Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biomol Struct Dyn ; 41(19): 9503-9522, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-36326488

RESUMO

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.


Assuntos
Infertilidade Masculina , Polimorfismo de Nucleotídeo Único , Masculino , Humanos , Aurora Quinase C/genética , Aurora Quinase C/metabolismo , Polimorfismo de Nucleotídeo Único/genética , Sêmen/metabolismo , Espermatozoides/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo
2.
Reproduction ; 164(4): V5-V7, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-36125382

RESUMO

In brief: The Aurora protein kinases have critical functions in controlling oocyte meiotic maturation. In this study, we describe an assay for examining their activation state in oocytes and establish the best working doses of three commonly used inhibitors. Abstract: Several small molecule inhibitors exist for targeting Aurora kinase proteins in somatic cells. From this point of view, we evaluate the specificity of these inhibitors in mouse oocytes, and we demonstrate that MLN 8237 and AZD 1152 are specific for Aurora kinase A and Aurora kinase C, respectively, only when used at low concentrations.


Assuntos
Aurora Quinase A , Meiose , Animais , Aurora Quinase A/metabolismo , Aurora Quinase C/metabolismo , Camundongos , Oócitos/metabolismo , Proteínas Quinases/metabolismo
3.
Aging Cell ; 20(11): e13489, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34704342

RESUMO

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.


Assuntos
Envelhecimento/metabolismo , Aurora Quinase B/metabolismo , Pontos de Checagem da Fase M do Ciclo Celular/genética , Meiose/genética , Reprodução/genética , Transdução de Sinais/genética , Fuso Acromático/metabolismo , Envelhecimento/genética , Aneuploidia , Animais , Aurora Quinase B/genética , Aurora Quinase C/genética , Aurora Quinase C/metabolismo , Segregação de Cromossomos/genética , Cromossomos de Mamíferos/metabolismo , Feminino , Deleção de Genes , Idade Materna , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oócitos/metabolismo
4.
Exp Cell Res ; 407(2): 112803, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34461108

RESUMO

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.


Assuntos
Aurora Quinase C/metabolismo , Biomarcadores Tumorais/metabolismo , Células Epiteliais/patologia , Regulação Neoplásica da Expressão Gênica , Meiose , Neoplasias/patologia , Retina/patologia , Apoptose , Aurora Quinase C/genética , Biomarcadores Tumorais/genética , Movimento Celular , Proliferação de Células , Células Cultivadas , Segregação de Cromossomos , Células Epiteliais/metabolismo , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Prognóstico , Retina/metabolismo , Taxa de Sobrevida , Transcriptoma
5.
BMC Cancer ; 21(1): 596, 2021 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-34030642

RESUMO

BACKGROUND: Phosphorylation of NF-kappaB inhibitor alpha (IκBα) is key to regulation of NF-κB transcription factor activity in the cell. Several sites of IκBα phosphorylation by members of the IκB kinase family have been identified, but phosphorylation of the protein by other kinases remains poorly understood. We investigated a new phosphorylation site on IκBα and identified its biological function in breast cancer cells. METHODS: Previously, we observed that aurora kinase (AURK) binds IκBα in the cell. To identify the domains of IκBα essential for phosphorylation by AURK, we performed kinase assays with a series of IκBα truncation mutants. AURK significantly promoted activation of IκBα at serine 32 but not serine 36; by contrast, IκB kinase (IKK) family proteins activated both of these residues. We also confirmed phosphorylation of IκBα by matrix-assisted laser-desorption/ionization time-of-flight mass spectrometry (MALDI-TOF/TOF MS) and nano-liquid chromatography hybrid quadrupole orbitrap mass spectrometer (nanoLC-MS/MS; Q-Exactive). RESULTS: We identified two novel sites of serine phosphorylation, S63 and S262. Alanine substitution of S63 and S262 (S63A and S262A) of IκBα inhibited proliferation and suppressed p65 transcription activity. In addition, S63A and/or S262A of IκBα regulated apoptotic and necroptotic effects in breast cancer cells. CONCLUSIONS: Phosphorylation of IκBα by AURK at novel sites is related to the apoptosis and necroptosis pathways in breast cancer cells.


Assuntos
Aurora Quinase C/metabolismo , Neoplasias da Mama/patologia , Inibidor de NF-kappaB alfa/metabolismo , Necroptose , Sítios de Ligação/genética , Feminino , Humanos , Células MCF-7 , Mutagênese Sítio-Dirigida , Inibidor de NF-kappaB alfa/genética , Inibidor de NF-kappaB alfa/isolamento & purificação , NF-kappa B/metabolismo , Fosforilação , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Espectrometria de Massas em Tandem/métodos
6.
Reproduction ; 161(2): R13-R35, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33170803

RESUMO

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.


Assuntos
Infertilidade Feminina , ATPases Associadas a Diversas Atividades Celulares , Aneuploidia , Aurora Quinase C/genética , Aurora Quinase C/metabolismo , Proteínas de Ciclo Celular/genética , Segregação de Cromossomos , Feminino , Humanos , Infertilidade Feminina/genética , Masculino , Meiose , Proteínas Nucleares , Gravidez , Espermatozoides/metabolismo , Transativadores , Tubulina (Proteína)
7.
Nat Commun ; 10(1): 3166, 2019 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-31320618

RESUMO

Aurora kinases B and C (AURKB/AURKC) are activated by binding to the C-terminal domain of INCENP. Full activation requires phosphorylation of two serine residues of INCENP that are conserved through evolution, although the mechanism of this activation has not been explained. Here we present crystal structures of the fully active complex of AURKC bound to INCENP, consisting of phosphorylated, activated, AURKC and INCENP phosphorylated on its TSS motif, revealing the structural and biochemical mechanism of synergistic activation of AURKC:INCENP. The structures show that TSS motif phosphorylation stabilises the kinase activation loop of AURKC. The TSS motif phosphorylations alter the substrate-binding surface consistent with a mechanism of altered kinase substrate selectivity and stabilisation of the protein complex against unfolding. We also analyse the binding of the most specific available AURKB inhibitor, BRD-7880, and demonstrate that the well-known Aurora kinase inhibitor VX-680 disrupts binding of the phosphorylated INCENP TSS motif.


Assuntos
Aurora Quinase B/metabolismo , Aurora Quinase C/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Cristalografia por Raios X , Ativação Enzimática , Humanos , Fosforilação , Piperazinas/farmacologia , Ligação Proteica/efeitos dos fármacos , Domínios Proteicos , Dobramento de Proteína , Serina/metabolismo
8.
Curr Biol ; 28(21): 3458-3468.e5, 2018 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-30415701

RESUMO

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.


Assuntos
Aurora Quinase A/genética , Aurora Quinase B/genética , Aurora Quinase C/genética , Meiose , Oócitos/metabolismo , Aneuploidia , Animais , Aurora Quinase A/metabolismo , Aurora Quinase B/metabolismo , Aurora Quinase C/metabolismo , Segregação de Cromossomos/genética , Feminino , Fertilidade/genética , Camundongos
9.
Virology ; 523: 41-51, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30077875

RESUMO

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.


Assuntos
Aurora Quinase B/genética , Aurora Quinase C/genética , Linfócitos T CD4-Positivos/metabolismo , DNA (Citosina-5-)-Metiltransferases/genética , Epigênese Genética , HIV-1/fisiologia , Interações Hospedeiro-Patógeno , Adulto , Aurora Quinase B/metabolismo , Aurora Quinase C/metabolismo , Linfócitos T CD4-Positivos/virologia , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Perfilação da Expressão Gênica , Voluntários Saudáveis , Humanos , Ativação Linfocitária , Análise em Microsséries , Cultura Primária de Células , Transdução de Sinais , Internalização do Vírus , Latência Viral , Replicação Viral , DNA Metiltransferase 3B
10.
Int J Mol Sci ; 19(1)2017 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-29283376

RESUMO

Aurora kinases (AKs) are serine/threonine kinases that are essential for cell division. Humans have three AK genes: AKA, AKB, and AKC. AKA is required for centrosome assembly, centrosome separation, and bipolar spindle assembly, and its mutation leads to abnormal spindle morphology. AKB is required for the spindle checkpoint and proper cytokinesis, and mutations cause chromosome misalignment and cytokinesis failure. AKC is expressed in germ cells, and has a role in meiosis analogous to that of AKB in mitosis. Mutation of any of the three isoforms can lead to cancer. AK proteins possess divergent N- and C-termini and a conserved central catalytic domain. We examined the evolution of the AK gene family using an identity matrix and by building a phylogenetic tree. The data suggest that AKA is the vertebrate ancestral gene, and that AKB and AKC resulted from gene duplication in placental mammals. In a nonsynonymous/synonymous rate substitution analysis, we found that AKB experienced the strongest, and AKC the weakest, purifying selection. Both the N- and C-termini and regions within the kinase domain experienced differential selection among the AK isoforms. These differentially selected sequences may be important for species specificity and isoform specificity, and are therefore potential therapeutic targets.


Assuntos
Aurora Quinase A/genética , Aurora Quinase B/genética , Aurora Quinase C/genética , Citocinese , Mitose , Seleção Genética , Sequência de Aminoácidos , Animais , Aurora Quinase A/metabolismo , Aurora Quinase B/metabolismo , Aurora Quinase C/metabolismo , Centrossomo/ultraestrutura , Evolução Molecular , Duplicação Gênica , Expressão Gênica , Humanos , Meiose , Filogenia , Plantas/classificação , Plantas/genética , Domínios Proteicos , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Fuso Acromático/metabolismo , Fuso Acromático/ultraestrutura
11.
Biol Reprod ; 96(6): 1197-1209, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28575288

RESUMO

During oocyte meiotic maturation, Aurora kinase C (AURKC) is required to accomplish many critical functions including destabilizing erroneous kinetochore-microtubule (K-MT)attachments and regulating bipolar spindle assembly. How localized activity of AURKC is regulated in mammalian oocytes, however, is not fully understood. Female gametes from many species, including mouse, contain stores of maternal transcripts that are required for downstream developmental events. We show here that depletion of maternal RNA in mouse oocytes resulted in impaired meiotic progression, increased incidence of chromosome misalignment and abnormal spindle formation at metaphase I (Met I), and cytokinesis defects. Importantly, depletion of maternal RNA perturbed the localization and activity of AURKC within the chromosomal passenger complex (CPC). These perturbations were not observed when translation was inhibited by cycloheximide (CHX) treatment. These results demonstrate a translation-independent function of maternal RNA to regulate AURKC-CPC function in mouse oocytes.


Assuntos
Aurora Quinase C/metabolismo , Oócitos/fisiologia , Biossíntese de Proteínas/fisiologia , RNA Mensageiro Estocado/fisiologia , Animais , Aurora Quinase C/genética , Clonagem Molecular , Feminino , Meiose/fisiologia , Camundongos
12.
Mol Biol Cell ; 28(17): 2233-2240, 2017 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-28659416

RESUMO

Aneuploidy is the leading genetic abnormality contributing to infertility, and chromosome segregation errors are common during female mammalian meiosis I (MI). Previous results indicate that haspin kinase regulates resumption of meiosis from prophase arrest, chromosome condensation, and kinetochore-microtubule attachments during early prometaphase of MI. Here we report that haspin inhibition in late prometaphase I causes acceleration of MI, bypass of the spindle assembly checkpoint (SAC), and loss of interchromatid axis-localized Aurora kinase C. Meiotic cells contain a second chromosomal passenger complex (CPC) population, with Aurora kinase B (AURKB) bound to INCENP. Haspin inhibition in oocytes from Aurkc-/- mice, where AURKB is the sole CPC kinase, does not alter MI completion timing, and no change in localization of the SAC protein, MAD2, is observed. These data suggest that AURKB on the interchromatid axis is not needed for SAC activation and illustrate a key difference between the functional capacities of the two AURK homologues.


Assuntos
Aurora Quinase C/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Anáfase , Aneuploidia , Animais , Aurora Quinase B/genética , Aurora Quinase B/metabolismo , Aurora Quinase C/genética , Segregação de Cromossomos , Feminino , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Cinetocoros/metabolismo , Pontos de Checagem da Fase M do Ciclo Celular/fisiologia , Proteínas Mad2/metabolismo , Meiose/fisiologia , Camundongos , Oócitos/metabolismo , Prometáfase , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética
13.
Curr Biol ; 27(10): 1462-1476.e5, 2017 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-28502659

RESUMO

In mammalian females, germ cells remain arrested as primordial follicles. Resumption of meiosis is heralded by germinal vesicle breakdown, condensation of chromosomes, and their eventual alignment on metaphase plates. At the first meiotic division, anaphase-promoting complex/cyclosome associated with Cdc20 (APC/CCdc20) activates separase and thereby destroys cohesion along chromosome arms. Because cohesion around centromeres is protected by shugoshin-2, sister chromatids remain attached through centromeric/pericentromeric cohesin. We show here that, by promoting proteolysis of cyclins and Cdc25B at the germinal vesicle (GV) stage, APC/C associated with the Cdh1 protein (APC/CCdh1) delays the increase in Cdk1 activity, leading to germinal vesicle breakdown (GVBD). More surprisingly, by moderating the rate at which Cdk1 is activated following GVBD, APC/CCdh1 creates conditions necessary for the removal of shugoshin-2 from chromosome arms by the Aurora B/C kinase, an event crucial for the efficient resolution of chiasmata.


Assuntos
Ciclossomo-Complexo Promotor de Anáfase/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cromossomos , Meiose , Animais , Subunidade Apc2 do Ciclossomo-Complexo Promotor de Anáfase/metabolismo , Aurora Quinase B/metabolismo , Aurora Quinase C/metabolismo , Proteína Quinase CDC2/metabolismo , Proteínas Cdc20/fisiologia , Proteínas Cdh1/metabolismo , Centrômero , Proteínas Cromossômicas não Histona/metabolismo , Feminino , Centro Germinativo , Masculino , Camundongos , Camundongos Knockout , Modelos Teóricos , Separase/metabolismo , Fosfatases cdc25/fisiologia , Coesinas
14.
Protein Cell ; 8(9): 662-674, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28434146

RESUMO

Coordination of cell division and cell fate is crucial for the successful development of mammalian early embryos. Aurora kinases are evolutionarily conserved serine/threonine kinases and key regulators of mitosis. Aurora kinase B (AurkB) is ubiquitously expressed while Aurora kinase C (AurkC) is specifically expressed in gametes and preimplantation embryos. We found that increasing AurkC level in one blastomere of the 2-cell embryo accelerated cell division and decreasing AurkC level slowed down mitosis. Changing AurkB level had the opposite effect. The kinase domains of AurkB and AurkC were responsible for their different ability to phosphorylate Histone H3 Serine 10 (H3S10P) and regulate metaphase timing. Using an Oct4-photoactivatable GFP fusion protein (Oct4-paGFP) and fluorescence decay after photoactivation assay, we found that AurkB overexpression reduced Oct4 retention in the nucleus. Finally, we show that blastomeres with higher AurkC level elevated pluripotency gene expression, which were inclined to enter the inner cell mass lineage and subsequently contributed to the embryo proper. Collectively, our results are the first demonstration that the activity of mitotic kinases can influence cell fate decisions in mammalian preimplantation embryos and have important implications to assisted reproduction.


Assuntos
Aurora Quinase B/metabolismo , Aurora Quinase C/metabolismo , Blastocisto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Histonas/metabolismo , Animais , Camundongos , Fosforilação/fisiologia
15.
Mol Hum Reprod ; 23(6): 406-416, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28369513

RESUMO

STUDY QUESTION: Are single nucleotide variants (SNVs) in Aurora kinases B and C (AURKB, AURKC) associated with risk of aneuploid conception? SUMMARY ANSWER: Two SNVs were found in patients with extreme aneuploid concepti rates with respect to their age; one variant, AURKC p.I79V, is benign, while another, AURKB p.L39P, is a potential gain-of-function mutant with increased efficiency in promoting chromosome alignment. WHAT IS KNOWN ALREADY: Maternal age does not always predict aneuploidy risk, and rare gene variants can be drivers of disease. The AURKB and AURKC regulate chromosome segregation, and are associated with reproductive impairments in mouse and human. STUDY DESIGN, SIZE, DURATION: An extreme phenotype sample selection scheme was performed for variant discovery. Ninety-six DNA samples were from young patients with higher than average embryonic aneuploidy rates and an additional 96 DNA samples were from older patients with lower than average aneuploidy rates. PARTICIPANTS/MATERIALS, SETTING, METHODS: Using the192 DNA samples, the coding regions of AURKB and AURKC were sequenced using next generation sequencing. To assess biological significance, we expressed complementary RNA encoding the human variants in mouse oocytes. Assays such as determining subcellular localization and assessing catalytic activity were performed to determine alterations in protein function during meiosis. MAIN RESULTS AND THE ROLE OF CHANCE: Ten SNVs were identified using three independent variant-calling methods. Two of the SNVs (AURKB p.L39P and AURKC p.I79V) were non-synonymous and identified by at least two variant-identification methods. The variant encoding AURKC p.I79V, identified in a young woman with a higher than average rate of aneuploid embryos, showed wild-type localization pattern and catalytic activity. On the other hand, the variant encoding AURKB p.L39P, identified in an older woman with lower than average rates of aneuploid embryos, increased the protein's ability to regulate alignment of chromosomes at the metaphase plate. These experiments were repeated three independent times using 2-3 mice for each trial. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: Biological significance of the human variants was assessed in an in vitro mouse oocyte model where the variants are over-expressed. Therefore, the human protein may not function identically to the mouse homolog, or the same in mouse oocytes as in human oocytes. Furthermore, supraphysiological expression levels may not accurately reflect endogenous activity. Moreover, the evaluated variants were identified in one patient each, and no trial linking the SNV to pregnancy outcomes was conducted. Finally, the patient aneuploidy rates were established by performing comprehensive chromosome screening in blastocysts, and because of the link between female gamete aneuploidy giving rise to aneuploid embryos, we evaluate the role of the variants in Meiosis I. However, it is possible that the chromosome segregation mistake arose during Meiosis II or in mitosis in the preimplantation embryo. Their implications in human female meiosis and aneuploidy risk remain to be determined. WIDER IMPLICATIONS OF THE FINDINGS: The data provide evidence that gene variants exist in reproductively younger or advanced aged women that are predictive of the risk of producing aneuploid concepti in humans. Furthermore, a single amino acid in the N-terminus of AURKB is a gain-of-function mutant that could be protective of euploidy. STUDY FUNDING/COMPETING INTERESTS: This work was supported by a Research Grant from the American Society of Reproductive Medicine and support from the Charles and Johanna Busch Memorial Fund at Rutgers, the State University of NJ to K.S. and the Foundation for Embryonic Competence, Inc to N.T. The authors declare no conflicts of interest.


Assuntos
Aneuploidia , Aurora Quinase B/genética , Aurora Quinase C/genética , Oócitos/metabolismo , Polimorfismo de Nucleotídeo Único , Animais , Aurora Quinase B/metabolismo , Aurora Quinase C/metabolismo , Segregação de Cromossomos , Cromossomos Humanos Par 17/química , Cromossomos Humanos Par 19/química , Embrião de Mamíferos , Feminino , Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Meiose/genética , Camundongos , Oócitos/patologia , Gravidez
16.
J Cell Sci ; 129(19): 3648-3660, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27562071

RESUMO

Meiotic oocytes lack classic centrosomes and, therefore, bipolar spindle assembly depends on clustering of acentriolar microtubule-organizing centers (MTOCs) into two poles. However, the molecular mechanism regulating MTOC assembly into two poles is not fully understood. The kinase haspin (also known as GSG2) is required to regulate Aurora kinase C (AURKC) localization at chromosomes during meiosis I. Here, we show that inhibition of haspin perturbed MTOC clustering into two poles and the stability of the clustered MTOCs. Furthermore, we show that AURKC localizes to MTOCs in mouse oocytes. Inhibition of haspin perturbed the localization of AURKC at MTOCs, and overexpression of AURKC rescued the MTOC-clustering defects in haspin-inhibited oocytes. Taken together, our data uncover a role for haspin as a regulator of bipolar spindle assembly by regulating AURKC function at acentriolar MTOCs in oocytes.


Assuntos
Aurora Quinase C/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Centro Organizador dos Microtúbulos/metabolismo , Oócitos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Metáfase , Camundongos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Transporte Proteico , Fuso Acromático/metabolismo
17.
PLoS One ; 11(6): e0157305, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27332895

RESUMO

Aurora-C, a member of the Aurora kinase family that can complement Aurora-B function in mitosis is either moderately expressed or repressed in most adult somatic tissues but is active in early embryonic development and expressed at elevated levels in multiple human cancers. Aurora-C overexpression reportedly plays a role in tumorigenic transformation. We performed detailed characterization of Aurora-C interactions with members of the Chromosome Passenger Complex (CPC), Survivin and Inner Centromere Protein (INCENP) in reference to known Aurora-B interactions to understand the functional significance of Aurora-C overexpression in human cancer cells. The results revealed that silencing of Aurora-C or -B individually does not affect localization of the other kinase and the two kinases exist predominantly in independent complexes in vivo. Presence of Aurora-C and -B in molecular complexes of varying as well as overlapping sizes and co-existence in INCENP overexpressing cells indicated oligomerization of ternary complexes under different physiological conditions in vivo. Furthermore, Aurora-C and -B stabilized INCENP through interaction with and phosphorylation of the IN box domain while Aurora-C was activated following Survivin phosphorylation on Serine 20. Phosphorylation of Survivin residue Serine 20 by Aurora-C and -B appears important for proper chromosome segregation. Taken together, our study suggests that Aurora-C, expressed at low levels in somatic cells, functions as a catalytic component of the CPC together with Aurora-B through mitosis. Elevated expression of Aurora-C in cancer cells alters the structural and functional characteristics of the Aurora-B-CPC leading to chromosomal instability.


Assuntos
Aurora Quinase B/metabolismo , Aurora Quinase C/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Sequência de Aminoácidos , Aurora Quinase B/química , Aurora Quinase C/química , Centrifugação com Gradiente de Concentração , Proteínas Cromossômicas não Histona/química , Segregação de Cromossomos , Sequência Conservada , Citocinese , Inativação Gênica , Células HeLa , Humanos , Imunoprecipitação , Modelos Biológicos , Fosforilação , Fosfosserina/metabolismo , Ligação Proteica , Domínios Proteicos , Estabilidade Proteica , Survivina
18.
Int J Mol Sci ; 17(6)2016 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-27275820

RESUMO

Genomic instability is a hallmark of human cancer and an enabling factor for the genetic alterations that drive cancer development. The processes involved in genomic instability resemble those of meiosis, where genetic material is interchanged between homologous chromosomes. In most types of human cancer, epigenetic changes, including hypomethylation of gene promoters, lead to the ectopic expression of a large number of proteins normally restricted to the germ cells of the testis. Due to the similarities between meiosis and genomic instability, it has been proposed that activation of meiotic programs may drive genomic instability in cancer cells. Some germ cell proteins with ectopic expression in cancer cells indeed seem to promote genomic instability, while others reduce polyploidy and maintain mitotic fidelity. Furthermore, oncogenic germ cell proteins may indirectly contribute to genomic instability through induction of replication stress, similar to classic oncogenes. Thus, current evidence suggests that testis germ cell proteins are implicated in cancer development by regulating genomic instability during tumorigenesis, and these proteins therefore represent promising targets for novel therapeutic strategies.


Assuntos
Expressão Ectópica do Gene , Regulação Neoplásica da Expressão Gênica , Instabilidade Genômica , Células Germinativas/metabolismo , Neoplasias/genética , Testículo/metabolismo , Animais , Aurora Quinase C/metabolismo , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Cromossomos/genética , Cromossomos/metabolismo , Replicação do DNA , Humanos , Masculino , Meiose/genética , Neoplasias/metabolismo , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Poliploidia , Ligação Proteica , Recombinação Genética , Estresse Fisiológico , Complexo Sinaptonêmico/metabolismo
19.
Cell Cycle ; 15(11): 1450-61, 2016 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-27096707

RESUMO

Studies using in vitro cultured oocytes have indicated that the protein phosphatase 2A (PP2A), a major serine/threonine protein phosphatase, participates in multiple steps of meiosis. Details of oocyte maturation regulation by PP2A remain unclear and an in vivo model can provide more convincing information. Here, we inactivated PP2A by mutating genes encoding for its catalytic subunits (PP2Acs) in mouse oocytes. We found that eliminating both PP2Acs caused female infertility. Oocytes lacking PP2Acs failed to complete 1(st) meiotic division due to chromosome misalignment and abnormal spindle assembly. In mitosis, PP2A counteracts Aurora kinase B/C (AurkB/C) to facilitate correct kinetochore-microtubule (KT-MT) attachment. In meiosis I in oocyte, we found that PP2Ac deficiency destabilized KT-MT attachments. Chemical inhibition of AurkB/C in PP2Ac-null oocytes partly restored the formation of lateral/merotelic KT-MT attachments but not correct KT-MT attachments. Taken together, our findings demonstrate that PP2Acs are essential for chromosome alignments and regulate the formation of correct KT-MT attachments in meiosis I in oocytes.


Assuntos
Cinetocoros/metabolismo , Meiose , Microtúbulos/metabolismo , Oócitos/metabolismo , Proteína Fosfatase 2/genética , Subunidades Proteicas/genética , Sequência de Aminoácidos , Animais , Aurora Quinase B/genética , Aurora Quinase B/metabolismo , Aurora Quinase C/genética , Aurora Quinase C/metabolismo , Cromossomos de Mamíferos/metabolismo , Cromossomos de Mamíferos/ultraestrutura , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Infertilidade Feminina/genética , Infertilidade Feminina/metabolismo , Cinetocoros/ultraestrutura , Camundongos , Camundongos Transgênicos , Microtúbulos/ultraestrutura , Mitose , Oócitos/ultraestrutura , Cultura Primária de Células , Proteína Fosfatase 2/metabolismo , Subunidades Proteicas/metabolismo , Transdução de Sinais , Fuso Acromático/metabolismo , Fuso Acromático/ultraestrutura
20.
Hum Mol Genet ; 25(13): 2698-2711, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27106102

RESUMO

Aneuploidy is the leading genetic abnormality that leads to miscarriage, and it is caused by a failure of accurate chromosome segregation during gametogenesis or early embryonic divisions. Aurora kinase C (AURKC) is essential for formation of euploid sperm in humans because mutations in AURKC are correlated with macrozoospermia and these sperm are tetraploid. These mutations are currently the most frequent mutations that cause macrozoospermia and result from an inability to complete meiosis I (MI). Three of these mutations AURKC c.144delC (AURKC p.L49Wfs22), AURKC c.686G > A (AURKC p.C229Y) and AURKC c.744C > G (AURKC p.Y248*) occur in the coding region of the gene and are the focus of this study. By expressing these alleles in oocytes isolated from Aurkc-/- mice, we show that the mutations have different effects on AURKC function during MI. AURKC p.L49Wfs22 is a loss-of-function mutant that perturbs localization of the chromosomal passenger complex (CPC), AURKC p.C229Y is a hypomorph that cannot fully support cell-cycle progression, and AURKC p.Y248* fails to localize and function with the CPC to support chromosome segregation yet retains catalytic activity in the cytoplasm. Finally, we show that these variants of AURKC cause meiotic failure and polyploidy due to a failure in AURKC-CPC function that results in metaphase chromosome misalignment. This study is the first to assess the function of mutant alleles of AURKC that affect human fertility in a mammalian meiotic system.


Assuntos
Aurora Quinase C/genética , Aurora Quinase C/metabolismo , Alelos , Aneuploidia , Animais , Segregação de Cromossomos , Cromossomos , Humanos , Infertilidade Masculina/genética , Masculino , Mamíferos , Meiose , Camundongos , Mutação , Espermatozoides
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA