Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros










Intervalo de año de publicación
1.
Biology (Basel) ; 13(3)2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38534420

RESUMEN

E. sinensis is an animal model for studying the reproduction and development of crustaceans. In this study, we knocked down the Es-Kif2a gene by injecting dsRNA into E. sinensis and inhibited Es-Plk1 gene expression by injecting PLK1 inhibitor BI6727 into E. sinensis. Then, the cell proliferation level, apoptosis level, and PI3K/AKT signaling expression level were detected. Our results showed that the proliferation level of spermatogenic cells decreased, while the apoptosis level increased after Es-Kif2a knockdown or Es-Plk1 inhibition. In order to verify whether these changes are caused by regulating the PI3K/AKT pathway, we detected the expression of PI3K and AKT proteins after Es-Kif2a knockdown or Es-Plk1 inhibition. Western Blot showed that in both the Es-Kif2a knockdown group and the Es-Plk1 inhibition group, the expression of PI3K and AKT proteins decreased. In addition, immunofluorescence showed that Es-KIF2A and Es-PLK1 proteins were co-localized during E. sinensis spermatogenesis. To further explore the upstream and downstream relationship between Es-KIF2A and Es-PLK1, we detected the expression level of Es-PLK1 after Es-Kif2a knockdown as well as the expression level of Es-KIF2A after Es-Plk1 inhibition. Western Blot showed that the expression of Es-PLK1 decreased after Es-Kif2a knockdown, while there was no significant change of Es-KIF2A after Es-Plk1 inhibition, indicating that Es-PLK1 may be a downstream factor of Es-KIF2A. Taken together, these results suggest that Es-KIF2A upregulates the PI3K/AKT signaling pathway through Es-PLK1 during the spermatogenesis of E. sinensis, thereby affecting the proliferation and apoptosis levels of spermatogenic cells.

2.
Cell Rep ; 42(10): 113257, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37851573

RESUMEN

Nociceptive axons undergo remodeling as they innervate their targets during development and in response to environmental insults and pathological conditions. How is nociceptive morphogenesis regulated? Here, we show that the microtubule destabilizer kinesin family member 2A (Kif2a) is a key regulator of nociceptive terminal structures and pain sensitivity. Ablation of Kif2a in sensory neurons causes hyperinnervation and hypersensitivity to noxious stimuli in young adult mice, whereas touch sensitivity and proprioception remain unaffected. Computational modeling predicts that structural remodeling is sufficient to explain the phenotypes. Furthermore, Kif2a deficiency triggers a transcriptional response comprising sustained upregulation of injury-related genes and homeostatic downregulation of highly specific channels and receptors at the late stage. The latter effect can be predicted to relieve the hyperexcitability of nociceptive neurons, despite persisting morphological aberrations, and indeed correlates with the resolution of pain hypersensitivity. Overall, we reveal a critical control node defining nociceptive terminal structure, which is regulating nociception.


Asunto(s)
Cinesinas , Nocicepción , Proteínas Represoras , Animales , Ratones , Cinesinas/genética , Neuronas/fisiología , Dolor , Proteínas Represoras/genética
3.
Mol Genet Genomic Med ; 11(10): e2225, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37331001

RESUMEN

BACKGROUND: KIF2A-related tubulinopathy (MIM: #615411) is a very rare disorder that was clinically characterized as microcephaly, epilepsy, motor developmental disorder (MDD), and various malformations of cortical development, but intellectual disability (ID) or global developmental delay (GDD) was rarely reported in the patients. METHODS: Quad whole-exome sequencing (WES) was performed on the proband, the older brother, and their parents. Sanger sequencing was used to verify the candidate gene variant. RESULTS: The proband, a 23-month-old boy, was previously diagnosed with GDD, and his brother, aged nine years, had ID; both were born to a healthy couple. Quad-WES detected a novel heterozygous KIF2A variant, c.1318G>A (p.G440R), in both the brothers but not in the parents. In-silico analysis revealed that the variants G440R and G318R (which were previously reported in the only reported patient with GDD) lead to markedly enlarged side chains and hinder ATP's emplacement in the NBD pocket. CONCLUSIONS: The type of KIF2A variants that sterically hinder ATP emplacing in KIF2A NBD pocket may be associated with the intellectual disability phenotype; however, further studies are needed. Findings in this case also suggest a rare parental germline mosaicism of KIF2A G440R.

4.
EMBO J ; 42(11): e112953, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37014312

RESUMEN

Microtubule (MT) minus ends are stabilized by CAMSAP family proteins at noncentrosomal MT-organizing centers. Despite progress in identifying diverse positive regulators, knowledge on the negative regulation of the MT minus-end distribution is lacking. Here, we identify CEP170B as a MT minus-end-binding protein that colocalizes with the microtubule-stabilizing complex at the cortical patches. CEP170B depends on the scaffold protein liprin-α1 for its cortical targeting and requires liprin-α1-bound PP2A phosphatase for its MT localization. CEP170B excludes CAMSAPs-stabilized MT minus ends from the cell periphery in HeLa cells and the basal cortex in human epithelial cells and is required for directional vesicle trafficking and cyst formation in 3D culture. Reconstitution experiments demonstrate that CEP170B autonomously tracks growing MT minus ends and blocks minus-end growth. Furthermore, CEP170B in a complex with the kinesin KIF2A acts as a potent MT minus-end depolymerase capable of antagonizing the stabilizing effect of CAMSAPs. Our study uncovers an antagonistic mechanism for controlling the spatial distribution of MT minus ends, which contributes to the establishment of polarized MT network and cell polarity.


Asunto(s)
Proteínas Asociadas a Microtúbulos , Microtúbulos , Humanos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Células HeLa , Microtúbulos/metabolismo , Proteínas del Citoesqueleto/metabolismo , Cinesinas/genética , Cinesinas/metabolismo
5.
Mol Ther Nucleic Acids ; 31: 197-210, 2023 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-36700049

RESUMEN

Synovial fibroblasts are the active and aggressive drivers in the progression of arthritis, but the cellular and molecular mechanisms remain unknown. Here, our results showed that regulator of G protein signaling 12 (RGS12) maintained ciliogenesis in synovial fibroblasts, which is critical for the development of inflammatory arthritis. Deletion of RGS12 led to a significant decrease in ciliogenesis, adhesion, migration, and secretion of synovial fibroblasts. Mechanistically, RGS12 overexpression in synovial fibroblasts increased the length and number of cilia but decreased the protein level of kinesin family member 2A (KIF2A). The results of LC-MS analyses showed that RGS12 interacted with MYC binding protein 2 to enhance its ubiquitination activity, through which the KIF2A protein was degraded in synovial fibroblasts. Moreover, overexpression of KIF2A blocked the increases in cilia length and number. Mice with RGS12 deficiency or treatment of RGS12 shRNA nanoparticles significantly decreased the clinical score, paw swelling, synovitis, and cartilage destruction during inflammatory arthritis by inhibiting the activation of synovial fibroblasts. Therefore, this study provides evidence that RGS12 activates synovial fibroblasts' pathological function via promoting MCYBP2-mediated degradation of KIF2A and ciliogenesis. Our data suggest that RGS12 may be a potential drug target for the treatment of inflammatory arthritis.

6.
Open Med (Wars) ; 17(1): 1780-1787, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36447525

RESUMEN

Cervical cancer is the fourth most common cancer and the fourth leading cause of cancer death in women. Human papillomavirus (HPV16) E6/E7 heterogenous expression in C33A cells increased the mRNA and protein levels of KIF2A, while siRNA deletion of endogenous E6/E7 reduced the mRNA and protein levels of KIF2A in SiHa cells. KIF2A promoted cell migration and invasion, and regulated the expression of epithelial-mesenchymal transition-related proteins in C33A and SiHa cells. The exogenous expression of E6/E7 in C33A cells increased the phosphorylation of Akt, ERK, and JNK. However, Akt (API-2) and ERK (PD98059) inhibitors had no effect on the increase in KIF2A expression induced by E6/E7, while JNK inhibitors (JNK-IN-8 and SP600125) blocked the increase in KIF2A expression induced by E6/E7. The exogenous expression of E6/E7 increased the levels of transcription factor c-Jun, which is the classic substrate of JNK. Knockdown of c-Jun reduced the increase in KIF2A expression induced by E6/E7. In summary, KIF2A plays a key role in the motility and metastasis of cervical cancer. HPV16 E6/E7 can increase the levels of transcription factor c-Jun by activating the JNK signal, thereby up-regulating the transcriptional expression of KIF2A.

7.
Allergy Asthma Clin Immunol ; 18(1): 55, 2022 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-35718777

RESUMEN

BACKGROUND: The microtubule-dependent molecular motor protein Kinesin Family Member 2A (KIF2A) is down-regulated in asthmatic human airway epithelium. However, little is known about the roles of KIF2A as well as the possible underlying mechanisms in asthma. METHODS: House dust mite (HDM) extract was administered to establish a murine model of asthma. The expression of KIF2A, IL-33 and the autophagy pathways were detected. The plasmid pCMV-KIF2A was used to overexpress KIF2A in the airway epithelial cells in vitro and in vivo. IL-4, IL-5, IL-33 and other cytokines in bronchoalveolar lavage fluid (BALF) and lung tissues homogenates were measured. RESULTS: In response to the challenge of house dust mite (HDM) in vitro and in vivo, airway epithelial cells displayed decreased production of KIF2A. Meanwhile, autophagy and IL-33 were increased in HMD-treated epithelial cells. Mechanistically, KIF2A decreased autophagy via suppressing mTORC1 pathway in HDM-treated epithelial cells, which contributed to the reduced production of IL-33. Moreover, in vivo KIF2A transfection reduced IL-33 and autophagy in the lung, leading to the attenuation of allergic asthma. CONCLUSION: KIF2A suppressed mTORC1-mediated autophagy and decreased the production of epithelial-derived cytokine IL-33 in allergic airway inflammation. These data indicate that KIF2A may be a novel target in allergic asthma.

8.
Cancer Cell Int ; 22(1): 81, 2022 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-35164763

RESUMEN

BACKGROUND: Breast cancer (BC) threatens the health of women around the world. Researchers have proved that hsa_circ_0005505 (circ_IRAK3) facilitates BC cell invasion and migration, but the regulatory mechanisms of circ_IRAK3 in BC remain mostly unknown. We aim to explore a new mechanism by which circ_IRAK3 promotes BC progression. METHODS: Levels of circ_IRAK3, microRNA (miR)-603, and kinesin family member 2A (KIF2A) mRNA in BC tissues and cells were examined by quantitative real-time polymerase chain reaction (qRT-PCR). The cell cycle progression, colony formation, and proliferation of BC cells were evaluated by flow cytometry, plate clone, or 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazoliumbromide (MTT) assays. The migration, invasion, and apoptosis of BC cells were determined by transwell or flow cytometry assays. Several protein levels were detected using western blotting. The targeting relationship between circ_IRAK3 or KIF2A and miR-603 was verified via dual-luciferase reporter assay. The role of circ_IRAK3 in vivo was verified by xenograft assay. RESULTS: We observed higher levels of circ_IRAK3 in BC tissues and cell lines than their respective controls. Functional experiments presented that circ_IRAK3 silencing induced BC cell apoptosis, curbed cell proliferation, migration, and invasion in vitro, and decreased tumor growth in vivo. Mechanistically, circ_IRAK3 could modulate kinesin family member 2A (KIF2A) expression through acting as a microRNA (miR)-603 sponge. miR-603 silencing impaired the effects of circ_IRAK3 inhibition on the malignant behaviors of BC cells. Also, the repressive effects of miR-603 mimic on the malignant behaviors of BC cells were weakened by KIF2A overexpression. CONCLUSIONS: circ_IRAK3 exerted a promoting effect on BC progression by modulating the miR-603/KIF2A axis, providing a piece of novel evidence for circ_IRAK3 as a therapeutic target for BC.

9.
eNeuro ; 8(5)2021.
Artículo en Inglés | MEDLINE | ID: mdl-34404749

RESUMEN

In recent years there has been extensive research on malformations of cortical development (MCDs) that result in clinical features like developmental delay, intellectual disability, and drug-resistant epilepsy (DRE). Various studies highlighted the contribution of microtubule-associated genes (including tubulin and kinesin encoding genes) in MCD development. It has been reported that de novo mutations in KIF2A, a member of the kinesin-13 family, are linked to brain malformations and DRE. Although it is known that KIF2A functions by regulating microtubule depolymerization via an ATP-driven process, in vivo implications of KIF2A loss of function remain partly unclear. Here, we present a novel kif2a knock-out zebrafish model, showing hypoactivity, habituation deficits, pentylenetetrazole-induced seizure susceptibility and microcephaly, as well as neuronal cell proliferation defects and increased apoptosis. Interestingly, kif2a-/- larvae survived until adulthood and were fertile. Notably, our kif2a zebrafish knock-out model demonstrated many phenotypic similarities to KIF2A mouse models. This study provides valuable insights into the functional importance of kif2a in zebrafish and phenotypical hallmarks related to KIF2A mutations. Ultimately, this model could be used in a future search for more effective therapies that alleviate the clinical symptoms typically associated with MCDs.


Asunto(s)
Epilepsia , Discapacidad Intelectual , Animales , Epilepsia/genética , Cinesinas/genética , Ratones , Proteínas Represoras , Tubulina (Proteína) , Pez Cebra
10.
Ann Med ; 53(1): 693-706, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34024242

RESUMEN

BACKGROUND: Circular RNA microarray analysis showed hsa_circ_0010235 (circ_0010235) was highly upregulated in non-small-cell lung cancer (NSCLC) patients; however, its role in carcinogenesis and development of NSCLC cells was unrevealed. Here, we intended to investigate role and mechanism of circ_0010235 in NSCLC proliferation, migration and invasion. METHODS AND RESULTS: Expression of circ_0010235, microRNA (miR)-338-3p and kinesin family member 2A (KIF2A) was detected by quantitative real-time PCR, western blotting and immunohistochemistry (IHC). Cell progression was measured by cell-counting kit-8 assay, 5-ethynyl-2-deoxyuridine (EdU) assay, flow cytometry, transwell assay, western blotting, IHC and xenograft experiment. The relationship among circ_0010235, miR-338-3p and KIF2A was determined by dual-luciferase reporter assay, RNA immunoprecipitation and Pearson's correlation analysis. Expression of circ_0010235 was increased in human NSCLC tissues and cells, accompanied with miR-338-3p downregulation and KIF2A upregulation. Essentially, circ_0010235 could sponge miR-338-3p via target binding, and miR-338-3p downstream targeted KIF2A. Functionally, exhaustion of circ_0010235 induced apoptosis rate of NSCLC cells and curbed cell viability, EdU incorporation, migration rate and invasion rate, accompanied with higher E-cadherin and lower N-cadherin expression. Additionally, re-expression of miR-338-3p prompted above similar effects in NSCLC cells in vitro. Contrarily, miR-338-3p blockage partially counteract the effects of circ_0010235 exhaustion; plus, restoration of KIF2A could attenuate miR-338-3p role, as well. Notably, interfering circ_0010235 delayed tumour growth of NSCLC cells by promoting miR-338-3p and E-cadherin expression, and depressing KIF2A, ki-67 and N-cadherin expression. CONCLUSIONS: circ_0010235 could be a novel identified oncogenic circRNA in NSCLC, and targeting miR-338-3p/KIF2A axis was one regulatory mechanism underlying circ_0010235.KEY MESSAGECirc_0010235 was an upregulated circRNA in NSCLC patients and cells.Interfering circ_0010235 restrained NSCLC cell proliferation and metastasis in vitro and in vivo.miR-338-3p per se suppressed NSCLC in vitro and its downregulation diminished the tumour-suppressive role of circ_0010235 blockage in NSCLC cells.miR-338-3p could downstream target KIF2A and be sponged by circ_0010235.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , MicroARNs , Cadherinas , Carcinogénesis , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Proliferación Celular/genética , Humanos , Cinesinas/genética , Neoplasias Pulmonares/genética , MicroARNs/genética , ARN Circular
11.
Development ; 148(4)2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-33531432

RESUMEN

KIF2A is a kinesin motor protein with essential roles in neural progenitor division and axonal pruning during brain development. However, how different KIF2A alternative isoforms function during development of the cerebral cortex is not known. Here, we focus on three Kif2a isoforms expressed in the developing cortex. We show that Kif2a is essential for dendritic arborization in mice and that the functions of all three isoforms are sufficient for this process. Interestingly, only two of the isoforms can sustain radial migration of cortical neurons; a third isoform, lacking a key N-terminal region, is ineffective. By proximity-based interactome mapping for individual isoforms, we identify previously known KIF2A interactors, proteins localized to the mitotic spindle poles and, unexpectedly, also translation factors, ribonucleoproteins and proteins that are targeted to organelles, prominently to the mitochondria. In addition, we show that a KIF2A mutation, which causes brain malformations in humans, has extensive changes to its proximity-based interactome, with depletion of mitochondrial proteins identified in the wild-type KIF2A interactome. Our data raises new insights about the importance of alternative splice variants during brain development.


Asunto(s)
Diferenciación Celular/genética , Movimiento Celular/genética , Regulación de la Expresión Génica , Cinesinas/genética , Neuronas/citología , Neuronas/metabolismo , Proteínas Represoras/genética , Empalme Alternativo , Animales , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Perfilación de la Expresión Génica , Cinesinas/metabolismo , Ratones , Mutación , Neurogénesis/genética , Proteómica/métodos , Isoformas de ARN , Proteínas Represoras/metabolismo
12.
Am J Med Genet A ; 185(4): 1113-1119, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33506645

RESUMEN

Cortical dysplasia, complex, with other brain malformations 3 (CDCBM3) is a rare autosomal dominant syndrome caused by Kinesin family Member 2A (KIF2A) gene mutation. Patients with CDCBM3 exhibit posterior dominant agyria/pachygyria with severe motor dysfunction. Here, we report an 8-year-old boy with CDCBM3 showing a typical, but relatively mild, clinical presentation of CDCBM3 features. Whole-exome sequencing identified a heterozygous mutation of NM_001098511.2:c.1298C>A [p.(Ser433Tyr)]. To our knowledge, the mutation has never been reported previously. The variant was located distal to the nucleotide binding domain (NBD), in which previously-reported variants in CDCBM3 patients have been located. The computational structural analysis showed the p.433 forms the pocket with NBD. Variants in KIF2A have been reported in the NBD for CDCBM3, in the kinesin motor 3 domain, but not in the NBD in epilepsy, and outside of the kinesin motor domain in autism spectrum syndrome, respectively. Our patient has a variant, that is not in the NBD but at the pocket with the NBD, resulting in a clinical features of CDCBM3 with mild symptoms. The clinical findings of patients with KIF2A variants appear restricted to the central nervous system and facial anomalies. We can call this spectrum "KIF2A syndrome" with variable severity.


Asunto(s)
Epilepsia/genética , Cinesinas/genética , Malformaciones del Desarrollo Cortical/genética , Proteínas Asociadas a Microtúbulos/genética , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Niño , Epilepsia/diagnóstico , Epilepsia/diagnóstico por imagen , Epilepsia/patología , Heterocigoto , Humanos , Cinesinas/ultraestructura , Masculino , Malformaciones del Desarrollo Cortical/diagnóstico , Malformaciones del Desarrollo Cortical/diagnóstico por imagen , Malformaciones del Desarrollo Cortical/patología , Proteínas Asociadas a Microtúbulos/ultraestructura , Mutación Missense/genética , Conformación Proteica , Tubulina (Proteína)/genética , Secuenciación del Exoma
13.
Cancer Biother Radiopharm ; 36(7): 556-567, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32833501

RESUMEN

Background: Temozolomide (TMZ) resistance is a serious hindrance in clinical chemotherapy for glioma. Circular RNA homeodomain interacting protein kinase 3 (circHIPK3) can be involved in regulating the progression of glioma, but the molecular mechanism of circHIPK3 in TMZ-resistant-glioma is completely unclear. Materials and Methods: The levels of circRNA, miRNA, and mRNA were examined using quantitative real-time polymerase chain reaction. 3-(4,5-dimethylthiazol-2-y1)-2, 5-diphenyl tetrazolium bromide assay was used for assessing the half inhibitory concentration (IC50) of TMZ and cell proliferation. Cell apoptosis and metastasis (migration and invasion) were detected by flow cytometry and transwell assay, respectively. Western blot and dual-luciferase reporter assay were performed several times to analyze the expression levels of associated proteins and the targeted relation. Results: The upregulation of circHIPK3 was found in TMZ-resistant glioma tissues and cells. Both circHIPK3 knockdown and kinesin family member 2A (KIF2A) inhibition could facilitate TMZ sensitivity and apoptosis but repress proliferation and metastasis in TMZ-resistant glioma cells. CircHIPK3 targeted microRNA-524-5p (miR-524-5p) and KIF2A functioned as a downstream target of miR-524-5p. Decrease of miR-524-5p relieved the effects of si-circHIPK3 on TMZ-resistant glioma cells by upregulating KIF2A. Downregulation of circHIPK3 refrained the phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT) signal pathway partly through miR-524-5p/KIF2A axis. Conclusions: Knockdown of circHIPK3 promoted TMZ sensitivity in glioma by modulating proliferation, metastasis, and apoptosis through miR-524-5p/KIF2A-mediated PI3K/AKT pathway. CircHIPK3 may be the potential target for the diagnosis and therapy of TMZ-resistant glioma.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Cinesinas/metabolismo , MicroARNs/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Circular/genética , Temozolomida/farmacología , Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Glioma/genética , Glioma/metabolismo , Glioma/patología , Humanos , Cinesinas/genética , MicroARNs/genética , ARN Circular/metabolismo
14.
Cell Mol Life Sci ; 78(4): 1765-1779, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32789689

RESUMEN

Maintaining the integrity of the mitotic spindle in metaphase is essential to ensure normal cell division. We show here that depletion of microtubule-associated protein ATIP3 reduces metaphase spindle length. Mass spectrometry analyses identified the microtubule minus-end depolymerizing kinesin Kif2A as an ATIP3 binding protein. We show that ATIP3 controls metaphase spindle length by interacting with Kif2A and its partner Dda3 in an Aurora kinase A-dependent manner. In the absence of ATIP3, Kif2A and Dda3 accumulate at spindle poles, which is consistent with reduced poleward microtubule flux and shortening of the spindle. ATIP3 silencing also limits Aurora A localization to the poles. Transfection of GFP-Aurora A, but not kinase-dead mutant, rescues the phenotype, indicating that ATIP3 maintains Aurora A activity on the poles to control Kif2A targeting and spindle size. Collectively, these data emphasize the pivotal role of Aurora kinase A and its mutual regulation with ATIP3 in controlling spindle length.


Asunto(s)
Aurora Quinasa A/genética , Cinesinas/genética , Fosfoproteínas/genética , Huso Acromático/genética , Proteínas Supresoras de Tumor/genética , Células HeLa , Humanos , Metafase , Proteínas Asociadas a Microtúbulos/genética , Microtúbulos/genética , Mitosis/genética
15.
Neurobiol Aging ; 96: 104-108, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32977080

RESUMEN

The levels of tau phosphorylation differ between sexes in Alzheimer's disease (AD). Transcriptome-wide associations of sex by disease interaction could indicate whether specific genes underlie sex differences in tau pathology; however, no such study has been reported yet. We report the first analysis of the effect of the interaction between disease status and sex on differential gene expression, meta-analyzing transcriptomic data from the 3 largest publicly available case-control studies (N = 785) in the brain to date. A total of 128 genes, significantly associated with sex-AD interactions, were enriched in phosphoproteins (false discovery rate (FDR) = 0.001). High and consistent associations were found for the overexpressions of NCL (FDR = 0.002), whose phosphorylated protein generates an epitope against neurofibrillary tangles and KIF2A (FDR = 0.005), a microtubule-associated motor protein gene. Transcriptome-wide interaction analyses suggest sex-modulated tau phosphorylation, at sites like Thr231, Ser199, or Ser202 that could increase the risk of women to AD and indicate sex-specific strategies for intervention and prevention.


Asunto(s)
Enfermedad de Alzheimer/genética , Perfilación de la Expresión Génica , Estudios de Asociación Genética , Caracteres Sexuales , Transcriptoma/genética , Proteínas tau/metabolismo , Enfermedad de Alzheimer/etiología , Epítopos , Femenino , Humanos , Cinesinas/genética , Masculino , Ovillos Neurofibrilares/genética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Riesgo , Nucleolina
16.
Cancer Manag Res ; 12: 6629-6640, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32801897

RESUMEN

OBJECTIVE: Gastric cancer (GC) is a gastrointestinal tumor. This study is aimed to explore the regulatory mechanism of long non-coding RNA BLACAT1 (BLACAT1)/microRNA-149-5p (miR-149-5p)/KIF2A cascade on GC. METHODS: The expression of BLACAT1, miR-149-5p and KIF2A in GC was detected by qRT-PCR. The proliferation, migration and invasion of GC cells in vitro were analyzed by MTT, wound-healing and transwell assay, respectively. The xenograft tumor model was constructed in nude mice to confirm the inhibition effect of BLACAT1 knockdown on GC in vivo. Then, dual-luciferase reporter assay was used to detect the interactions among BLACAT1, miR-149-5p and KIF2A. Western blot assay was performed to determine the protein expression of KIF2A. RESULTS: The expression of BLACAT1 and KIF2A was up-regulated in GC, but miR-149-5p expression was down-regulated. Silencing of BLACAT1 retarded the proliferation, migration and invasion of GC cells in vitro and the growth of tumor xenograft in vivo. Moreover, BLACAT1 acted as the molecular sponge of miR-149-5p to up-regulate KIF2A expression. At last, feedback experiments suggested that BLACAT1 accelerated the proliferation, migration and invasion of GC cells by regulating miR-149-5p/KIF2A axis. CONCLUSION: BLACAT1 facilitated the tumorigenesis of GC through regulating miR-149-5p/KIF2A axis, which indicated BLACAT1/miR-149-5p/KIF2A cascade may be a new therapeutic target.

17.
J Cell Mol Med ; 24(16): 9114-9124, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32638511

RESUMEN

Abnormal expression of neuropilin and tolloid-like 1 (NETO1) has been detected in some human carcinomas. However, the expression of NETO1 and the underlying mechanism in epithelial ovarian cancer (EOC) remain unknown. In this study, we found that a higher NETO1 expression in EOC tissue samples compared to normal ovarian tissue samples was significantly correlated with worse overall survival. Additionally, Cox regression analysis suggested that NETO 1 was independently associated with overall survival. NETO1 overexpression enhanced the EOC cells' migration and invasion capability in vitro via regulation of actin cytoskeleton. Mechanistically, silencing NETO1 reduced the expression of ß-tubulin, F-actin and KIF2A. In conclusion, our results demonstrated the critical role of NETO1 in EOC invasion, and therapies aimed at inhibiting its expression or activity might significantly control EOC growth, invasion and metastatic dissemination.


Asunto(s)
Carcinoma Epitelial de Ovario/metabolismo , Neuropilinas/metabolismo , Neoplasias Ováricas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Carcinoma Epitelial de Ovario/patología , Línea Celular Tumoral , Movimiento Celular/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Cinesinas/metabolismo , Persona de Mediana Edad , Neoplasias Ováricas/patología , Tubulina (Proteína)/metabolismo
18.
Cancer Manag Res ; 12: 3731-3739, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32547209

RESUMEN

AIM: Kinesin family member 2A (KIF2A) is a member of the kinesin-13 superfamily protein. KIF2A played a role in the development of many tumors. However, the role of KIF2A in esophageal squamous cell carcinoma (ESCC) remains unclear. In this study, we aimed to investigate the role of KIF2A in ESCC. METHODS: We used bioinformatics analysis to study the expression levels and prognosis of KIF2A in ESCC and normal tissues. We also used our own samples to verify the results by immunohistochemistry. Then, the biological functions of KIF2A in ESCC was studied by cell experiments and animal experiments. RESULTS: Both the TCGA database and our samples showed that KIF2A was relatively highly expressed in ESCC tissues and was significantly associated with disease-free survival (P =0.037) in TCGA database. Colony formation assay, CCK8 and Western blotting results showed that knockdown of KIF2A can significantly reduce colony forming ability and proliferation ability. The results of animal experiments showed that knocking down KIF2A can significantly reduce the tumor volume of mice. CONCLUSION: KIF2A might be used as a prognostic factor for ESCC, and knockdown of KIF2A could inhibit ESCC proliferation in vitro and in vivo, respectively. KIF2A could serve as a potential prognostic biomarker and therapeutic target for future ESCC.

19.
Am J Cancer Res ; 10(4): 1130-1139, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32368390

RESUMEN

Circular RNAs (circRNAs) have been shown to regulate the development and progression of various cancers. However, the expression and function of circRNAs in papillary thyroid cancer (PTC) remain largely unknown. This study is aimed to investigate the potential roles of circEIF3I in PTC and elucidate the functional mechanism. We found that the expression of circEIF3I was significantly upregulated in PTC tissues compared to adjacent normal tissues. CircEIF3I expression was positively associated with tumor size, TNM stage and metastasis. CircEIF3I knockdown remarkably suppressed the proliferation, migration and invasion of PTC cells. Mechanistically, circEIF3I promoted KIF2A expression through competitively interacting with miR-149. In conclusion, circEIF3I upregulation in PTC tissues facilitates KIF2A expression by inhibiting miR-149, leading to malignant progression of PTC. This study suggested circEIF3I/miR-149/KIF2A axis might be a potential therapeutic target.

20.
Biomed Pharmacother ; 125: 109840, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32106376

RESUMEN

Kinesin family protein 2A (KIF2A), an M-type nonmotile microtubule depolymerase, plays essential roles in development and progression of various human cancers. However, its exact function and the underlying mechanism in tumorigenesis of gastric cancer (GC) haven't been fully elucidated. In the present study, KIF2A was overexpressed in human GC and predicted poor prognosis according to the results of GEPIA analysis. KIF2A was also observed to be upregulated in 82 GC samples compared with paired pericarcinoma tissues. Its overexpression was associated with tumor metastasis (P = 0.047) and Ⅲ stage GC (P = 0.0267). The mRNA and protein expression levels of KIF2A were significantly suppressed in KIF2A specific siRNA transfected GC cells compared with the wild-type and negative control (NC) siRNA transfected cells. Furthermore, the effects of KIF2A on the growth, migration, invasion, and apoptosis of GC cell were evaluated in vitro and the underlying mechanisms were explored. It was found that silencing KIF2A effectively induced the apoptosis, and inhibited the proliferation, migration and invasion capacities of GC cells. Western blot analysis demonstrated that silencing of KIF2A significantly decreased the expression levels of AKT, Cyclin D1 and S6K. Moreover, bioinformatics analysis showed that the promoter (from -414 to -407bp) of KIF2A has the ability to bind to transcription factor ETV4, which was confirmed by bi-luciferase reporter assay using 293T cells. The level of ETV4 was upregulated and positively correlated with KIF2A in human GC tissues. Our results also proved that ETV4 upregulated the expression of KIF2A and blocked the decline of proliferation induced by KIF2A knockdown in MKN-45 and AGS cells. In summary, KIF2A is upregulated by transcription factor ETV4, and its knockdown can effectively inhibit the proliferation and induce the apoptosis of GC cells through the AKT signaling pathway in GC cells, implying that the inhibition of KIF2A expression is a potential target for GC therapy.


Asunto(s)
Cinesinas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-ets/metabolismo , Neoplasias Gástricas/metabolismo , Apoptosis/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Progresión de la Enfermedad , Humanos , Invasividad Neoplásica , Transducción de Señal , Neoplasias Gástricas/patología , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA