Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Dev Biol ; 504: 49-57, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37741309

RESUMEN

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.


Asunto(s)
Proteínas Supresoras de Tumor , Pez Cebra , Animales , Humanos , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , ARN Guía de Sistemas CRISPR-Cas , Sistema Nervioso Central/metabolismo , Movimiento Celular/genética , ARN Mensajero , Mamíferos/metabolismo
2.
Neural Regen Res ; 18(12): 2727-2732, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37449637

RESUMEN

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.

3.
BMC Biol ; 21(1): 95, 2023 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-37095525

RESUMEN

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.


Asunto(s)
Edición Génica , Pez Cebra , Animales , Pez Cebra/genética , Natronobacterium/genética , Natronobacterium/metabolismo , ADN , Proteínas Argonautas/genética , Expresión Génica
4.
J Biol Chem ; 299(3): 103020, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36791914

RESUMEN

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.


Asunto(s)
Células-Madre Neurales , Pez Cebra , Animales , Ratas , Diferenciación Celular/fisiología , Proliferación Celular , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo , Células-Madre Neurales/metabolismo , Factor de Transcripción PAX6/genética , Factor de Transcripción PAX6/metabolismo , Factores de Transcripción/metabolismo , Pez Cebra/metabolismo
5.
Neurosci Lett ; 784: 136742, 2022 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-35716963

RESUMEN

Posttranslational modification (PTM) of tubulin proteins is involved in microtubule dynamics. Acetylation, an important alpha-tubulin PTM, which is regarded as a hallmark event of stable microtubules, often occurs in neurogenesis and axon outgrowth. GCN5/KAT2A is a well-known histone acetyltransferase and has also been reported to hold the activity of nonhistone acetyltransferases, such as acetylated tubulin (Ace-tubulin). In this study, we investigated the role of GCN5/KAT2A in axon growth and neurogenesis. E18 cortical neurons obtained from day 18 embryos of pregnant Sprague-Dawley (SD) rats were cultured and transfected with GCN5 siRNA or treated with the GCN5 inhibitor MB-3. Neural stem cells (NSCs) derived from the cerebral cortexes of E14 SD rats were cultured and differentiated. During differentiation, MB-3 was applied to investigate the effect of GCN5 dysfunction on neurogenesis. The axonal length and the ratio and distribution of acetylated and tyrosinated tubulin (Tyr-tubulin) were evaluated by immunostaining assay. The expression levels of Nestin, Tuj1, acetylated tubulin, and tyrosinated tubulin proteins were analyzed by Western blotting assays. In primary neurons, both GCN5 siRNA and MB-3 treatment reduced acetylated tubulin protein, changed the ratio of acetylated and tyrosinated tubulin, and decreased axonal length. During NSC differentiation, MB-3 application reduced axon outgrowth, decreased acetylated tubulin and altered the distribution of acetylated tubulin and tyrosinated tubulin. This study revealed for the first time that the acetyltransferase GCN5/KAT2A could contribute to axon outgrowth by altering the ratio and distribution of acetylated tubulin.


Asunto(s)
Axones , Histona Acetiltransferasas , Microtúbulos , Neurogénesis , Tubulina (Proteína) , Acetilación , Animales , Axones/metabolismo , Células Cultivadas , Histona Acetiltransferasas/genética , Microtúbulos/metabolismo , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Tubulina (Proteína)/metabolismo
6.
Front Surg ; 9: 1081823, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36733676

RESUMEN

Background: Pathological spine fractures caused by metastases of lung cancer have brought great suffering to patients. Percutaneous kyphoplasty (PKP) has been considered a preferred alternative for painful spinal metastases. The clinical efficacy and safety of PKP for metastatic spinal lesions are urgently to be evaluated. Methods: A cohort study was conducted on 54 cases with pathologic spine fractures caused by metastasis of lung cancer. The correction of kyphosis was assessed by the Cobb angle. The life dependence and quality of the patients were evaluated by the Barthel Index of activities of daily living (ADL) and the quality-adjusted life year (QALY). Patients' survival was carefully recorded. Results: PKP significantly corrected the kyphosis compared with conservative treatment. The ratio of moderate dependence after fracture was clearly increased by PKP. QALY indicated a better life quality brought by PKP. However, PKP could not improve the survival rate of patients. Conclusion: PKP can be used as an effective palliative care treatment for patients with metastatic pathologic spinal fractures of lung cancer.

7.
Front Cell Dev Biol ; 9: 768970, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34820384

RESUMEN

Cadherins play an important role in tissue homeostasis, as they are responsible for cell-cell adhesion during embryogenesis, tissue morphogenesis, and differentiation. In this study, we identified Cadherin-12 (CDH12), which encodes a type II classical cadherin, as a gene that promotes neurite outgrowth in an in vitro model of neurons with differentiated intrinsic growth ability. First, the effects of CDH12 on neurons were evaluated via RNA interference, and the results indicated that the knockdown of CDH12 expression restrained the axon extension of E18 neurons. The transcriptome profile of neurons with or without siCDH12 treatment revealed a set of pathways positively correlated with the effect of CDH12 on neurite outgrowth. We further revealed that CDH12 affected Rac1/Cdc42 phosphorylation in a PKA-dependent manner after testing using H-89 and 8-Bromo-cAMP sodium salt. Moreover, we investigated the expression of CDH12 in the brain, spinal cord, and dorsal root ganglia (DRG) during development using immunofluorescence staining. After that, we explored the effects of CDH12 on neurite outgrowth in vivo. A zebrafish model of CDH12 knockdown was established using the NgAgo-gDNA system, and the vital role of CDH12 in peripheral neurogenesis was determined. In summary, our study is the first to report the effect of CDH12 on axonal extension in vitro and in vivo, and we provide a preliminary explanation for this mechanism.

8.
FASEB J ; 35(5): e21526, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33813773

RESUMEN

Discovering safe and effective drugs that promote neuron regeneration is an essential strategy for the recovery of central nervous system injuries. In this study, we found that L-leucine, an essential amino acid obtained from both supplements and food sources, could dramatically boost axonal outgrowth and regeneration. First, the effects of L-leucine on neurons were evaluated by cell apoptosis, survival, and death assays, and the results showed no changes in these processes after treatment. By live cell imaging, L-leucine was found to remarkably increase axonal length and growth velocity after axotomy. We also verified that L-leucine enhanced p-mTOR/p-S6K activation in neurons by testing with an mTOR inhibitor, rapamycin. Thereafter, we investigated the effects of L-leucine on the spinal cord injury in vivo. A mouse model of spinal cord hemi-section was established, and L-leucine was administered by tail intravenous injection. Basso mouse scale values revealed that L-leucine could improve functional recovery after injury. It was also notable that L-leucine treatment promoted axon growth across chondroitin sulfate proteoglycan (CSPG) areas. Furthermore, we used CSPGs as inhibitory environmental cues and clarified that L-leucine significantly enhanced axonal outgrowth and regeneration by promoting p-mTOR and p-S6K activation. Therefore, our study is the first to report that L-leucine promotes axonal regeneration in vitro and in vivo and could be candidate drug for axonal re-growth and nervous functional recovery.


Asunto(s)
Leucina/farmacología , Regeneración Nerviosa , Proyección Neuronal , Neuronas/citología , Recuperación de la Función , Traumatismos de la Médula Espinal/terapia , Serina-Treonina Quinasas TOR/metabolismo , Animales , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos ICR , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología , Serina-Treonina Quinasas TOR/genética
9.
Front Cell Dev Biol ; 8: 593234, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33585441

RESUMEN

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.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...