Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Cell ; 184(5): 1245-1261.e21, 2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33636132

RESUMO

How early events in effector T cell (TEFF) subsets tune memory T cell (TMEM) responses remains incompletely understood. Here, we systematically investigated metabolic factors in fate determination of TEFF and TMEM cells using in vivo pooled CRISPR screening, focusing on negative regulators of TMEM responses. We found that amino acid transporters Slc7a1 and Slc38a2 dampened the magnitude of TMEM differentiation, in part through modulating mTORC1 signaling. By integrating genetic and systems approaches, we identified cellular and metabolic heterogeneity among TEFF cells, with terminal effector differentiation associated with establishment of metabolic quiescence and exit from the cell cycle. Importantly, Pofut1 (protein-O-fucosyltransferase-1) linked GDP-fucose availability to downstream Notch-Rbpj signaling, and perturbation of this nutrient signaling axis blocked terminal effector differentiation but drove context-dependent TEFF proliferation and TMEM development. Our study establishes that nutrient uptake and signaling are key determinants of T cell fate and shape the quantity and quality of TMEM responses.


Assuntos
Aminoácidos/metabolismo , Linfócitos T CD8-Positivos/citologia , Memória Imunológica , Transdução de Sinais , Sistemas de Transporte de Aminoácidos/metabolismo , Animais , Linfócitos T CD8-Positivos/imunologia , Sistemas CRISPR-Cas , Ciclo Celular , Diferenciação Celular , Modelos Animais de Doenças , Feminino , Técnicas de Introdução de Genes , Coriomeningite Linfocítica/imunologia , Masculino , Camundongos , Camundongos Transgênicos , Células Precursoras de Linfócitos T/citologia
2.
Genes Dev ; 35(5-6): 335-353, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33602870

RESUMO

mSWI/SNF or BAF chromatin regulatory complexes are dosage-sensitive regulators of human neural development frequently mutated in autism spectrum disorders and intellectual disability. Cell cycle exit and differentiation of neural stem/progenitor cells is accompanied by BAF subunit switching to generate neuron-specific nBAF complexes. We manipulated the timing of BAF subunit exchange in vivo and found that early loss of the npBAF subunit BAF53a stalls the cell cycle to disrupt neurogenesis. Loss of BAF53a results in decreased chromatin accessibility at specific neural transcription factor binding sites, including the pioneer factors SOX2 and ASCL1, due to Polycomb accumulation. This results in repression of cell cycle genes, thereby blocking cell cycle progression and differentiation. Cell cycle block upon Baf53a deletion could be rescued by premature expression of the nBAF subunit BAF53b but not by other major drivers of proliferation or differentiation. WNT, EGF, bFGF, SOX2, c-MYC, or PAX6 all fail to maintain proliferation in the absence of BAF53a, highlighting a novel mechanism underlying neural progenitor cell cycle exit in the continued presence of extrinsic proliferative cues.


Assuntos
Actinas/metabolismo , Ciclo Celular/genética , Córtex Cerebelar/embriologia , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Actinas/genética , Animais , Sítios de Ligação/genética , Células Cultivadas , Cromatina/genética , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/genética , Embrião de Mamíferos , Deleção de Genes , Genes cdc/genética , Camundongos , Neurogênese/genética , Proteínas do Grupo Polycomb/metabolismo , Fatores de Transcrição/metabolismo
3.
Proc Natl Acad Sci U S A ; 118(7)2021 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-33558234

RESUMO

Using a gain-of-function screen in Drosophila, we identified the Krüppel-like factor Cabut (Cbt) as a positive regulator of cell cycle gene expression and cell proliferation. Enforced cbt expression is sufficient to induce an extra cell division in the differentiating fly wing or eye, and also promotes intestinal stem cell divisions in the adult gut. Although inappropriate cell proliferation also results from forced expression of the E2f1 transcription factor or its target, Cyclin E, Cbt does not increase E2F1 or Cyclin E activity. Instead, Cbt regulates a large set of E2F1 target genes independently of E2F1, and our data suggest that Cbt acts via distinct binding sites in target gene promoters. Although Cbt was not required for cell proliferation during wing or eye development, Cbt is required for normal intestinal stem cell divisions in the midgut, which expresses E2F1 at relatively low levels. The E2F1-like functions of Cbt identify a distinct mechanism for cell cycle regulation that may be important in certain normal cell cycles, or in cells that cycle inappropriately, such as cancer cells.


Assuntos
Ciclo Celular/genética , Proteínas de Drosophila/metabolismo , Fatores de Transcrição/metabolismo , Animais , Olho Composto de Artrópodes/citologia , Olho Composto de Artrópodes/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster , Fator de Transcrição E2F1/genética , Fator de Transcrição E2F1/metabolismo , Mutação com Ganho de Função , Teste de Complementação Genética , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Fatores de Transcrição/genética
4.
Int J Mol Sci ; 25(17)2024 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-39273443

RESUMO

Vascular smooth muscle cells (SMCs) can transition between a quiescent contractile or "differentiated" phenotype and a "proliferative-dedifferentiated" phenotype in response to environmental cues, similar to what in occurs in the wound healing process observed in fibroblasts. When dysregulated, these processes contribute to the development of various lung and cardiovascular diseases such as Chronic Obstructive Pulmonary Disease (COPD). Long non-coding RNAs (lncRNAs) have emerged as key modulators of SMC differentiation and phenotypic changes. In this study, we examined the expression of lncRNAs in primary human pulmonary artery SMCs (hPASMCs) during cell-to-cell contact-induced SMC differentiation. We discovered a novel lncRNA, which we named Differentiation And Growth Arrest-Related lncRNA (DAGAR) that was significantly upregulated in the quiescent phenotype with respect to proliferative SMCs and in cell-cycle-arrested MRC5 lung fibroblasts. We demonstrated that DAGAR expression is essential for SMC quiescence and its knockdown hinders SMC differentiation. The treatment of quiescent SMCs with the pro-inflammatory cytokine Tumor Necrosis Factor (TNF), a known inducer of SMC dedifferentiation and proliferation, elicited DAGAR downregulation. Consistent with this, we observed diminished DAGAR expression in pulmonary arteries from COPD patients compared to non-smoker controls. Through pulldown experiments followed by mass spectrometry analysis, we identified several proteins that interact with DAGAR that are related to cell differentiation, the cell cycle, cytoskeleton organization, iron metabolism, and the N-6-Methyladenosine (m6A) machinery. In conclusion, our findings highlight DAGAR as a novel lncRNA that plays a crucial role in the regulation of cell proliferation and SMC differentiation. This paper underscores the potential significance of DAGAR in SMC and fibroblast physiology in health and disease.


Assuntos
Diferenciação Celular , Proliferação de Células , Fibroblastos , Miócitos de Músculo Liso , RNA Longo não Codificante , Humanos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Fibroblastos/metabolismo , Diferenciação Celular/genética , Miócitos de Músculo Liso/metabolismo , Proliferação de Células/genética , Artéria Pulmonar/metabolismo , Artéria Pulmonar/citologia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/citologia , Doença Pulmonar Obstrutiva Crônica/metabolismo , Doença Pulmonar Obstrutiva Crônica/genética , Doença Pulmonar Obstrutiva Crônica/patologia , Células Cultivadas
5.
New Phytol ; 237(5): 1652-1666, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36451535

RESUMO

The processes that contribute to plant organ morphogenesis are spatial-temporally organized. Within the meristem, mitosis produces new cells that subsequently engage in cell expansion and differentiation programs. The latter is frequently accompanied by endoreplication, being an alternative cell cycle that replicates the DNA without nuclear division, causing a stepwise increase in somatic ploidy. Here, we show that the Arabidopsis SCL28 transcription factor promotes organ growth by modulating cell expansion dynamics in both root and leaf cells. Gene expression studies indicated that SCL28 regulates members of the SIAMESE/SIAMESE-RELATED (SIM/SMR) family, encoding cyclin-dependent kinase inhibitors with a role in promoting mitotic cell cycle (MCC) exit and endoreplication, both in response to developmental and environmental cues. Consistent with this role, mutants in SCL28 displayed reduced endoreplication, both in roots and leaves. We also found evidence indicating that SCL28 co-expresses with and regulates genes related to the biogenesis, assembly, and remodeling of the cytoskeleton and cell wall. Our results suggest that SCL28 controls, not only cell proliferation as reported previously but also cell expansion and differentiation by promoting MCC exit and endoreplication and by modulating aspects of the biogenesis, assembly, and remodeling of the cytoskeleton and cell wall.


Assuntos
Proteínas de Arabidopsis , Arabidopsis , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Endorreduplicação , Regulação da Expressão Gênica de Plantas , Mitose
6.
Neurochem Res ; 48(3): 996-1008, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36436172

RESUMO

The orphan receptor, G protein-coupled receptor 137 (GPR137), is an integral membrane protein involved in several types of cancer. GPR137 is expressed ubiquitously, including in the central nervous system (CNS). We established a GPR137 knockout (KO) neuro2A cell line to analyze GPR137 function in neuronal cells. KO cells were generated by genome editing using clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 and cultured as single cells by limited dilution. Rescue cells were then constructed to re-express GPR137 in GPR137 KO neuro2A cells using an expression vector with an EF1-alpha promoter. GPR137 KO cells increased cellular proliferation and decreased neurite outgrowth (i.e., a lower level of neuronal differentiation). Furthermore, GPR137 KO cells exhibited increased expression of a cell cycle regulator, cyclin D1, and decreased expression of a neuronal differentiation marker, NeuroD1. Additionally, GPR137 KO cells exhibited lower expression levels of the neurite outgrowth markers STAT3 and GAP43. These phenotypes were all abrogated in the rescue cells. In conclusion, GPR137 deletion increased cellular proliferation and decreased neuronal differentiation, suggesting that GPR137 promotes cell cycle exit and neuronal differentiation in neuro2A cells. Regulation of neuronal differentiation by GPR137 could be vital to constructing neuronal structure during brain development.


Assuntos
Diferenciação Celular , Receptores Acoplados a Proteínas G , Animais , Camundongos , Ciclo Celular , Diferenciação Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Sistemas CRISPR-Cas , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
7.
Genes Dev ; 29(3): 322-36, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25644606

RESUMO

Dicentric chromosomes are unstable products of erroneous DNA repair events that can lead to further genome rearrangements and extended gene copy number variations. During mitosis, they form anaphase bridges, resulting in chromosome breakage by an unknown mechanism. In budding yeast, dicentrics generated by telomere fusion break at the fusion, a process that restores the parental karyotype and protects cells from rare accidental telomere fusion. Here, we observed that dicentrics lacking telomere fusion preferentially break within a 25- to 30-kb-long region next to the centromeres. In all cases, dicentric breakage requires anaphase exit, ruling out stretching by the elongated mitotic spindle as the cause of breakage. Instead, breakage requires cytokinesis. In the presence of dicentrics, the cytokinetic septa pinch the nucleus, suggesting that dicentrics are severed after actomyosin ring contraction. At this time, centromeres and spindle pole bodies relocate to the bud neck, explaining how cytokinesis can sever dicentrics near centromeres.


Assuntos
Centrômero/genética , Quebra Cromossômica , Cromossomos Fúngicos/genética , Citocinese , Saccharomyces cerevisiae/genética , Telômero/metabolismo , Divisão do Núcleo Celular , Mitose
8.
Int J Mol Sci ; 24(2)2023 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-36675024

RESUMO

Cell division regulators play a vital role in neural progenitor cell (NPC) proliferation and differentiation. Cell division cycle 25C (CDC25C) is a member of the CDC25 family of phosphatases which positively regulate cell division by activating cyclin-dependent protein kinases (CDKs). However, mice with the Cdc25c gene knocked out were shown to be viable and lacked the apparent phenotype due to genetic compensation by Cdc25a and/or Cdc25b. Here, we investigate the function of Cdc25c in developing rat brains by knocking down Cdc25c in NPCs using in utero electroporation. Our results indicate that Cdc25c plays an essential role in maintaining the proliferative state of NPCs during cortical development. The knockdown of Cdc25c causes early cell cycle exit and the premature differentiation of NPCs. Our study uncovers a novel role of CDC25C in NPC division and cell fate determination. In addition, our study presents a functional approach to studying the role of genes, which elicit genetic compensation with knockout, in cortical neurogenesis by knocking down in vivo.


Assuntos
Proteínas de Ciclo Celular , Células-Tronco Neurais , Neurogênese , Fosfatases cdc25 , Animais , Ratos , Fosfatases cdc25/genética , Fosfatases cdc25/metabolismo , Ciclo Celular , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/genética , Quinases Ciclina-Dependentes/metabolismo , Regulação para Baixo/genética , Neurogênese/genética , Neurogênese/fisiologia , Células-Tronco Neurais/metabolismo
9.
Development ; 144(19): 3454-3464, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28851709

RESUMO

The termination of the proliferation of Drosophila neural stem cells, also known as neuroblasts (NBs), requires a 'decommissioning' phase that is controlled in a lineage-specific manner. Most NBs, with the exception of those of the mushroom body (MB), are decommissioned by the ecdysone receptor and mediator complex, causing them to shrink during metamorphosis, followed by nuclear accumulation of Prospero and cell cycle exit. Here, we demonstrate that the levels of Imp and Syp RNA-binding proteins regulate NB decommissioning. Descending Imp and ascending Syp expression have been shown to regulate neuronal temporal fate. We show that Imp levels decline slower in the MB than in other central brain NBs. MB NBs continue to express Imp into pupation, and the presence of Imp prevents decommissioning partly by inhibiting the mediator complex. Late-larval induction of transgenic Imp prevents many non-MB NBs from decommissioning in early pupae. Moreover, the presence of abundant Syp in aged NBs permits Prospero accumulation that, in turn, promotes cell cycle exit. Together, our results reveal that progeny temporal fate and progenitor decommissioning are co-regulated in protracted neuronal lineages.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Células-Tronco Neurais/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Animais Geneticamente Modificados , Núcleo Celular/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Larva/metabolismo , Complexo Mediador/metabolismo , Modelos Biológicos , Corpos Pedunculados/citologia , Corpos Pedunculados/metabolismo , Células-Tronco Neurais/citologia , Ligação Proteica , Pupa/metabolismo , Proteínas de Ligação a RNA/genética
10.
Int J Mol Sci ; 19(5)2018 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-29734659

RESUMO

Direct reprogramming of fibroblasts into induced cardiomyocytes (iCMs) holds a great promise for regenerative medicine and has been studied in several major directions. However, cell-cycle regulation, a fundamental biological process, has not been investigated during iCM-reprogramming. Here, our time-lapse imaging on iCMs, reprogrammed by Gata4, Mef2c, and Tbx5 (GMT) monocistronic retroviruses, revealed that iCM-reprogramming was majorly initiated at late-G1- or S-phase and nearly half of GMT-reprogrammed iCMs divided soon after reprogramming. iCMs exited cell cycle along the process of reprogramming with decreased percentage of 5-ethynyl-20-deoxyuridine (EdU)⁺/α-myosin heavy chain (αMHC)-GFP⁺ cells. S-phase synchronization post-GMT-infection could enhance cell-cycle exit of reprogrammed iCMs and yield more GFPhigh iCMs, which achieved an advanced reprogramming with more expression of cardiac genes than GFPlow cells. However, S-phase synchronization did not enhance the reprogramming with a polycistronic-viral vector, in which cell-cycle exit had been accelerated. In conclusion, post-infection synchronization of S-phase facilitated the early progression of GMT-reprogramming through a mechanism of enhanced cell-cycle exit.


Assuntos
Pontos de Checagem do Ciclo Celular/genética , Diferenciação Celular/genética , Reprogramação Celular/genética , Miócitos Cardíacos/citologia , Animais , Ciclo Celular/genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Camundongos , Miócitos Cardíacos/metabolismo , Medicina Regenerativa/tendências
11.
Genesis ; 55(4)2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28196404

RESUMO

p57Kip2 (p57) is a maternally expressed imprinted gene regulating growth arrest which belongs to the CIP/KIP family of cyclin-dependent kinase inhibitors. While initially identified as a cell cycle arrest protein through inhibition of cyclin and cyclin-dependent kinase complexes, p57 activity has also been linked to differentiation, apoptosis, and senescence. In addition, p57 has recently been shown to be involved in tumorigenesis and cell fate decisions in stem cells. Yet, p57 function in adult tissues remains poorly characterized due to the perinatal lethality of p57 knock-out mice. To analyze p57 tissue-specific activity, we generated a conditional mouse line (p57FL-ILZ/+ ) by flanking the coding exons 2-3 by LoxP sites. To track p57-expressing or mutant cells, the p57FL-ILZ allele also contains an IRES-linked ß-galactosidase reporter inserted in the 3' UTR of the gene. Here, we show that the ß-galactosidase reporter expression pattern recapitulates p57 tissue specificity during development and in postnatal mice. Furthermore, we crossed the p57FL-ILZ/+ mice with PGK-Cre mice to generate p57cKO-ILZ/+ animals with ubiquitous loss of p57. p57cKO-ILZ/+ mice display developmental phenotypes analogous to previously described p57 knock-outs. Thus, p57FL-ILZ/+ is a new genetic tool allowing expression and functional conditional analyses of p57.


Assuntos
Inibidor de Quinase Dependente de Ciclina p57/genética , Marcação de Genes/métodos , Mutação , Alelos , Animais , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p57/metabolismo , Embrião de Mamíferos/metabolismo , Feminino , Genes Reporter , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Fenótipo , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
12.
Biochem Biophys Res Commun ; 466(2): 147-54, 2015 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-26363457

RESUMO

Mammalian cardiomyocytes actively proliferate during embryonic stages, following which they exit their cell cycle after birth, and the exit is maintained. Previously, we showed that two inhibitory systems (the G1-phase inhibitory system: repression of cyclin D1 expression; the M-phase inhibitory system: inhibition of CDK1 activation) maintain the cell cycle exit of mouse adult cardiomyocytes. We also showed that two CDK inhibitors (CKIs), p21(Cip1) and p27(Kip1), regulate the cell cycle exit in a portion of postnatal cardiomyocytes. It remains unknown whether the two inhibitory systems are involved in the cell cycle exit of postnatal cardiomyocytes and whether p21(Cip1) and p27(Kip1) also inhibit entry to M-phase. Here, we showed that more than 40% of cardiomyocytes entered an additional cell cycle by induction of cyclin D1 expression at postnatal stages, but M-phase entry was inhibited in the majority of cardiomyocytes. Marked cell cycle progression and endoreplication were observed in cardiomyocytes of p21(Cip1) knockout mice at 4 weeks of age. In addition, tri- and tetranucleated cardiomyocytes increased significantly in p21(Cip1) knockout mice. These data showed that the G1-phase inhibitory system and two CKIs (p21(Cip1) and p27(Kip1)) inhibit entry to an additional cell cycle in postnatal cardiomyocytes, and that the M-phase inhibitory system and p21(Cip1) inhibit M-phase entry of cardiomyocytes which have entered the additional cell cycle.


Assuntos
Ciclo Celular , Quinases Ciclina-Dependentes/antagonistas & inibidores , Miócitos Cardíacos/citologia , Inibidores de Proteínas Quinases/farmacologia , Animais , Proliferação de Células , Ciclina D1/biossíntese , Inibidor de Quinase Dependente de Ciclina p21/genética , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout
13.
Biochem Biophys Res Commun ; 443(3): 1105-9, 2014 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-24380855

RESUMO

Mammalian cardiomyocytes actively proliferate during embryonic stages, following which cardiomyocytes exit their cell cycle after birth. The irreversible cell cycle exit inhibits cardiac regeneration by the proliferation of pre-existing cardiomyocytes. Exactly how the cell cycle exit occurs remains largely unknown. Previously, we showed that cyclin E- and cyclin A-CDK activities are inhibited before the CDKs levels decrease in postnatal stages. This result suggests that factors such as CDK inhibitors (CKIs) inhibit CDK activities, and contribute to the cell cycle exit. In the present study, we focused on a Cip/Kip family, which can inhibit cyclin E- and cyclin A-CDK activities. Expression of p21(Cip1) and p27(Kip1) but not p57(Kip2) showed a peak around postnatal day 5, when cyclin E- and cyclin A-CDK activities start to decrease. p21(Cip1) and p27(Kip1) bound to cyclin E, cyclin A and CDK2 at postnatal stages. Cell cycle distribution patterns of postnatal cardiomyocytes in p21(Cip1) and p27(Kip1) knockout mice showed failure in the cell cycle exit at G1-phase, and endoreplication. These results indicate that p21(Cip1) and p27(Kip) play important roles in the cell cycle exit of postnatal cardiomyocytes.


Assuntos
Ciclo Celular , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Mamíferos/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Animais , Regulação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL
14.
Methods Cell Biol ; 182: 187-197, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38359976

RESUMO

Replication stress risks genomic integrity. Depending on the level, replication stress can lead to slower progression through S phase and entry into G2 phase with DNA damage. In G2 phase, cells either recover and eventually enter mitosis or permanently withdraw from the cell cycle. Here we describe a method to detect cell cycle distribution, replication stress and cell cycle exit from G2 phase using fluorescence microscopy. We provide a script to automate the analysis using ImageJ. The focus has been to make a script and setup that is accessible to people without extensive computer knowledge.


Assuntos
Fase G2 , Mitose , Humanos , Ciclo Celular/genética , Dano ao DNA , Microscopia de Fluorescência , Replicação do DNA
15.
Front Pharmacol ; 14: 1242109, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37795025

RESUMO

Introduction: Methamphetamine (METH) abuse by pregnant drug addicts causes toxic effects on fetal neurodevelopment; however, the mechanism underlying such effect of METH is poorly understood. Methods: In the present study, we applied three-dimensional (3D) neurospheres derived from the embryonic rat hippocampal tissue to investigate the effect of METH on neurodevelopment. Through the combination of whole genome transcriptional analyses, the involved cell signalings were identified and investigated. Results: We found that METH treatment for 24 h significantly and concentration-dependently reduced the size of neurospheres. Analyses of genome-wide transcriptomic profiles found that those down-regulated differentially expressed genes (DEGs) upon METH exposure were remarkably enriched in the cell cycle progression. By measuring the cell cycle and the expression of cell cycle-related checkpoint proteins, we found that METH exposure significantly elevated the percentage of G0/G1 phase and decreased the levels of the proteins involved in the G1/S transition, indicating G0/G1 cell cycle arrest. Furthermore, during the early neurodevelopment stage of neurospheres, METH caused aberrant cell differentiation both in the neurons and astrocytes, and attenuated migration ability of neurospheres accompanied by increased oxidative stress and apoptosis. Conclusion: Our findings reveal that METH induces an aberrant cell cycle arrest and neuronal differentiation, impairing the coordination of migration and differentiation of neurospheres.

16.
Brain Res ; 1807: 148317, 2023 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-36898477

RESUMO

To analyze the role of syndecan-3 (SDC3), a heparan sulfate proteoglycan, in cerebellum development, we examined the effect of SDC3 on the transition from cell cycle exit to the initial differentiation stage of cerebellar granule cell precursors (CGCPs). First, we examined SDC3 localization in the developing cerebellum. SDC3 was mainly localized to the inner external granule layer where the transition from the cell cycle exit to the initial differentiation of CGCPs occurs. To examine how SDC3 regulates the cell cycle exit of CGCPs, we performed SDC3-knockdown (SDC3-KD) and -overexpression (Myc-SDC3) assays using primary CGCPs. SDC3-KD significantly increased the ratio of p27Kip1+ cells to total cells at day 3 in vitro (DIV3) and 4, but Myc-SDC3 reduced that at DIV3. Regarding the cell cycle exit efficiency using 24 h-labelled bromodeoxyuridine (BrdU) and a marker of cell cycling, Ki67, SDC3-KD significantly increased cell cycle exit efficiency (Ki67-; BrdU+ cells/BrdU+ cells) in primary CGCP at DIV4 and 5, but Myc-SDC3 reduced that at DIV4 and 5. However, SDC3-KD and Myc-SDC3 did not affect the efficiency of the final differentiation from CGCPs to granule cells at DIV3-5. Furthermore, the ratio of CGCPs in the cell cycle exiting stage to total cells, identified by initial differentiation markers TAG1 and Ki67 (TAG1+; Ki67+ cells), was considerably decreased by SDC3-KD at DIV4, but increased by Myc-SDC3 at DIV4 and 5. Altogether, these results indicate that SDC3 regulates the timing of the transition from the cell cycle exit stage to the initial differentiation stage of CGCP.


Assuntos
Cerebelo , Camundongos , Animais , Bromodesoxiuridina/metabolismo , Antígeno Ki-67/metabolismo , Sindecana-3/metabolismo , Cerebelo/metabolismo , Diferenciação Celular , Ciclo Celular/fisiologia
17.
Methods Mol Biol ; 2583: 13-23, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36418722

RESUMO

Cell cycle progression is a vital aspect of neural development. Repeated cell division in neural progenitor populations amplifies the numbers of specific cell types and is required to prevent growth failure that manifests as microcephaly. Regulated cycling is also required for cell fate specification. Analysis of cell cycle states is a valuable tool to understand the mechanisms underlying brain growth. Here we describe the preparation of cells for immunofluorescent-stained samples and flow cytometry and how to analyze cell cycle progression and cell cycle exit in progenitors. We describe methods as applied to analysis of cerebellar granule neuron progenitors (CGNPs), but similar methods in brain sections can also be applied to other brain neural progenitor populations, such as the hippocampus and subventricular zone.


Assuntos
Microcefalia , Humanos , Citometria de Fluxo , Coloração e Rotulagem , Interneurônios , Divisão Celular
18.
Methods Cell Biol ; 175: 235-249, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36967143

RESUMO

Joubert syndrome (JS) is an autosomal recessive ciliopathy that mainly affects the morphogenesis of the cerebellum and brain stem. To date, mutations in at least 39 genes have been identified in JS; all these gene-encoding proteins are involved in the biogenesis of the primary cilium and centrioles. Recent studies using the mouse model carrying deleted or mutated JS-related genes exhibited cerebellar hypoplasia with a reduction in neurogenesis; however, investigating specific neuronal behaviors during their development in vivo remains challenging. Here, we describe an in vivo cerebellar electroporation technique that can be used to deliver plasmids carrying GFP and/or shRNAs into the major cerebellar cell type, granule neurons, from their progenitor state to their maturation in a spatiotemporal-specific manner. By combining this method with cerebellar immunostaining and EdU incorporation, these approaches enable the investigation of the cell-autonomous effect of JS-related genes in granule neuron progenitors, including the pathogenesis of ectopic neurons and the defects in neuronal differentiation. This approach provides information toward understanding the multifaceted roles of JS-related genes during cerebellar development in vivo.


Assuntos
Anormalidades Múltiplas , Anormalidades do Olho , Doenças Renais Císticas , Camundongos , Animais , Cerebelo/metabolismo , Cerebelo/patologia , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/metabolismo , Anormalidades Múltiplas/patologia , Doenças Renais Císticas/genética , Doenças Renais Císticas/metabolismo , Doenças Renais Císticas/patologia , Anormalidades do Olho/genética , Anormalidades do Olho/metabolismo , Anormalidades do Olho/patologia , Retina , Neurônios/metabolismo , Diferenciação Celular/genética , Proteínas , Proliferação de Células/genética , Eletroporação
19.
Front Physiol ; 13: 796190, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35153829

RESUMO

Myoblast differentiation is an ordered multistep process that includes withdrawal from the cell cycle, elongation, and fusion to form multinucleated myotubes. Id3, a member of the Id family, plays a crucial role in cell cycle exit and differentiation. However, in muscle cells after differentiation induction, the detailed mechanisms that diminish Id3 function and cause the cells to withdraw from the cell cycle are unknown. Induction of myoblast differentiation resulted in decreased expression of Id3 and increased expression of XBP1u, and XBP1u accelerated proteasomal degradation of Id3 in C2C12 cells. The expression levels of the cyclin-dependent kinase inhibitors p21, p27, and p57 were not increased after differentiation induction of XBP1-knockdown C2C12 cells. Moreover, knockdown of Id3 rescued myogenic differentiation of XBP1-knockdown C2C12 cells. Taken together, these findings provide evidence that XBP1u regulates cell cycle exit after myogenic differentiation induction through interactions with Id3. To the best of our knowledge, this is the first report of the involvement of XBP1u in myoblast differentiation. These results indicate that XBP1u may act as a "regulator" of myoblast differentiation under various physiological conditions.

20.
Neural Regen Res ; 17(1): 130-136, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34100448

RESUMO

Our previous study showed that cell cycle exit and neuronal differentiation 1 (CEND1) may participate in neural stem cell cycle exit and oriented differentiation. However, whether CEND1-transfected neural stem cells can improve the prognosis of traumatic brain injury remained unclear. In this study, we performed quantitative proteomic analysis and found that after traumatic brain injury, CEND1 expression was downregulated in mouse brain tissue. Three days after traumatic brain injury, we transplanted CEND1-transfected neural stem cells into the area surrounding the injury site. We found that at 5 weeks after traumatic brain injury, transplantation of CEND1-transfected neural stem cells markedly alleviated brain atrophy and greatly improved neurological function. In vivo and in vitro results indicate that CEND1 overexpression inhibited the proliferation of neural stem cells, but significantly promoted their neuronal differentiation. Additionally, CEND1 overexpression reduced protein levels of Notch1 and cyclin D1, but increased levels of p21 in CEND1-transfected neural stem cells. Treatment with CEND1-transfected neural stem cells was superior to similar treatment without CEND1 transfection. These findings suggest that transplantation of CEND1-transfected neural stem cells is a promising cell therapy for traumatic brain injury. This study was approved by the Animal Ethics Committee of the School of Biomedical Engineering of Shanghai Jiao Tong University, China (approval No. 2016034) on November 25, 2016.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA