an immortalized human DP cell line, SV40T-hTERT-DPC, by introducing human telomerase reverse transcriptase (hTERT) gene into the transformed cell line, SV40T-DPC. In this study, we co-transfected the simian virus 40 large T antigen (SV40T-Ag) and hTERT into DP cells from scalp hair follicles from a male with androgenetic alopecia and established five immortalized DP cell lines and named KNU-101, KNU-102, KNU-103, KNU-201 and KNU-202. We then evaluated tumorigenicity, expression of DP markers, responses to androgen, Wnt3a and BMP4, and expression of DP signature genes. These cell lines displayed early passage morphology and maintained responses to androgen, Wnt and BMP. Furthermore, these cell lines expressed DP markers and DP signature genes. KNU cell lines established in this study are potentially useful sources for hair research.


Assuntos
Alopecia/genética , Derme/metabolismo , Efeito Fundador , Folículo Piloso/metabolismo , Células A549 , Alopecia/metabolismo , Alopecia/patologia , Animais , Biglicano/genética , Biglicano/metabolismo , Biomarcadores/metabolismo , Proteína Morfogenética Óssea 4/farmacologia , Carcinogênese/genética , Carcinogênese/metabolismo , Carcinogênese/patologia , Linhagem Celular Transformada , Derme/patologia , Di-Hidrotestosterona/farmacologia , Feminino , Expressão Gênica , Folículo Piloso/efeitos dos fármacos , Folículo Piloso/patologia , Humanos , Queratina-8/genética , Queratina-8/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Couro Cabeludo/metabolismo , Couro Cabeludo/patologia , Versicanas/genética , Versicanas/metabolismo , Proteína Wnt3A/farmacologia
15.
J Cell Physiol ; 232(5): 1104-1113, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27575935

RESUMO

In canonical pathway, Wnt3A has been known to stabilize ß-catenin through the dissociation between ß-catenin and glycogen synthase kinase-3ß (GSK-3ß) that suppresses the phosphorylation and degradation of ß-catenin. In non-canonical signaling pathway, Wnt was known to activate Rho GTPases and to induce cell migration. The cross-talk between canonical and non-canonical pathways by Wnt signaling; however, has not been fully elucidated. Here, we revealed that Wnt3A induces not only the phosphorylation of GSK-3ß and accumulation of ß-catenin but also RhoA activation in RAW264.7 and HEK293 cells. Notably, sh-RhoA and Tat-C3 abolished both the phosphorylation of GSK-3ß and accumulation of ß-catenin. Y27632, an inhibitor of Rho-associated coiled coil kinase (ROCK) and si-ROCK inhibited both GSK-3ß phosphorylation and ß-catenin accumulation. Furthermore, active domain of ROCK directly phosphorylated the purified recombinant GSK-3ß in vitro. In addition, Wnt3A-induced cell proliferation and migration, which were inhibited by Tat-C3 and Y27632. Taken together, we propose the cross-talk between canonical and non-canonical signaling pathways of Wnt3A, which induces GSK-3ß phosphorylation and ß-catenin accumulation through RhoA and ROCK activation. J. Cell. Physiol. 232: 1104-1113, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Glicogênio Sintase Quinase 3 beta/metabolismo , Proteína Wnt3A/farmacologia , beta Catenina/metabolismo , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Amidas/farmacologia , Animais , Movimento Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Quimiocinas/metabolismo , Células HEK293 , Humanos , Camundongos , Fosforilação/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Piridinas/farmacologia , Células RAW 264.7 , Proteínas Recombinantes de Fusão/farmacologia , Quinases Associadas a rho/antagonistas & inibidores , Proteína rhoA de Ligação ao GTP/antagonistas & inibidores
16.
Stem Cells ; 34(5): 1263-72, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26840479

RESUMO

Intravenous administration of bone marrow derived mesenchymal stem cells (MSCs) has been shown to reduce blood brain barrier compromise and improve neurocognition following traumatic brain injury (TBI). These effects occur in the absence of engraftment and differentiation of these cells in the injured brain. Recent studies have shown that soluble factors produced by MSCs mediate a number of the therapeutic effects. In this study, we sought to determine if intravenous administration of MSCs (IV-MSCs) could enhance hippocampal neurogenesis following TBI. Our results demonstrate that IV-MSC treatment attenuates loss of neural stem cells and promotes hippocampal neurogenesis in TBI injured mice. As Wnt signaling has been implicated in neurogenesis, we measured circulating Wnt3a levels in serum following IV-MSC administration and found a significant increase in Wnt3a. Concurrent with this increase, we detected increased activation of the Wnt/ß-catenin signaling pathway in hippocampal neurons. Furthermore, IV recombinant Wnt3a treatment provided neuroprotection, promoted neurogenesis, and improved neurocognitive function in TBI injured mice. Taken together, our results demonstrate a role for Wnt3a in the therapeutic potential of MSCs and identify Wnt3a as a potential stand-alone therapy or as part of a combination therapeutic strategy for the treatment of TBI. Stem Cells 2016;34:1263-1272.


Assuntos
Lesões Encefálicas Traumáticas/tratamento farmacológico , Cognição , Células-Tronco Mesenquimais/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Recuperação de Função Fisiológica , Proteína Wnt3A/metabolismo , Proteína Wnt3A/uso terapêutico , Administração Intravenosa , Animais , Lesões Encefálicas Traumáticas/patologia , Lesões Encefálicas Traumáticas/fisiopatologia , Sobrevivência Celular/efeitos dos fármacos , Cognição/efeitos dos fármacos , Hipocampo/patologia , Hipocampo/fisiopatologia , Humanos , Pulmão/metabolismo , Transplante de Células-Tronco Mesenquimais , Camundongos Endogâmicos C57BL , Modelos Biológicos , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Neurogênese/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Recuperação de Função Fisiológica/efeitos dos fármacos , Resultado do Tratamento , Via de Sinalização Wnt/efeitos dos fármacos , Proteína Wnt3A/sangue , Proteína Wnt3A/farmacologia
17.
EMBO Rep ; 16(4): 490-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25656539

RESUMO

Wnt signaling stimulates cell proliferation by promoting the G1/S transition of the cell cycle through ß-catenin/TCF4-mediated gene transcription. However, Wnt signaling peaks in mitosis and contributes to the stabilization of proteins other than ß-catenin, a pathway recently introduced as Wnt-dependent stabilization of proteins (Wnt/STOP). Here, we show that Wnt/STOP regulated by basal Wnt signaling during a normal cell cycle is required for proper spindle microtubule assembly and for faithful chromosome segregation during mitosis. Consequently, inhibition of basal Wnt signaling results in increased microtubule assembly rates, abnormal mitotic spindle formation and the induction of aneuploidy in human somatic cells.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Mitose , Fuso Acromático/metabolismo , Fatores de Transcrição/genética , Via de Sinalização Wnt , Proteína Wnt3A/metabolismo , beta Catenina/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Segregação de Cromossomos , Proteínas Desgrenhadas , Regulação da Expressão Gênica , Células HCT116 , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Proteínas Relacionadas a Receptor de LDL/genética , Proteínas Relacionadas a Receptor de LDL/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Estabilidade Proteica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Fuso Acromático/ultraestrutura , Fator de Transcrição 4 , Fatores de Transcrição/metabolismo , Proteína Wnt3A/antagonistas & inibidores , Proteína Wnt3A/farmacologia , beta Catenina/metabolismo
18.
J Biol Chem ; 290(1): 157-67, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-25336648

RESUMO

Focal radiotherapy for cancer patients has detrimental effects on bones within the radiation field and the primary clinical signs of bone damage include the loss of functional osteoblasts. We reported previously that daily injection of parathyroid hormone (PTH, 1-34) alleviates radiation-induced osteopenia in a preclinical radiotherapy model by improving osteoblast survival. To elucidate the molecular mechanisms, we irradiated osteoblastic UMR 106-01 cells and calvarial organ culture and demonstrated an anti-apoptosis effect of PTH1-34 on these cultures. Inhibitor assay indicated that PTH exerts its radioprotective action mainly through protein kinase A/ß-catenin pathway. γ-H2AX foci staining and comet assay revealed that PTH efficiently promotes the repair of DNA double strand breaks (DSBs) in irradiated osteoblasts via activating the ß-catenin pathway. Interestingly, Wnt3a alone also blocked cell death and accelerated DNA repair in primary osteoprogenitors, osteoblastic and osteocytic cells after radiation through the canonical signaling. Further investigations revealed that both Wnt3a and PTH increase the amount of Ku70, a core protein for initiating the assembly of DSB repair machinery, in osteoblasts after radiation. Moreover, down-regulation of Ku70 by siRNA abrogated the prosurvival effect of PTH and Wnt3a on irradiated osteoblasts. In summary, our results identify a novel role of PTH and canonical Wnt signaling in regulating DSB repair machinery and apoptosis in osteoblasts and shed light on using PTH1-34 or Wnt agonist as possible therapy for radiation-induced osteoporosis.


Assuntos
Apoptose/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , Protetores contra Radiação/farmacologia , Animais , Animais Recém-Nascidos , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Apoptose/efeitos da radiação , Diferenciação Celular , Linhagem Celular Tumoral , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA/efeitos da radiação , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Autoantígeno Ku , Osteoblastos/citologia , Osteoblastos/efeitos da radiação , Osteócitos/citologia , Osteócitos/efeitos dos fármacos , Osteócitos/efeitos da radiação , Ratos , Proteínas Recombinantes/farmacologia , Transdução de Sinais , Crânio/citologia , Crânio/efeitos dos fármacos , Crânio/efeitos da radiação , Técnicas de Cultura de Tecidos , Proteína Wnt3A/metabolismo , Proteína Wnt3A/farmacologia , Raios X , beta Catenina/genética , beta Catenina/metabolismo
19.
Biochim Biophys Acta ; 1852(5): 839-50, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25558817

RESUMO

Aberrant fibroblast growth factor (FGF) signaling disturbs chondrocyte differentiation in skeletal dysplasia, but the mechanisms underlying this process remain unclear. Recently, FGF was found to activate canonical WNT/ß-catenin pathway in chondrocytes via Erk MAP kinase-mediated phosphorylation of WNT co-receptor Lrp6. Here, we explore the cellular consequences of such a signaling interaction. WNT enhanced the FGF-mediated suppression of chondrocyte differentiation in mouse limb bud micromass and limb organ cultures, leading to inhibition of cartilage nodule formation in micromass cultures, and suppression of growth in cultured limbs. Simultaneous activation of the FGF and WNT/ß-catenin pathways resulted in loss of chondrocyte extracellular matrix, expression of genes typical for mineralized tissues and alteration of cellular shape. WNT enhanced the FGF-mediated downregulation of chondrocyte proteoglycan and collagen extracellular matrix via inhibition of matrix synthesis and induction of proteinases involved in matrix degradation. Expression of genes regulating RhoA GTPase pathway was induced by FGF in cooperation with WNT, and inhibition of the RhoA signaling rescued the FGF/WNT-mediated changes in chondrocyte cellular shape. Our results suggest that aberrant FGF signaling cooperates with WNT/ß-catenin in suppression of chondrocyte differentiation.


Assuntos
Cartilagem/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Condrócitos/efeitos dos fármacos , Fatores de Crescimento de Fibroblastos/farmacologia , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animais , Western Blotting , Cartilagem/citologia , Cartilagem/metabolismo , Diferenciação Celular/genética , Linhagem Celular Tumoral , Células Cultivadas , Condrócitos/metabolismo , Sinergismo Farmacológico , Fator 2 de Crescimento de Fibroblastos/farmacologia , Células HEK293 , Humanos , Botões de Extremidades/efeitos dos fármacos , Botões de Extremidades/embriologia , Botões de Extremidades/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Microscopia Confocal , Modelos Biológicos , Ratos , Receptores de Fatores de Crescimento de Fibroblastos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transcriptoma/efeitos dos fármacos , Transcriptoma/genética , Proteínas Wnt/genética , Proteínas Wnt/farmacologia , Proteína Wnt3A/farmacologia , beta Catenina/genética
20.
Glia ; 64(3): 350-62, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26462610

RESUMO

Myelination in corpus callosum plays important role for normal brain functions by transferring neurological information between various brain regions. However, the factors controlling expression of myelin genes in myelination are poorly understood. Here, CXXC5, a recently identified protein with CXXC-type zinc finger DNA binding motif, was characterized as a transcriptional activator of major myelin genes. We identified expression of CXXC5 expression was increased by Wnt/ß-catenin signaling. CXXC5 specifically expressed in the white matter induced expression of myelin genes through the direct binding of CXXC DNA-binding motif of CXXC5 on the MBP promoter. During the differentiation of neural stem cells (NSCs) of CXXC5(-/-) mice, the expressions of myelin genes were simultaneously reduced. The CXXC5(-/-) mice exhibited severely reduction of myelin genes expression in corpus callosum as well as abnormalities in myelin structure. The disrupted structural integrity of myelin in the CXXC5(-/-) mice resulted in reduced electrical conduction amplitudes at corpus callosum. These findings indicate that the regulation of myelin genes expression by CXXC5 is important for forming myelin structure involved with axonal electrical signal transfer in the corpus callosum.


Assuntos
Diferenciação Celular/genética , Regulação da Expressão Gênica/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Bainha de Mielina/metabolismo , Oligodendroglia/fisiologia , Potenciais de Ação/genética , Animais , Animais Recém-Nascidos , Axônios/metabolismo , Axônios/ultraestrutura , Células Cultivadas , Corpo Caloso/crescimento & desenvolvimento , Corpo Caloso/metabolismo , Proteínas de Ligação a DNA , Embrião de Mamíferos , Proteína Glial Fibrilar Ácida/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Transgênicos , Proteína Básica da Mielina/genética , Proteína Básica da Mielina/metabolismo , Proteína Proteolipídica de Mielina/genética , Proteína Proteolipídica de Mielina/metabolismo , Bainha de Mielina/genética , Condução Nervosa/genética , Células-Tronco Neurais , Oligodendroglia/ultraestrutura , Fatores de Transcrição , Via de Sinalização Wnt/genética , Proteína Wnt3A/farmacologia , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA