Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur J Neurosci ; 59(12): 3389-3402, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38663879

RESUMO

Neurons are post-mitotic cells, with microtubules playing crucial roles in axonal transport and growth. Kinesin family member 2c (KIF2C), a member of the Kinesin-13 family, possesses the ability to depolymerize microtubules and is involved in remodelling the microtubule lattice. Myocyte enhancer factor 2c (MEF2C) was initially identified as a regulator of muscle differentiation but has recently been associated with neurological abnormalities such as severe cognitive impairment, stereotyping, epilepsy and brain malformations when mutated or deleted. However, further investigation is required to determine which target genes MEF2C acts upon to influence neuronal function as a transcription regulator. Our data demonstrate that knockdown of both Mef2c and Kif2c significantly impacts spinal motor neuron development and behaviour in zebrafish. Luciferase reporter assays and chromosome immunoprecipitation assays, along with down/upregulated expression analysis, revealed that MFE2C functions as a novel transcription regulator for the Kif2c gene. Additionally, the knockdown of either Mef2c or Kif2c expression in E18 cortical neurons substantially reduces the number of primary neurites and axonal branches during neuronal development in vitro without affecting neurite length. Finally, depletion of Kif2c eliminated the effects of overexpression of Mef2c on the neurite branching. Based on these findings, we provided novel evidence demonstrating that MEF2C regulates the transcription of the Kif2c gene thereby influencing the axonal branching.


Assuntos
Axônios , Cinesinas , Fatores de Transcrição MEF2 , Peixe-Zebra , Animais , Fatores de Transcrição MEF2/metabolismo , Fatores de Transcrição MEF2/genética , Cinesinas/metabolismo , Cinesinas/genética , Axônios/metabolismo , Axônios/fisiologia , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Neurônios Motores/metabolismo , Neurogênese/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos
2.
Dev Biol ; 504: 49-57, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37741309

RESUMO

SAM and SH3 domain-containing 1 (SASH1), a member of the SLy protein family, is a tumor suppressor gene that has been studied for its association with various cancers. SASH1 is highly expressed in the mammalian central nervous system, particularly in glial cells, and is expressed in the central nervous system during zebrafish embryo development. However, SASH1's role in brain development has rarely been investigated. In this study, Morpholino oligonucleotides (MO) were used to down-regulate sash1a expression in zebrafish to observe morphological changes in the brain. Three transgenic zebrafish lines, Tg(gfap:eGFP), Tg(hb9:eGFP), and Tg(coro1a:eGFP) were selected to observe changes in glial cells, neurons, and immune cells after sash1a knockdown. Our results showed that the number of microglia residing in the developmental brain was reduced, whereas the axonal growth of caudal primary motor neurons was unaffected by sash1a downregulation. And more significantly, the gfap + glia presented abnormal arrangements and disordered orientations in sash1a morphants. The similar phenotype was verified in the mutation induced by the injection of cas9 mRNA and sash1a sgRNA. We further performed behavioral experiments in zebrafish larvae that had been injected with sash1a MO at one-cell stage, and found them exhibiting abnormal behavior trajectories. Moreover, injecting the human SASH1 mRNA rescued these phenomena in sash1a MO zebrafish. In summary, our study revealed that the downregulation of SASH1 leads to malformations in the embryonic brain and disorganization of glial cell marshalling, suggesting that SASH1 plays an important role in the migration of glial cells during embryonic brain development.


Assuntos
Proteínas Supressoras de Tumor , Peixe-Zebra , Animais , Humanos , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , RNA Guia de Sistemas CRISPR-Cas , Sistema Nervoso Central/metabolismo , Movimento Celular/genética , RNA Mensageiro , Mamíferos/metabolismo
3.
J Biol Chem ; 299(9): 105153, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37567476

RESUMO

Astrocyte activation and proliferation contribute to glial scar formation during spinal cord injury (SCI), which limits nerve regeneration. The long noncoding RNAs (lncRNAs) are involved in astrocyte proliferation and act as novel epigenetic regulators. Here, we found that lncRNA-LOC100909675 (LOC9675) expression promptly increased after SCI and that reducing its expression decreased the proliferation and migration of the cultured spinal astrocytes. Depletion of LOC9675 reduced astrocyte proliferation and facilitated axonal regrowth after SCI. LOC9675 mainly localized in astrocytic nuclei. We used RNA-seq to analyze gene expression profile alterations in LOC9675-depleted astrocytes and identified the cyclin-dependent kinase 1 (Cdk1) gene as a hub candidate. Our RNA pull-down and RNA immunoprecipitation assays showed that LOC9675 directly interacted with the transcriptional regulator CCCTC-binding factor (CTCF). Dual-luciferase reporter and chromatin immunoprecipitation assays, together with downregulated/upregulated expression investigation, revealed that CTCF is a novel regulator of the Cdk1 gene. Interestingly, we found that with the simultaneous overexpression of CTCF and LOC9675 in astrocytes, the Cdk1 transcript was restored to the normal level. We then designed the deletion construct of LOC9675 by removing its interacting region with CTCF and found this effect disappeared. A transcription inhibition assay using actinomycin D revealed that LOC9675 could stabilize Cdk1 mRNA, while LOC9675 depletion or binding with CTCF reduced Cdk1 mRNA stability. These data suggest that the cooperation between CTCF and LOC9675 regulates Cdk1 transcription at a steady level, thereby strictly controlling astrocyte proliferation. This study provides a novel perspective on the regulation of the Cdk1 gene transcript by lncRNA LOC9675.

4.
Neural Regen Res ; 18(12): 2727-2732, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37449637

RESUMO

Fidgetin, a microtubule-severing enzyme, regulates neurite outgrowth, axonal regeneration, and cell migration by trimming off the labile domain of microtubule polymers. Because maintenance of the microtubule labile domain is essential for axon initiation, elongation, and navigation, it is of interest to determine whether augmenting the microtubule labile domain via depletion of fidgetin serves as a therapeutic approach to promote axonal regrowth in spinal cord injury. In this study, we constructed rat models of spinal cord injury and sciatic nerve injury. Compared with spinal cord injury, we found that expression level of tyrosinated microtubules in the labile portion of microtubules continuously increased, whereas fidgetin decreased after peripheral nerve injury. Depletion of fidgetin enhanced axon regeneration after spinal cord injury, whereas expression level of end binding protein 3 (EB3) markedly increased. Next, we performed RNA interference to knockdown EB3 or fidgetin. We found that deletion of EB3 did not change fidgetin expression. Conversely, deletion of fidgetin markedly increased expression of tyrosinated microtubules and EB3. Deletion of fidgetin increased the amount of EB3 at the end of neurites and thereby increased the level of tyrosinated microtubules. Finally, we deleted EB3 and overexpressed fidgetin. We found that fidgetin trimmed tyrosinated tubulins by interacting with EB3. When fidgetin was deleted, the labile portion of microtubules was elongated, and as a result the length of axons and number of axon branches were increased. These findings suggest that fidgetin can be used as a novel therapeutic target to promote axonal regeneration after spinal cord injury. Furthermore, they reveal an innovative mechanism by which fidgetin preferentially severs labile microtubules.

5.
Metab Brain Dis ; 38(7): 2369-2381, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37256467

RESUMO

Neuropsychiatric disorders have a high incidence worldwide. Kinesins, a family of microtubule-based molecular motor proteins, play essential roles in intracellular and axonal transport. Variants of kinesins have been found to be related to many diseases, including neurodevelopmental/neurodegenerative disorders. Kinesin-12 (also known as Kif15) was previously found to affect the frequency of both directional microtubule transports. However, whether Kif15 deficiency impacts mood in mice is yet to be investigated. In this study, we used the CRISPR/Cas9 method to obtain Kif15-/- mice. In behavioral tests, Kif15-/- female mice exhibited prominent depressive characteristics. Further studies showed that the expression of BDNF was significantly decreased in the frontal cortex, corpus callosum, and hippocampus of Kif15-/- mice, along with the upregulation of Interleukin-6 and Interleukin-1ß in the corpus callosum. In addition, the expression patterns of AnkG were notably changed in the developing brain of Kif15-/- mice. Based on our previous studies, we suggested that this appearance of altered AnkG was due to the maladjustment of the microtubule patterns induced by Kif15 deficiency. The distribution of PSD95 in neurites notably decreased after cultured neurons treated with the Kif15 inhibitor, but total PSD95 protein level was not impacted, which revealed that Kif15 may contribute to PSD95 transportation. This study suggested that Kif15 may serve as a potential target for future depression studies.


Assuntos
Depressão , Cinesinas , Animais , Feminino , Camundongos , Depressão/genética , Cinesinas/genética , Cinesinas/metabolismo , Microtúbulos/metabolismo , Neurônios/metabolismo
6.
BMC Biol ; 21(1): 95, 2023 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-37095525

RESUMO

BACKGROUND: Natronobacterium gregoryi Argonaute (NgAgo) was found to reduce mRNA without generating detectable DNA double-strand breaks in a couple of endogenous genes in zebrafish, suggesting its potential as a tool for gene knockdown. However, little is known about how it interacts with nucleic acid molecules to interfere with gene expression. RESULTS: In this study, we first confirmed that coinjection of NgAgo and gDNA downregulated target genes, generated gene-specific phenotypes and verified some factors (including 5' phosphorylation, GC ratio, and target positions) of gDNAs affecting gene downregulation. Therein, the sense and antisense gDNAs were equally effective, suggesting that NgAgo possibly binds to DNA. NgAgo-VP64 with gDNAs targeting promoters upregulated the target genes, further providing evidence that NgAgo interacts with genomic DNA and controls gene transcription. Finally, we explain the downregulation of NgAgo/gDNA target genes by interference with the process of gene transcription, which differs from that of morpholino oligonucleotides. CONCLUSIONS: The present study provides conclusions that NgAgo may target genomic DNA and that target positions and the gDNA GC ratio influence its regulation efficiency.


Assuntos
Edição de Genes , Peixe-Zebra , Animais , Peixe-Zebra/genética , Natronobacterium/genética , Natronobacterium/metabolismo , DNA , Proteínas Argonautas/genética , Expressão Gênica
7.
Zebrafish ; 20(1): 10-18, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36795618

RESUMO

The swim bladder functions to maintain the fish balance at a certain position under water. Although the motoneuron-dependent swim-up behavior is important for swim bladder inflation, the underlying molecular mechanism remains largely unknown. We generated a sox2 KO zebrafish using TALEN and found that the posterior chamber of the swim bladder was uninflated. The tail flick and the swim-up behavior were absent in the mutant zebrafish embryos and the behavior could not be accomplished. As the tail flick behavior is absent, the mutant larvae therefore cannot reach the water surface to gulp air, ultimately leading to the uninflation of the swim bladder. To understand the mechanism underlying the swim-up defects, we crossed the sox2 null allele in the background of Tg(huc:eGFP) and Tg(hb9:GFP). The deficiency of sox2 in zebrafish resulted in abnormal motoneuron axons in the regions of trunk, tail, and swim bladder. To identify the downstream target gene of sox2 to control the motor neuron development, we performed RNA sequencing on the transcriber of mutant embryos versus wild type embryos and found that the axon guidance pathway was abnormal in the mutant embryos. RT-PCR demonstrated that the expression of sema3bl, ntn1b, and robo2 were decreased in the mutants.


Assuntos
Fatores de Transcrição SOX , Proteínas de Peixe-Zebra , Peixe-Zebra , Animais , Embrião não Mamífero/fisiologia , Organogênese , Bexiga Urinária , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , Locomoção , Fatores de Transcrição SOX/genética
8.
J Biol Chem ; 299(3): 103020, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36791914

RESUMO

Neural stem cells (NSCs) proliferation and differentiation rely on proper expression and posttranslational modification of transcription factors involved in the determination of cell fate. Further characterization is needed to connect modifying enzymes with their transcription factor substrates in the regulation of these processes. Here, we demonstrated that the inhibition of KAT2A, a histone acetyltransferase, leads to a phenotype of small eyes in the developing embryo of zebrafish, which is associated with enhanced proliferation and apoptosis of NSCs in zebrafish eyes. We confirmed that this phenotype is mediated by the elevated level of PAX6 protein. We further verified that KAT2A negatively regulates PAX6 at the protein level in cultured neural stem cells of rat cerebral cortex. We revealed that PAX6 is a novel acetylation substrate of KAT2A and the acetylation of PAX6 promotes its ubiquitination mediated by the E3 ligase RNF8 that facilitated PAX6 degradation. Our study proposes that KAT2A inhibition results in accelerated proliferation, delayed differentiation, or apoptosis, depending on the context of PAX6 dosage. Thus, the KAT2A/PAX6 axis plays an essential role to keep a balance between the self-renewal and differentiation of NSCs.


Assuntos
Células-Tronco Neurais , Peixe-Zebra , Animais , Ratos , Diferenciação Celular/fisiologia , Proliferação de Células , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Células-Tronco Neurais/metabolismo , Fator de Transcrição PAX6/genética , Fator de Transcrição PAX6/metabolismo , Fatores de Transcrição/metabolismo , Peixe-Zebra/metabolismo
9.
Exp Neurol ; 361: 114315, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36586551

RESUMO

Neurons require a constant increase in protein synthesis during axonal growth and regeneration. AKT-mTOR is a central pathway for mammalian cell survival and regeneration. Fidgetin (Fign) is an ATP-dependent microtubule (MT)-severing enzyme whose functions are associated with neurite outgrowth, axon regeneration and cell migration. Although most previous studies have indicated that depletion of Fign is involved in those biological activities by increasing labile MT mass, it remains unknown whether mTOR activation contributes to this process. Here, we showed that depletion of Fign enhanced p-mTOR/p-S6K activation, and the mTOR inhibitor Rapamycin inhibited axon outgrowth and p-rpS6 activation. We then investigated the effects of neuronal-specific Fign deletion in a rat spinal cord hemisection model by injecting syn-GFP Fign shRNA virus. BBB values revealed an improvement in functional recovery. The p-mTOR was activated along with neuronal Fign depletion. The syn-mCherry virus showed more sprouting neurites entering the injury region, which was confirmed by immunostaining GAP43 protein. Further, we showed that Fign siRNA treatment promoted axon outgrowth and branching, whose underlying mechanism was firstly attributed to local activation of the mTOR pathway, and increased MT dynamicity. Finally, considering L-leucine, promotes axonal growth and neuronal survival, we applied L-leucine with Fign depletion after spinal cord injury or in chondroitin sulfate proteoglycan inhibitory molecules. The phenomenon of synergistically augmented axon regeneration was observed. In summary, our results indicated a novel local mTOR pathway for fidgetin to impact axon growth and provided a combined strategy in SCI.


Assuntos
Axônios , Traumatismos da Medula Espinal , Ratos , Animais , Axônios/fisiologia , Regeneração Nervosa/fisiologia , Leucina/metabolismo , Leucina/farmacologia , Neurônios/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Mamíferos
10.
CNS Neurosci Ther ; 29(1): 228-238, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36286186

RESUMO

AIMS: This study aimed to evaluate the effects of the depletion of SAM and SH3 domain-containing protein 1 (SASH1) on functional recovery after spinal cord injury (SCI) and to investigate the possible mechanism of SASH1 knockdown in astrocytes facilitating axonal growth. METHODS: SCI model was established in adult rats. SASH1 small interfering RNA (siSASH1) was used to investigate its function. Hindlimb motor function was evaluated by the Basso-Bresnahan-Beattie (BBB) assay. The gene expressions were evaluated by the methods of qRT-PCR, Western-blotting, ELISA, and immunohistochemistry. RESULTS: SASH1 knockdown improved the BBB scores after SCI and significantly reduced GFAP expression. In cultured spinal astrocytes, siSASH1 treatment decreased interferon-γ release and increased brain-derived neurotrophic factor (BDNF) release. When cocultured with SASH1-knockdown astrocytes, axonal growth increased. The neuronal tropomyosin receptor kinase B (BDNF receptor) expression increased, especially in the axonal tips. SASH1 expression increased while NSCs differentiated into glial cells, instead of neurons. After SASH1 depletion, differentiated NSCs maintained a higher level of Nestin protein and an increase in BDNF release. CONCLUSIONS: These results indicate that SASH1 acts as an astrocytic differentiation-maintaining protein, and SASH1 downregulation limits glial activation and contributes toward functional recovery after SCI.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Astrócitos , Traumatismos da Medula Espinal , Animais , Ratos , Astrócitos/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Diferenciação Celular , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/fisiologia , RNA Interferente Pequeno/genética , Medula Espinal/metabolismo , Traumatismos da Medula Espinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética
11.
J Mol Cell Biol ; 14(5)2022 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-35704676

RESUMO

High-altitude cerebral edema (HACE) is a potentially fatal encephalopathy associated with a time-dependent exposure to the hypobaric hypoxia of altitude. The formation of HACE is affected by both vasogenic and cytotoxic edema. The over-activated microglia potentiate the damage of blood-brain barrier (BBB) and exacerbate cytotoxic edema. In light with the activation of microglia in HACE, we aimed to investigate whether the over-activated microglia were the key turning point of acute mountain sickness to HACE. In in vivo experiments, by exposing mice to hypobaric hypoxia (7000 m above sea level) to induce HACE model, we found that microglia were activated and migrated to blood vessels. Microglia depletion by PLX5622 obviously relieved brain edema. In in vitro experiments, we found that hypoxia induced cultured microglial activation, leading to the destruction of endothelial tight junction and astrocyte swelling. Up-regulated nuclear respiratory factor 1 (NRF1) accelerated pro-inflammatory factors through transcriptional regulation on nuclear factor kappa B p65 (NF-κB p65) and mitochondrial transcription factor A (TFAM) in activated microglia under hypoxia. NRF1 also up-regulated phagocytosis by transcriptional regulation on caveolin-1 (CAV-1) and adaptor-related protein complex 2 subunit beta (AP2B1). The present study reveals a new mechanism in HACE: hypoxia over-activates microglia through up-regulation of NRF1, which both induces inflammatory response through transcriptionally activating NF-κB p65 and TFAM, and enhances phagocytic function through up-regulation of CAV-1 and AP2B1; hypoxia-activated microglia destroy the integrity of BBB and release pro-inflammatory factors that eventually induce HACE.


Assuntos
Doença da Altitude , Edema Encefálico , Complexo 2 de Proteínas Adaptadoras/metabolismo , Altitude , Doença da Altitude/complicações , Animais , Edema Encefálico/complicações , Edema Encefálico/metabolismo , Caveolina 1/metabolismo , Hipóxia/complicações , Camundongos , Microglia/metabolismo , NF-kappa B/metabolismo , Fator 1 Nuclear Respiratório/metabolismo
12.
Artigo em Inglês | MEDLINE | ID: mdl-33069857

RESUMO

During development, highly dynamic reconstruction of microtubules is involved in many cellular processes, including cell division, migration, morphological changes, and material transportation within cells. Microtubule severing proteins (MSPs), with the function of cutting microtubules into short parts, are important regulators in the reconstruction of microtubule arrays. Fidgetin (fign) and its family members fidgetin like 1 (fignl1) and fignl2 are MSPs, and knowledge on the expression patterns of fign family members will benefit our understanding of their primary roles in one specific stage during development. In this study, we compared the evolutionary relationships of fign family members and found that fignl2 is closer to fign than fignl1. We utilized the zebrafish model and in situ hybridization (ISH) to parallelly identify the expression features of fign family members. Our findings revealed that before 12 h post fertilization (hpf), the expression patterns of fign and fignl1 and fignl2 genes were similar, but differences arose thereafter. Fignl2 transcripts were present in more tissues and organs of zebrafish after 12 hpf and potentially exhibited more ubiquitous functions. This study is the first to assess systematic comparable data on the expression patterns of fign family members during development.


Assuntos
Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Associadas aos Microtúbulos/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Sequência de Aminoácidos , Animais , Biologia Computacional , Proteínas Associadas aos Microtúbulos/química , Proteínas Associadas aos Microtúbulos/metabolismo
13.
Front Cell Dev Biol ; 8: 593234, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33585441

RESUMO

Previously, fidgetin (fign) and its family members fidgetin-like 1 (fignl1) and fidgetin-like 2 (fignl2) were found to be highly expressed during zebrafish brain development, suggesting their functions in the nervous system. In this study, we report the effects of loss-of-function of these genes on development. We designed and identified single-guide RNAs targeted to generate fign, fignl1, and fignl2 mutants and then observed the overall morphological and behavioral changes. Our findings showed that while fign and fignl1 null mutants displayed no significant defects, fignl2 null zebrafish mutants displayed pericardial edema, reduced heart rate, and smaller eyes; fignl2 null mutants responded to the light-darkness shift with a lower swimming velocity. fignl2 mRNAs were identified in vascular endothelial cells by in situ hybridization and re-analysis of an online dataset of single-cell RNAseq results. Finally, we used morpholino oligonucleotides to confirm that fignl2 knockdown resulted in severe heart edema, which was caused by abnormal vascular branching. The zebrafish fignl2 morphants also showed longer axonal length and more branches of caudal primary neurons. Taken together, we summarize that Fignl2 functions on cellular branches in endothelial cells and neurons. This study reported for the first time that the microtubule-severing protein Fignl2 contributes to cell branching during development.

14.
BMC Dev Biol ; 19(1): 25, 2019 12 30.
Artigo em Inglês | MEDLINE | ID: mdl-31884948

RESUMO

BACKGROUND: Rab proteins are GTPases responsible for intracellular vesicular trafficking regulation. Rab11 proteins, members of the Rab GTPase family, are known to regulate vesicular recycling during embryonic development. In zebrafish, there are 3 rab11 paralogues, known as rab11a, rab11ba and rab11bb, sharing high identity with each other. However, the expression analysis of rab11 is so far lacking. RESULTS: Here, by phylogeny analysis, we found the three rab11 genes are highly conserved especially for their GTPase domains. We examined the expression patterns of rab11a, rab11ba and rab11bb using RT-PCR and in situ hybridization. We found that all the three genes were highly enriched in the central nervous system, but in different areas of the brain. Apart from brain, rab11a was also expressed in caudal vein, pronephric duct, proctodeum, pharyngeal arches and digestive duct, rab11ba was detected to express in muscle, and rab11bb was expressed in kidney, fin and spinal cord. Different from rab11a and rab11ba, which both have maternal expressions in embryos, rab11bb only expresses during 24hpf to 96hpf. CONCLUSIONS: Our results suggest that rab11 genes play important but distinct roles in the development of the nervous system in zebrafish. The findings could provide new evidences for better understanding the functions of rab11 in the development of zebrafish embryos.


Assuntos
Peixe-Zebra/embriologia , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/metabolismo , Sequência Conservada , Feminino , Trato Gastrointestinal/embriologia , Trato Gastrointestinal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Herança Materna , Família Multigênica , Domínios Proteicos , Distribuição Tecidual , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/química , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo , Proteínas rab de Ligação ao GTP/química
15.
Cell Death Dis ; 10(6): 417, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-31138780

RESUMO

SAM and SH3 domain-containing 1 (SASH1), a scaffold protein, is regarded as a tumor suppressor. Recent studies have verified the decreased expression of SASH1 in many tumors. Our previous clinical investigation found that SASH1 was widely expressed in normal brain tissues but reduced or absent in glioma tissues. However, the functions of SASH1 in normal astrocytes and the reasons for the reductions in SASH1 levels in glioma tissues are unclear. In this study, we found that in astrocytes, SASH1 functions in cell adhesion. We observed that knockdown of SASH1 expression in cultured astrocytes significantly decreased cell adhesion and increased invasion. Conversely, overexpression of SASH1 in C6 cells markedly promoted cell adhesion and decreased cell invasion. In addition, we found that the expression level of one member of the integrin family, integrin ß8, was significantly reduced in SASH1-downregulated astrocytes and elevated in SASH1-upregulated C6 cells. Furthermore, the results of methylation and ChIP assays showed that the methylation level of the SASH1 gene was markedly higher in C6 cells than in astrocytes and that HMGB1 could bind to the CpG islands of the SASH1 gene. HMGB1 overexpression in astrocytes significantly increased the methylation level of the SASH1 gene. This study reveals, for the first time, that HMGB1 contributes to the methylation of the SASH1 gene, and our findings suggest that methylation downregulates the expression of the SASH1 gene and later reduces integrin ß8 expression, thereby reducing cell adhesion and promoting cell migration.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Astrócitos/metabolismo , Adesão Celular/genética , Glioma/metabolismo , Proteína HMGB1/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Ilhas de CpG/genética , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Ontologia Genética , Proteína HMGB1/genética , Cadeias beta de Integrinas/genética , Cadeias beta de Integrinas/metabolismo , Metilação , RNA-Seq , Ratos , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/genética
16.
Traffic ; 20(1): 71-81, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30411440

RESUMO

KIF15, the vertebrate kinesin-12, is best known as a mitotic motor protein, but continues to be expressed in neurons. Like KIF11 (the vertebrate kinesin-5), KIF15 interacts with microtubules in the axon to limit their sliding relative to one another. Unlike KIF11, KIF15 also regulates interactions between microtubules and actin filaments at sites of axonal branch formation and in growth cones. Our original work on these motors was done on cultured rat neurons, but we are now using zebrafish to extend these studies to an in vivo model. We previously studied kif15 in zebrafish by injecting splice-blocking morpholinos injected into embryos. Consistent with the cell culture work, these studies demonstrated that axons grow faster and longer when KIF15 levels are reduced. In the present study, we applied CRISPR/Cas9-based knockout technology to create kif15 mutants and labeled neurons with Tg(mnx1:GFP) transgene or transient expression of elavl3:EGFP-alpha tubulin. We then compared by live imaging the homozygotic, heterozygotic mutants to their wildtype siblings to ascertain the effects of depletion of kif15 during Caudal primary motor neuron and Rohon-Beard (R-B) sensory neuron development. The results showed, compared to the kif15 wildtype, the number of branches was reduced while axon outgrowth was accelerated in kif15 homozygotic and heterozygotic mutants. In R-B sensory neurons, after laser irradiation, injured axons with loss of kif15 displayed significantly greater regenerative velocity. Given these results and the fact that kif15 drugs are currently under development, we posit kif15 as a novel target for therapeutically augmenting regeneration of injured axons.


Assuntos
Cinesinas/genética , Mutação , Regeneração Nervosa , Crescimento Neuronal , Proteínas de Peixe-Zebra/genética , Animais , Sistemas CRISPR-Cas , Neurônios Motores/citologia , Neurônios Motores/metabolismo , Neurônios Motores/fisiologia , Peixe-Zebra
17.
J Biol Chem ; 294(8): 2732-2743, 2019 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-30593510

RESUMO

Zebrafish gata4/5/6 genes encode transcription factors that lie on the apex of the regulatory hierarchy in primitive myelopoiesis. However, little is known about the roles of microRNAs in gata4/5/6-regulated processes. Performing RNA-Seq deep sequencing analysis of the expression changes of microRNAs in gata4/5/6-knockdown embryos, we identified miR-210-5p as a regulator of zebrafish primitive myelopoiesis. Knocking down gata4/5/6 (generating gata5/6 morphants) significantly increased miR-210-5p expression, whereas gata4/5/6 overexpression greatly reduced its expression. Consistent with inhibited primitive myelopoiesis in the gata5/6 morphants, miR-210-5p overexpression repressed primitive myelopoiesis, indicated by reduced numbers of granulocytes and macrophages. Moreover, knocking out miR-210 partially rescued the defective primitive myelopoiesis in zebrafish gata4/5/6-knockdown embryos. Furthermore, we show that the restrictive role of miR-210-5p in zebrafish primitive myelopoiesis is due to impaired differentiation of hemangioblast into myeloid progenitor cells. By comparing the set of genes with reduced expression levels in the gata5/6 morphants to the predicted target genes of miR-210-5p, we found that foxj1b and slc3a2a, encoding a forkhead box transcription factor and a solute carrier family 3 protein, respectively, are two direct downstream targets of miR-210-5p that mediate its inhibitory roles in zebrafish primitive myelopoiesis. In summary, our results reveal that miR-210-5p has an important role in the genetic network controlling zebrafish primitive myelopoiesis.


Assuntos
Embrião não Mamífero/citologia , Regulação da Expressão Gênica no Desenvolvimento , Inativação Gênica , MicroRNAs/genética , Mielopoese , RNA Mensageiro/antagonistas & inibidores , Proteínas de Peixe-Zebra/antagonistas & inibidores , Peixe-Zebra/embriologia , Animais , Embrião não Mamífero/metabolismo , Fatores de Transcrição Forkhead/antagonistas & inibidores , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Cadeia Pesada da Proteína-1 Reguladora de Fusão/antagonistas & inibidores , Cadeia Pesada da Proteína-1 Reguladora de Fusão/genética , Cadeia Pesada da Proteína-1 Reguladora de Fusão/metabolismo , Fatores de Transcrição GATA/antagonistas & inibidores , Fatores de Transcrição GATA/genética , Fatores de Transcrição GATA/metabolismo , Fator de Transcrição GATA5/antagonistas & inibidores , Fator de Transcrição GATA5/genética , Fator de Transcrição GATA5/metabolismo , Redes Reguladoras de Genes , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
18.
J Mol Endocrinol ; 2018 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-30400066

RESUMO

Hypoxia-inducible factor-1 (HIF1) is a critical transcription factor involved in cell response to hypoxia. Under physiological conditions, its a subunit is rapidly degraded in most tissues except testes. HIF1 is stably expressed in Leydig cells, which are the main source of testosterone for male, and might bind to the promoter region of steroidogenic acute regulatory protein (Star), which is necessary for the testosterone synthesis, according to software analysis. This study aims to identify the binding sites of HIF1 on Star promoter and its transcriptional regulation of Star to affect testosterone synthesis. Testosterone level and steroid synthesis-related proteins were determined in male Balb/C mice exposed to hypoxia (8% O2). While HIF1 was upregulated, the testosterone level was significantly decreased. This was further confirmed by in vitro experiments with rat primary Leydig cells or TM3 cells exposed to hypoxia (1% O2), CoCl2 or DFX to raise HIF1. The decline of testosterone was reversed by pregnenolone but not cAMP, indicating the cholesterol transport disorder as the main cause. In agreement, StAR expression level was decreased in response to HIF1, while 3b-hydroxysteroid dehydrogenase, 17b-hydroxysteroid dehydrogenase and p450scc did not exhibit significant changes. By ChIP, EMSA supershift and dual-luciferase reporter assays, HIF1 was found to bind to the Star promoter region and repress the expression of StAR. Mutation assays identified three HIF1-binding sites on mouse Star promoter. These findings indicate that HIF1 represses Star transcription through directly binding to the Star promoter at -2082/-2078, -2064/-2060 and -1910/-1906, leading to the negative regulation of testosterone synthesis.

19.
Gene ; 668: 204-210, 2018 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-29783071

RESUMO

Calpain 3 (Capn3), a skeletal muscle-specific member of the calpain family, executes some non-proteolytic functions besides its role as a Ca2+-regulated proteolytic enzyme. Previously, we found that changes in Capn3 expression were linearly correlated with the degree of muscular atrophy following reversible sciatic nerve injury and that knockdown of Capn3 gene expression promoted myoblast differentiation. While the regulation of capn3 gene expression is interesting, transcriptional regulation of Capn3 is still unclear. In the present study, we provided experimental data showing that the myogenic enhancer factor 2A (MEF2A) regulated Capn3 gene expression. Firstly, the luciferase reporter assay and EMSA were performed and showed that ectopic expression of the Mef2a gene could bind to the predicted site of the Capn3 promoter region. Furthermore, in the L6 myoblast differentiation model in vitro, Capn3 gene expression was shown to be positively associated with the level of Mef2a by qRT-PCR, western-blotting, and immunocytochemistry. The Capn3 protein level decreased as MEF2A decreased when induced by Mef2a siRNA transfection in L6 myoblasts. Finally, the results of ChIP indicated that MEF2A occupied the promoter region of the Capn3 gene in rat denervated gastrocnemius muscle tissue. Based on these results, we proposed that MEF2A is a transcriptional regulator for Capn3 gene expression.


Assuntos
Calpaína/genética , Regulação da Expressão Gênica , Isoenzimas/genética , Proteínas Musculares/genética , Mioblastos Esqueléticos/metabolismo , Animais , Sítios de Ligação , Diferenciação Celular , Linhagem Celular , Células HEK293 , Humanos , Fatores de Transcrição MEF2/metabolismo , Músculo Esquelético/metabolismo , Mioblastos Esqueléticos/citologia , Regiões Promotoras Genéticas , Ratos
20.
Oncotarget ; 8(10): 16401-16413, 2017 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-28146428

RESUMO

Male fertility disorders play a key role in half of all infertility cases. Reduction in testosterone induced by hypoxia might cause diseases in reproductive system and other organs. Hypoxic exposure caused a significant decrease of NRF1. Software analysis reported that the promoter region of steroidogenic acute regulatory protein (StAR) contained NRF1 binding sites, indicating NRF1 promoted testicular steroidogenesis. The purpose of this study is to determine NRF1 is involved in testosterone synthesis; and under hypoxia, the decrease of testosterone synthesis is caused by lower expression of NRF1. We designed both in vivo and in vitro experiments. Under hypoxia, the expressions of NRF1 in Leydig cells and testosterone level were significantly decreased both in vivo and in vitro. Overexpression and interference NRF1 could induced StAR and testosterone increased and decreased respectively. ChIP results confirmed the binding of NRF1 to StAR promoter region. In conclusion, decline of NRF1 expression downregulated the level of StAR, which ultimately resulted in a reduction in testosterone synthesis.


Assuntos
Hipóxia/metabolismo , Células Intersticiais do Testículo/metabolismo , Fator 1 Nuclear Respiratório/metabolismo , Fosfoproteínas/biossíntese , Testosterona/biossíntese , Animais , Hipóxia Celular/fisiologia , Regulação para Baixo , Humanos , Hipóxia/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fator 1 Nuclear Respiratório/biossíntese , Análise de Sequência com Séries de Oligonucleotídeos , Fosfoproteínas/antagonistas & inibidores , Fosfoproteínas/genética , Regiões Promotoras Genéticas , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...