Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
EMBO Rep ; 25(3): 1256-1281, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38429579

RESUMO

The plant homeodomain zinc-finger protein, PHF6, is a transcriptional regulator, and PHF6 germline mutations cause the X-linked intellectual disability (XLID) Börjeson-Forssman-Lehmann syndrome (BFLS). The mechanisms by which PHF6 regulates transcription and how its mutations cause BFLS remain poorly characterized. Here, we show genome-wide binding of PHF6 in the developing cortex in the vicinity of genes involved in central nervous system development and neurogenesis. Characterization of BFLS mice harbouring PHF6 patient mutations reveals an increase in embryonic neural stem cell (eNSC) self-renewal and a reduction of neural progenitors. We identify a panel of Ephrin receptors (EphRs) as direct transcriptional targets of PHF6. Mechanistically, we show that PHF6 regulation of EphR is impaired in BFLS mice and in conditional Phf6 knock-out mice. Knockdown of EphR-A phenocopies the PHF6 loss-of-function defects in altering eNSCs, and its forced expression rescues defects of BFLS mice-derived eNSCs. Our data indicate that PHF6 directly promotes Ephrin receptor expression to control eNSC behaviour in the developing brain, and that this pathway is impaired in BFLS.


Assuntos
Epilepsia , Face/anormalidades , Dedos/anormalidades , Transtornos do Crescimento , Hipogonadismo , Deficiência Intelectual , Deficiência Intelectual Ligada ao Cromossomo X , Obesidade , Humanos , Camundongos , Animais , Deficiência Intelectual/genética , Proteínas Repressoras , Deficiência Intelectual Ligada ao Cromossomo X/genética , Deficiência Intelectual Ligada ao Cromossomo X/metabolismo , Epilepsia/genética , Epilepsia/metabolismo , Fatores de Transcrição
2.
Curr Top Membr ; 92: 99-123, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38007271

RESUMO

Chloride intracellular channel 1 (CLIC1) has emerged as a therapeutic target in various cancers. CLIC1 promotes cell cycle progression and cancer stem cell (CSC) self-renewal. Furthermore, CLIC1 is shown to play diverse roles in proliferation, cell volume regulation, tumour invasion, migration, and angiogenesis. In glioblastoma (GB), CLIC1 facilitates the G1/S phase transition and tightly regulates glioma stem-like cells (GSCs), a rare population of self-renewing CSCs with central roles in tumour resistance to therapy and tumour recurrence. CLIC1 is found as either a monomeric soluble protein or as a non-covalent dimeric protein that can form an ion channel. The ratio of dimeric to monomeric protein is altered in GSCs and depends on the cell redox state. Elucidating the mechanisms underlying the alterations in CLIC1 expression and structural transitions will further our understanding of its role in GSC biology. This review will highlight the role of CLIC1 in GSCs and its significance in facilitating different hallmarks of cancer.


Assuntos
Glioblastoma , Humanos , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Canais de Cloreto/metabolismo , Canais de Cloreto/uso terapêutico
3.
EMBO Rep ; 21(12): e49499, 2020 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-33047485

RESUMO

The function and maintenance of muscle stem cells (MuSCs) are tightly regulated by signals originating from their niche environment. Skeletal myofibers are a principle component of the MuSC niche and are in direct contact with the muscle stem cells. Here, we show that Myf6 establishes a ligand/receptor interaction between muscle stem cells and their associated muscle fibers. Our data show that Myf6 transcriptionally regulates a broad spectrum of myokines and muscle-secreted proteins in skeletal myofibers, including EGF. EGFR signaling blocks p38 MAP kinase-induced differentiation of muscle stem cells. Homozygous deletion of Myf6 causes a significant reduction in the ability of muscle to produce EGF, leading to a deregulation in EGFR signaling. Consequently, although Myf6-knockout mice are born with a normal muscle stem cell compartment, they undergo a progressive reduction in their stem cell pool during postnatal life due to spontaneous exit from quiescence. Taken together, our data uncover a novel role for Myf6 in promoting the expression of key myokines, such as EGF, in the muscle fiber which prevents muscle stem cell exhaustion by blocking their premature differentiation.


Assuntos
Fatores de Regulação Miogênica , Células-Tronco , Animais , Diferenciação Celular/genética , Homozigoto , Camundongos , Músculo Esquelético , Fatores de Regulação Miogênica/genética , Deleção de Sequência
4.
Mol Cell ; 47(3): 457-68, 2012 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-22771117

RESUMO

In skeletal myogenesis, the transcription factor MyoD activates distinct transcriptional programs in progenitors compared to terminally differentiated cells. Using ChIP-Seq and gene expression analyses, we show that in primary myoblasts, Snail-HDAC1/2 repressive complex binds and excludes MyoD from its targets. Notably, Snail binds E box motifs that are G/C rich in their central dinucleotides, and such sites are almost exclusively associated with genes expressed during differentiation. By contrast, Snail does not bind the A/T-rich E boxes associated with MyoD targets in myoblasts. Thus, Snai1-HDAC1/2 prevent MyoD occupancy on differentiation-specific regulatory elements, and the change from Snail to MyoD binding often results in enhancer switching during differentiation. Furthermore, we show that a regulatory network involving myogenic regulatory factors (MRFs), Snai1/2, miR-30a, and miR-206 acts as a molecular switch that controls entry into myogenic differentiation. Together, these results reveal a regulatory paradigm that directs distinct gene expression programs in progenitors versus terminally differentiated cells.


Assuntos
Elementos Facilitadores Genéticos/fisiologia , Desenvolvimento Muscular/genética , Proteína MyoD/metabolismo , Mioblastos Esqueléticos/fisiologia , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Diferenciação Celular/genética , Camundongos , Dados de Sequência Molecular , Proteína MyoD/química , Proteína MyoD/genética , Mioblastos Esqueléticos/citologia , Cultura Primária de Células , Ligação Proteica/genética , Fatores de Transcrição da Família Snail , Fatores de Transcrição/química , Fatores de Transcrição/genética , Transcrição Gênica/fisiologia
5.
Hum Mol Genet ; 24(16): 4573-83, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26002103

RESUMO

Defects in mitochondrial fission and cyclin dependent kinase 5 (CDK5) activation are early events that precede neuronal loss following NMDA-induced neuronal death. Here, we report that the cytoplasmic CDK5 tightly regulates mitochondrial morphology defects associated with NMDA-induced neuronal injury via regulation of the mitochondrial fission protein, dynamin-related protein 1 (DRP1). We show that DRP1 is a direct target of CDK5. CDK5-mediated phosphorylation of DRP1 at a conserved Serine residue, S585, is elevated at the mitochondria and is associated with increased mitochondrial fission. Ectopic expression of a cytoplasmic CDK5 or mutant DRP1-S585D results in increased mitochondrial fragmentation in primary neurons. Conversely, expression of a dominant negative form of cytoplasmic CDK5 or mutant DRP1-S585A results in elongated mitochondria. In addition, pharmacological inhibition of CDK5 by Roscovitine inhibits DRP1 phosphorylation and mitochondrial fission associated with NMDA-induced neuronal loss. Importantly, conditional deletion of CDK5 significantly attenuates DRP1 phosphorylation at S585 and rescues mitochondrial fission defects in neurons exposed to NMDA. Our studies delineate an important mechanism by which CDK5 regulates mitochondrial morphology defects associated with neuronal injury.


Assuntos
Quinase 5 Dependente de Ciclina/metabolismo , Dinaminas/metabolismo , Mitocôndrias/metabolismo , N-Metilaspartato/toxicidade , Neurônios/metabolismo , Substituição de Aminoácidos , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Quinase 5 Dependente de Ciclina/genética , Dinaminas/genética , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/patologia , Mutação de Sentido Incorreto , Neurônios/patologia , Fosforilação
6.
J Pathol ; 240(4): 381-383, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27538356

RESUMO

Chordoma is a rare primary bone cancer with limited treatment options. Surgical resection followed by radiotherapy has proven effective; however, when, in 30-40% of patients, tumours recur and metastasize, a high level of resistance to chemotherapies leaves these patients with a dearth of treatment options. Recent work published in the Journal of Pathology by Scheipl et al describing a focused compound drug screen highlights the significance of epidermal growth factor receptor (EGFR) signalling in chordoma, and shows potential for EGFR inhibitors as a way forward for developing an effective treatment for chordoma. Importantly, combining EGFR inhibitors with a MET inhibitor induces a synergistic effect on growth inhibition of resistant chordoma cells, highlighting the significance of combined EGFR and MET inhibitors as a potential avenue to defeat chemoresistance in chordoma patients. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Cordoma/tratamento farmacológico , Receptores ErbB/antagonistas & inibidores , Antineoplásicos/farmacologia , Humanos , Recidiva Local de Neoplasia/tratamento farmacológico , Reino Unido
7.
Neurobiol Dis ; 96: 227-235, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27633282

RESUMO

Intellectual disability encompasses a large set of neurodevelopmental disorders of cognition that are more common in males than females. Although mutations in over 100 X-linked genes associated to intellectual disability have been identified, only a few X-linked intellectual disability proteins have been intensively studied. Hence, the molecular mechanisms underlying the majority of X-linked intellectual disability disorders remain poorly understood. A substantial fraction of X-linked intellectual disability genes encode nuclear proteins, suggesting that elucidating their functions in the regulation of transcription may provide novel insights into the pathogenesis of intellectual disability. Recent studies have uncovered mechanisms by which mutations of the gene encoding plant homeodomain (PHD)-like finger protein 6 (PHF6) contribute to the pathogenesis of the X-linked intellectual disability disorder Börjeson-Forssman-Lehmann syndrome (BFLS). PHF6 plays a critical role in the migration of neurons in the mouse cerebral cortex in vivo, and patient-specific mutations disrupt the ability of PHF6 to promote neuronal migration. Interestingly, PHF6 physically associates with the PAF1 transcriptional elongation complex and thereby drives neuronal migration in the cerebral cortex. PHF6 also interacts with the NuRD chromatin remodeling complex and with the nucleolar transcriptional regulator UBF, though the biological role of these interactions remains to be characterized. In other studies, PHF6 mRNA has been identified as the target of the microRNA miR-128 in the cerebral cortex, providing new insights into regulation of PHF6 function in neuronal migration. Importantly, deregulation of PHF6 function in neuronal migration triggers the formation of white matter heterotopias that harbor neuronal hyperexcitability, which may be relevant to the pathogenesis of intellectual disability and seizures in BFLS. Collectively, these studies are beginning to provide key insights into the molecular pathogenesis of BFLS.


Assuntos
Proteínas de Transporte/genética , Córtex Cerebral/metabolismo , Epilepsia/genética , Epilepsia/patologia , Face/anormalidades , Dedos/anormalidades , Transtornos do Crescimento/genética , Transtornos do Crescimento/patologia , Hipogonadismo/genética , Hipogonadismo/patologia , Deficiência Intelectual Ligada ao Cromossomo X/genética , Deficiência Intelectual Ligada ao Cromossomo X/patologia , Obesidade/genética , Obesidade/patologia , Animais , Córtex Cerebral/patologia , Epilepsia/complicações , Face/patologia , Dedos/patologia , Transtornos do Crescimento/complicações , Humanos , Hipogonadismo/complicações , Deficiência Intelectual/etiologia , Deficiência Intelectual/genética , Deficiência Intelectual Ligada ao Cromossomo X/complicações , Mutação/genética , Obesidade/complicações , Proteínas Repressoras
8.
J Neurosci ; 32(23): 7806-18, 2012 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-22674257

RESUMO

Malignant gliomas, including glioblastoma multiforme, constitute the most common and aggressive primary brain tumors in adults. The transcription factor signal transducer and activator of transcription 3 (STAT3) plays an essential role in glioblastoma pathogenesis downstream of the major oncogenic protein epidermal growth factor receptor variant III (EGFRvIII). However, the critical gene targets of STAT3 that mediate EGFRvIII-induced glial transformation have remained unknown. Here, we identify inducible nitric oxide synthase (iNOS) as a novel target gene of STAT3 in EGFRvIII-expressing mouse astrocytes. Endogenous STAT3 occupies the endogenous iNOS promoter and stimulates iNOS transcription in EGFRvIII-expressing astrocytes. STAT3 does not appear to control iNOS transcription in astrocytes deficient in the major glioblastoma tumor suppressor protein phosphatase and tensin homolog (PTEN), suggesting that STAT3 regulates iNOS transcription specifically in EGFRvIII-expressing astrocytes. Importantly, inhibition of iNOS by distinct approaches, including knockdown by RNA interference, reduces cell population growth and invasiveness of EGFRvIII-expressing astrocytes. In addition, upon iNOS knockdown or administration of a small-molecule inhibitor of iNOS, EGFRvIII-expressing astrocytes form smaller tumors in vivo. These findings suggest that inhibition of iNOS may have potential therapeutic value for EGFRvIII-activated brain tumors.


Assuntos
Proliferação de Células , Transformação Celular Neoplásica/patologia , Receptores ErbB/fisiologia , Neuroglia/fisiologia , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/fisiologia , Fator de Transcrição STAT3/fisiologia , Transdução de Sinais/fisiologia , Animais , Animais Geneticamente Modificados , Astrócitos/efeitos dos fármacos , Astrócitos/fisiologia , Sítios de Ligação , Transformação Celular Neoplásica/efeitos dos fármacos , Células Cultivadas , Imunoprecipitação da Cromatina , Inibidores Enzimáticos/farmacologia , Glioblastoma/genética , Glioblastoma/patologia , Imuno-Histoquímica , Lentivirus/genética , Camundongos , Óxido Nítrico Sintase Tipo II/genética , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/fisiologia , Plasmídeos/genética , Reação em Cadeia da Polimerase , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica
9.
Nat Commun ; 14(1): 535, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36726011

RESUMO

Adult stem cells are indispensable for tissue regeneration, but their function declines with age. The niche environment in which the stem cells reside plays a critical role in their function. However, quantification of the niche effect on stem cell function is lacking. Using muscle stem cells (MuSC) as a model, we show that aging leads to a significant transcriptomic shift in their subpopulations accompanied by locus-specific gain and loss of chromatin accessibility and DNA methylation. By combining in vivo MuSC transplantation and computational methods, we show that the expression of approximately half of all age-altered genes in MuSCs from aged male mice can be restored by exposure to a young niche environment. While there is a correlation between gene reversibility and epigenetic alterations, restoration of gene expression occurs primarily at the level of transcription. The stem cell niche environment therefore represents an important therapeutic target to enhance tissue regeneration in aging.


Assuntos
Células-Tronco Adultas , Músculo Esquelético , Masculino , Camundongos , Animais , Músculo Esquelético/metabolismo , Fibras Musculares Esqueléticas , Células-Tronco/metabolismo , Envelhecimento/fisiologia
10.
J Biol Chem ; 286(6): 4772-82, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21041314

RESUMO

Mitochondrial dynamics have been extensively studied in the context of classical cell death models involving Bax-mediated cytochrome c release. Excitotoxic neuronal loss is a non-classical death signaling pathway that occurs following overactivation of glutamate receptors independent of Bax activation. Presently, the role of mitochondrial dynamics in the regulation of excitotoxicity remains largely unknown. Here, we report that NMDA-induced excitotoxicity results in defects in mitochondrial morphology as evident by the presence of excessive fragmented mitochondria, cessation of mitochondrial fusion, and cristae dilation. Up-regulation of the mitochondrial inner membrane GTPase, Opa1, is able to restore mitochondrial morphology and protect neurons against excitotoxic injury. Opa1 functions downstream of the calcium-dependent protease, calpain. Inhibition of calpain activity by calpastatin, an endogenous calpain inhibitor, significantly rescued mitochondrial defects and maintained neuronal survival. Opa1 was required for calpastatin-mediated neuroprotection because the enhanced survival found following NMDA-induced toxicity was significantly reduced upon loss of Opa1. Our results define a mechanism whereby breakdown of the mitochondrial network mediated through loss of Opa1 function contributes to neuronal death following excitotoxic neuronal injury. These studies suggest Opa1 as a potential therapeutic target to promote neuronal survival following acute brain damage and neurodegenerative diseases.


Assuntos
Cerebelo/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Membranas Mitocondriais/metabolismo , Proteínas Mitocondriais/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Animais , Lesões Encefálicas/genética , Lesões Encefálicas/metabolismo , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Cerebelo/citologia , Citotoxinas/efeitos adversos , Citotoxinas/farmacologia , Agonistas de Aminoácidos Excitatórios/efeitos adversos , Agonistas de Aminoácidos Excitatórios/farmacologia , GTP Fosfo-Hidrolases/genética , Camundongos , Proteínas Mitocondriais/genética , N-Metilaspartato/efeitos adversos , N-Metilaspartato/farmacologia , Proteínas do Tecido Nervoso/genética , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Neurônios/citologia
11.
Methods Mol Biol ; 2515: 1-15, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35776342

RESUMO

Defects in mitochondrial oxidative phosphorylation have been observed in numerous neurodegenerative disorders and are linked to bioenergetic crises leading to neuronal death. The distinct metabolic profile of neurons is predominantly oxidative, which is characterized by the oxidation of glucose or its metabolites in the mitochondria to produce ATP. This process involves the tricarboxylic acid cycle, electron transfer in the respiratory chain, and oxygen consumption. Therefore, measurement of oxygen consumption rates (OCR) can be accurately applied to assess the rate of mitochondrial respiration. In this chapter, we describe our optimized protocol for the assessment of OCR specifically in primary mouse cerebellar granule neurons (CGN). The protocol includes isolation and manipulation of mouse CGNs followed by real-time assessment of mitochondrial OCR using a Seahorse XFe96 extracellular flux analyzer.


Assuntos
Respiração Celular , Mitocôndrias , Animais , Camundongos , Mitocôndrias/metabolismo , Neurônios/metabolismo , Fosforilação Oxidativa , Respiração
12.
Methods Mol Biol ; 2515: 297-308, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35776359

RESUMO

Neurogenesis is outlined as a process in which new neurons are generated from neural stem cells (NSCs). This process comprises proliferation and fate specification of NSCs, migration of newborn neurons, and their maturation. Defects in embryonic neurogenesis have emerged as a key mechanism underlying neurodevelopmental disorders such as autism spectrum disorders and intellectual disability. An impairment in neurogenesis has also been observed in neurodegenerative disorders such as Huntington's disease. Transgenic animal models of neurodevelopmental and neurodegenerative diseases have been developed which serve as invaluable tools to investigate the early mechanisms of disease pathogenesis. In this chapter, we describe our optimized method to obtain and maintain reproducible neurosphere cultures from transgenic or patient mouse models followed by characterization of NSCs by flow cytometry.


Assuntos
Células-Tronco Embrionárias Murinas , Células-Tronco Neurais , Animais , Biomarcadores , Citometria de Fluxo , Camundongos , Neurogênese
13.
Methods Mol Biol ; 2515: 343-354, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35776362

RESUMO

The generation of new neurons in the adult brain throughout life is integral to brain plasticity and repair. Adult neural stem cells (aNSCs), present in the subventricular zone (SVZ) of the lateral ventricle wall and the subgranular zone (SGZ) of the hippocampal dentate gyrus, divide symmetrically or asymmetrically to maintain the stem cell pool or become committed progenitors and differentiate into various cell lineages. Depletion or dysregulation of aNSCs impairs proper brain connectivity and function and can contribute to several brain diseases including cognitive and neurodegenerative disorders and brain cancer. In this chapter, we present our optimized method to obtain and maintain reproducible neurosphere cultures from the adult mouse brain followed by evaluation of self-renewal using the extreme limiting dilution assay (ELDA) software. We use this assay routinely on aNSCs obtained from patient mouse models to generate log fraction plots and provide confidence intervals for all limiting dilution assay (LDA) data. At the same time, given the low number of NSCs required for the completion of the ELDA experiment, it is feasible to employ this approach to conduct high-content compound screening for therapeutic interventions aimed at enhancing the stem cell pool or combating a cohort of genetic and epigenetic disorders.


Assuntos
Células-Tronco Adultas , Células-Tronco Neurais , Animais , Encéfalo , Humanos , Ventrículos Laterais , Camundongos , Neurônios
14.
STAR Protoc ; 3(3): 101554, 2022 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-35880130

RESUMO

Improper or aberrant protein-protein interactions can lead to severe human diseases including cancer. Here, we describe an adapted proximity ligation assay (PLA) protocol for the assessment of galectin-1-HOXA5 interaction in brain tumor stem cells (BTSCs). We detail the steps for culturing and preparation of BTSCs followed by PLA and detection of protein interactions in situ using fluorescent microscopy. This PLA protocol is optimized specifically for BTSCs and includes key controls for effective result analysis. For complete details on the use and execution of this protocol, please refer to Sharanek et al. (2021).


Assuntos
Encéfalo , Mapeamento de Interação de Proteínas , Humanos , Microscopia de Fluorescência/métodos , Células-Tronco Neoplásicas , Mapeamento de Interação de Proteínas/métodos
15.
Methods Cell Biol ; 170: 47-58, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35811103

RESUMO

Brain tumor stem cells (BTSCs) are a rare population of self-renewing stem cells that are cultured as spheres and are often slow growing compared to other mammalian cell lines. Analysis of BTSC proteome requires careful handling as well as techniques that can be applied to small quantities of cell material. Subcellular fractionation is a widely used technique to assess protein localization. Although proteins are often destined to a defined cell compartment via a signal peptide such as mitochondrial or nuclear localization signals, the recruitment of a protein from one compartment to another can occur as a result of post-translational modification and/or structural variations in response to intracellular and extracellular stimuli. These events assign different functions to a protein making the study of protein localization a useful approach for better understanding of its role in disease progression. Sequential centrifugation remains a simple and versatile fractionation method for proteomic analysis. It can also be applied for diverse downstream applications such as multi-omics using pure nuclear fractions or metabolomic studies on isolated mitochondria. In this chapter, we describe our optimized protocol for subcellular fractionation of BTSC spheres in which we use a commercially available kit with additional centrifugation steps. We provide details on BTSC maintenance and handling, fractionation protocol and evaluation of fraction purity.


Assuntos
Células-Tronco Neoplásicas , Proteômica , Animais , Encéfalo/metabolismo , Fracionamento Celular/métodos , Núcleo Celular/metabolismo , Mamíferos/metabolismo , Células-Tronco Neoplásicas/patologia , Proteoma/metabolismo , Proteômica/métodos , Frações Subcelulares/metabolismo
16.
Biochim Biophys Acta ; 1802(1): 162-6, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19747972

RESUMO

Mitochondria are highly dynamic organelles that undergo constant cycles of fusion and fission. An additional level of regulation of mitochondrial function, which is particularly important in neurons, is their active transport along microtubules. Recent evidence suggests that the mitochondrial fusion/fission machinery as well as the molecular motors responsible for their movement constitute powerful regulatory control points that directly impact metabolism and regulation of cell death. This is true for not only apoptosis, but also for excitotoxicity where calcium overload is a major component of the cell death process. In this review, we will describe the molecular mechanisms regulating fusion and fission and how this impinges on cell survival in the context of acute neuronal injury.


Assuntos
Mitocôndrias/metabolismo , Animais , Apoptose , Morte Celular , Sobrevivência Celular , Fusão de Membrana , Modelos Biológicos , Neurônios/metabolismo
17.
Cell Rep ; 36(9): 109647, 2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34469737

RESUMO

Brain tumor stem cells (BTSCs) and intratumoral heterogeneity represent major challenges in glioblastoma therapy. Here, we report that the LGALS1 gene, encoding the carbohydrate binding protein, galectin1, is a key regulator of BTSCs and glioblastoma resistance to therapy. Genetic deletion of LGALS1 alters BTSC gene expression profiles and results in downregulation of gene sets associated with the mesenchymal subtype of glioblastoma. Using a combination of pharmacological and genetic approaches, we establish that inhibition of LGALS1 signaling in BTSCs impairs self-renewal, suppresses tumorigenesis, prolongs lifespan, and improves glioblastoma response to ionizing radiation in preclinical animal models. Mechanistically, we show that LGALS1 is a direct transcriptional target of STAT3 with its expression robustly regulated by the ligand OSM. Importantly, we establish that galectin1 forms a complex with the transcription factor HOXA5 to reprogram the BTSC transcriptional landscape. Our data unravel an oncogenic signaling pathway by which the galectin1/HOXA5 complex maintains BTSCs and promotes glioblastoma.


Assuntos
Neoplasias Encefálicas/metabolismo , Galectina 1/metabolismo , Glioblastoma/metabolismo , Células-Tronco Neoplásicas/metabolismo , Transcrição Gênica , Idoso , Animais , Antineoplásicos/farmacologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/radioterapia , Calixarenos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Autorrenovação Celular , Receptores ErbB/genética , Receptores ErbB/metabolismo , Galectina 1/antagonistas & inibidores , Galectina 1/genética , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Glioblastoma/patologia , Glioblastoma/radioterapia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Masculino , Camundongos SCID , Pessoa de Meia-Idade , Mutação , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/efeitos da radiação , Tolerância a Radiação , Radiossensibilizantes/farmacologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Nat Commun ; 11(1): 4116, 2020 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-32807793

RESUMO

Glioblastoma contains a rare population of self-renewing brain tumor stem cells (BTSCs) which are endowed with properties to proliferate, spur the growth of new tumors, and at the same time, evade ionizing radiation (IR) and chemotherapy. However, the drivers of BTSC resistance to therapy remain unknown. The cytokine receptor for oncostatin M (OSMR) regulates BTSC proliferation and glioblastoma tumorigenesis. Here, we report our discovery of a mitochondrial OSMR that confers resistance to IR via regulation of oxidative phosphorylation, independent of its role in cell proliferation. Mechanistically, OSMR is targeted to the mitochondrial matrix via the presequence translocase-associated motor complex components, mtHSP70 and TIM44. OSMR interacts with NADH ubiquinone oxidoreductase 1/2 (NDUFS1/2) of complex I and promotes mitochondrial respiration. Deletion of OSMR impairs spare respiratory capacity, increases reactive oxygen species, and sensitizes BTSCs to IR-induced cell death. Importantly, suppression of OSMR improves glioblastoma response to IR and prolongs lifespan.


Assuntos
Glioblastoma/metabolismo , Células-Tronco Neoplásicas/metabolismo , Radiação Ionizante , Receptores de Oncostatina M/metabolismo , Animais , Morte Celular/efeitos da radiação , Linhagem Celular Tumoral , Proliferação de Células/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Células Cultivadas , Imunofluorescência , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Masculino , Camundongos , Camundongos SCID , NADH Desidrogenase/genética , NADH Desidrogenase/metabolismo , Células-Tronco Neoplásicas/efeitos da radiação , Oncostatina M/metabolismo , Estresse Oxidativo/efeitos da radiação , Receptores de Oncostatina M/genética , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos da radiação
19.
J Neurosci ; 28(24): 6068-78, 2008 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-18550749

RESUMO

Despite the importance of Mcl-1, an anti-apoptotic Bcl-2 family member, in the regulation of apoptosis, little is known regarding its role in nervous system development and injury-induced neuronal cell death. Because germline deletion of Mcl-1 results in peri-implantation lethality, we address the function of Mcl-1 in the nervous system using two different conditional Mcl-1 mouse mutants in the developing nervous system. Here, we show for the first time that Mcl-1 is required for neuronal development. Neural precursors within the ventricular zone and newly committed neurons in the cortical plate express high levels of Mcl-1 throughout cortical neurogenesis. Loss of Mcl-1 in neuronal progenitors results in widespread apoptosis. Double labeling with active caspase 3 and Tuj1 reveals that newly committed Mcl1 deficient neurons undergo apoptosis as they commence migration away from the ventricular zone. Examination of neural progenitor differentiation in vitro demonstrated that cell death in the absence of Mcl1 is cell autonomous. Although conditional deletion of Mcl-1 in cultured neurons does not trigger apoptosis, loss of Mcl-1 sensitizes neurons to an acute DNA damaging insult. Indeed, the rapid reduction of Mcl-1 mRNA and protein levels are early events after DNA damage in neurons, and maintaining high Mcl-1 levels can protect neurons against death. Together, our results are the first to demonstrate the requirement of Mcl-1, an anti-apoptotic Bcl-2 family protein, for cortical neurogenesis and the survival of neurons after DNA damage.


Assuntos
Apoptose/fisiologia , Sistema Nervoso Central/embriologia , Dano ao DNA/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neurônios/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Fatores Etários , Animais , Apoptose/genética , Caspase 3/metabolismo , Diferenciação Celular , Movimento Celular/genética , Movimento Celular/fisiologia , Proliferação de Células , Células Cultivadas , Sistema Nervoso Central/citologia , Dano ao DNA/genética , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Transgênicos , Mutação/genética , Proteína de Sequência 1 de Leucemia de Células Mieloides , Proteínas do Tecido Nervoso/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Células-Tronco/fisiologia , Transfecção/métodos , Tubulina (Proteína)/metabolismo
20.
Cell Rep ; 25(6): 1404-1414.e6, 2018 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-30403997

RESUMO

Mutations of the transcriptional regulator PHF6 cause the X-linked intellectual disability disorder Börjeson-Forssman-Lehmann syndrome (BFLS), but the pathogenesis of BFLS remains poorly understood. Here, we report a mouse model of BFLS, generated using a CRISPR-Cas9 approach, in which cysteine 99 within the PHD domain of PHF6 is replaced with phenylalanine (C99F). Mice harboring the patient-specific C99F mutation display deficits in cognitive functions, emotionality, and social behavior, as well as reduced threshold to seizures. Electrophysiological studies reveal that the intrinsic excitability of entorhinal cortical stellate neurons is increased in PHF6 C99F mice. Transcriptomic analysis of the cerebral cortex in C99F knockin mice and PHF6 knockout mice show that PHF6 promotes the expression of neurogenic genes and represses synaptic genes. PHF6-regulated genes are also overrepresented in gene signatures and modules that are deregulated in neurodevelopmental disorders of cognition. Our findings advance our understanding of the mechanisms underlying BFLS pathogenesis.


Assuntos
Epilepsia/patologia , Face/anormalidades , Dedos/anormalidades , Transtornos do Crescimento/patologia , Hipogonadismo/patologia , Deficiência Intelectual Ligada ao Cromossomo X/patologia , Obesidade/patologia , Animais , Sequência de Bases , Encéfalo/patologia , Proteínas de Transporte/genética , Cognição , Modelos Animais de Doenças , Suscetibilidade a Doenças , Emoções , Epilepsia/genética , Face/patologia , Dedos/patologia , Regulação da Expressão Gênica , Transtornos do Crescimento/genética , Hipogonadismo/genética , Relações Interpessoais , Masculino , Deficiência Intelectual Ligada ao Cromossomo X/genética , Camundongos , Camundongos Mutantes , Neurônios/metabolismo , Neurônios/patologia , Obesidade/genética , Proteínas Repressoras , Convulsões/patologia , Sinapses/metabolismo , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA