Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
J Biomol Struct Dyn ; 41(19): 9503-9522, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-36326488

RESUMEN

Aurora Kinase C (AURKC) is considered an important element in Chromosome Passenger Complex (CPC), its interaction with Inner Centromere Protein (INCENP) plays a critical role in the establishment and the recruitment of a stable CPC during spermatogenesis. Genetic variations of AURKC gene are susceptible to impact AURKC-INCENP interaction, which may affect CPC stability and predispose male subjects to macrozoospermia. In this study, we systematically applied computational approaches using different bioinformatic tools to predict the effect of missense SNPs reported on AURKC gene, we selected the deleterious ones and we introduced their corresponding amino acid substitutions on AURKC protein structure. Then we did a protein-protein docking between AURKC variants and INCENP followed by a structural assessment of each resulting complex using PRODIGY server, Yassara view, Ligplot + and we choose the complexes of the most impactful variants for molecular dynamics (MD) simulation study. Seventeen missense SNPs of AURKC were identified as deleterious between all reported ones. All of them were located on relatively conserved positions on AURKC protein according to Consurf server. Only the four missense SNPs; E91K, D166V, D221Y and G235V were ranked as the most impactful ones and were chosen for MD simulation. D221Y and G235V were responsible for the most remarkable changes on AURKC-INCENP structural stability, therefore, they were selected as the most deleterious ones. Experimental studies are recommended to test the actual effect of these two variants and their actual impact on the morphology of sperm cells.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Infertilidad Masculina , Polimorfismo de Nucleótido Simple , Masculino , Humanos , Aurora Quinasa C/genética , Aurora Quinasa C/metabolismo , Polimorfismo de Nucleótido Simple/genética , Semen/metabolismo , Espermatozoides/metabolismo , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo
2.
Aging Cell ; 20(11): e13489, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34704342

RESUMEN

A hallmark of advanced maternal age is a significant increase in meiotic chromosome segregation errors, resulting in early miscarriages and congenital disorders. These errors most frequently occur during meiosis I (MI). The spindle assembly checkpoint (SAC) prevents chromosome segregation errors by arresting the cell cycle until proper chromosome alignment is achieved. Unlike in mitosis, the SAC in oocytes is desensitized, allowing chromosome segregation in the presence of improperly aligned chromosomes. Whether SAC integrity further deteriorates with advancing maternal age, and if this decline contributes to increased segregation errors remains a fundamental question. In somatic cells, activation of the SAC depends upon Aurora kinase B (AURKB), which functions to monitor kinetochore-microtubule attachments and recruit SAC regulator proteins. In mice, oocyte-specific deletion of AURKB (Aurkb cKO) results in an increased production of aneuploid metaphase II-arrested eggs and premature age-related infertility. Here, we aimed to understand the cause of the short reproductive lifespan and hypothesized that SAC integrity was compromised. In comparing oocytes from young and sexually mature Aurkb cKO females, we found that SAC integrity becomes compromised rapidly with maternal age. We show that the increased desensitization of the SAC is driven by reduced expression of MAD2, ZW10 and Securin proteins, key contributors to the SAC response pathway. The reduced expression of these proteins is the result of altered protein homeostasis, likely caused by the accumulation of reactive oxygen species. Taken together, our results demonstrate a novel function for AURKB in preserving the female reproductive lifespan possibly by protecting oocytes from oxidative stress.


Asunto(s)
Envejecimiento/metabolismo , Aurora Quinasa B/metabolismo , Puntos de Control de la Fase M del Ciclo Celular/genética , Meiosis/genética , Reproducción/genética , Transducción de Señal/genética , Huso Acromático/metabolismo , Envejecimiento/genética , Aneuploidia , Animales , Aurora Quinasa B/genética , Aurora Quinasa C/genética , Aurora Quinasa C/metabolismo , Segregación Cromosómica/genética , Cromosomas de los Mamíferos/metabolismo , Femenino , Eliminación de Gen , Edad Materna , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oocitos/metabolismo
3.
Sci Rep ; 11(1): 17444, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34465813

RESUMEN

Glioblastoma multiforme (GBM) is the most frequent type of primary astrocytomas. We examined the association between single nucleotide polymorphisms (SNPs) in Aurora kinase A (AURKA), Aurora kinase B (AURKB), Aurora kinase C (AURKC) and Polo-like kinase 1 (PLK1) mitotic checkpoint genes and GBM risk by qPCR genotyping. In silico analysis was performed to evaluate effects of polymorphic biological sequences on protein binding motifs. Chi-square and Fisher statistics revealed a significant difference in genotypes frequencies between GBM patients and controls for AURKB rs2289590 variant (p = 0.038). Association with decreased GBM risk was demonstrated for AURKB rs2289590 AC genotype (OR = 0.54; 95% CI = 0.33-0.88; p = 0.015). Furthermore, AURKC rs11084490 CG genotype was associated with lower GBM risk (OR = 0.57; 95% CI = 0.34-0.95; p = 0.031). Bioinformatic analysis of rs2289590 polymorphic region identified additional binding site for the Yin-Yang 1 (YY1) transcription factor in the presence of C allele. Our results indicated that rs2289590 in AURKB and rs11084490 in AURKC were associated with a reduced GBM risk. The present study was performed on a less numerous but ethnically homogeneous population. Hence, future investigations in larger and multiethnic groups are needed to strengthen these results.


Asunto(s)
Aurora Quinasa A/genética , Aurora Quinasa B/genética , Aurora Quinasa C/genética , Biomarcadores de Tumor/genética , Proteínas de Ciclo Celular/genética , Glioblastoma/patología , Polimorfismo de Nucleótido Simple , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Femenino , Estudios de Seguimiento , Genotipo , Glioblastoma/genética , Humanos , Puntos de Control de la Fase M del Ciclo Celular , Masculino , Persona de Mediana Edad , Pronóstico , Adulto Joven , Quinasa Tipo Polo 1
4.
Exp Cell Res ; 407(2): 112803, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34461108

RESUMEN

Aberrant expression of meiosis-specific genes in cancer has recently emerged as a driver of some cancer formation. Aurora kinase C (AURKC) is a member of the Aurora kinase family of proteins known to regulate chromosome segregation during cell divisions. AURKC is normally expressed in meiotic cells; however, elevated levels of AURKC mRNA and protein are frequently measured in cancer cells. To understand the function of AURKC in cancer cells, expression was induced in noncancerous, human retina pigmented epithelial cells. While AURKC expression did not alter cell proliferation over 72 h, it did increase cell migration and anchorage independent growth in soft agar suggesting an oncogenic role in mitotically dividing cells. To evaluate AURKC as a potential therapeutic target, a frameshift mutation in the gene was introduced in U2OS osteosarcoma cells using CRISPR-Cas9 technology resulting in a premature stop codon. Cancer cells lacking AURKC displayed no change in cell proliferation over 72 h but did migrate less and formed fewer colonies in soft agar. Whole transcriptome sequencing analysis uncovered over 400 differentially expressed genes in U2OS cells with and without AURKC. GO analysis revealed alterations in proteinaceous extracellular matrix genes including COL1A1. These data indicate that therapeutics targeting AURKC could decrease cancer cell metastasis and disease progression. Because AURKC is transcriptionally silenced in normal mitotic cells, its disruption could specifically target cancer cells limiting the toxic side effects associated with current therapeutics.


Asunto(s)
Aurora Quinasa C/metabolismo , Biomarcadores de Tumor/metabolismo , Células Epiteliales/patología , Regulación Neoplásica de la Expresión Génica , Meiosis , Neoplasias/patología , Retina/patología , Apoptosis , Aurora Quinasa C/genética , Biomarcadores de Tumor/genética , Movimiento Celular , Proliferación Celular , Células Cultivadas , Segregación Cromosómica , Células Epiteliales/metabolismo , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Pronóstico , Retina/metabolismo , Tasa de Supervivencia , Transcriptoma
5.
PLoS Genet ; 17(4): e1009327, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33901174

RESUMEN

The Aurora protein kinases are well-established regulators of spindle building and chromosome segregation in mitotic and meiotic cells. In mouse oocytes, there is significant Aurora kinase A (AURKA) compensatory abilities when the other Aurora kinase homologs are deleted. Whether the other homologs, AURKB or AURKC can compensate for loss of AURKA is not known. Using a conditional mouse oocyte knockout model, we demonstrate that this compensation is not reciprocal because female oocyte-specific knockout mice are sterile, and their oocytes fail to complete meiosis I. In determining AURKA-specific functions, we demonstrate that its first meiotic requirement is to activate Polo-like kinase 1 at acentriolar microtubule organizing centers (aMTOCs; meiotic spindle poles). This activation induces fragmentation of the aMTOCs, a step essential for building a bipolar spindle. We also show that AURKA is required for regulating localization of TACC3, another protein required for spindle building. We conclude that AURKA has multiple functions essential to completing MI that are distinct from AURKB and AURKC.


Asunto(s)
Aurora Quinasa A/genética , Proteínas de Ciclo Celular/genética , Proteínas Fetales/genética , Meiosis/genética , Proteínas Asociadas a Microtúbulos/genética , Oocitos/crecimiento & desarrollo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Animales , Aurora Quinasa B/genética , Aurora Quinasa C/genética , División del Núcleo Celular/genética , Segregación Cromosómica/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Ratones , Centro Organizador de los Microtúbulos/metabolismo , Oocitos/metabolismo , Huso Acromático/genética , Polos del Huso/genética , Quinasa Tipo Polo 1
6.
J Assist Reprod Genet ; 38(6): 1571-1588, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33725274

RESUMEN

PURPOSE: Is it possible to eliminate metastasised chronic myeloid leukaemia (CML) and acute myeloid leukaemia (AML) cells from ovarian cortex fragments by inhibition of Aurora B/C kinases (AURKB/C) without compromising ovarian tissue or follicles? METHODS: Human ovarian cortex tissue with experimentally induced tumour foci of CML, AML and primary cells of AML patients were exposed to a 24h treatment with 1 µM GSK1070916, an AURKB/C inhibitor, to eliminate malignant cells by invoking mitotic catastrophe. After treatment, the inhibitor was removed, followed by an additional culture period of 6 days to allow any remaining tumour cells to form new foci. Ovarian tissue integrity after treatment was analysed by four different assays. Appropriate controls were included in all experiments. RESULTS: Foci of metastasised CML and AML cells in ovarian cortex tissue were severely affected by a 24h ex vivo treatment with an AURKB/C inhibitor, leading to the formation of multi-nuclear syncytia and large-scale apoptosis. Ovarian tissue morphology and viability was not compromised by the treatment, as no significant difference was observed regarding the percentage of morphologically normal follicles, follicular viability, glucose uptake or in vitro growth of small follicles between ovarian cortex treated with 1 µM GSK1070916 and the control. CONCLUSION: Purging of CML/AML metastases in ovarian cortex is possible by targeting the Mitotic Catastrophe Signalling Pathway using GSK1070916 without affecting the ovarian tissue. This provides a therapeutic strategy to prevent reintroduction of leukaemia and enhances safety of autotransplantation in leukaemia patients currently considered at high risk for ovarian involvement.


Asunto(s)
Aurora Quinasa B/genética , Aurora Quinasa C/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mieloide Aguda/genética , Apoptosis/efectos de los fármacos , Compuestos Aza/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Criopreservación , Femenino , Humanos , Indoles/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Leucemia Mielógena Crónica BCR-ABL Positiva/terapia , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/terapia , Mitosis/efectos de los fármacos , Mitosis/genética , Metástasis de la Neoplasia , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/crecimiento & desarrollo , Transducción de Señal/efectos de los fármacos , Trasplante Autólogo/normas
7.
Reproduction ; 161(2): R13-R35, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33170803

RESUMEN

Idiopathic or 'unexplained' infertility represents as many as 30% of infertility cases worldwide. Conception, implantation, and term delivery of developmentally healthy infants require chromosomally normal (euploid) eggs and sperm. The crux of euploid egg production is error-free meiosis. Pathologic genetic variants dysregulate meiotic processes that occur during prophase I, meiotic resumption, chromosome segregation, and in cell cycle regulation. This dysregulation can result in chromosomally abnormal (aneuploid) eggs. In turn, egg aneuploidy leads to a broad range of clinical infertility phenotypes, including primary ovarian insufficiency and early menopause, egg fertilization failure and embryonic developmental arrest, or recurrent pregnancy loss. Therefore, maternal genetic variants are emerging as infertility biomarkers, which could allow informed reproductive decision-making. Here, we select and deeply examine human genetic variants that likely cause dysregulation of critical meiotic processes in 14 female infertility-associated genes: SYCP3, SYCE1, TRIP13, PSMC3IP, DMC1, MCM8, MCM9, STAG3, PATL2, TUBB8, CEP120, AURKB, AURKC, andWEE2. We discuss the function of each gene in meiosis, explore genotype-phenotype relationships, and delineate the frequencies of infertility-associated variants.


Asunto(s)
Infertilidad Femenina , ATPasas Asociadas con Actividades Celulares Diversas , Aneuploidia , Aurora Quinasa C/genética , Aurora Quinasa C/metabolismo , Proteínas de Ciclo Celular/genética , Segregación Cromosómica , Femenino , Humanos , Infertilidad Femenina/genética , Masculino , Meiosis , Proteínas Nucleares , Embarazo , Espermatozoides/metabolismo , Transactivadores , Tubulina (Proteína)
8.
Best Pract Res Clin Endocrinol Metab ; 34(6): 101473, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33183966

RESUMEN

Spermatozoa are polarized cells with a head and a flagellum joined by the connecting piece. Head integrity is critical for normal sperm function, and head defects consistently lead to male infertility. Abnormalities of the sperm head are among the most severe and characteristic sperm defects. Patients presenting with a monomorphic head sperm defects such as globozoospermia or marcrozoospermia were analyzed permitting to identify several key genes for spermatogenesis such as AURKC and DPY19L2. The study of patients with other specific sperm head defects such as acephalic spermatozoa have also enabled the identification of new infertility genes such as SUN5. Here, we review the genetic causes leading to morphological defects of sperm head. Advances in the genetics of male infertility are necessary to improve the management of infertility and will pave the road towards future strategies of treatments, especially for patients with the most severe phenotype as sperm head defects.


Asunto(s)
Cabeza del Espermatozoide/patología , Espermatozoides/anomalías , Teratozoospermia/genética , Aurora Quinasa C/genética , Humanos , Infertilidad Masculina/genética , Infertilidad Masculina/patología , Masculino , Proteínas de la Membrana/genética , Cabeza del Espermatozoide/metabolismo , Espermatogénesis/genética , Espermatozoides/metabolismo , Espermatozoides/patología , Teratozoospermia/patología
9.
J Cell Sci ; 133(23)2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-33172986

RESUMEN

Precise control of chromosome dynamics during meiosis is critical for fertility. A gametocyte undergoing meiosis coordinates formation of the synaptonemal complex (SC) to promote efficient homologous chromosome recombination. Subsequent disassembly of the SC occurs prior to segregation of homologous chromosomes during meiosis I. We examined the requirements of the mammalian Aurora kinases (AURKA, AURKB and AURKC) during SC disassembly and chromosome segregation using a combination of chemical inhibition and gene deletion approaches. We find that both mouse and human spermatocytes fail to disassemble SC lateral elements when the kinase activity of AURKB and AURKC are chemically inhibited. Interestingly, both Aurkb conditional knockout and Aurkc knockout mouse spermatocytes successfully progress through meiosis, and the mice are fertile. In contrast, Aurkb, Aurkc double knockout spermatocytes fail to coordinate disassembly of SC lateral elements with chromosome condensation and segregation, resulting in delayed meiotic progression. In addition, deletion of Aurkb and Aurkc leads to an accumulation of metaphase spermatocytes, chromosome missegregation and aberrant cytokinesis. Collectively, our data demonstrate that AURKB and AURKC functionally compensate for one another ensuring successful mammalian spermatogenesis.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Aurora Quinasa B , Aurora Quinasa C , Meiosis , Oocitos , Espermatogénesis , Animales , Aurora Quinasa B/genética , Aurora Quinasa C/genética , Segregación Cromosómica/genética , Humanos , Masculino , Ratones , Espermatocitos , Espermatogénesis/genética
10.
Andrologia ; 52(11): e13868, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33118205

RESUMEN

Macrozoospermia is associated with severe male infertility. To date, the only gene implicated in this phenotype is the Aurora Kinase C gene. We report in this work the genetic screening of AURKC mutations in 34 patients with macrozoospermia among 3,536 Algerian infertile men. Nineteen patients (56%) were homozygotes for the c.144delC mutation, eight (23.52%) homozygotes for the c.744C>G (p.Y248*) mutation and two (5.88%) compound heterozygotes. No AURKC mutation was identified in five patients (14.7%). Interestingly and although it is generally accepted that nearly all positive mutated AURKC patients have close to 100% large-head spermatozoa, our results showed that 11 patients with AURKC mutations (32.35%) had large-headed spermatozoa lower than 70% (7 with c.144delC and 4 with p.Y248*), and no mutation was found in 2 patients who had >70% of macrocephalic spermatozoa. Twenty ICSI attempts were performed before genetic screening resulting in 39 embryos but no pregnancy was obtained. The sequencing of AURKC exons 3 and 6 is appropriate as a first-line genetic exploration in these patients to avoid unsuccessful ICSI attempts. A percentage of large head spermatozoa beyond 25% and a percentage of multiflagellar spermatozoa beyond 10% are predictive of a positive mutation diagnosis.


Asunto(s)
Infertilidad Masculina , Aurora Quinasa C/genética , Homocigoto , Humanos , Infertilidad Masculina/genética , Masculino , Mutación , Espermatozoides
11.
Andrologia ; 52(9): e13663, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32478938

RESUMEN

Among causes of infertility, teratozoospermia is characterised by a percentage of morphologically abnormal spermatozoa >4%. Macrozoospermia, one form of monomorphic teratozoospermia, is observed in <1% of cases of male infertility and is described as approximately 100% large-headed and/or multitailed spermatozoa. This study reports that an infertile man with large-head spermatozoa presenting compound heterozygosity aurora kinase C (AURKC) mutations (c.382C>T, c.572C>T) by whole-exome sequencing. Consequently, both two novel AURKC mutations had high probability of damage-causing and conserved across species and extremely low allele frequency in the population. Flow cytometry analysis revealed a high ratio of sperm DNA fragmentation. Two intracytoplasmic sperm injection (ICSI) procedures were attempted for the patient, but all were unsuccessful. These results indicate that sequence analysis should be performed for the variants of AURKC in Chinese patients with macrozoospermia.


Asunto(s)
Infertilidad Masculina , Teratozoospermia , Aurora Quinasa C/genética , Humanos , Infertilidad Masculina/genética , Masculino , Mutación , Espermatozoides , Teratozoospermia/genética
12.
Andrologia ; 52(7): e13619, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32399982

RESUMEN

Macrozoospermia is a rare syndrome. The key marker of the disease is a high percentage of spermatozoa with abnormal phenotypes namely enlarged head and multiple tails. The presence of at least 70% of spermatozoa with a large head is usually associated with Aurora kinase C gene (AURKC) mutations. We sought to assess AURKC as a potential genetic actor of macrozoospermia in a sample of infertile Egyptian men. We recruited 30 patients and conducted a clinical examination, semen analysis, and DNA sequencing and RFLP for AURKC. We diagnosed 17 patients with characteristic macrozoospermia and classified them into eight severe and nine mild cases. We detected genetic variants of AURKC in five patients (29.4%): Three patients with severe macrozoospermia had c.144delC mutations in exon 3 (37.5% of the severe), and two mild cases had c.1157G>A polymorphism in the 3' UTR (22.2% of the mild). A successful intracytoplasmic sperm injection (ICSI) was achieved only with a severe macrozoospermia patient without apparent AURKC mutation. The present study is the first report to link macrozoospermia and AURKC mutations in Egypt. The study recommends macrozoospermia patients to perform AURKC gene analysis and attempt ICSI, even those with a high percentage of large head spermatozoa.


Asunto(s)
Infertilidad Masculina , Aurora Quinasa C/genética , Análisis Mutacional de ADN , Egipto , Humanos , Infertilidad Masculina/genética , Masculino , Espermatozoides
13.
Rev Int Androl ; 18(4): 159-163, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31455599

RESUMEN

A Chilean 35-year-old male patient with a history of primary infertility made an appointment at the Unit of Reproductive Medicine at Clínica Las Condes, Santiago, Chile. Multiple semen analyses revealed abnormal sperm morphology as the most prevalent finding. Multiflagellated and macrocephalic spermatozoa were observed and indicated a possible macrozoospermic phenotype. The constant presence of abnormal sperm morphology led the scope of the study to include Aurora Kinase C (AURKC) gene sequencing. The patient was diagnosed with a homozygous mutation of this gene. The mutation was detected in exon 6, type c.744C>G+/+ (P.Y248*) variant. As previously described in the Human Gene Mutation Database (HGMD), this pathogenic variant is associated with macrozoospermia. Although this mutation is not the most frequently observed, it is the first of its kind reported in Latin America.


Asunto(s)
Aurora Quinasa C/genética , Infertilidad Masculina/genética , Espermatozoides/anomalías , Adulto , Chile , Exones , Humanos , Infertilidad Masculina/diagnóstico , América Latina , Masculino , Mutación , Teratozoospermia/genética
14.
PLoS Genet ; 15(12): e1008414, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31830031

RESUMEN

Human nondisjunction errors in oocytes are the leading cause of pregnancy loss, and for pregnancies that continue to term, the leading cause of intellectual disabilities and birth defects. For the first time, we have conducted a candidate gene and genome-wide association study to identify genes associated with maternal nondisjunction of chromosome 21 as a first step to understand predisposing factors. A total of 2,186 study participants were genotyped on the HumanOmniExpressExome-8v1-2 array. These participants included 749 live birth offspring with standard trisomy 21 and 1,437 parents. Genotypes from the parents and child were then used to identify mothers with nondisjunction errors derived in the oocyte and to establish the type of error (meiosis I or meiosis II). We performed a unique set of subgroup comparisons designed to leverage our previous work suggesting that the etiologies of meiosis I and meiosis II nondisjunction differ for trisomy 21. For the candidate gene analysis, we selected genes associated with chromosome dynamics early in meiosis and genes associated with human global recombination counts. Several candidate genes showed strong associations with maternal nondisjunction of chromosome 21, demonstrating that genetic variants associated with normal variation in meiotic processes can be risk factors for nondisjunction. The genome-wide analysis also suggested several new potentially associated loci, although follow-up studies using independent samples are required.


Asunto(s)
Síndrome de Down/genética , Estudio de Asociación del Genoma Completo/métodos , No Disyunción Genética/genética , Aurora Quinasa C/genética , Proteínas de Transporte de Catión/genética , Niño , Síndrome de Down/etnología , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Meiosis , Madres , Oocitos , Estados Unidos/etnología , Factor A de Crecimiento Endotelial Vascular/genética
15.
J Assist Reprod Genet ; 36(3): 529-534, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30594972

RESUMEN

PURPOSE: Macrozoospermia is a rare sperm morphologic abnormality associated with male infertility and is characterized by a high percentage of spermatozoa with large irregular heads. The aim of this study was to identify the genetic cause of an infertile male with macrozoospermia from a consanguineous family. METHODS: Whole-exome sequencing (WES) was performed using peripheral blood genomic DNA from the patient and his parents. RESULTS: WES analysis of the patient with macrozoospermia from a consanguineous family allowed the identification of a novel homozygous missense variant in the AURKC gene (c.269G>A). Bioinformatics analysis also suggested this variant a pathogenic mutation. Quantitative real-time PCR analysis showed that the mRNA level of AURKC is significantly decreased in the patient compared with his father. Moreover, no embryos were available for transfer after ICSI. CONCLUSIONS: These results further support the important role of AURKC in male infertility and guide the practitioner in optimal decision making for patients with macrozoospermia.


Asunto(s)
Aurora Quinasa C/genética , Infertilidad Masculina/genética , Teratozoospermia/genética , Adulto , Homocigoto , Humanos , Infertilidad Masculina/patología , Masculino , Mutación/genética , Linaje , Cabeza del Espermatozoide/patología , Espermatozoides/crecimiento & desarrollo , Espermatozoides/patología , Teratozoospermia/patología , Secuenciación del Exoma
16.
Klin Onkol ; 31(6): 429-433, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30545223

RESUMEN

BACKGROUND: Cancer testis antigens (CTAs) are considered cancer bio-markers due to their highly specific expression pattern in human malignancies and near absence from normal somatic tissues. Their specific expression has made them potential targets for early dia-gnosis, assessment of patients prognosis and treatment of cancer in recent years. Lactobacilli are a group of probio-tics with anti-cancer, immunomodulatory and other beneficial features. These bacteria have been shown to alter expression of several cancer-related genes. AIM: We investigated the effect of Lactobacillus rhamnosus GG supernatant (LRS) and Lactobacillus crispatus SJ-3C-US supernatant (LCS) on expression of four CTAs (TSGA10, AURKC, OIP5 and AKAP4) in HeLa cell line after synchronization using MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay and quantitative real-time polymerase chain reaction. RESULTS: LRS and LCS inhibited HeLa cell growth after 24 h as demonstrated by MTT assay. Expressions of all CTAs were down-regulated after treatment with both supernatants. CONCLUSION: This study showed the role of Lactobacilli in down-regulation of CTAs genes. Such expression change might be involved in the anticancer effects of these Lactobacilli. The underlying mechanisms of these observations are not clear but epigenetic modulatory mechanisms may participate in this process. Future studies are needed to assess functional roles of Lactobacilli in modulation of other cancer-related genes. Key words: probio-tic - cancer testis antigen - bio-marker - HeLa cell line.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/genética , Aurora Quinasa C/genética , Proteínas Cromosómicas no Histona/genética , Lacticaseibacillus rhamnosus , Lactobacillus crispatus , Proteínas/genética , Proteínas de Ciclo Celular , Proteínas del Citoesqueleto , Regulación hacia Abajo , Células HeLa , Humanos
17.
Curr Biol ; 28(21): 3458-3468.e5, 2018 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-30415701

RESUMEN

Errors in chromosome segregation during female meiosis I occur frequently, and aneuploid embryos account for 1/3 of all miscarriages in humans [1]. Unlike mitotic cells that require two Aurora kinase (AURK) homologs to help prevent aneuploidy (AURKA and AURKB), mammalian germ cells also require a third (AURKC) [2, 3]. AURKA is the spindle-pole-associated homolog, and AURKB/C are the chromosome-localized homologs. In mitosis, AURKB has essential roles as the catalytic subunit of the chromosomal passenger complex (CPC), regulating chromosome alignment, kinetochore-microtubule attachments, cohesion, the spindle assembly checkpoint, and cytokinesis [4, 5]. In mouse oocyte meiosis, AURKC takes over as the predominant CPC kinase [6], although the requirement for AURKB remains elusive [7]. In the absence of AURKC, AURKB compensates, making defining potential non-overlapping functions difficult [6, 8]. To investigate the role(s) of AURKB and AURKC in oocytes, we analyzed oocyte-specific Aurkb and Aurkc single- and double-knockout (KO) mice. Surprisingly, we find that double KO female mice are fertile. We demonstrate that, in the absence of AURKC, AURKA localizes to chromosomes in a CPC-dependent manner. These data suggest that AURKC prevents AURKA from localizing to chromosomes by competing for CPC binding. This competition is important for adequate spindle length to support meiosis I. We also describe a unique requirement for AURKB to negatively regulate AURKC to prevent aneuploidy. Together, our work reveals oocyte-specific roles for the AURKs in regulating each other's localization and activity. This inter-kinase regulation is critical to support wild-type levels of fecundity in female mice.


Asunto(s)
Aurora Quinasa A/genética , Aurora Quinasa B/genética , Aurora Quinasa C/genética , Meiosis , Oocitos/metabolismo , Aneuploidia , Animales , Aurora Quinasa A/metabolismo , Aurora Quinasa B/metabolismo , Aurora Quinasa C/metabolismo , Segregación Cromosómica/genética , Femenino , Fertilidad/genética , Ratones
18.
Virology ; 523: 41-51, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30077875

RESUMEN

Several studies have related epigenetic mechanisms to HIV-1 latency. However, the epigenetic modifications of the host cell genome involved in the early stages of HIV-1 infection remain unclear. This study aimed to investigate epigenetic factors that are regulated at the beginning of HIV-1 infection in activated and resting CD4+ T cells. We analyzed the gene expression of 84 epigenetic targets, global DNA methylation, and HIV-1 replication kinetics for 36 h after infecting CD4+ T cells obtained from the blood of twelve healthy donors. The epigenetic targets aurora kinase B (AURKB), aurora kinase C (AURKC) and DNA methyltransferase 3B (DNMT3B), and the global DNA methylation profile are regulated during HIV-1 replication in CD4+ T cells, and this regulation can be influenced by the activation state of the cell at the time of infection. Approaches that affect the expression of these epigenetic targets could help current strategies to suppress HIV-1 replication.


Asunto(s)
Aurora Quinasa B/genética , Aurora Quinasa C/genética , Linfocitos T CD4-Positivos/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , Epigénesis Genética , VIH-1/fisiología , Interacciones Huésped-Patógeno , Adulto , Aurora Quinasa B/metabolismo , Aurora Quinasa C/metabolismo , Linfocitos T CD4-Positivos/virología , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Perfilación de la Expresión Génica , Voluntarios Sanos , Humanos , Activación de Linfocitos , Análisis por Micromatrices , Cultivo Primario de Células , Transducción de Señal , Internalización del Virus , Latencia del Virus , Replicación Viral , ADN Metiltransferasa 3B
19.
Artículo en Inglés | MEDLINE | ID: mdl-29307372

RESUMEN

Resveratrol (RSV) is a naturally occurring polyphenolic phytoalexin possessing numerous health-promoting effects. Chromosomal instability (CIN), usually results from defective spindle assembly checkpoint (SAC), is a major contributor to many diseases. While it's recently recognized that RSV exhibits a nonlinear dose response for disease prevention, whether it's the case for its role in CIN remains unknown. Here, we investigated the potential of a broad range of RSV concentrations (0.01-100µM) on CIN and the underlying mechanisms in human normal colon epithelial NCM460 cells. CIN was measured by cytokinesis-block micronucleus assay; mitotic fidelity was determined by aberrant mitosis analysis; SAC activity was assessed by nocodazole-challenge assay, and the expression of SAC genes was examined by RT-qPCR. We found that 0.1µM RSV significantly reduced CIN (P<0.01), while 100µM RSV significantly induced it (P<0.05). Mitotic infidelity was significantly prevented by 0.1µM RSV but promoted by 100µM RSV (P<0.05 for both). Moreover, the function of SAC was sustained and impaired by 0.1µM and 100µM RSV, respectively. Several SAC genes, including Aurora-B, Aurora-C, Plk-1 and CENP-E, were significantly up-regulated and down-regulated by 0.1µM and 100µM RSV, respectively (P<0.05). In conclusion, RSV exhibited a biphasic dose-dependent effect on CIN that was exerted via the regulation of mitotic fidelity through the SAC network. The health implications of these findings were summarized.


Asunto(s)
Colon/citología , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Resveratrol/toxicidad , Aurora Quinasa B/genética , Aurora Quinasa C/genética , Proteínas de Ciclo Celular/genética , Línea Celular , Inestabilidad Cromosómica , Proteínas Cromosómicas no Histona/genética , Colon/efectos de los fármacos , Colon/metabolismo , Relación Dosis-Respuesta a Droga , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Quinasa Tipo Polo 1
20.
Front Biosci (Elite Ed) ; 10(1): 143-154, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28930610

RESUMEN

Wilms' tumor (WT) is the most frequent renal cancer in childhood, the occurrence of which is characterized by a relatively low frequency of associated mutations. While epigenetic alterations have been postulated to play a relevant role in the emergence of this tumor, the mechanisms involved in WT development remain largely unknown. In this study, the DNA methylation profile of WT was characterized with Beadchip array. Comparisons between WT with normal kidney identified 827 differentially methylated regions, most of which were attributable in hypermethylation in CpG islands. Among affected genes, WT1 and TP73 showed altered enhancers where hypermethylation was validaded by pyrosequencing. Thirty differentially methylated regions (DMRs) were identified in WT as compared to normal kidney, two of which were previously described. Two novel DMRs, located in RPS6KA4/MIR1237 and the AURKC promoter, were found to be hypermethylated in WT. Altogether, our data reinforced the relevance of alterations of DNA methylation in WT, highlighting the complex nature of these alterations that affect promoter regions as well as enhancers, UTRs and gene bodies.


Asunto(s)
Aurora Quinasa C/genética , Metilación de ADN , MicroARNs/genética , Regiones Promotoras Genéticas , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética , Tumor de Wilms/genética , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...