Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Neurosci ; 43(4): 540-558, 2023 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-36460463

RESUMEN

In the CNS, oligodendrocyte progenitor cells (OPCs) differentiate into mature oligodendrocytes to generate myelin, an essential component for normal nervous system function. OPC differentiation is driven by signaling pathways, such as mTOR, which functions in two distinct complexes: mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2), containing Raptor or Rictor, respectively. In the current studies, mTORC2 signaling was selectively deleted from OPCs in PDGFRα-Cre X Rictorfl/fl mice. This study examined developmental myelination in male and female mice, comparing the impact of mTORC2 deletion in the corpus callosum and spinal cord. In both regions, Rictor loss in OPCs resulted in early reduction in myelin RNAs and proteins. However, these deficits rapidly recovered in spinal cord, where normal myelin was noted at P21 and P45. By contrast, the losses in corpus callosum resulted in severe hypomyelination and increased unmyelinated axons. The hypomyelination may result from decreased oligodendrocytes in the corpus callosum, which persisted in animals as old as postnatal day 350. The current studies focus on uniquely altered signaling pathways following mTORC2 loss in developing oligodendrocytes. A major mTORC2 substrate is phospho-Akt-S473, which was significantly reduced throughout development in both corpus callosum and spinal cord at all ages measured, yet this had little impact in spinal cord. Loss of mTORC2 signaling resulted in decreased expression of actin regulators, such as gelsolin in corpus callosum, but only minimal loss in spinal cord. The current study establishes a regionally specific role for mTORC2 signaling in OPCs, particularly in the corpus callosum.SIGNIFICANCE STATEMENT mTORC1 and mTORC2 signaling has differential impact on myelination in the CNS. Numerous studies identify a role for mTORC1, but deletion of Rictor (mTORC2 signaling) in late-stage oligodendrocytes had little impact on myelination in the CNS. However, the current studies establish that deletion of mTORC2 signaling from oligodendrocyte progenitor cells results in reduced myelination of brain axons. These studies also establish a regional impact of mTORC2, with little change in spinal cord in these conditional Rictor deletion mice. Importantly, in both brain and spinal cord, mTORC2 downstream signaling targets were impacted by Rictor deletion. Yet, these signaling changes had little impact on myelination in spinal cord, while they resulted in long-term alterations in myelination in brain.


Asunto(s)
Células Precursoras de Oligodendrocitos , Animales , Femenino , Masculino , Ratones , Diferenciación Celular/fisiología , Sistema Nervioso Central/fisiología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Ratones Noqueados , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
2.
J Neurosci ; 41(9): 1864-1877, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33478987

RESUMEN

The actin cytoskeleton is crucial for oligodendrocyte differentiation and myelination. Here we show that p21-activated kinase 1 (PAK1), a well-known actin regulator, promotes oligodendrocyte morphologic change and myelin production in the CNS. A combination of in vitro and in vivo models demonstrated that PAK1 is expressed throughout the oligodendrocyte lineage with highest expression in differentiated oligodendrocytes. Inhibiting PAK1 early in oligodendrocyte development decreased oligodendrocyte morphologic complexity and altered F-actin spreading at the tips of oligodendrocyte progenitor cell processes. Constitutively activating AKT in oligodendrocytes in male and female mice, which leads to excessive myelin wrapping, increased PAK1 expression, suggesting an impact of PAK1 during active myelin wrapping. Furthermore, constitutively activating PAK1 in oligodendrocytes in zebrafish led to an increase in myelin internode length while inhibiting PAK1 during active myelination decreased internode length. As myelin parameters influence conduction velocity, these data suggest that PAK1 may influence communication within the CNS. These data support a model in which PAK1 is a positive regulator of CNS myelination.SIGNIFICANCE STATEMENT Myelin is a critical component of the CNS that provides metabolic support to neurons and also facilitates communication between cells in the CNS. Recent data demonstrate that actin dynamics drives myelin wrapping, but how actin is regulated during myelin wrapping is unknown. The authors investigate the role of the cytoskeletal modulator PAK1 during differentiation and myelination by oligodendrocytes, the myelinating cells of the CNS. They demonstrate that PAK1 promotes oligodendrocyte differentiation and myelination by modulating the cytoskeleton and thereby internode length, thus playing a critical role in the function of the CNS.


Asunto(s)
Vaina de Mielina/metabolismo , Neurogénesis/fisiología , Oligodendroglía/citología , Oligodendroglía/metabolismo , Quinasas p21 Activadas/metabolismo , Animales , Diferenciación Celular/fisiología , Femenino , Masculino , Ratones , Ratas , Ratas Sprague-Dawley , Pez Cebra
3.
J Neurosci ; 41(40): 8321-8337, 2021 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-34417330

RESUMEN

In demyelinating diseases, such as multiple sclerosis, primary loss of myelin and subsequent neuronal degeneration throughout the CNS impair patient functionality. While the importance of mechanistic target of rapamycin (mTOR) signaling during developmental myelination is known, no studies have yet directly examined the function of mTOR signaling specifically in the oligodendrocyte (OL) lineage during remyelination. Here, we conditionally deleted Mtor from adult oligodendrocyte precursor cells (OPCs) using Ng2-CreERT in male adult mice to test its function in new OLs responsible for remyelination. During early remyelination after cuprizone-induced demyelination, mice lacking mTOR in adult OPCs had unchanged OL numbers but thinner myelin. Myelin thickness recovered by late-stage repair, suggesting a delay in myelin production when Mtor is deleted from adult OPCs. Surprisingly, loss of mTOR in OPCs had no effect on efficiency of remyelination after lysophosphatidylcholine lesions in either the spinal cord or corpus callosum, suggesting that mTOR signaling functions specifically in a pathway dysregulated by cuprizone to promote remyelination efficiency. We further determined that cuprizone and inhibition of mTOR cooperatively compromise metabolic function in primary rat OLs undergoing differentiation. Together, our results support the conclusion that mTOR signaling in OPCs is required to overcome the metabolic dysfunction in the cuprizone-demyelinated adult brain.SIGNIFICANCE STATEMENT Impaired remyelination by oligodendrocytes contributes to the progressive pathology in multiple sclerosis, so it is critical to identify mechanisms of improving remyelination. The goal of this study was to examine mechanistic target of rapamycin (mTOR) signaling in remyelination. Here, we provide evidence that mTOR signaling promotes efficient remyelination of the brain after cuprizone-mediated demyelination but has no effect on remyelination after lysophosphatidylcholine demyelination in the spinal cord or brain. We also present novel data revealing that mTOR inhibition and cuprizone treatment additively affect the metabolic profile of differentiating oligodendrocytes, supporting a mechanism for the observed remyelination delay. These data suggest that altered metabolic function may underlie failure of remyelination in multiple sclerosis lesions and that mTOR signaling may be of therapeutic potential for promoting remyelination.


Asunto(s)
Encéfalo/metabolismo , Cuprizona/toxicidad , Células Precursoras de Oligodendrocitos/metabolismo , Remielinización/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Encéfalo/efectos de los fármacos , Quelantes/toxicidad , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas Sprague-Dawley , Remielinización/efectos de los fármacos , Serina-Treonina Quinasas TOR/genética
4.
J Neurosci ; 40(15): 2993-3007, 2020 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-32139584

RESUMEN

During differentiation, oligodendrocyte precursor cells (OPCs) extend a network of processes that make contact with axons and initiate myelination. Recent studies revealed that actin polymerization is required for initiation of myelination whereas actin depolymerization promotes myelin wrapping. Here, we used primary OPCs in culture isolated from neonatal rat cortices of both sexes and young male and female mice with oligodendrocyte-specific deletion of mechanistic target of rapamycin (mTOR) to demonstrate that mTOR regulates expression of specific cytoskeletal targets and actin reorganization in oligodendrocytes during developmental myelination. Loss or inhibition of mTOR reduced expression of profilin2 and ARPC3, actin polymerizing factors, and elevated levels of active cofilin, which mediates actin depolymerization. The deficits in actin polymerization were revealed in reduced phalloidin and deficits in oligodendrocyte cellular branching complexity at the peak of morphologic differentiation and a delay in initiation of myelination. We further show a critical role for mTOR in expression and localization of myelin basic protein (Mbp) mRNA and MBP protein to the cellular processes where it is necessary at the myelin membrane for axon wrapping. Mbp mRNA transport deficits were confirmed by single molecule RNA FISH. Moreover, expression of the kinesin family member 1B, an Mbp mRNA transport protein, was reduced in CC1+ cells in the mTOR cKO and in mTOR inhibited oligodendrocytes undergoing differentiation in vitro These data support the conclusion that mTOR regulates both initiation of myelination and axon wrapping by targeting cytoskeletal reorganization and MBP localization to oligodendrocyte processes.SIGNIFICANCE STATEMENT Myelination is essential for normal CNS development and adult axon preservation and function. The mechanistic target of rapamycin (mTOR) signaling pathway has been implicated in promoting CNS myelination; however, there is a gap in our understanding of the mechanisms by which mTOR promotes developmental myelination through regulating specific downstream targets. Here, we present evidence that mTOR promotes the initiation of myelination through regulating specific cytoskeletal targets and cellular process expansion by oligodendrocyte precursor cells as well as expression and cellular localization of myelin basic protein.


Asunto(s)
Citoesqueleto/genética , Vaina de Mielina/genética , Oligodendroglía , Serina-Treonina Quinasas TOR/fisiología , Complejo 2-3 Proteico Relacionado con la Actina/genética , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Axones , Diferenciación Celular/genética , Cinesinas/genética , Cinesinas/metabolismo , Ratones , Ratones Noqueados , Proteína Básica de Mielina/genética , Proteína Proteolipídica de la Mielina/genética , Proteína Proteolipídica de la Mielina/metabolismo , Oligodendroglía/ultraestructura , Ratas , Ratas Sprague-Dawley , Células Madre , Serina-Treonina Quinasas TOR/genética , Pez Cebra
5.
Carcinogenesis ; 41(9): 1294-1305, 2020 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-31958132

RESUMEN

The insulin receptor gene (INSR) undergoes alternative splicing to give rise to two functionally related, but also distinct, isoforms IR-A and IR-B, which dictate proliferative and metabolic regulations, respectively. Previous studies identified the RNA-binding protein CUGBP1 as a key regulator of INSR splicing. In this study, we show that the differential splicing of INSR occurs more frequently in breast cancer than in non-tumor breast tissues. In breast cancer cell lines, the IR-A:IR-B ratio varies in different molecular subtypes, knockdown or overexpression of CUGBP1 gene in breast cancer cells altered IR-A:IR-B ratio through modulation of IR-A expression, thereby reversed or enhanced the insulin-induced oncogenic behavior of breast cancer cells, respectively. Our data revealed the predominant mitogenic role of IR-A isoform in breast cancer and depicted a novel interplay between INSR and CUGBP1, implicating CUGBP1 and IR-A isoform as the potential therapeutic targets and biomarkers for breast cancer.


Asunto(s)
Antígenos CD/genética , Neoplasias de la Mama/patología , Proteínas CELF1/metabolismo , Empalme del ARN , Receptor ErbB-2/metabolismo , Receptor de Insulina/genética , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/clasificación , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proteínas CELF1/genética , Femenino , Estudios de Seguimiento , Regulación Neoplásica de la Expresión Génica , Humanos , Pronóstico , Isoformas de Proteínas
6.
Glia ; 68(6): 1274-1290, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31904150

RESUMEN

Oligodendrocyte precursor cells (OPCs) differentiate and mature into oligodendrocytes, which produce myelin in the central nervous system. Prior studies have shown that the mechanistic target of rapamycin (mTOR) is necessary for proper myelination of the mouse spinal cord and that bone morphogenetic protein (BMP) signaling inhibits oligodendrocyte differentiation, in part by promoting expression of inhibitor of DNA binding 2 (Id2). Here we provide evidence that mTOR functions specifically in the transition from early stage OPC to immature oligodendrocyte by downregulating BMP signaling during postnatal spinal cord development. When mTOR is deleted from the oligodendrocyte lineage, expression of the FK506 binding protein 1A (FKBP12), a suppressor of BMP receptor activity, is reduced, downstream Smad activity is increased and Id2 expression is elevated. Additionally, mTOR inhibition with rapamycin in differentiating OPCs alters the transcriptional complex present at the Id2 promoter. Deletion of mTOR in oligodendroglia in vivo resulted in fewer late stage OPCs and fewer newly formed oligodendrocytes in the spinal cord with no effect on OPC proliferation or cell cycle exit. Finally, we demonstrate that inhibiting BMP signaling rescues the rapamycin-induced deficit in myelin protein expression. We conclude that mTOR promotes early oligodendrocyte differentiation by suppressing BMP signaling in OPCs.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular/fisiología , Oligodendroglía/metabolismo , Sirolimus/metabolismo , Médula Espinal/metabolismo , Animales , Ciclo Celular/fisiología , Sistema Nervioso Central/metabolismo , Ratones , Proteínas de la Mielina/metabolismo , Neurogénesis , Transducción de Señal/fisiología , Células Madre/citología , Células Madre/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
7.
J Neurosci ; 37(31): 7534-7546, 2017 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-28694334

RESUMEN

Although the mammalian target of rapamycin (mTOR) is an essential regulator of developmental oligodendrocyte differentiation and myelination, oligodendrocyte-specific deletion of tuberous sclerosis complex (TSC), a major upstream inhibitor of mTOR, surprisingly also leads to hypomyelination during CNS development. However, the function of TSC has not been studied in the context of remyelination. Here, we used the inducible Cre-lox system to study the function of TSC in the remyelination of a focal, lysolecithin-demyelinated lesion in adult male mice. Using two different mouse models in which Tsc1 is deleted by Cre expression in oligodendrocyte progenitor cells (OPCs) or in premyelinating oligodendrocytes, we reveal that deletion of Tsc1 affects oligodendroglia differently depending on the stage of the oligodendrocyte lineage. Tsc1 deletion from NG2+ OPCs accelerated remyelination. Conversely, Tsc1 deletion from proteolipid protein (PLP)-positive oligodendrocytes slowed remyelination. Contrary to developmental myelination, there were no changes in OPC or oligodendrocyte numbers in either model. Our findings reveal a complex role for TSC in oligodendrocytes during remyelination in which the timing of Tsc1 deletion is a critical determinant of its effect on remyelination. Moreover, our findings suggest that TSC has different functions in developmental myelination and remyelination.SIGNIFICANCE STATEMENT Myelin loss in demyelinating disorders such as multiple sclerosis results in disability due to loss of axon conductance and axon damage. Encouragingly, the nervous system is capable of spontaneous remyelination, but this regenerative process often fails. Many chronically demyelinated lesions have oligodendrocyte progenitor cells (OPCs) within their borders. It is thus of great interest to elucidate mechanisms by which we might enhance endogenous remyelination. Here, we provide evidence that deletion of Tsc1 from OPCs, but not differentiating oligodendrocytes, is beneficial to remyelination. This finding contrasts with the loss of oligodendroglia and hypomyelination seen with Tsc1 or Tsc2 deletion in the oligodendrocyte lineage during CNS development and points to important differences in the regulation of developmental myelination and remyelination.


Asunto(s)
Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Fibras Nerviosas Mielínicas/patología , Oligodendroglía/metabolismo , Oligodendroglía/patología , Células Madre/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Axones , Diferenciación Celular/fisiología , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Fibras Nerviosas Mielínicas/metabolismo , Regeneración Nerviosa/fisiología , Células Madre/patología , Proteína 1 del Complejo de la Esclerosis Tuberosa
8.
Breast Cancer Res ; 20(1): 138, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30458886

RESUMEN

BACKGROUND: Early analyses of human breast cancer identified high expression of the insulin-like growth factor type 1 receptor (IGF-1R) correlated with hormone receptor positive breast cancer and associated with a favorable prognosis, whereas low expression of IGF-1R correlated with triple negative breast cancer (TNBC). We previously demonstrated that the IGF-1R acts as a tumor and metastasis suppressor in the Wnt1 mouse model of TNBC. The mechanisms for how reduced IGF-1R contributes to TNBC phenotypes is unknown. METHODS: We analyzed the METABRIC dataset to further stratify IGF-1R expression with patient survival and specific parameters of TNBC. To investigate molecular events associated with the loss of IGF-1R function in breast tumor cells, we inhibited IGF-1R in human cell lines using an IGF-1R blocking antibody and analyzed MMTV-Wnt1-mediated mouse tumors with reduced IGF-1R function through expression of a dominant-negative transgene. RESULTS: Our analysis of the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) dataset revealed association between low IGF-1R and reduced overall patient survival. IGF-1R expression was inversely correlated with patient survival even within hormone receptor-positive breast cancers, indicating reduced overall patient survival with low IGF-1R was not due simply to low IGF-1R expression within TNBCs. Inhibiting IGF-1R in either mouse or human tumor epithelial cells increased reactive oxygen species (ROS) production and activation of the endoplasmic reticulum stress response. IGF-1R inhibition in tumor epithelial cells elevated interleukin (IL)-6 and C-C motif chemokine ligand 2 (CCL2) expression, which was reversed by ROS scavenging. Moreover, the Wnt1/dnIGF-1R primary tumors displayed a tumor-promoting immune phenotype. The increased CCL2 promoted an influx of CD11b+ monocytes into the primary tumor that also had increased matrix metalloproteinase (MMP)-2, MMP-3, and MMP-9 expression. Increased MMP activity in the tumor stroma was associated with enhanced matrix remodeling and collagen deposition. Further analysis of the METABRIC dataset revealed an increase in IL-6, CCL2, and MMP-9 expression in patients with low IGF-1R, consistent with our mouse tumor model and data in human breast cancer cell lines. CONCLUSIONS: Our data support the hypothesis that reduction of IGF-1R function increases cellular stress and cytokine production to promote an aggressive tumor microenvironment through infiltration of immune cells and matrix remodeling.


Asunto(s)
Citocinas/metabolismo , Estrés del Retículo Endoplásmico , Neoplasias Mamarias Experimentales/patología , Receptores de Somatomedina/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Animales , Línea Celular Tumoral , Conjuntos de Datos como Asunto , Femenino , Humanos , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/virología , Virus del Tumor Mamario del Ratón/genética , Virus del Tumor Mamario del Ratón/patogenicidad , Ratones , Ratones Transgénicos , Receptor IGF Tipo 1 , Transducción de Señal , Microambiente Tumoral , Proteína Wnt1/genética
9.
Matern Child Health J ; 22(11): 1668-1675, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29978309

RESUMEN

Objectives The first 6 weeks postpartum represent a time of significant adjustment and learning for parents. Healthcare providers accurately understanding parents' needs and preferences regarding support, education, and services during this critical time is essential for optimizing maternal and infant health. The first objective of this study was to explore parents' experiences adjusting to the parenting role during the first 6 weeks postpartum. The second objective was to elicit from parents where and how they sought support and information during the early postpartum period, and what hindered this process. Methods Five focus groups were conducted with 33 mothers and fathers of young children, stratified by ethnicity, education, and income. An interdisciplinary team thematically coded verbatim transcripts and identified emergent themes. Results Main themes included low confidence in parenting and sifting through parenting information. Additional themes included communicating with partner about changing roles, breastfeeding, maternal mental health, and maternal postpartum recovery. Low parenting confidence was closely linked with information seeking, yet participants expressed being overwhelmed by the task of managing conflicting parenting information. Women reported that providers focused on infant needs, leaving them feeling unprepared for their own mental and physical health needs. Conclusions for Practice Parents report extensive needs for education and support in the early postpartum period, yet also report feeling overwhelmed by the quantity of parenting information available. These findings suggest parents need and desire reliable healthcare education after discharge that includes parent health and adjustment.


Asunto(s)
Padre/psicología , Madres/psicología , Responsabilidad Parental/psicología , Periodo Posparto , Apoyo Social , Adolescente , Adulto , Lactancia Materna/psicología , Crianza del Niño , Femenino , Grupos Focales , Humanos , Lactante , Masculino , Padres/educación , Adulto Joven
10.
J Neurosci Res ; 94(12): 1421-1433, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27557736

RESUMEN

There are many lines of evidence indicating that oligodendrocyte progenitor cells and oligodendrocyte populations in the central nervous system (CNS) are heterogeneous based on their developmental origins as well as from morphological and molecular criteria. Whether these distinctions reflect functional heterogeneity is less clear and has been the subject of considerable debate. Recent findings, particularly from knockout mouse models, have provided new evidence for regional variations in myelination phenotypes, particularly between brain and spinal cord. These data raise the possibility that oligodendrocytes in these regions have different functional capacities and/or ability to compensate for loss of a specific gene. The goal of this review is to briefly revisit the evidence for oligodendrocyte heterogeneity and then to present data from transgenic and demyelinating mouse models suggesting functional heterogeneity in myelination, demyelination, and remyelination in the CNS and, finally, to discuss the implications of these findings for human diseases. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Enfermedades del Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Oligodendroglía/patología , Animales , Humanos , Ratones , Ratones Transgénicos , Células-Madre Neurales/patología
11.
J Neurosci ; 34(13): 4453-65, 2014 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-24671992

RESUMEN

Prior studies support a role for mammalian target of rapamycin (mTOR) signaling in oligodendrocyte differentiation and myelination. Here we use Cre-recombinase driven by the CNP promoter to generate a mouse line with oligodendrocyte-specific knockdown of mTOR (mTOR cKO) in the CNS. We provide evidence that mTOR is necessary for proper oligodendrocyte differentiation and myelination in the spinal cord. Specifically, the number of mature oligodendrocytes was reduced, and the initiation and extent of myelination were impaired during spinal cord development. Consistent with these data, myelin protein expression, including myelin basic protein, proteolipid protein, myelin oligodendrocyte glycoprotein, and myelin-associated glycoprotein, was delayed in the spinal cord. Hypomyelination of the spinal cord persisted into adulthood, as did the reduction in numbers of mature oligodendrocytes. In the cortex, the structure of myelin appeared normal during development and in the adult; however, myelin protein expression was delayed during development and was abnormal in the adult. Myelin basic protein was significantly reduced in all regions at postnatal day 25. These data demonstrate that mTOR promotes oligodendrocyte differentiation and CNS myelination in vivo and show that the requirement for mTOR varies by region with the spinal cord most dependent on mTOR.


Asunto(s)
Diferenciación Celular/genética , Sistema Nervioso Central/citología , Regulación del Desarrollo de la Expresión Génica/fisiología , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Serina-Treonina Quinasas TOR/fisiología , 2',3'-Nucleótido Cíclico 3'-Fosfodiesterasa/genética , Factores de Edad , Animales , Animales Recién Nacidos , Recuento de Células , Sistema Nervioso Central/crecimiento & desarrollo , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Técnicas In Vitro , Integrasas/genética , Integrasas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de la Mielina/metabolismo , Vaina de Mielina/ultraestructura , Oligodendroglía/ultraestructura , Serina-Treonina Quinasas TOR/deficiencia , Serina-Treonina Quinasas TOR/genética
12.
J Neurosci ; 34(13): 4466-80, 2014 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-24671993

RESUMEN

During CNS development, oligodendrocytes, the myelinating glia of the CNS, progress through multiple transitory stages before terminating into fully mature cells. Oligodendrocyte differentiation and myelination is a tightly regulated process requiring extracellular signals to converge to elicit specific translational and transcriptional changes. Our lab has previously shown that the protein kinases, Akt and mammalian Target of Rapamycin (mTOR), are important regulators of CNS myelination in vivo. mTOR functions through two distinct complexes, mTOR complex 1 (mTORC1) and mTORC2, by binding to either Raptor or Rictor, respectively. To establish whether the impact of mTOR on CNS myelination results from unique functions of mTORC1 or mTORC2 during CNS myelination, we conditionally ablated either Raptor or Rictor in the oligodendrocyte lineage, in vivo. We show that Raptor (mTORC1) is a positive regulator of developmental CNS mouse myelination when mTORC2 is functional, whereas Rictor (mTORC2) ablation has a modest positive effect on oligodendrocyte differentiation, and very little effect on myelination, when mTORC1 is functional. Also, we show that loss of Raptor in oligodendrocytes results in differential dysmyelination in specific areas of the CNS, with the greatest impact on spinal cord myelination.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Portadoras/metabolismo , Diferenciación Celular/fisiología , Sistema Nervioso Central/citología , Cuerpo Calloso/citología , Vaina de Mielina/metabolismo , Oligodendroglía/fisiología , 2',3'-Nucleótido Cíclico 3'-Fosfodiesterasa/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas Portadoras/genética , Recuento de Células , Diferenciación Celular/genética , Sistema Nervioso Central/fisiología , Cuerpo Calloso/ultraestructura , Femenino , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Vaina de Mielina/ultraestructura , Oligodendroglía/metabolismo , Oligodendroglía/ultraestructura , Proteína Asociada al mTOR Insensible a la Rapamicina , Proteína Reguladora Asociada a mTOR , Transducción de Señal/fisiología , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
13.
J Biol Chem ; 289(8): 4626-33, 2014 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-24398690

RESUMEN

The objective of this study was to employ genetically engineered IGF-II analogs to establish which receptor(s) mediate the stemness promoting actions of IGF-II on mouse subventricular zone neural precursors. Neural precursors from the subventricular zone were propagated in vitro in culture medium supplemented with IGF-II analogs. Cell growth and identity were analyzed using sphere generation and further analyzed by flow cytometry. F19A, an analog of IGF-II that does not bind the IGF-2R, stimulated an increase in the proportion of neural stem cells (NSCs) while decreasing the proportion of the later stage progenitors at a lower concentration than IGF-II. V43M, which binds to the IGF-2R with high affinity but which has low binding affinity to the IGF-1R and to the A isoform of the insulin receptor (IR-A) failed to promote NSC growth. The positive effects of F19A on NSC growth were unaltered by the addition of a functional blocking antibody to the IGF-1R. Altogether, these data lead to the conclusion that IGF-II promotes stemness of NSCs via the IR-A and not through activation of either the IGF-1R or the IGF-2R.


Asunto(s)
Factor II del Crecimiento Similar a la Insulina/análogos & derivados , Factor II del Crecimiento Similar a la Insulina/farmacología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Receptor de Insulina/metabolismo , Animales , Biomarcadores/metabolismo , Proliferación Celular/efectos de los fármacos , Citometría de Flujo , Humanos , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Ligandos , Ratones , Ratones Endogámicos C57BL , Unión Proteica/efectos de los fármacos , Receptor IGF Tipo 2/metabolismo , Esferoides Celulares/citología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo
14.
J Biol Chem ; 289(13): 9430-9, 2014 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-24509861

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a member of the TNF family. The interaction of TRAIL with death receptor 4 (DR4) and DR5 can trigger apoptotic cell death. The aim of this study was to investigate the role of TRAIL signaling in neonatal hypoxia-ischemia (HI). Using a neonatal mouse model of HI, mRNA, and protein expression of TRAIL, DR5 and the TRAIL decoy receptors osteoprotegerin (OPG), mDcTRAILR1, and mDcTRAILR2 were determined. In vitro, mRNA expression of these genes was measured in primary neurons and oligodendrocyte progenitor cells (OPCs) after inflammatory cytokine (TNF-α/IFN-γ) treatment and/or oxygen and glucose deprivation (OGD). The toxicity of these various paradigms was also measured. The expression of TRAIL, DR5, OPG, and mDcTRAILR2 was significantly increased after HI. In vitro, inflammatory cytokines and OGD treatment significantly induced mRNAs for TRAIL, DR5, OPG, and mDcTRAILR2 in primary neurons and of TRAIL and OPG in OPCs. TRAIL protein was expressed primarily in microglia and astroglia, whereas DR5 co-localized with neurons and OPCs in vivo. OGD enhanced TNF-α/IFN-γ toxicity in both neuronal and OPC cultures. Recombinant TRAIL exerted toxicity alone or in combination with OGD and TNF-α/IFN-γ in primary neurons but not in OPC cultures. The marked increases in the expression of TRAIL and its receptors after cytokine exposure and OGD in primary neurons and OPCs were similar to those found in our animal model of neonatal HI. The toxicity of TRAIL in primary neurons suggests that TRAIL signaling participates in neonatal brain injury after inflammation and HI.


Asunto(s)
Sistema Nervioso Central/patología , Hipoxia/patología , Isquemia/patología , Transducción de Señal , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/deficiencia , Hipoxia/metabolismo , Inflamación/metabolismo , Inflamación/patología , Interferón gamma/farmacología , Isquemia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/patología , Oligodendroglía/patología , Oxígeno/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Transducción de Señal/efectos de los fármacos , Células Madre/efectos de los fármacos , Células Madre/patología , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Factor de Necrosis Tumoral alfa/farmacología
15.
Artículo en Inglés | MEDLINE | ID: mdl-38503504

RESUMEN

Myelination has evolved as a mechanism to ensure fast and efficient propagation of nerve impulses along axons. Within the central nervous system (CNS), myelination is carried out by highly specialized glial cells, oligodendrocytes. The formation of myelin is a prolonged aspect of CNS development that occurs well into adulthood in humans, continuing throughout life in response to injury or as a component of neuroplasticity. The timing of myelination is tightly tied to the generation of oligodendrocytes through the differentiation of their committed progenitors, oligodendrocyte precursor cells (OPCs), which reside throughout the developing and adult CNS. In this article, we summarize our current understanding of some of the signals and pathways that regulate the differentiation of OPCs, and thus the myelination of CNS axons.


Asunto(s)
Diferenciación Celular , Vaina de Mielina , Oligodendroglía , Oligodendroglía/fisiología , Oligodendroglía/citología , Humanos , Animales , Vaina de Mielina/fisiología , Vaina de Mielina/metabolismo , Transducción de Señal , Sistema Nervioso Central/fisiología , Axones/fisiología , Axones/metabolismo
16.
Stem Cells ; 30(6): 1265-76, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22593020

RESUMEN

Insulin-like growth factor (IGF)-I and IGF-II regulate brain development and growth through the IGF type 1 receptor (IGF-1R). Less appreciated is that IGF-II, but not IGF-I, activates a splice variant of the insulin receptor (IR) known as IR-A. We hypothesized that IGF-II exerts distinct effects from IGF-I on neural stem/progenitor cells (NSPs) via its interaction with IR-A. Immunofluorescence revealed high IGF-II in the medial region of the subventricular zone (SVZ) comprising the neural stem cell niche, with IGF-II mRNA predominant in the adjacent choroid plexus. The IGF-1R and the IR isoforms were differentially expressed with IR-A predominant in the medial SVZ, whereas the IGF-1R was more abundant laterally. Similarly, IR-A was more highly expressed by NSPs, whereas the IGF-1R was more highly expressed by lineage restricted cells. In vitro, IGF-II was more potent in promoting NSP expansion than either IGF-I or standard growth medium. Limiting dilution and differentiation assays revealed that IGF-II was superior to IGF-I in promoting stemness. In vivo, NSPs propagated in IGF-II migrated to and took up residence in periventricular niches while IGF-I-treated NSPs predominantly colonized white matter. Knockdown of IR or IGF-1R using shRNAs supported the conclusion that the IGF-1R promotes progenitor proliferation, whereas the IR is important for self-renewal. Q-PCR revealed that IGF-II increased Oct4, Sox1, and FABP7 mRNA levels in NSPs. Our data support the conclusion that IGF-II promotes the self-renewal of neural stem/progenitors via the IR. By contrast, IGF-1R functions as a mitogenic receptor to increase precursor abundance.


Asunto(s)
Factor II del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Animales , Diferenciación Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Factor I del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/genética , Ratones , Transducción de Señal
17.
J Mammary Gland Biol Neoplasia ; 17(2): 119-23, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22678420

RESUMEN

Originally adapted from the neurosphere assay, the nonadherent mammosphere assay has been utilized to assess early progenitor/stem cell frequency in a given population of mammary epithelial cells. This method has also been used to measure the frequency of tumorsphere initiating cells in both primary mammary tumors as well as in tumor cell lines. Although, the mammosphere assay has been used extensively in the mammary gland field, a standard method of quantifying and analyzing sphere growth in this assay has remained undefined. Here, we discuss the use and benefit of using a limiting dilution analysis to quantify sphere-forming frequency in primary mammary epithelial cells grown in nonadherent conditions.


Asunto(s)
Neoplasias de la Mama/patología , Glándulas Mamarias Humanas/patología , Células Madre Neoplásicas/patología , Ensayo de Tumor de Célula Madre , Animales , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Microambiente Celular , Epitelio/patología , Femenino , Humanos , Glándulas Mamarias Animales/patología , Células Tumorales Cultivadas
18.
Endocr Relat Cancer ; 30(10)2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37490874

RESUMEN

Despite decades of research presenting insulin-like growth factor-1 receptor (IGF1R) as an attractive target for cancer therapy, IGF1R inhibitors ultimately failed in clinical trials. This was surprising due to the known cancer-promoting functions of IGF1R, including stimulation of cell invasion, proliferation, and survival. Discourse in the literature has acknowledged that a lack of patient stratification may have impacted the success of IGF1R-inhibitor trials. This argument alludes to the possibility that IGF1R function may be contingent on tumor type and cellular composition. Looking into the known roles of IGF1R, it becomes clear that this receptor interacts with a multitude of different proteins and even has tumor-suppressing functions. IGF1R is implicated in both cell-cell and cell-surface adhesion dynamics, and the effects of either IGF1R downregulation or pharmacological inhibition on cellular adhesion remain poorly understood. In turn, adhesion receptors modulate IGF1R signaling. In addition, our understanding of IGF1R function in tumor-associated immune and stromal cells is lacking, which could contribute to the overwhelming failure of IGF1R inhibitors in the clinic. In this review, we re-investigate clinical trial data to make connections between the failure of these drugs in human cancer patients and the understudied facets of IGF1R function. We describe lesser-known and potentially tumor-suppressive functions of IGF1R that include promoting cell-cell adhesion through E-cadherin, augmenting a pro-inflammatory macrophage phenotype, and stimulating B cells to produce immunoglobulins. We also highlight the important role of adhesion receptors in regulating IGF1R function, and we use this information to infer stratification criteria for selecting patients that might benefit from IGF1R inhibitors.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina , Neoplasias , Receptor IGF Tipo 1 , Humanos , Cadherinas/genética , Cadherinas/metabolismo , Cadherinas/farmacología , Línea Celular Tumoral , Proliferación Celular , Factor I del Crecimiento Similar a la Insulina/metabolismo , Integrinas , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Transducción de Señal
19.
Neurotrauma Rep ; 4(1): 433-446, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37435356

RESUMEN

Myelin loss and oligodendrocyte death are well documented in patients with traumatic brain injury (TBI), as well as in experimental animal models after moderate-to-severe TBI. In comparison, mild TBI (mTBI) does not necessarily result in myelin loss or oligodendrocyte death, but causes structural alterations in the myelin. To gain more insight into the impact of mTBI on oligodendrocyte lineage in the adult brain, we subjected mice to mild lateral fluid percussion injury (mFPI) and characterized the early impact (1 and 3 days post-injury) on oligodendrocytes in the corpus callosum using multiple oligodendrocyte lineage markers (platelet-derived growth factor receptor [PDGFR]-α, glutathione S-transferase [GST]-π, CC1, breast carcinoma-amplified sequence 1 [BCAS1], myelin basic protein [MBP], myelin-associated glycoprotein [MAG], proteolipid protein [PLP], and FluoroMyelin™). Two regions of the corpus callosum in relation to the impact site were analyzed: areas near (focal) and anterior (distal) to the impact site. mFPI did not result in oligodendrocyte death in either the focal or distal corpus callosum, nor impact on oligodendrocyte precursors (PDGFR-α+) and GST-π+ oligodendrocyte numbers. In the focal but not distal corpus callosum, mFPI caused a decrease in CC1+ as well as BCAS1+ actively myelinating oligodendrocytes and reduced FluoroMyelin intensity without altering myelin protein expression (MBP, PLP, and MAG). Disruption in node-paranode organization and loss of Nav1.6+ nodes were observed in both the focal and distal regions, even in areas without obvious axonal damage. Altogether, our study shows regional differences in mature and myelinating oligodendrocyte in response to mFPI. Further, mFPI elicits a widespread impact on node-paranode organization that affects regions both close to and remotely located from the site of injury.

20.
Glia ; 60(11): 1684-95, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22836368

RESUMEN

Extrinsic factors including growth factors influence decisions of oligodendrocyte progenitor cells (OPCs) to continue cell cycle progression or exit the cell cycle and terminally differentiate into oligodendrocytes capable of producing myelin. Multiple studies have elucidated how the G1/S transition is regulated in OPCs; however, little is known about how S phase progression and the G2/M transition are regulated in these cells. Herein, we report that insulin-like growth factor (IGF)-I coordinates with FGF-2 to promote S phase progression but regulates G2/M progression independently. During S phase, IGF-I/FGF-2 enhances protein expression of cyclin A and cdk2, and further increases effective complex formation resulting in enhanced cdk2 activity. Surprisingly, however, OPCs exposed to FGF-2 in the absence of IGF-I fail to traverse through G2/M. Consistent with this observation, OPCs exposed to IGF-I, but not FGF-2, increase cell number over 48 h. IGF-I enhances cdk1 kinase activity during G2/M by promoting nuclear localization of cyclin B/cdk1 as well as of Cdc25C, an activator of cdk1. IGF-I also induces phosphorylation of histone 3 indicating traverse of cells through mitosis. Finally, we demonstrate that IGF-I-mediated G2/M regulation requires mammalian target of rapamycin activity. These data support an important function for IGF-I in G2/M progression in OPCs.


Asunto(s)
Ciclo Celular/fisiología , Factor I del Crecimiento Similar a la Insulina/farmacología , Células-Madre Neurales/metabolismo , Oligodendroglía/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Proteína Quinasa CDC2/metabolismo , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Células Cultivadas , Factor 2 de Crecimiento de Fibroblastos/farmacología , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Oligodendroglía/citología , Oligodendroglía/efectos de los fármacos , Fosforilación , Prosencéfalo/citología , Prosencéfalo/efectos de los fármacos , Prosencéfalo/metabolismo , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA