Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 149(21)2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36196625

RESUMEN

Migration of myoblasts derived from the occipital somites is essential for tongue morphogenesis. However, the molecular mechanisms of myoblast migration remain elusive. In this study, we report that deletion of Isl1 in the mouse mandibular epithelium leads to aglossia due to myoblast migration defects. Isl1 regulates the expression pattern of chemokine ligand 12 (Cxcl12) in the first branchial arch through the Shh/Wnt5a cascade. Cxcl12+ mesenchymal cells in Isl1ShhCre embryos were unable to migrate to the distal region, but instead clustered in a relatively small proximal domain of the mandible. CXCL12 serves as a bidirectional cue for myoblasts expressing its receptor CXCR4 in a concentration-dependent manner, attracting Cxcr4+ myoblast invasion at low concentrations but repelling at high concentrations. The accumulation of Cxcl12+ mesenchymal cells resulted in high local concentrations of CXCL12, which prevented Cxcr4+ myoblast invasion. Furthermore, transgenic activation of Ihh alleviated defects in tongue development and rescued myoblast migration, confirming the functional involvement of Hedgehog signaling in tongue development. In summary, this study provides the first line of genetic evidence that the ISL1/SHH/CXCL12 axis regulates myoblast migration during tongue development.


Asunto(s)
Quimiocina CXCL12 , Proteínas Hedgehog , Proteínas con Homeodominio LIM , Transducción de Señal , Lengua , Factores de Transcripción , Animales , Ratones , Movimiento Celular/genética , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Ligandos , Transducción de Señal/genética , Lengua/embriología , Proteínas con Homeodominio LIM/genética , Factores de Transcripción/genética , Quimiocina CXCL12/genética
2.
J Neurosci ; 43(24): 4405-4417, 2023 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-37188512

RESUMEN

Although NG2 is known to be selectively expressed in oligodendrocyte precursor cells (OPCs) for many years, its expressional regulation and functional involvement in oligodendrocyte differentiation have remained elusive. Here, we report that the surface-bound NG2 proteoglycan can physically bind to PDGF-AA and enhances PDGF receptor alpha (PDGFRα) activation of downstream signaling. During differentiation stage, NG2 protein is cleaved by A disintegrin and metalloproteinase with thrombospondin motifs type 4 (Adamts4), which is highly upregulated in differentiating OPCs but gradually downregulated in mature myelinating oligodendrocytes. Genetic ablation of Adamts4 gene impedes NG2 proteolysis, leading to elevated PDGFRα signaling but impaired oligodendrocyte differentiation and axonal myelination in both sexes of mice. Moreover, Adamts4 deficiency also lessens myelin repair in adult brain tissue following Lysophosphatidylcholine-induced demyelination. Thus, Adamts4 could be a potential therapeutic target for enhancing oligodendrocyte differentiation and axonal remyelination in demyelinating diseases.SIGNIFICANCE STATEMENT NG2 is selectively expressed in OPCs and downregulated during differentiation stage. To date, the molecular mechanism underlying the progressive removal of NG2 surface proteoglycan in differentiating OPCs has been unknown. In this study, we demonstrate that ADAMTS4 released by differentiating OPCs cleaves surface NG2 proteoglycan, attenuates PDGFRα signaling, and accelerates oligodendrocyte differentiation. In addition, our study also suggests ADAMTS4 as a potential therapeutic target for promoting myelin recovery in demyelinating diseases.


Asunto(s)
Enfermedades Desmielinizantes , Remielinización , Masculino , Femenino , Ratones , Animales , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas , Vaina de Mielina/metabolismo , Proteoglicanos/genética , Oligodendroglía/metabolismo , Diferenciación Celular/fisiología , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/metabolismo
3.
J Biol Chem ; 299(8): 105034, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37442233

RESUMEN

Lung branching morphogenesis relies on a complex coordination of multiple signaling pathways and transcription factors. Here, we found that ablation of the LIM homeodomain transcription factor Islet1 (Isl1) in lung epithelium resulted in defective branching morphogenesis and incomplete formation of five lobes. A reduction in mesenchymal cell proliferation was observed in Isl1ShhCre lungs. There was no difference in apoptosis between the wild-type (ShhCre) and Isl1ShhCre embryos. RNA-Seq and in situ hybridization analysis showed that Shh, Ptch1, Sox9, Irx1, Irx2, Tbx2, and Tbx3 were downregulated in the lungs of Isl1ShhCre embryos. ChIP assay implied the Shh gene served as a direct target of ISL1, since the transcription factor ISL1 could bind to the Shh epithelial enhancer sequence (MACS1). Also, activation of the Hedgehog pathway via ectopic gene expression rescued the defects caused by Isl1 ablation, confirming the genetic integration of Hedgehog signaling. In conclusion, our works suggest that epithelial Isl1 regulates lung branching morphogenesis through administrating the Shh signaling mediated epithelial-mesenchymal communications.


Asunto(s)
Proteínas Hedgehog , Pulmón , Factores de Transcripción , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Pulmón/crecimiento & desarrollo , Pulmón/metabolismo , Morfogénesis , Transducción de Señal/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Ratones
4.
Glia ; 72(7): 1304-1318, 2024 07.
Artículo en Inglés | MEDLINE | ID: mdl-38546197

RESUMEN

Oligodendrocyte differentiation and myelination in the central nervous system are controlled and coordinated by a complex gene regulatory network that contains several transcription factors, including Zfp488 and Nkx2.2. Despite the proven role in oligodendrocyte differentiation little is known about the exact mode of Zfp488 and Nkx2.2 action, including their target genes. Here, we used overexpression of Zfp488 and Nkx2.2 in differentiating CG4 cells to identify aspects of the oligodendroglial expression profile that depend on these transcription factors. Although both transcription factors are primarily described as repressors, the detected changes argue for an additional function as activators. Among the genes activated by both Zfp488 and Nkx2.2 was the G protein-coupled receptor Gpr37 that is important during myelination. In agreement with a positive effect on Gpr37 expression, downregulation of the G protein-coupled receptor was observed in Zfp488- and in Nkx2.2-deficient oligodendrocytes in the mouse. We also identified several potential regulatory regions of the Gpr37 gene. Although Zfp488 and Nkx2.2 both bind to one of the regulatory regions downstream of the Gpr37 gene in vivo, none of the regulatory regions was activated by either transcription factor alone. Increased activation by Zfp488 or Nkx2.2 was only observed in the presence of Sox10, a transcription factor continuously present in oligodendroglial cells. Our results argue that both Zfp488 and Nkx2.2 also act as transcriptional activators during oligodendrocyte differentiation and cooperate with Sox10 to allow the expression of Gpr37 as a modulator of the myelination process.


Asunto(s)
Diferenciación Celular , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio , Oligodendroglía , Receptores Acoplados a Proteínas G , Factores de Transcripción SOXE , Factores de Transcripción , Animales , Femenino , Masculino , Ratones , Diferenciación Celular/fisiología , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Factores de Transcripción SOXE/metabolismo , Factores de Transcripción SOXE/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética
5.
J Neurosci ; 42(30): 5860-5869, 2022 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-35760530

RESUMEN

Thyroid hormone (TH) controls the timely differentiation of oligodendrocytes (OLs), and its deficiency can delay myelin development and cause mental retardation. Previous studies showed that the active TH T3 is converted from its prohormone T4 by the selenoprotein DIO2, whose mRNA is primarily expressed in astrocytes in the CNS. In the present study, we discovered that SECISBP2L is highly expressed in differentiating OLs and is required for DIO2 translation. Conditional knock-out (CKO) of Secisbp2l in OL lineage resulted in a decreased level of DIO2 and T3, accompanied by impaired OL differentiation, hypomyelination and motor deficits in both sexes of mice. Moreover, the defective differentiation of OLs in Secisbp2l mutants can be alleviated by T3 or its analog, but not the prohormone T4. The present study has provided strong evidence for the autonomous regulation of OL differentiation by its intrinsic T3 production mediated by the novel SECISBP2L-DIO2-T3 pathway during myelin development.SIGNIFICANCE STATEMENT Secisbp2l is specifically expressed in differentiating oligodendrocytes (OLs) and is essential for selenoprotein translation in OLs. Secisbp2l regulates Dio2 translation for active thyroid hormone (TH) T3 production in the CNS. Autonomous regulation of OLs differentiation via SECISBP2L-DIO2-T3 pathway.


Asunto(s)
Neurogénesis , Oligodendroglía , Selenoproteínas , Animales , Diferenciación Celular , Femenino , Yoduro Peroxidasa , Masculino , Ratones , Vaina de Mielina/metabolismo , Oligodendroglía/citología , Oligodendroglía/metabolismo , Selenoproteínas/biosíntesis , Selenoproteínas/genética , Hormonas Tiroideas , Yodotironina Deyodinasa Tipo II
6.
Glia ; 71(11): 2499-2510, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37278537

RESUMEN

In the central nervous system, oligodendrocytes (OLs) produce myelin sheaths that provide trophic support to neuronal axons and increase the propagation speed of action potential. OLs are constantly generated from OL precursor cells (OPCs) throughout life span. The production of myelinating OLs consists of three canonical stages: OPCs, newly-formed OLs (NFOs), and mature myelinating OLs. Recently, single-cell RNA transcriptomic analyses identified a new population of oligodendroglial cells, namely differentiation committed OPCs (COPs). COPs represent a critical intermediate population between OPCs and NFOs, as revealed by specific expression of G-protein coupled receptor 17 (GPR17). The dysregulation of COPs leads to the remyelination failure in demyelinating diseases and impairs the replacement of lost myelin sheaths due to aging. Hence, understanding the development of COPs and their underlying regulatory network will be helpful in establishing new strategies for promoting myelin repair in demyelinating diseases. This review summarizes the current knowledge on the development and functions of COPs under both physiological and pathological conditions. Overall, COPs function as "checkpoints" to prevent inappropriate precocious OL differentiation and myelination through expressing distinct regulatory factors. Deepening our understanding of COPs may not only advance our knowledge of how OL lineage progresses during development, but also open the door to new treatments for demyelinating diseases.


Asunto(s)
Enfermedades Desmielinizantes , Células Precursoras de Oligodendrocitos , Humanos , Enfermedades Desmielinizantes/patología , Células Precursoras de Oligodendrocitos/metabolismo , Oligodendroglía/metabolismo , Vaina de Mielina/metabolismo , Sistema Nervioso Central/metabolismo , Diferenciación Celular/fisiología , Antioxidantes , Receptores Acoplados a Proteínas G/metabolismo
7.
Int J Mol Sci ; 24(12)2023 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-37373466

RESUMEN

Oligodendrocytes generate myelin sheaths vital for the formation, health, and function of the central nervous system. Mounting evidence suggests that receptor tyrosine kinases (RTKs) are crucial for oligodendrocyte differentiation and myelination in the CNS. It was recently reported that discoidin domain receptor 1 (Ddr1), a collagen-activated RTK, is expressed in oligodendrocyte lineage. However, its specific expression stage and functional role in oligodendrocyte development in the CNS remain to be determined. In this study, we report that Ddr1 is selectively upregulated in newly differentiated oligodendrocytes in the early postnatal CNS and regulates oligodendrocyte differentiation and myelination. Ddr1 knock-out mice of both sexes displayed compromised axonal myelination and apparent motor dysfunction. Ddr1 deficiency alerted the ERK pathway, but not the AKT pathway in the CNS. In addition, Ddr1 function is important for myelin repair after lysolecithin-induced demyelination. Taken together, the current study described, for the first time, the role of Ddr1 in myelin development and repair in the CNS, providing a novel molecule target for the treatment of demyelinating diseases.


Asunto(s)
Receptor con Dominio Discoidina 1 , Vaina de Mielina , Oligodendroglía , Animales , Femenino , Masculino , Ratones , Diferenciación Celular , Sistema Nervioso Central , Receptor con Dominio Discoidina 1/genética , Receptor con Dominio Discoidina 1/metabolismo , Ratones Noqueados , Vaina de Mielina/metabolismo , Neurogénesis , Oligodendroglía/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo
8.
Int J Mol Sci ; 24(17)2023 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-37686400

RESUMEN

In the vertebrate brain, sensory experience plays a crucial role in shaping thalamocortical connections for visual processing. However, it is still not clear how visual experience influences tissue homeostasis and neurogenesis in the developing thalamus. Here, we reported that the majority of SOX2-positive cells in the thalamus are differentiated neurons that receive visual inputs as early as stage 47 Xenopus. Visual deprivation (VD) for 2 days shifts the neurogenic balance toward proliferation at the expense of differentiation, which is accompanied by a reduction in nuclear-accumulated ß-catenin in SOX2-positive neurons. The knockdown of ß-catenin decreases the expression of SOX2 and increases the number of progenitor cells. Coimmunoprecipitation studies reveal the evolutionary conservation of strong interactions between ß-catenin and SOX2. These findings indicate that ß-catenin interacts with SOX2 to maintain homeostatic neurogenesis during thalamus development.


Asunto(s)
Evolución Biológica , beta Catenina , Animales , Homeostasis , Tálamo , Xenopus laevis
9.
J Neurosci ; 41(48): 9872-9890, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34725188

RESUMEN

Oligodendrocytes are vulnerable to genetic and environmental insults and its injury leads to demyelinating diseases. The roles of ErbB receptors in maintaining the CNS myelin integrity are largely unknown. Here, we overactivate ErbB receptors that mediate signaling of either neuregulin (NRG) or epidermal growth factor (EGF) family growth factors and found their synergistic activation caused deleterious outcomes in white matter. Sustained ErbB activation induced by the tetracycline-dependent mouse tool Plp-tTA resulted in demyelination, axonal degeneration, oligodendrocyte precursor cell (OPC) proliferation, astrogliosis, and microgliosis in white matter. Moreover, there was hypermyelination before these inflammatory pathologic events. In contrast, sustained ErbB activation induced by another tetracycline-dependent mouse tool Sox10+/rtTA caused hypomyelination in the corpus callosum and optic nerve, which appeared to be a developmental deficit and did not associate with OPC regeneration, astrogliosis, or microgliosis. By tracing the differentiation states of cells expressing tetracycline-controlled transcriptional activator (tTA)/reverse tTA (rtTA)-dependent transgene or pulse-labeled reporter proteins in vitro and in vivo, we found that Plp-tTA targeted mainly mature oligodendrocytes (MOs), whereas Sox10+/rtTA targeted OPCs and newly-formed oligodendrocytes (NFOs). The distinct phenotypes of mice with ErbB overactivation induced by Plp-tTA and Sox10+/rtTA consolidated their nonoverlapping targeting preferences in the oligodendrocyte lineage, and enabled us to demonstrate that ErbB overactivation in MOs induced necroptosis that caused inflammatory demyelination, whereas in OPCs induced apoptosis that caused noninflammatory hypomyelination. Early interference with aberrant ErbB activation ceased oligodendrocyte deaths and restored myelin development in both mice. This study suggests that aberrant ErbB activation is an upstream pathogenetic mechanism of demyelinating diseases, providing a potential therapeutic target.SIGNIFICANCE STATEMENT Primary oligodendropathy is one of the etiologic mechanisms for multiple sclerosis, and oligodendrocyte necroptosis is a pathologic hallmark in the disease. Moreover, the demyelinating disease is now a broad concept that embraces schizophrenia, in which white matter lesions are an emerging feature. ErbB overactivation has been implicated in schizophrenia by genetic analysis and postmortem studies. This study suggests the etiologic implications of ErbB overactivation in myelin pathogenesis and elucidates the pathogenetic mechanisms.


Asunto(s)
Enfermedades Desmielinizantes/patología , Receptores ErbB/metabolismo , Neurogénesis/fisiología , Células Precursoras de Oligodendrocitos/patología , Oligodendroglía/patología , Animales , Apoptosis/fisiología , Enfermedades Desmielinizantes/metabolismo , Femenino , Masculino , Ratones , Necroptosis/fisiología , Oligodendroglía/metabolismo , Sustancia Blanca/patología
10.
Glia ; 70(3): 590-601, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34889481

RESUMEN

Myelin sheathes ensure the rapid conduction of neural impulse and provide nutritional support for neurons. Myelin sheathes are formed by differentiated oligodendrocytes (OLs) in the central nervous system. During OL development, the differentiation of oligodendrocyte progenitor cells (OPCs) into mature OLs is controlled by both positive differentiation factors (drivers) and negative regulatory factors (brakes). Previous studies have suggested Id2 and Id4 as the key negative factors for OL differentiation. However, these conclusions were mainly based on in vitro studies and the reported OL phenotype in Id4 mutants appear to be mild. In this study, we systematically investigated the in vivo function of Id2 and Id4 genes in OL differentiation in their genetic mutants and in embryonic chicken spinal cord. Our results showed that disruption of Id4 has no effect on OL differentiation and maturation, whereas Id2 mutants and Id2/Id4 compound mutants display a mild and transient precocity of OL differentiation. In agreement with these loss-of-function studies, Id2, but not Id4, is weakly expressed in OPCs. Despite their minor roles in OL differentiation, forced expression of Id2 and Id4 in embryonic chicken spinal cords strongly inhibit the differentiation of OPCs. Taken together, our detailed functional and expressional studies strongly suggest that Id2 and Id4 are not the major in vivo repressors of OPC differentiation during animal development, shedding new light on the molecular regulation of early OL development.


Asunto(s)
Células Precursoras de Oligodendrocitos , Oligodendroglía , Animales , Diferenciación Celular/fisiología , Sistema Nervioso Central/metabolismo , Neurogénesis , Células Precursoras de Oligodendrocitos/metabolismo , Oligodendroglía/metabolismo , Factores de Transcripción/metabolismo
11.
Glia ; 70(11): 2093-2107, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35775976

RESUMEN

In humans, loss-of-function mutations of Kcnj10 in SeSAME/EAST syndrome, which encodes the inwardly rectifying K+ channel 4.1 (Kir 4.1), causes progressive neurological decline. Despite its rich expression in oligodendrocyte (OL) lineage cells and an emerging link with demyelinating disease, the function of Kir 4.1 in OLs is unclear. Here we show a novel role of Kir 4.1 in OL development. Kir 4.1 expression is markedly greater in OLs than in OL precursor cells (OPCs), and the down-regulation of Kir 4.1 impairs OL maturation by affecting OPC differentiation. Interestingly, Kir 4.1 regulates the intracellular pH of OPCs and OLs via the Na+ /H+ exchanger, which underlies impeded OPC differentiation by Kir 4.1 inhibition. Furthermore, Kir 4.1 regulates GSK3ß and SOX10, two molecules critical to OPC development. Collectively, our work opens a new avenue to understanding the functions of Kir 4.1 and intracellular pH in OLs.


Asunto(s)
Células Precursoras de Oligodendrocitos , Canales de Potasio de Rectificación Interna , Humanos , Concentración de Iones de Hidrógeno , Neurogénesis/fisiología , Células Precursoras de Oligodendrocitos/metabolismo , Oligodendroglía/metabolismo , Potasio/metabolismo , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo
12.
Mol Med ; 28(1): 50, 2022 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-35508991

RESUMEN

BACKGROUND: The conversion of astrocytes activated by nerve injuries to oligodendrocytes is not only beneficial to axonal remyelination, but also helpful for reversal of glial scar. Recent studies have shown that pathological niche promoted the Sox10-mediated astrocytic transdifferentiation to oligodendrocytes. The extracellular factors underlying the cell fate switching are not known. METHODS: Astrocytes were obtained from mouse spinal cord dissociation culture and purified by differential adherent properties. The lineage conversion of astrocytes into oligodendrocyte lineage cells was carried out by Sox10-expressing virus infection both in vitro and in vivo, meanwhile, epidermal growth factor (EGF) and epidermal growth factor receptor (EGFR) inhibitor Gefitinib were adopted to investigate the function of EGF signaling in this fate transition process. Pharmacological inhibition analyses were performed to examine the pathway connecting the EGF with the expression of oligodendrogenic genes and cell fate transdifferentiation. RESULTS: EGF treatment facilitated the Sox10-induced transformation of astrocytes to O4+ induced oligodendrocyte precursor cells (iOPCs) in vitro. The transdifferentiation of astrocytes to iOPCs went through two distinct but interconnected processes: (1) dedifferentiation of astrocytes to astrocyte precursor cells (APCs); (2) transformation of APCs to iOPCs, EGF signaling was involved in both processes. And EGF triggered astrocytes to express oligodendrogenic genes Olig1 and Olig2 by activating extracellular signal-regulated kinase 1 and 2 (Erk1/2) pathway. In addition, we discovered that EGF can enhance astrocyte transdifferentiation in injured spinal cord tissues. CONCLUSIONS: These findings provide strong evidence that EGF facilitates the transdifferentiation of astrocytes to oligodendrocytes, and suggest that targeting the EGF-EGFR-Erk1/2 signaling axis may represent a novel therapeutic strategy for myelin repair in injured central nervous system (CNS) tissues.


Asunto(s)
Astrocitos , Factor de Crecimiento Epidérmico , Animales , Astrocitos/metabolismo , Diferenciación Celular , Células Cultivadas , Factor de Crecimiento Epidérmico/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/metabolismo , Ratones , Oligodendroglía/metabolismo
13.
Biochem Biophys Res Commun ; 599: 156-163, 2022 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-35202849

RESUMEN

Primitive Endoderm (PrE) is an extraembryonic structure derived from inner cell mass (ICM) in the blastocysts. Its interaction with the epiblast is critical to sustain embryonic growth and embryonic pattern. In this study, we reported a simple and efficient method to induce the differentiation of mouse Embryonic Stem Cells (mESCs) into PrE cells. In the process of ESC monolayer adherent culture, 1 µM atRA and 10 µM CHIR inducers were used to activate RA and Wnt signaling pathways respectively. After 9 days of differentiation, the proportion of PrE cells was up to 85%. Further studies indicated that Wnt signaling pathway acted as a switch that RA induces mESCs differentiation between SMC and PrE cell. In the presence of only RA signaling, mESCs adopted the fate of smooth muscle cells (SMCs); Simultaneous activation of the Wnt signaling pathway changed the differentiation fate of mESCs into PrE cells. This efficient induction method can provide new cellular resources and models for relevant studies of PrE.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Endodermo/citología , Células Madre Embrionarias de Ratones/citología , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Células Madre Embrionarias de Ratones/fisiología , Piridinas/farmacología , Pirimidinas/farmacología , Tretinoina/farmacología , Vía de Señalización Wnt/efectos de los fármacos
14.
J Biol Chem ; 295(7): 1879-1888, 2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-31932307

RESUMEN

The homeodomain protein NK2 homeobox 2 (NKX2-2) is a transcription factor that plays a critical role in the control of cell fate specification and differentiation in many tissues. In the developing central nervous system, this developmentally important transcription factor functions as a transcriptional repressor that governs oligodendrocyte (OL) differentiation and myelin gene expression, but the roles of various NKX2-2 structural domains in this process are unclear. In this study, using in situ hybridization, immunofluorescence, and coimmunoprecipitation, we determined the structural domains that mediate the repressive functions of murine NKX2-2 and identified the transcriptional corepressors that interact with it in OL cells. Through in ovo electroporation in embryonic chicken spinal cords, we demonstrate that the N-terminal Tinman domain and C-terminal domain synergistically promote OL differentiation by recruiting distinct transcriptional corepressors, including enhancer of split Groucho 3 (GRG3), histone deacetylase 1 (HDAC1), and DNA methyltransferase 3 α (DNMT3A). We also observed that the NK2-specific domain suppresses the function of the C-terminal domain in OL differentiation. These findings delineate the distinct NKX2-2 domains and their roles in OL differentiation and suggest that NKX2-2 regulates differentiation by repressing gene expression via multiple cofactors and molecular mechanisms.


Asunto(s)
Diferenciación Celular/genética , Proteínas de Homeodominio/genética , Neurogénesis/genética , Proteínas de Pez Cebra/genética , Animales , Embrión de Pollo , Proteínas Co-Represoras/genética , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Regulación del Desarrollo de la Expresión Génica/genética , Histona Desacetilasa 1/genética , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio/química , Ratones , Oligodendroglía/metabolismo , Regiones Promotoras Genéticas/genética , Dominios Proteicos/genética , Factores de Transcripción/genética
15.
Reprod Fertil Dev ; 33(16): 810-816, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34758896

RESUMEN

Atypical protein serine kinase RIOK3 is involved in cellular invasion and survival. The spatiotemporal expression pattern and regulatory mechanisms controlling expression of Riok3 were investigated in the rat ovary during the periovulatory period. Immature female rats (22-23 days old) were treated with pregnant mare's serum gonadotropin (PMSG) to stimulate follicular development, followed 48h later by injection with human chorionic gonadotrophin (hCG). Ovaries, granulosa cells, or theca-interstitial cells were collected at various times after hCG administration. Both real-time polymerase chain reaction (PCR) and in situ hybridisation analysis revealed that Riok3 was highly induced in both granulosa cells and theca-interstitial cells by hCG. Riok3 expression was induced in theca-interstitial cells at 4h after hCG. However, the expression of Riok3 mRNA was stimulated in granulosa cells at 8h. Both protein kinase C inhibitor (GF109203) and the protein kinase A inhibitor (H89) could block the stimulation of Riok3 mRNA by hCG. Furthermore, Riok3 induction is dependent on new protein synthesis. Inhibition of prostaglandin synthesis or progesterone action did not alter Riok3 mRNA expression, whereas inhibition of the epidermal growth factor (EGF) pathway downregulated Riok3 expression. In conclusion, our findings suggest that the induction of the RIOK3 may be important for ovulation and luteinisation.


Asunto(s)
Luteinización/metabolismo , Ovario/metabolismo , Ovulación/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Femenino , Gonadotropinas Equinas/farmacología , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/metabolismo , Luteinización/efectos de los fármacos , Luteinización/genética , Ovario/efectos de los fármacos , Ovulación/efectos de los fármacos , Ovulación/genética , Proteínas Serina-Treonina Quinasas/genética , Ratas , Transducción de Señal/efectos de los fármacos
16.
Glia ; 68(2): 422-434, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31605511

RESUMEN

Elucidation of signaling pathways that control oligodendrocyte (OL) development is a prerequisite for developing novel strategies for myelin repair in neurological diseases. Despite the extensive work outlining the importance of Hedgehog (Hh) signaling in the commitment and generation of OL progenitor cells (OPCs), there are conflicting reports on the role of Hh signaling in regulating OL differentiation and maturation. In the present study, we systematically investigated OPC specification and differentiation in genetically modified mouse models of Smoothened (Smo), an essential component of the Hh signaling pathway in vertebrates. Through conditional gain-of-function strategy, we demonstrated that hyperactivation of Smo in neural progenitors induced transient ectopic OPC generation and precocious OL differentiation accompanied by the co-induction of Olig2 and Nkx2.2. After the commitment of OL lineage, Smo activity is not required for OL differentiation, and sustained expression of Smo in OPCs stimulated cell proliferation but inhibited terminal differentiation. These findings have uncovered the stage-specific regulation of OL development by Smo-mediated Hh signaling, providing novel insights into the molecular regulation of OL differentiation and myelin repair.


Asunto(s)
Proteínas Hedgehog/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Oligodendroglía/metabolismo , Médula Espinal/metabolismo , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Proteína Homeobox Nkx-2.2 , Ratones Transgénicos , Vaina de Mielina/metabolismo , Transducción de Señal/fisiología , Células Madre/metabolismo
17.
Cell Mol Neurobiol ; 40(4): 653-662, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-31782037

RESUMEN

In the developing central nervous system, the terminal differentiation of oligodendrocytes (OLs) is regulated by both extrinsic and intrinsic factors. Recent studies have suggested that the Notch-Hes signaling pathway influences the maturation of oligodendrocytes in culture and during development. However, the specific Notch receptors and their downstream effectors Hes genes that are involved in oligodendrocyte maturation have not been investigated systematically. In this study, we showed that Notch1 and Notch3 are expressed in oligodendrocyte precursor cells (OPCs) during gliogenesis, and Hes5 is the major Notch downstream transcription factor that is transiently expressed in OPCs. Overexpression of Notch intracellular domain (NICD) and Hes5 proteins in embryonic chicken spinal cord suppressed both the endogenous and Sox10-induced Mbp gene expression. Unexpectedly, overexpression of NICD/Hes5 did not inhibit Sox10 induction of Olig2 expression and Myrf induced Mbp expression, suggesting the differential inhibitory effects of NICD/Hes5 signaling on Sox10 activation of myelin-related genes and early progenitor genes.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Receptores Notch/metabolismo , Factores de Transcripción SOXE/antagonistas & inhibidores , Transducción de Señal , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Pollos , Regulación del Desarrollo de la Expresión Génica , Ratones Noqueados , Proteína Básica de Mielina/metabolismo , Oligodendroglía/metabolismo , Receptores Notch/genética , Factores de Transcripción SOXE/metabolismo , Médula Espinal/embriología , Médula Espinal/metabolismo , Células Madre/metabolismo
18.
J Neurosci ; 38(44): 9505-9513, 2018 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-30242047

RESUMEN

PDGFRα, specifically expressed by immature oligodendrocyte progenitor cells (OPCs) in the CNS, plays a critical role in OPC proliferation and migration. However, it has been uncertain whether all cells of oligodendrocyte lineage are derived from the PDGFRα-expressing OPCs. In the present study, we uncovered a PDGFRα-independent oligodendrocyte lineage in the developing cortex. This OPC subpopulation originates from the local ventricular/subventricular zone after birth and contributes to the earliest mature oligodendrocytes in the cortex. PDGFRα signaling does not regulate the generation and differentiation of cortical OPCs. Fate-mapping studies in the PDGFRαCreER; Sox10-GFP/tdTom double-transgenic mice of either sex have further corroborated the PDGFRα-independent oligodendrocyte lineage. This study provides additional missing genetic evidence for PDGFRα-independent oligodendrocyte lineage in the developing hindbrain.SIGNIFICANCE STATEMENT This is the first report of a subpopulation of oligodendrocyte lineage in the developing mouse cortex independent of PDGFRα signaling. These oligodendrocyte progenitor cells are generated from the local ventral ventricular zone/subventricular zone after birth, and contribute to the earliest mature oligodendrocytes in the cortex.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Corteza Cerebral/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Rombencéfalo/metabolismo , Animales , Corteza Cerebral/embriología , Corteza Cerebral/crecimiento & desarrollo , Femenino , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Embarazo , Rombencéfalo/embriología , Rombencéfalo/crecimiento & desarrollo
19.
J Neurosci ; 38(46): 9829-9839, 2018 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-30249802

RESUMEN

Myelin sheath formed by oligodendrocytes (OLs) is essential for the rapid propagation of action potentials in the vertebrate CNS. Myelin regulatory factor (MYRF) is one of the critical factors that control OL differentiation and myelin maintenance. Previous studies showed that MYRF is a membrane-bound transcription factor associated with the endoplasmic reticulum (ER). After self-cleavage, the N-fragment of MYRF is released from the ER and translocated into the nucleus where it functions as a transcription factor to activate myelin gene expression. At present, it remains unknown whether MYRF self-cleavage and functional activation can be regulated during OL differentiation. Here, we report that TMEM98, an ER-associated transmembrane protein, is capable of binding to the C-terminal of MYRF and inhibiting its self-cleavage and N-fragment nuclear translocation. In the developing CNS, TMEM98 is selectively expressed in early maturing OLs in mouse pups of either sex. Forced expression of TMEM98 in embryonic chicken spinal cord of either sex suppresses endogenous OL differentiation and MYRF-induced ectopic expression of myelin genes. These results suggest that TMEM98, through inhibiting the self-cleavage of MYRF, functions as a negative feedback regulator of MYRF in oligodendrocyte differentiation and myelination.SIGNIFICANCE STATEMENT MYRF protein is initially synthesized as an ER-associated membrane protein that undergoes autoproteolytic cleavage to release the N-fragment, which is then transported into the nucleus and activates the transcription of myelin genes. To date, the molecular mechanisms that regulate the self-cleavage and function of MYRF in regulating oligodendrocyte differentiation have remained unknown. In this study, we present the molecular and functional evidence that TMEM98 membrane protein physically interacts with MYRF in the ER and subsequently blocks its self-cleavage, N-terminal nuclear translocation, and functional activation of myelin gene expression. To our knowledge, this is the first report on the regulation of MYRF self-proteolytic activity and function by an interacting protein, providing new insights into the molecular regulation of OL differentiation and myelinogenesis.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas de la Membrana/metabolismo , Oligodendroglía/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular , Pollos , Retículo Endoplásmico/metabolismo , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Transgénicos , Vaina de Mielina/metabolismo , Unión Proteica/fisiología
20.
J Neurosci ; 38(6): 1575-1587, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29326173

RESUMEN

Bergmann glia facilitate granule neuron migration during development and maintain the cerebellar organization and functional integrity. At present, molecular control of Bergmann glia specification from cerebellar radial glia is not fully understood. In this report, we show that ZEB2 (aka, SIP1 or ZFHX1B), a Mowat-Wilson syndrome-associated transcriptional regulator, is highly expressed in Bergmann glia, but hardly detectable in astrocytes in the cerebellum. The mice lacking Zeb2 in cerebellar radial glia exhibit severe deficits in Bergmann glia specification, and develop cerebellar cortical lamination dysgenesis and locomotion defects. In developing Zeb2-mutant cerebella, inward migration of granule neuron progenitors is compromised, the proliferation of glial precursors is reduced, and radial glia fail to differentiate into Bergmann glia in the Purkinje cell layer. In contrast, Zeb2 ablation in granule neuron precursors or oligodendrocyte progenitors does not affect Bergmann glia formation, despite myelination deficits caused by Zeb2 mutation in the oligodendrocyte lineage. Transcriptome profiling identified that ZEB2 regulates a set of Bergmann glia-related genes and FGF, NOTCH, and TGFß/BMP signaling pathway components. Our data reveal that ZEB2 acts as an integral regulator of Bergmann glia formation ensuring maintenance of cerebellar integrity, suggesting that ZEB2 dysfunction in Bergmann gliogenesis might contribute to motor deficits in Mowat-Wilson syndrome.SIGNIFICANCE STATEMENT Bergmann glia are essential for proper cerebellar organization and functional circuitry, however, the molecular mechanisms that control the specification of Bergmann glia remain elusive. Here, we show that transcriptional factor ZEB2 is highly expressed in mature Bergmann glia, but not in cerebellar astrocytes. The mice lacking Zeb2 in cerebellar radial glia, but not oligodendrocyte progenitors or granular neuron progenitors, exhibit severe defects in Bergmann glia formation. The orderly radial scaffolding formed by Bergmann glial fibers critical for cerebellar lamination was not established in Zeb2 mutants, displaying motor behavior deficits. This finding demonstrates a previously unrecognized critical role for ZEB2 in Bergmann glia specification, and points to an important contribution of ZEB2 dysfunction to cerebellar motor disorders in Mowat-Wilson syndrome.


Asunto(s)
Cerebelo/citología , Cerebelo/crecimiento & desarrollo , Neurogénesis/genética , Neurogénesis/fisiología , Neuroglía/fisiología , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/fisiología , Animales , Astrocitos/fisiología , Recuento de Células , Cerebelo/fisiología , Facies , Perfilación de la Expresión Génica , Enfermedad de Hirschsprung/genética , Discapacidad Intelectual/genética , Locomoción/fisiología , Ratones , Ratones Transgénicos , Microcefalia/genética , Células-Madre Neurales/fisiología , Oligodendroglía/fisiología , Células de Purkinje/fisiología , Transcriptoma/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA