Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Intervalo de año de publicación
1.
Kidney Int ; 104(5): 995-1007, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37598857

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) resulting from pathogenic variants in PKD1 and PKD2 is the most common form of PKD, but other genetic causes tied to primary cilia function have been identified. Biallelic pathogenic variants in the serine/threonine kinase NEK8 cause a syndromic ciliopathy with extra-kidney manifestations. Here we identify NEK8 as a disease gene for ADPKD in 12 families. Clinical evaluation was combined with functional studies using fibroblasts and tubuloids from affected individuals. Nek8 knockout mouse kidney epithelial (IMCD3) cells transfected with wild type or variant NEK8 were further used to study ciliogenesis, ciliary trafficking, kinase function, and DNA damage responses. Twenty-one affected monoallelic individuals uniformly exhibited cystic kidney disease (mostly neonatal) without consistent extra-kidney manifestations. Recurrent de novo mutations of the NEK8 missense variant p.Arg45Trp, including mosaicism, were seen in ten families. Missense variants elsewhere within the kinase domain (p.Ile150Met and p.Lys157Gln) were also identified. Functional studies demonstrated normal localization of the NEK8 protein to the proximal cilium and no consistent cilia formation defects in patient-derived cells. NEK8-wild type protein and all variant forms of the protein expressed in Nek8 knockout IMCD3 cells were localized to cilia and supported ciliogenesis. However, Nek8 knockout IMCD3 cells expressing NEK8-p.Arg45Trp and NEK8-p.Lys157Gln showed significantly decreased polycystin-2 but normal ANKS6 localization in cilia. Moreover, p.Arg45Trp NEK8 exhibited reduced kinase activity in vitro. In patient derived tubuloids and IMCD3 cells expressing NEK8-p.Arg45Trp, DNA damage signaling was increased compared to healthy passage-matched controls. Thus, we propose a dominant-negative effect for specific heterozygous missense variants in the NEK8 kinase domain as a new cause of PKD.


Asunto(s)
Enfermedades Renales Poliquísticas , Riñón Poliquístico Autosómico Dominante , Animales , Humanos , Recién Nacido , Ratones , Proteínas Portadoras/metabolismo , Cilios/patología , Riñón/metabolismo , Mutación , Quinasas Relacionadas con NIMA/genética , Quinasas Relacionadas con NIMA/metabolismo , Enfermedades Renales Poliquísticas/genética , Riñón Poliquístico Autosómico Dominante/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Serina/genética , Serina/metabolismo , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo
2.
Cell Commun Signal ; 21(1): 209, 2023 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-37596667

RESUMEN

Radiotherapy and chemotherapy remain the mainstay of treatment for colorectal cancer (CRC), although their efficacy is limited. A detailed understanding of the molecular mechanisms underlying CRC progression could lead to the development of new therapeutic strategies. Although it has been established that MYC signaling is dysregulated in various human cancers, direct targeting MYC remains challenging due to its "undruggable" protein structure. Post-translational modification of proteins can affect their stability, activation, and subcellular localization. Hence, targeting the post-translational modification of MYC represents a promising approach to disrupting MYC signaling. Herein, we revealed that NEK8 positively regulates CRC progression by phosphorylating c-MYC protein at serine 405, which exhibited enhanced stability via polyubiquitination. Our findings shed light on the role of NEK8/MYC signaling in CRC progression, offering a novel and helpful target for colorectal cancer treatment. Video Abstract.


Asunto(s)
Neoplasias Colorrectales , Quinasas Relacionadas con NIMA , Proteínas Proto-Oncogénicas c-myc , Humanos , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-myc/genética , Serina , Transducción de Señal
3.
Mol Genet Genomic Med ; 11(4): e2135, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36756677

RESUMEN

BACKGROUND: Renal-hepatic-pancreatic dysplasia type 2 (RHPD2) is a rare condition that has been described in the literature disproportionately in perinatal losses. The main features of liver and kidney involvement are well described, with cardiac malformations and cardiomyopathy adding additional variation to the phenotype. Many patients reported are within larger cohorts of congenital anomalies of kidney and urinary tract (CAKUT) or liver failure, and with minimal phenotypic and clinical course data. METHODS: An independent series of phenotypes and prognosis was aggregated from the literature. In this literature review, we describe an additional patient with RHPD2, provide a clinical update on the oldest known living patient, and report the cumulative phenotypes from the existing published patients. RESULTS: With now examining the 17 known patients in the literature, 13 died within the perinatal period-pregnancy to one year of life. Of the four cases living past the first year of life, one case died at 5 years secondary to renal failure, the other at 30 months secondary to liver and kidney failure. Two are currently alive and well at one year and 13 years. Two cases have had transplantation with one resulting in long-term survival. CONCLUSIONS: These patients serve to expand the existing phenotype of RHPD2 as a perinatal lethal condition into a pediatric disorder with variable expressivity. Additionally, we introduce the consideration of transplantation and outcomes within this cohort and future patients.


Asunto(s)
Anomalías Múltiples , Sistema Urinario , Embarazo , Femenino , Humanos , Riñón/anomalías , Sistema Urinario/anomalías , Hígado
4.
Nephron ; 147(3-4): 229-233, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36215968

RESUMEN

INTRODUCTION: Nephronophthisis (NPHP) is a group of autosomal recessive renal diseases characterized by a reduced ability of the kidneys to concentrate solutes, chronic tubulointerstitial nephritis, and cystic kidney disease. It represents the most common genetic cause of childhood renal failure. To date, around 20 different genes, encoding primary cilia proteins, have been linked to NPHP. These contribute to one-third of cases with NPHP while the majority of patients remain molecularly undiagnosed. MATERIALS AND METHODS: Whole exome sequencing (WES) was carried out on a 2-year-old Lebanese boy with infantile NPHP characterized by multicystic kidney dysplasia, kidney insufficiency, and enlarged kidneys in addition to chronic anemia. The candidate variant, detected by WES, was then tested in the patient and his parents by Sanger sequencing. Copy number variation (CNV) analysis was subsequently performed in the proband. RESULTS: Our studies enabled the detection of a heterozygous de novo variant in NEK8 (NM_178170: p.Arg45Trp) in the proband. CNV analysis excluded the presence of big deletions or insertions in this gene. CONCLUSION: Here we report a de novo heterozygous variant in the NEK8 gene in infantile NPHP. This variant was previously detected at a de novo state in a patient presenting with the same clinical features as the proband. This suggests that autosomal dominant forms of NEK8-linked nephropathies may exist. Reporting further patients with NEK8 mutations is essential to confirm these findings and assess whether dominant forms of the disease are restricted to a specific mutational spot or are linked to variants scattered throughout the NEK8 gene.


Asunto(s)
Enfermedades Renales Poliquísticas , Proteínas Quinasas , Masculino , Humanos , Preescolar , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Quinasas Relacionadas con NIMA/genética , Variaciones en el Número de Copia de ADN , Enfermedades Renales Poliquísticas/genética , Mutación
5.
J Cell Mol Med ; 25(18): 8748-8763, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34374193

RESUMEN

Glioma is the most common malignancy of the nervous system with high rates of recurrence and mortality, even after surgery. The 5-year survival rate is only about 5%. NEK8 is involved in multiple biological processes in a variety of cancers; however, its role in glioma is still not clear. In the current study, we evaluated the prognostic value of NEK8, as well as its role in the pathogenesis of glioma. Using a bioinformatics approach and RNA-seq data from public databases, we found that NEK8 expression is elevated in glioma tissues; we further verified this result by RT-PCR, Western blotting and immunochemistry using clinical samples. Functional enrichment analyses of genes with correlated expression indicated that elevated NEK8 expression is associated with increased immune cell infiltration in glioma and may affect the tumour microenvironment via the regulation of DNA damage/repair. Survival analyses revealed that high levels of NEK8 are associated with a poorer prognosis; higher WHO grade, IDH status, 1p/19q codeletion, age and NEK8 were identified as an independent prognostic factor. These findings support the crucial role of NEK8 in the progression of glioma via effects on immune cell infiltration and suggest that it is a new prognostic biomarker.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Biología Computacional/métodos , Glioma , Quinasas Relacionadas con NIMA/metabolismo , Glioma/inmunología , Glioma/metabolismo , Glioma/terapia , Humanos , Infiltración Leucémica/patología , Pronóstico , Tasa de Supervivencia , Microambiente Tumoral
6.
Nephrol Dial Transplant ; 32(10): 1665-1675, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28387813

RESUMEN

BACKGROUND: In the absence of cytogenetic abnormality, fetuses with congenital anomalies of the kidney and urinary tract (CAKUT) with/without other structural anomalies show a higher likelihood of monogenic causes; however, defining the underlying pathology can be challenging. Here, we investigate the value of whole-exome sequencing (WES) in fetuses with CAKUT but normal findings upon karyotyping and chromosome microarray analysis. METHODS: WES was performed on DNA from the cord blood of 30 fetuses with unexplained CAKUT with/without other structural anomalies. In the first 23 cases, sequencing was initially performed on fetal DNA only; for the remaining seven cases, the trio of fetus, mother and father was sequenced simultaneously. RESULTS: Of the 30 cases, pathogenic variants were identified in 4 (13%) (UMOD, NEK8, HNF1B and BBS2) and incidental variants in 2 (7%) (HSPD1 and GRIN2B). Furthermore, two of the above four cases had other anomalies in addition to CAKUT. Thus, the detection rate was only 2/22 (9.1%) for isolated CAKUT and 2/8 (25%) for CAKUT with other abnormalities. CONCLUSIONS: Applying WES to the prenatal diagnostic approach in CAKUT fetuses with or without other anomalies allows for an accurate and early etiology-based diagnosis and improved clinical management. To expedite interpretation of the results, trio sequencing should be employed; however, interpretation may nevertheless be compromised by incomplete coverage of all relevant genes.


Asunto(s)
Exoma , Anomalías Urogenitales/genética , Adulto , Secuencia de Aminoácidos , Amniocentesis , Secuencia de Bases , Análisis Mutacional de ADN , Femenino , Feto , Humanos , Riñón/anomalías , Riñón/diagnóstico por imagen , Técnicas de Diagnóstico Molecular , Embarazo , Ultrasonografía Prenatal , Sistema Urinario/anomalías , Sistema Urinario/diagnóstico por imagen , Anomalías Urogenitales/diagnóstico por imagen , Secuenciación del Exoma , Adulto Joven
7.
Cell Cycle ; 16(4): 335-347, 2017 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-27892797

RESUMEN

Proteins essential for homologous recombination play a pivotal role in the repair of DNA double strand breaks, DNA inter-strand crosslinks and replication fork stability. Defects in homologous recombination also play a critical role in the development of cancer and the sensitivity of these cancers to chemotherapy. RAD51, an essential factor for homologous recombination and replication fork protection, accumulates and forms immunocytochemically detectable nuclear foci at sites of DNA damage. To identify kinases that may regulate RAD51 localization to sites of DNA damage, we performed a human kinome siRNA library screen, using DNA damage-induced RAD51 foci formation as readout. We found that NEK8, a NIMA family kinase member, is required for efficient DNA damage-induced RAD51 foci formation. Interestingly, knockout of Nek8 in murine embryonic fibroblasts led to cellular sensitivity to the replication inhibitor, hydroxyurea, and inhibition of the ATR kinase. Furthermore, NEK8 was required for proper replication fork protection following replication stall with hydroxyurea. Loading of RAD51 to chromatin was decreased in NEK8-depleted cells and Nek8-knockout cells. Single-molecule DNA fiber analyses revealed that nascent DNA tracts were degraded in the absence of NEK8 following treatment with hydroxyurea. Consistent with this, Nek8-knockout cells showed increased chromosome breaks following treatment with hydroxyurea. Thus, NEK8 plays a critical role in replication fork stability through its regulation of the DNA repair and replication fork protection protein RAD51.


Asunto(s)
Daño del ADN , Replicación del ADN , Quinasas Relacionadas con NIMA/metabolismo , Recombinasa Rad51/metabolismo , Inestabilidad Genómica , Células HeLa , Recombinación Homóloga/genética , Humanos , Quinasas Relacionadas con NIMA/deficiencia , ARN Interferente Pequeño/metabolismo , Estrés Fisiológico
8.
Am J Med Genet A ; 170(3): 750-3, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26697755

RESUMEN

We studied two brothers who presented in the newborn period with cardiac, renal, and hepatic anomalies that were initially suggestive of ALGS, although no mutations in JAG1 or NOTCH2 were identified. Exome sequencing demonstrated compound heterozygous mutations in the NEK8 gene (Never in mitosis A-related Kinase 8), a ciliary kinase indispensable for cardiac and renal development based on murine studies. The mutations included a c.2069_2070insC variant (p.Ter693LeufsTer86), and a c.1043C>T variant (p.Thr348Met) in the highly conserved RCC1 (Regulation of Chromosome Condensation 1) domain. The RCC1 domain is crucial for localization of the NEK8 protein to the centrosomes and cilia. Mutations in NEK8 have been previously reported in three fetuses (from a single family) with renal-hepatic-pancreatic dysplasia 2 (RHPD2), similar to Ivemark syndrome, and in three individuals with nephronophthisis (NPHP9). This is the third report of disease-causing mutations in the NEK8 gene in humans and only the second describing multi-organ involvement. The clinical features we describe differ from those in the previously published report in that (1) a pancreatic phenotype was not observed in the individuals reported here, (2) there were more prominent cardiac findings, (consistent with observations in murine models), and (3) we observed bile duct hypoplasia rather than ductal plate malformation. The patients reported here expand our understanding of the NEK8-associated phenotype. Our findings highlight the variable phenotypic expressivity and the spectrum of clinical manifestations due to mutations in the NEK8 gene.


Asunto(s)
Cardiopatías Congénitas/genética , Heterocigoto , Fallo Renal Crónico/genética , Hepatopatías/congénito , Mutación , Proteínas Quinasas/genética , Hermanos , Anomalías Múltiples , Exoma , Cardiopatías Congénitas/diagnóstico , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Recién Nacido , Riñón/anomalías , Fallo Renal Crónico/diagnóstico , Hígado/anomalías , Hepatopatías/diagnóstico , Masculino , Quinasas Relacionadas con NIMA , Páncreas/anomalías
9.
J Biol Chem ; 290(3): 1389-94, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25451921

RESUMEN

NEK8 (never in mitosis gene A (NIMA)-related kinase 8) is involved in cytoskeleton, cilia, and DNA damage response/repair. Abnormal expression and/or dysfunction of NEK8 are related to cancer development and progression. However, the mechanisms that regulate NEK8 are not well declared. We demonstrated here that pVHL may be involved in regulating NEK8. We found that CAK-I cells with wild-type vhl expressed a lower level of NEK8 than the cells loss of vhl, such as 786-O, 769-P, and A-498 cells. Moreover, pVHL overexpression down-regulated the NEK8 protein in 786-O cells, whereas pVHL knockdown up-regulated NEK8 in CAK-I cells. In addition, we found that the positive hypoxia response elements (HREs) are located in the promoter of the nek8 sequence and hypoxia could induce nek8 expression in different cell types. Consistent with this, down-regulation of hypoxia-inducible factors α (HIF-1α or HIF-2α) by isoform-specific siRNA reduced the ability of hypoxia inducing nek8 expression. In vivo, NEK8 and HIF-1α expression were increased in kidneys of rats subjected to an experimental hypoxia model of ischemia and reperfusion. Furthermore, NEK8 siRNA transfection significantly blocked pVHL-knockdown-induced cilia disassembling, through impairing the pVHL-knockdown-up-regulated NEK8 expression. These results support that nek8 may be a novel hypoxia-inducible gene. In conclusion, our findings show that nek8 may be a new HIF target gene and pVHL can down-regulate NEK8 via HIFs to maintain the primary cilia structure in human renal cancer cells.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Proteínas Quinasas/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Animales , Hipoxia de la Célula , Línea Celular Tumoral , Cilios/metabolismo , Perfilación de la Expresión Génica , Humanos , Riñón/enzimología , Neoplasias Renales/metabolismo , Masculino , Quinasas Relacionadas con NIMA , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión , Transfección
10.
Chongqing Medicine ; (36): 597-599,604, 2015.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-600493

RESUMEN

Objective To investigate the expression of Nek8 in esophageal squamous cell carcinoma(ESCC) tissues and cell line , and to evaluate its correlation with the clinicopathological features of ESCC and the survival rate of ESCC patients after operation . Methods The expression of Nek8 mRNA in human ESCC Eca109 cell line and two pairs of ESCC tissues and adjacent normal e‐sophageal mucosal epithelium were detected by semi‐quantitative reverse transcription polymerase chain reaction (RT‐PCR) .Immu‐nohistochemistry and tissue microarray technique were used to examine the expression of Nek 8 protein in ESCC tissues and tumor‐adjacent tissues .The correlation between Nek8 expression and clinicopathological features of ESCC and survival rate of ESCC pa‐tients was then analyzed .Results The expression of Nek8 mRNA was positive in Eca109 cells and two cases of ESCC tissue ,and it was negative in paired normal esophageal mucosal epithelium specimens .In tissue microarray ,the expression of Nek8 protein in ES‐CC tissues ,which was mainly in the cytoplasm ,was significantly higher than that in tumor‐adjacent tissues(P= 2 .16E‐13) .The high expression of Nek8 was associated with tumor size (P=0 .008) ,but not with sex ,age ,histological grade ,infiltration degree , lymph node metastasis ,and the survival rate(P>0 .05) .Conclusion The expression of Nek8 is up‐regulated in ESCC tissues and cell line ,and may be involved in tumorigenesis and development of ESCC .Nek8 could act as a potential biomarker for ESCC diag‐nose and target for therapy .

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA