Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Mol Biosci ; 11: 1328077, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38410188

RESUMO

Background: The mitotic kinesin, KIF18A, is required for proliferation of cancer cells that exhibit chromosome instability (CIN), implicating it as a promising target for treatment of a subset of aggressive tumor types. Determining regions of the KIF18A protein to target for inhibition will be important for the design and optimization of effective small molecule inhibitors. Methods: In this study, we used cultured cell models to investigate the effects of mutating S284 within the alpha-4 helix of KIF18A, which was previously identified as a phosphorylated residue. Results: Mutations in S284 cause relocalization of KIF18A from the plus-ends of spindle microtubules to the spindle poles. Furthermore, KIF18A S284 mutants display loss of KIF18A function and fail to support proliferation in CIN tumor cells. Interestingly, similar effects on KIF18A localization and function were seen after treatment of CIN cells with KIF18A inhibitory compounds that are predicted to interact with residues within the alpha-4 helix. Conclusion: These data implicate the KIF18A alpha-4 helix as an effective target for inhibition and demonstrate that small molecules targeting KIF18A selectively limit CIN tumor cell proliferation and result in phenotypically similar effects on mitosis at the single cell level compared to genetic perturbations.

2.
Mol Biol Cell ; 35(1): ar3, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37903223

RESUMO

Kinesins support many diverse cellular processes, including facilitating cell division through mechanical regulation of the mitotic spindle. However, how kinesin activity is controlled to facilitate this process is not well understood. Interestingly, posttranslational modifications have been identified within the enzymatic region of all 45 mammalian kinesins, but the significance of these modifications has gone largely unexplored. Given the critical role of the enzymatic region in facilitating nucleotide and microtubule binding, it may serve as a primary site for kinesin regulation. Consistent with this idea, a phosphomimetic mutation at S357 in the neck-linker of KIF18A alters the localization of KIF18A within the spindle from kinetochore microtubules to nonkinetochore microtubules at the periphery of the spindle. Changes in localization of KIF18A-S357D are accompanied by defects in mitotic spindle positioning and the ability to promote mitotic progression. This altered localization pattern is mimicked by a shortened neck-linker mutant, suggesting that KIF18A-S357D may cause the motor to adopt a shortened neck-linker-like state that decreases KIF18A accumulation at the plus-ends of kinetochore microtubules. These findings demonstrate that posttranslational modifications in the enzymatic region of kinesins could be important for biasing their localization to particular microtubule subpopulations.


Assuntos
Cinesinas , Cinetocoros , Microtúbulos , Animais , Humanos , Células HeLa , Cinesinas/metabolismo , Cinetocoros/metabolismo , Microtúbulos/metabolismo , Mitose , Fuso Acromático/metabolismo
3.
bioRxiv ; 2023 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-37905069

RESUMO

The mitotic kinesin, KIF18A, is required for proliferation of cancer cells that exhibit chromosome instability (CIN), implicating it as a promising target for treatment of a subset of aggressive tumor types. Determining regions of the KIF18A protein to target for inhibition will be important for the design and optimization of effective small molecule inhibitors. In this study, we investigated the effects of mutating S284 within the alpha-4 helix of KIF18A, which was previously identified as a phosphorylated residue. Mutations in S284 cause relocalization of KIF18A from the plus-ends of spindle microtubules to the spindle poles. Furthermore, KIF18A S284 mutants display loss of KIF18A function and fail to support proliferation in CIN tumor cells. Interestingly, similar effects on KIF18A localization and function were seen after treatment of CIN cells with KIF18A inhibitory compounds that are predicted to interact with residues within the alpha-4 helix. These data implicate the KIF18A alpha-4 helix as an effective target for inhibition and demonstrate that small molecules targeting KIF18A selectively limit CIN tumor cell proliferation and result in phenotypically similar effects on mitosis at the single cell level compared to genetic perturbations.

4.
bioRxiv ; 2023 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-37205510

RESUMO

Kinesins support many diverse cellular processes, including facilitating cell division through mechanical regulation of the mitotic spindle. However, how kinesin activity is controlled to facilitate this process is not well understood. Interestingly, post-translational modifications have been identified within the enzymatic region of all 45 mammalian kinesins, but the significance of these modifications has gone largely unexplored. Given the critical role of the enzymatic region in facilitating nucleotide and microtubule binding, it may serve as a primary site for kinesin regulation. Consistent with this idea, a phosphomimetic mutation at S357 in the neck-linker of KIF18A alters the localization of KIF18A within the spindle from kinetochore microtubules to peripheral microtubules. Changes in localization of KIF18A-S357D are accompanied by defects in mitotic spindle positioning and the ability to promote mitotic progression. This altered localization pattern is mimicked by a shortened neck-linker mutant, suggesting that KIF18A-S357D may cause the motor to adopt a shortened neck-linker like state that prevents KIF18A from accumulating at the plus-ends of kinetochore microtubules. These findings demonstrate that post-translational modifications in the enzymatic region of kinesins could be important for biasing their localization to particular microtubule subpopulations.

5.
Elife ; 112022 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-35730929

RESUMO

The chromokinesin KIF22 generates forces that contribute to mitotic chromosome congression and alignment. Mutations in the α2 helix of the motor domain of KIF22 have been identified in patients with abnormal skeletal development, and we report the identification of a patient with a novel mutation in the KIF22 tail. We demonstrate that pathogenic mutations do not result in a loss of KIF22's functions in early mitosis. Instead, mutations disrupt chromosome segregation in anaphase, resulting in reduced proliferation, abnormal daughter cell nuclear morphology, and, in a subset of cells, cytokinesis failure. This phenotype could be explained by a failure of KIF22 to inactivate in anaphase. Consistent with this model, constitutive activation of the motor via a known site of phosphoregulation in the tail phenocopied the effects of pathogenic mutations. These results suggest that the motor domain α2 helix may be an important site for regulation of KIF22 activity at the metaphase to anaphase transition. In support of this conclusion, mimicking phosphorylation of α2 helix residue T158 also prevents inactivation of KIF22 in anaphase. These findings demonstrate the importance of both the head and tail of the motor in regulating the activity of KIF22 and offer insight into the cellular consequences of preventing KIF22 inactivation and disrupting force balance in anaphase.


Assuntos
Anáfase , Segregação de Cromossomos , Proteínas de Ligação a DNA , Cinesinas , Proteínas Nucleares , Proteínas de Ligação a DNA/genética , Cinesinas/genética , Metáfase , Mitose , Mutação , Proteínas Nucleares/genética , Fuso Acromático
6.
Methods Mol Biol ; 2415: 139-149, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34972951

RESUMO

The chromokinesin KIF22 (Kid, kinesin-10 family) is the primary generator of polar ejection forces, which contribute to chromosome positioning and alignment in mitotic cells. Assessment of KIF22 function requires quantitative comparison of relative polar ejection forces between experimental conditions. This is facilitated by the generation of monopolar spindles to reduce the impact of bioriented microtubule attachment at kinetochores on chromosome positions and increase the dependence of chromosome positions on chromokinesin activity. Radial profile plots measure the intensity of chromatin signal in concentric circles around the poles of monopolar cells and represent an expedient quantitative measure of relative polar ejection forces. As such, this assay can be used to measure changes in polar ejection forces resulting from chromokinesin depletion or perturbation.


Assuntos
Cromossomos , Cinesinas , Cromossomos/genética , Proteínas de Ligação a DNA/genética , Cinetocoros , Microtúbulos , Mitose , Proteínas Nucleares/genética , Fuso Acromático
7.
Sci Adv ; 7(47): eabj9812, 2021 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-34797717

RESUMO

Kinesins are regulated in space and time to ensure activation only in the presence of cargo. Kinesin-binding protein (KIFBP), which is mutated in Goldberg-Shprintzen syndrome, binds to and inhibits the catalytic motor heads of 8 of 45 kinesin superfamily members, but the mechanism remains poorly defined. Here, we used cryo­electron microscopy and cross-linking mass spectrometry to determine high-resolution structures of KIFBP alone and in complex with two mitotic kinesins, revealing structural remodeling of kinesin by KIFBP. We find that KIFBP remodels kinesin motors and blocks microtubule binding (i) via allosteric changes to kinesin and (ii) by sterically blocking access to the microtubule. We identified two regions of KIFBP necessary for kinesin binding and cellular regulation during mitosis. Together, this work further elucidates the molecular mechanism of KIFBP-mediated kinesin inhibition and supports a model in which structural rearrangement of kinesin motor domains by KIFBP abrogates motor protein activity.

8.
J Cell Biol ; 220(11)2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34515734

RESUMO

Micronuclei, whole or fragmented chromosomes spatially separated from the main nucleus, are associated with genomic instability and have been identified as drivers of tumorigenesis. Paradoxically, Kif18a mutant mice produce micronuclei due to asynchronous segregation of unaligned chromosomes in vivo but do not develop spontaneous tumors. We report here that micronuclei in Kif18a mutant mice form stable nuclear envelopes. Challenging Kif18a mutant mice via deletion of the Trp53 gene led to formation of thymic lymphoma with elevated levels of micronuclei. However, loss of Kif18a had modest or no effect on survival of Trp53 homozygotes and heterozygotes, respectively. Micronuclei in cultured KIF18A KO cells form stable nuclear envelopes characterized by increased recruitment of nuclear envelope components and successful expansion of decondensing chromatin compared with those induced by nocodazole washout or radiation. Lagging chromosomes were also positioned closer to the main chromatin masses in KIF18A KO cells. These data suggest that not all micronuclei actively promote tumorigenesis.


Assuntos
Carcinogênese/genética , Núcleo Celular/genética , Cinesinas/genética , Membrana Nuclear/genética , Animais , Linhagem Celular , Cromatina/genética , Cromossomos/genética , Dano ao DNA/genética , Feminino , Instabilidade Genômica/genética , Humanos , Masculino , Camundongos
9.
Nat Commun ; 12(1): 1213, 2021 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-33619254

RESUMO

Chromosomal instability (CIN) is a hallmark of tumor cells caused by changes in the dynamics and control of microtubules that compromise the mitotic spindle. Thus, CIN cells may respond differently than diploid cells to treatments that target mitotic spindle regulation. Here, we test this idea by inhibiting a subset of kinesin motor proteins involved in mitotic spindle control. KIF18A is required for proliferation of CIN cells derived from triple negative breast cancer or colorectal cancer tumors but is not required in near-diploid cells. Following KIF18A inhibition, CIN tumor cells exhibit mitotic delays, multipolar spindles, and increased cell death. Sensitivity to KIF18A knockdown is strongly correlated with centrosome fragmentation, which requires dynamic microtubules but does not depend on bipolar spindle formation or mitotic arrest. Our results indicate the altered spindle microtubule dynamics characteristic of CIN tumor cells can be exploited to reduce the proliferative capacity of CIN cells.


Assuntos
Instabilidade Cromossômica , Cinesinas/metabolismo , Neoplasias/genética , Neoplasias/patologia , Pontos de Checagem do Ciclo Celular , Morte Celular , Linhagem Celular Tumoral , Proliferação de Células , Centrossomo/metabolismo , Humanos , Microtúbulos/metabolismo , Mitose , Modelos Biológicos , Nocodazol/farmacologia , Paclitaxel/farmacologia , Fuso Acromático/metabolismo
10.
Nature ; 590(7846): 486-491, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33505028

RESUMO

Selective targeting of aneuploid cells is an attractive strategy for cancer treatment1. However, it is unclear whether aneuploidy generates any clinically relevant vulnerabilities in cancer cells. Here we mapped the aneuploidy landscapes of about 1,000 human cancer cell lines, and analysed genetic and chemical perturbation screens2-9 to identify cellular vulnerabilities associated with aneuploidy. We found that aneuploid cancer cells show increased sensitivity to genetic perturbation of core components of the spindle assembly checkpoint (SAC), which ensures the proper segregation of chromosomes during mitosis10. Unexpectedly, we also found that aneuploid cancer cells were less sensitive than diploid cells to short-term exposure to multiple SAC inhibitors. Indeed, aneuploid cancer cells became increasingly sensitive to inhibition of SAC over time. Aneuploid cells exhibited aberrant spindle geometry and dynamics, and kept dividing when the SAC was inhibited, resulting in the accumulation of mitotic defects, and in unstable and less-fit karyotypes. Therefore, although aneuploid cancer cells could overcome inhibition of SAC more readily than diploid cells, their long-term proliferation was jeopardized. We identified a specific mitotic kinesin, KIF18A, whose activity was perturbed in aneuploid cancer cells. Aneuploid cancer cells were particularly vulnerable to depletion of KIF18A, and KIF18A overexpression restored their response to SAC inhibition. Our results identify a therapeutically relevant, synthetic lethal interaction between aneuploidy and the SAC.


Assuntos
Aneuploidia , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Neoplasias/patologia , Cariótipo Anormal/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Segregação de Cromossomos/efeitos dos fármacos , Diploide , Genes Letais , Humanos , Cinesinas/deficiência , Cinesinas/genética , Cinesinas/metabolismo , Neoplasias/genética , Fuso Acromático/efeitos dos fármacos , Mutações Sintéticas Letais/efeitos dos fármacos , Mutações Sintéticas Letais/genética , Fatores de Tempo
11.
Elife ; 92020 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-31958056

RESUMO

Kinesin-5 motors organize mitotic spindles by sliding apart microtubules. They are homotetramers with dimeric motor and tail domains at both ends of a bipolar minifilament. Here, we describe a regulatory mechanism involving direct binding between tail and motor domains and its fundamental role in microtubule sliding. Kinesin-5 tails decrease microtubule-stimulated ATP-hydrolysis by specifically engaging motor domains in the nucleotide-free or ADP states. Cryo-EM reveals that tail binding stabilizes an open motor domain ATP-active site. Full-length motors undergo slow motility and cluster together along microtubules, while tail-deleted motors exhibit rapid motility without clustering. The tail is critical for motors to zipper together two microtubules by generating substantial sliding forces. The tail is essential for mitotic spindle localization, which becomes severely reduced in tail-deleted motors. Our studies suggest a revised microtubule-sliding model, in which kinesin-5 tails stabilize motor domains in the microtubule-bound state by slowing ATP-binding, resulting in high-force production at both homotetramer ends.


Assuntos
Cinesinas/metabolismo , Microtúbulos/metabolismo , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Microscopia Crioeletrônica , Humanos , Hidrólise , Cinesinas/química , Cinesinas/ultraestrutura , Cinética , Ligação Proteica , Domínios Proteicos , Fuso Acromático/metabolismo
12.
J Cell Biol ; 218(4): 1148-1163, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30733233

RESUMO

Chromosome alignment at the equator of the mitotic spindle is a highly conserved step during cell division; however, its importance to genomic stability and cellular fitness is not understood. Normal mammalian somatic cells lacking KIF18A function complete cell division without aligning chromosomes. These alignment-deficient cells display normal chromosome copy numbers in vitro and in vivo, suggesting that chromosome alignment is largely dispensable for maintenance of euploidy. However, we find that loss of chromosome alignment leads to interchromosomal compaction defects during anaphase, abnormal organization of chromosomes into a single nucleus at mitotic exit, and the formation of micronuclei in vitro and in vivo. These defects slow cell proliferation and are associated with impaired postnatal growth and survival in mice. Our studies support a model in which the alignment of mitotic chromosomes promotes proper organization of chromosomes into a single nucleus and continued proliferation by ensuring that chromosomes segregate as a compact mass during anaphase.


Assuntos
Anáfase , Segregação de Cromossomos , Cromossomos Humanos , Fuso Acromático/fisiologia , Animais , Linhagem Celular , Proliferação de Células , Células Epiteliais/fisiologia , Humanos , Cinesinas/genética , Cinesinas/metabolismo , Camundongos Knockout , Epitélio Pigmentado da Retina/fisiologia , Fuso Acromático/genética , Fuso Acromático/metabolismo , Fatores de Tempo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
13.
J Cell Biol ; 218(4): 1218-1234, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30709852

RESUMO

Mitotic kinesins must be regulated to ensure a precise balance of spindle forces and accurate segregation of chromosomes into daughter cells. Here, we demonstrate that kinesin-binding protein (KBP) reduces the activity of KIF18A and KIF15 during metaphase. Overexpression of KBP disrupts the movement and alignment of mitotic chromosomes and decreases spindle length, a combination of phenotypes observed in cells deficient for KIF18A and KIF15, respectively. We show through gliding filament and microtubule co-pelleting assays that KBP directly inhibits KIF18A and KIF15 motor activity by preventing microtubule binding. Consistent with these effects, the mitotic localizations of KIF18A and KIF15 are altered by overexpression of KBP. Cells depleted of KBP exhibit lagging chromosomes in anaphase, an effect that is recapitulated by KIF15 and KIF18A overexpression. Based on these data, we propose a model in which KBP acts as a protein buffer in mitosis, protecting cells from excessive KIF18A and KIF15 activity to promote accurate chromosome segregation.


Assuntos
Anáfase , Núcleo Celular/enzimologia , Segregação de Cromossomos , Cromossomos Humanos , Cinesinas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Núcleo Celular/genética , Células HeLa , Humanos , Cinesinas/genética , Proteínas do Tecido Nervoso/genética , Epitélio Pigmentado da Retina/enzimologia , Transdução de Sinais
14.
Life Sci Alliance ; 2(1)2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30655363

RESUMO

KIF18A (kinesin-8) is required for mammalian mitotic chromosome alignment. KIF18A confines chromosome movement to the mitotic spindle equator by accumulating at the plus-ends of kinetochore microtubule bundles (K-fibers), where it functions to suppress K-fiber dynamics. It is not understood how the motor accumulates at K-fiber plus-ends, a difficult feat requiring the motor to navigate protein dense microtubule tracks. Our data indicate that KIF18A's relatively long neck linker is required for the motor's accumulation at K-fiber plus-ends. Shorter neck linker (sNL) variants of KIF18A display a deficiency in accumulation at the ends of K-fibers at the center of the spindle. Depletion of K-fiber-binding proteins reduces the KIF18A sNL localization defect, whereas their overexpression reduces wild-type KIF18A's ability to accumulate on this same K-fiber subset. Furthermore, single-molecule assays indicate that KIF18A sNL motors are less proficient in navigating microtubules coated with microtubule-associated proteins. Taken together, these results support a model in which KIF18A's neck linker length permits efficient navigation of obstacles to reach K-fiber ends during mitosis.


Assuntos
Cinesinas/metabolismo , Metáfase/fisiologia , Microtúbulos/metabolismo , Fuso Acromático/metabolismo , Cromossomos/metabolismo , Células HeLa , Humanos , Cinesinas/genética , Cinetocoros/metabolismo , Leupeptinas/farmacologia , Metáfase/efeitos dos fármacos , RNA Interferente Pequeno/genética , Transfecção
15.
Proc Natl Acad Sci U S A ; 115(8): E1779-E1788, 2018 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-29432173

RESUMO

Numerous posttranslational modifications have been described in kinesins, but their consequences on motor mechanics are largely unknown. We investigated one of these-acetylation of lysine 146 in Eg5-by creating an acetylation mimetic lysine to glutamine substitution (K146Q). Lysine 146 is located in the α2 helix of the motor domain, where it makes an ionic bond with aspartate 91 on the neighboring α1 helix. Molecular dynamics simulations predict that disrupting this bond enhances catalytic site-neck linker coupling. We tested this using structural kinetics and single-molecule mechanics and found that the K146Q mutation increases motor performance under load and coupling of the neck linker to catalytic site. These changes convert Eg5 from a motor that dissociates from the microtubule at low load into one that is more tightly coupled and dissociation resistant-features shared by kinesin 1. These features combined with the increased propensity to stall predict that the K146Q Eg5 acetylation mimetic should act in the cell as a "brake" that slows spindle pole separation, and we have confirmed this by expressing this modified motor in mitotically active cells. Thus, our results illustrate how a posttranslational modification of a kinesin can be used to fine tune motor behavior to meet specific physiological needs.


Assuntos
Cinesinas/química , Cinesinas/metabolismo , Mitose/fisiologia , Sequência de Aminoácidos , Fenômenos Biomecânicos , Células HeLa , Humanos , Modelos Moleculares , Mutação , Conformação Proteica
16.
Mol Cancer Res ; 16(4): 587-598, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29378907

RESUMO

Aggressive breast cancer is difficult to treat as it is unresponsive to many hormone-based therapies; therefore, it is imperative to identify novel, targetable regulators of progression. Long non-coding RNAs (lncRNA) are important regulators in breast cancer and have great potential as therapeutic targets; however, little is known about how the majority of lncRNAs function within breast cancer. This study characterizes a novel lncRNA, MANCR (mitotically-associated long noncoding RNA; LINC00704), which is upregulated in breast cancer patient specimens and cells. Depletion of MANCR in triple-negative breast cancer cells significantly decreases cell proliferation and viability, with concomitant increases in DNA damage. Transcriptome analysis, based on RNA sequencing, following MANCR knockdown reveals significant differences in the expression of >2,000 transcripts, and gene set enrichment analysis identifies changes in multiple categories related to cell-cycle regulation. Furthermore, MANCR expression is highest in mitotic cells by both RT-qPCR and RNA in situ hybridization. Consistent with a role in cell-cycle regulation, MANCR-depleted cells have a lower mitotic index and higher incidences of defective cytokinesis and cell death. Taken together, these data reveal a role for the novel lncRNA, MANCR, in genomic stability of aggressive breast cancer, and identify it as a potential therapeutic target.Implications: The novel lncRNA, MANCR (LINC00704), is upregulated in breast cancer and is functionally linked with cell proliferation, viability, and genomic stability. Mol Cancer Res; 16(4); 587-98. ©2018 AACR.


Assuntos
Neoplasias da Mama/genética , Mitose , RNA Longo não Codificante/genética , Regulação para Cima , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Instabilidade Genômica , Humanos , Análise de Sequência de RNA
17.
Cell Rep ; 20(9): 2044-2056, 2017 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-28854357

RESUMO

Oxidative damage to telomere DNA compromises telomere integrity. We recently reported that the DNA glycosylase NEIL3 preferentially repairs oxidative lesions in telomere sequences in vitro. Here, we show that loss of NEIL3 causes anaphase DNA bridging because of telomere dysfunction. NEIL3 expression increases during S phase and reaches maximal levels in late S/G2. NEIL3 co-localizes with TRF2 and associates with telomeres during S phase, and this association increases upon oxidative stress. Mechanistic studies reveal that NEIL3 binds to single-stranded DNA via its intrinsically disordered C terminus in a telomere-sequence-independent manner. Moreover, NEIL3 is recruited to telomeres through its interaction with TRF1, and this interaction enhances the enzymatic activity of purified NEIL3. Finally, we show that NEIL3 interacts with AP Endonuclease 1 (APE1) and the long-patch base excision repair proteins PCNA and FEN1. Taken together, we propose that NEIL3 protects genome stability through targeted repair of oxidative damage in telomeres during S/G2 phase.


Assuntos
Segregação de Cromossomos , Dano ao DNA , Reparo do DNA , Mitose , N-Glicosil Hidrolases/metabolismo , Fase S , Telômero/patologia , Linfócitos T CD4-Positivos/metabolismo , Pontos de Checagem do Ciclo Celular , Núcleo Celular/metabolismo , DNA/metabolismo , Técnicas de Silenciamento de Genes , Células HCT116 , Células HeLa , Humanos , Microtúbulos/metabolismo , N-Glicosil Hidrolases/química , Estresse Oxidativo , Ligação Proteica , Domínios Proteicos , Fuso Acromático/metabolismo
18.
Methods Mol Biol ; 1413: 253-62, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27193854

RESUMO

The alignment of chromosomes during metaphase is a hallmark of mitosis. For this reason, chromosome alignment has served as an informative functional assay for evaluating mitotic fidelity. The common approach of quantifying the number of mitotic cells with unaligned chromosomes within a population has led to the identification of many proteins required for this conserved process. However, more sensitive assays are now required to dissect the complex molecular control of chromosome alignment. In this chapter, we describe a microscopy-based method for objectively quantifying the distribution of fluorescently labeled chromosomes within the mitotic spindle that can be used to evaluate the extent of chromosome alignment within individual mitotic cells.


Assuntos
Pareamento Cromossômico , Mitose , Técnicas de Cultura de Células , Imunofluorescência , Células HeLa , Humanos , Cinetocoros/metabolismo , Microtúbulos/metabolismo , Imagem Molecular , Interferência de RNA , Fuso Acromático/metabolismo
19.
Dev Biol ; 402(2): 253-262, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25824710

RESUMO

Genome integrity in the developing germ line is strictly required for fecundity. In proliferating somatic cells and in germ cells, there are mitotic checkpoint mechanisms that ensure accurate chromosome segregation and euploidy. There is growing evidence of mitotic cell cycle components that are uniquely required in the germ line to ensure genome integrity. We previously showed that the primary phenotype of germ cell deficient 2 (gcd2) mutant mice is infertility due to germ cell depletion during embryogenesis. Here we show that the underlying mutation is a mis-sense mutation, R308K, in the motor domain of the kinesin-8 family member, KIF18A, a protein that is expressed in a variety of proliferative tissues and is a key regulator of chromosome alignment during mitosis. Despite the conservative nature of the mutation, we show that its functional consequences are equivalent to KIF18A deficiency in HeLa cells. We also show that somatic cells progress through mitosis, despite having chromosome alignment defects, while germ cells with similar chromosome alignment defects undergo mitotic arrest and apoptosis. Our data provide evidence for differential requirements for chromosome alignment in germ and somatic cells and show that Kif18a is one of a growing number of genes that are specifically required for cell cycle progression in proliferating germ cells.


Assuntos
Proteínas de Ciclo Celular/genética , Células Germinativas/fisiologia , Cinesinas/genética , Mitose/fisiologia , Animais , Apoptose/fisiologia , Western Blotting , Proteínas de Ciclo Celular/metabolismo , Clonagem Molecular , Citometria de Fluxo , Imunofluorescência , Inativação Gênica , Vetores Genéticos/genética , Células HeLa , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Imuno-Histoquímica , Hibridização In Situ , Marcação In Situ das Extremidades Cortadas , Cinesinas/metabolismo , Camundongos , Mitose/genética , Mutação de Sentido Incorreto/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...