Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neurosci ; 42(11): 2149-2165, 2022 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-35046122

RESUMO

During neuronal migration, forces generated by cytoplasmic dynein yank on microtubules extending from the centrosome into the leading process and move the nucleus along microtubules that extend behind the centrosome. Scaffolds, such as radial glia, guide neuronal migration outward from the ventricles, but little is known about the internal machinery that ensures that the soma migrates along its proper path rather than moving backward or off the path. Here we report that depletion of KIFC1, a minus-end-directed kinesin called HSET in humans, causes neurons to migrate off their appropriate path, suggesting that this molecular motor is what ensures fidelity of the trajectory of migration. For these studies, we used rat migratory neurons in vitro and developing mouse brain in vivo, together with RNA interference and ectopic expression of mutant forms of KIFC1. We found that crosslinking of microtubules into a nonsliding mode by KIFC1 is necessary for dynein-driven forces to achieve sufficient traction to thrust the soma forward. Asymmetric bouts of microtubule sliding driven by KIFC1 thereby enable the soma to tilt in one direction or another, thus providing midcourse corrections that keep the neuron on its appropriate trajectory. KIFC1-driven sliding of microtubules further assists neurons in remaining on their appropriate path by allowing the nucleus to rotate directionally as it moves, which is consistent with how we found that KIFC1 contributes to interkinetic nuclear migration at an earlier stage of neuronal development.SIGNIFICANCE STATEMENT Resolving the mechanisms of neuronal migration is medically important because many developmental disorders of the brain involve flaws in neuronal migration and because deployment of newly born neurons may be important in the adult for cognition and memory. Drugs that inhibit KIFC1 are candidates for chemotherapy and therefore should be used with caution if they are allowed to enter the brain.


Assuntos
Cinesinas , Microtúbulos , Animais , Movimento Celular , Dineínas do Citoplasma/metabolismo , Cinesinas/genética , Camundongos , Microtúbulos/metabolismo , Neurônios/fisiologia , Ratos , beta Carioferinas
2.
Int J Mol Sci ; 24(22)2023 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-38003261

RESUMO

Quadruple-negative breast cancer (QNBC) lacks traditional actionable targets, including androgen receptor (AR). QNBC disproportionately afflicts and impacts patients of African genetic ancestry. Kinesin family member C1 (KIFC1/HSET), a centrosome clustering protein that prevents cancer cells from undergoing centrosome-amplification-induced apoptosis, has been reported to be upregulated in TNBCs and African-American (AA) TNBCs. Herein, we analyzed KIFC1 RNA levels and their associations with clinical features and outcomes among AR-low and AR-high TNBC tumors in three distinct publicly available gene expression datasets and in the breast cancer gene expression database (bc-GenExMiner). KIFC1 levels were significantly higher in AR-low and basal-like TNBCs than in AR-high and non-basal-like TNBCs, irrespective of the stage, grade, tumor size, and lymph node status. KIFC1 levels were also upregulated in AR-low tumors relative to AR-high tumors among Black and premenopausal women with TNBC. High KIFC1 levels conferred significantly shorter overall survival, disease-free survival, and distant metastasis-free survival among AR-low and basal-like TNBC patients in Kaplan-Meier analyses. In conclusion, KIFC1 levels may be upregulated in AR-low tumors and, specifically, in those of African descent, wherein it may promote poor outcomes. KIFC1 may be an actionable cancer-cell-specific target for the AR-low TNBC subpopulation and could aid in alleviating racial disparities in TNBC outcomes.


Assuntos
Neoplasias de Mama Triplo Negativas , Feminino , Humanos , Família , Estimativa de Kaplan-Meier , Cinesinas/genética , Cinesinas/metabolismo , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Neoplasias de Mama Triplo Negativas/patologia
3.
EMBO Rep ; 21(6): e49234, 2020 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-32270908

RESUMO

Centrosome amplification is a hallmark of cancer, and centrosome clustering is essential for cancer cell survival. The mitotic kinesin HSET is an essential contributor to this process. Recent studies have highlighted novel functions for intraflagellar transport (IFT) proteins in regulating motors and mitotic processes. Here, using siRNA knock-down of various IFT proteins or AID-inducible degradation of endogenous IFT88 in combination with small-molecule inhibition of HSET, we show that IFT proteins together with HSET are required for efficient centrosome clustering. We identify a direct interaction between the kinesin HSET and IFT proteins, and we define how IFT proteins contribute to clustering dynamics during mitosis using high-resolution live imaging of centrosomes. Finally, we demonstrate the requirement of IFT88 for efficient centrosome clustering in a variety of cancer cell lines naturally harboring supernumerary centrosomes and its importance for cancer cell proliferation. Overall, our data unravel a novel role for the IFT machinery in centrosome clustering during mitosis in cells harboring supernumerary centrosomes.


Assuntos
Proteínas de Transporte , Centrossomo , Proteínas de Transporte/genética , Centrossomo/metabolismo , Análise por Conglomerados , Cinesinas/genética , Cinesinas/metabolismo , Mitose/genética
4.
EMBO Rep ; 19(2): 368-381, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29330318

RESUMO

Mitotic spindles assemble from two centrosomes, which are major microtubule-organizing centers (MTOCs) that contain centrioles. Meiotic spindles in oocytes, however, lack centrioles. In mouse oocytes, spindle microtubules are nucleated from multiple acentriolar MTOCs that are sorted and clustered prior to completion of spindle assembly in an "inside-out" mechanism, ending with establishment of the poles. We used HSET (kinesin-14) as a tool to shift meiotic spindle assembly toward a mitotic "outside-in" mode and analyzed the consequences on the fidelity of the division. We show that HSET levels must be tightly gated in meiosis I and that even slight overexpression of HSET forces spindle morphogenesis to become more mitotic-like: rapid spindle bipolarization and pole assembly coupled with focused poles. The unusual length of meiosis I is not sufficient to correct these early spindle morphogenesis defects, resulting in severe chromosome alignment abnormalities. Thus, the unique "inside-out" mechanism of meiotic spindle assembly is essential to prevent chromosomal misalignment and production of aneuploidy gametes.


Assuntos
Cromossomos , Meiose , Mitose , Oócitos , Fuso Acromático/metabolismo , Animais , Centrossomo , Segregação de Cromossomos , Expressão Gênica , Humanos , Cinesinas/genética , Cinesinas/metabolismo , Camundongos
5.
Bioorg Med Chem ; 28(1): 115154, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31753800

RESUMO

Although cancer cells often harbor supernumerary centrosomes, they form pseudo-bipolar spindles via centrosome clustering, instead of lethal multipolar spindles, and thus avoid cell death. Kinesin-14 HSET/KIFC1 is a crucial protein involved in centrosome clustering. Accordingly, a compound that targets HSET could potentially inhibit cancer cell proliferation in a targeted manner. Here, we report three natural compounds derived from Solidago altissima that restored the growth of fission yeast cells exhibiting lethal HSET overproduction (positive screening), namely solidagonic acid (SA) (1), kolavenic acid analog (KAA: a stereo isomer at C-9 and C-10 of 6ß-tigloyloxykolavenic acid) (2), and kolavenic acid (KA) (3). All three compounds suppressed fission yeast cell death and enabled reversion of the mitotic spindles from a monopolar to bipolar morphology. Compound 2, which exerted the strongest activity against HSET-overproducing yeast cells, also inhibited centrosome clustering in MDA-MB-231 human breast adenocarcinoma cells, which contained large numbers of supernumerary centrosomes. These natural compounds may be useful as bioprobes in studies of HSET function. Moreover, compound 2 is a prime contender in the development of novel agents for cancer treatment.


Assuntos
Diterpenos/farmacologia , Cinesinas/antagonistas & inibidores , Mitose/efeitos dos fármacos , Schizosaccharomyces/efeitos dos fármacos , Linhagem Celular Tumoral , Centrossomo/efeitos dos fármacos , Diterpenos/síntese química , Diterpenos/química , Relação Dose-Resposta a Droga , Humanos , Cinesinas/biossíntese , Estrutura Molecular , Schizosaccharomyces/crescimento & desenvolvimento , Fuso Acromático/efeitos dos fármacos , Relação Estrutura-Atividade
6.
Cancers (Basel) ; 16(18)2024 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-39335162

RESUMO

There are currently no approved targeted treatments for quadruple-negative breast cancer [QNBC; ER-/PR-/HER2-/androgen receptor (AR)-], a subtype of triple-negative breast cancer (TNBC). AR-low TNBC is more proliferative and clinically aggressive than AR-high TNBC. Centrosome amplification (CA), a cancer hallmark, is rampant in TNBC, where it induces spindle multipolarity-mediated cell death unless centrosome clustering pathways are co-upregulated to avert these sequelae. We recently showed that genes that confer CA and centrosome clustering are strongly overexpressed in AR-low TNBCs relative to AR-high TNBCs. However, the molecular mechanisms that index centrosome clustering to the levels of CA are undefined. We argue that FOXM1, a cell cycle-regulated oncogene, links the expression of genes that drive CA to the expression of genes that act at kinetochores and along microtubules to facilitate centrosome clustering. We provide compelling evidence that upregulation of the FOXM1-E2F1-ATAD2 oncogene triad in AR-low TNBC is accompanied by CA and the co-upregulation of centrosome clustering proteins such as KIFC1, AURKB, BIRC5, and CDCA8, conferring profound dysregulation of cell cycle controls. Targeting FOXM1 in AR-low TNBC may render cancer cells incapable of clustering their centrosomes and impair their ability to generate excess centrosomes. Hence, our review illuminates FOXM1 as a potential actionable target for AR-low TNBC.

7.
Adv Biol (Weinh) ; 5(6): e2000493, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33788418

RESUMO

Accurate segregation of chromosomes during anaphase relies on the central spindle and its regulators. A newly raised concept of the central spindle, the bridging fiber, shows that sliding of antiparallel microtubules (MTs) within the bridging fiber promotes chromosome segregation. However, the regulators of the bridging fiber and its regulatory mechanism on MTs sliding remain largely unknown. In this study, the non-motor microtubule-associated protein, hyaluronan-mediated motility receptor (HMMR), is identified as a novel regulator of the bridging fiber. It then identifies that HMMR regulates MTs sliding within the bridging fiber by cooperating with its binding partner HSET. By utilizing a laser-based cell ablation system and photoactivation approach, the study's results reveal that depletion of HMMR causes an inhibitory effect on MTs sliding within the bridging fiber and disrupts the forced uniformity on the kinetochore-attached microtubules-formed fibers (k-fibers). These are created by suppressing the dynamics of HSET, which functions in transiting the force from sliding of bridging MTs to the k-fiber. This study sheds new light on the novel regulatory mechanism of MTs sliding within the bridging fiber by HMMR and HSET and uncovers the role of HMMR in chromosome segregation during anaphase.


Assuntos
Segregação de Cromossomos , Fuso Acromático , Anáfase , Proteínas da Matriz Extracelular , Receptores de Hialuronatos , Microtúbulos
8.
J Pers Med ; 11(12)2021 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-34945833

RESUMO

The enigma of why some premalignant or pre-invasive breast lesions transform and progress while others do not remains poorly understood. Currently, no radiologic or molecular biomarkers exist in the clinic that can successfully risk-stratify high-risk lesions for malignant transformation or tumor progression as well as serve as a minimally cytotoxic actionable target for at-risk subpopulations. Breast carcinogenesis involves a series of key molecular deregulatory events that prompt normal cells to bypass tumor-suppressive senescence barriers. Kinesin family member C1 (KIFC1/HSET), which confers survival of cancer cells burdened with extra centrosomes, has been observed in premalignant and pre-invasive lesions, and its expression has been shown to correlate with increasing neoplastic progression. Additionally, KIFC1 has been associated with aggressive breast tumor molecular subtypes, such as basal-like and triple-negative breast cancers. However, the role of KIFC1 in malignant transformation and its potential as a predictive biomarker of neoplastic progression remain elusive. Herein, we review compelling evidence suggesting the involvement of KIFC1 in enabling pre-neoplastic cells to bypass senescence barriers necessary to become immortalized and malignant. We also discuss evidence inferring that KIFC1 levels may be higher in premalignant lesions with a greater inclination to transform and acquire aggressive tumor intrinsic subtypes. Collectively, this evidence provides a strong impetus for further investigation into KIFC1 as a potential risk-stratifying biomarker and minimally cytotoxic actionable target for high-risk patient subpopulations.

9.
Curr Biol ; 28(14): 2356-2362.e5, 2018 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-30017484

RESUMO

A common mitotic defect observed in cancer cells that possess supernumerary (more than two) centrosomes is multipolar spindle formation [1, 2]. Such structures are resolved into a bipolar geometry by minus-end-directed motor proteins, such as cytoplasmic dynein and the kinesin-14 HSET [3-8]. HSET is also thought to antagonize plus-end-directed kinesin-5 Eg5 to balance spindle forces [4, 5, 7, 9]. However, the biomechanics of this force opposition are unclear, as HSET has previously been defined as a non-processive motor [10-16]. Here, we use optical trapping to elucidate the mechanism of force generation by HSET. We show that a single HSET motor has a processive nature with the ability to complete multiple steps while trapped along a microtubule and when unloaded can move in both directions for microns. Compared to other kinesins, HSET has a relatively weak stall force of 1.1 pN [17, 18]. Moreover, HSET's tail domain and its interaction with the E-hook of tubulin are necessary for long-range motility. In vitro polarity-marked bundle assays revealed that HSET selectively generates force in anti-parallel bundles on the order of its stall force. When combined with varied ratios of Eg5, HSET adopts Eg5's directionality while acting as an antagonizing force brake, requiring at least a 10-fold higher Eg5 concentration to surpass HSET's sliding force. These results reveal HSET's ability to change roles within the spindle from acting as an adjustable microtubule slider and force regulator to a processive motor that aids in minus end focusing.


Assuntos
Centrossomo/metabolismo , Cinesinas/metabolismo , Microtúbulos/metabolismo , Dineínas/metabolismo , Fuso Acromático/metabolismo , Tubulina (Proteína)/metabolismo
10.
Future Med Chem ; 8(4): 463-89, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26976726

RESUMO

The kinesin class of microtubule-associated motor proteins present attractive anticancer targets owing to their roles in key functions in dividing cells. Two interpolar mitotic kinesins Eg5 and HSET have opposing motor functions in mitotic spindle assembly with respect to microtubule movement, but both offer opportunities to develop cancer selective therapeutic agents. Here, we summarize the progress to date in developing inhibitors of Eg5 and HSET, with an emphasis on structural biology insights into the binding modes of allosteric inhibitors, compound selectivity and mechanisms of action of different chemical scaffolds. We discuss translation of preclinical studies to clinical experience with Eg5 inhibitors, recent findings on potential resistance mechanisms and explore the implications for future anticancer drug development against these targets.


Assuntos
Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Cinesinas/antagonistas & inibidores , Mitose/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Antineoplásicos/química , Inibidores Enzimáticos/química , Humanos , Cinesinas/metabolismo , Modelos Moleculares , Conformação Molecular , Neoplasias/metabolismo
11.
Mater Sci Eng C Mater Biol Appl ; 62: 771-8, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26952483

RESUMO

Gene therapy is an optimistic approach in cancer treatment. However, for efficient delivery of gene materials, designing an appropriate vector is necessary. Polyelectrolyte complexes (PECs) of chitosan and dextran could be considered a proper nanoparticulate carrier for sensitive biomaterials. In this study, PECs of chitosan and thiolated dextran were used as either an injectable or oral gene delivery system. hSET1 antisense was loaded into the PECs to suppress proliferation of colon cancer cell line. The prepared nanoparticles have ~115nm diameter size and positive zeta potential with high mucoadhesion properties. They are able to protect antisense from degradation in serum and biorelevant fluids (FaSSIF and FaSSGF). Furthermore, prepared nanoparticles demonstrated superior cellular penetration and inhibitory effect on SW480 colon cancer cell proliferation. All nanoparticles significantly down regulated hSET1 in comparison with naked antisense. It can be concluded that thiolated PECs have potential use for injectable or oral delivery of nucleic acids such as antisense.


Assuntos
Dextranos/química , Portadores de Fármacos/química , Histona-Lisina N-Metiltransferase/metabolismo , Nanopartículas/química , Oligonucleotídeos Antissenso/metabolismo , Transfecção , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Quitosana/química , Colo/metabolismo , Regulação para Baixo/efeitos dos fármacos , Portadores de Fármacos/toxicidade , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Histona-Lisina N-Metiltransferase/genética , Humanos , Microscopia Confocal , Nanopartículas/toxicidade , Tamanho da Partícula , Espectroscopia de Infravermelho com Transformada de Fourier , Compostos de Sulfidrila/química
12.
Oncotarget ; 6(8): 6076-91, 2015 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-25788277

RESUMO

Human breast tumors harbor supernumerary centrosomes in almost 80% of tumor cells. Although amplified centrosomes compromise cell viability via multipolar spindles resulting in death-inducing aneuploidy, cancer cells tend to cluster extra centrosomes during mitosis. As a result cancer cells display bipolar spindle phenotypes to maintain a tolerable level of aneuploidy, an edge to their survival. HSET/KifC1, a kinesin-like minus-end directed microtubule motor has recently found fame as a crucial centrosome clustering molecule. Here we show that HSET promotes tumor progression via mechanisms independent of centrosome clustering. We found that HSET is overexpressed in breast carcinomas wherein nuclear HSET accumulation correlated with histological grade and predicted poor progression-free and overall survival. In addition, deregulated HSET protein expression was associated with gene amplification and/or translocation. Our data provide compelling evidence that HSET overexpression is pro-proliferative, promotes clonogenic-survival and enhances cell-cycle kinetics through G2 and M-phases. Importantly, HSET co-immunoprecipitates with survivin, and its overexpression protects survivin from proteasome-mediated degradation, resulting in its increased steady-state levels. We provide the first evidence of centrosome clustering-independent activities of HSET that fuel tumor progression and firmly establish that HSET can serve both as a potential prognostic biomarker and as a valuable cancer-selective therapeutic target.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Centrossomo/metabolismo , Cinesinas/biossíntese , Aneuploidia , Biomarcadores Tumorais/biossíntese , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Centrossomo/patologia , Progressão da Doença , Feminino , Células HeLa , Humanos , Proteínas Inibidoras de Apoptose/metabolismo , Microtúbulos/metabolismo , Gradação de Tumores , Survivina , Transfecção , Regulação para Cima
13.
Cytoskeleton (Hoboken) ; 70(9): 515-21, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24039245

RESUMO

Tip-tracking of kinesin-14 motor proteins is believed to be crucial for the assembly and maintenance of dynamic microtubule arrays. However, in contrast to other members of the kinesin-14 family, H. sapiens kinesin-14 HSET has so far never been observed to be prominently located at microtubule plus ends. Here, using an in vitro microtubule dynamics reconstitution assay we observe tip-tracking of GFP-HSET in the presence of H. sapiens EB1 (hsEB1). Tip-tracking depended on the SxIP-like motif in HSET as well as on the EB homology domain in hsEB1. D. melanogaster Ncd and S. pombe Klp2 tip-tracking reconstitution assays accompanied by kinesin-14 amino acid sequence comparisons suggest that SxIP-like motif mediated tip-tracking dependent on EB family proteins is conserved in the kinesin-14 family of molecular motors.


Assuntos
Cinesinas/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Sequência de Aminoácidos , Animais , Drosophila melanogaster , Humanos , Cinesinas/química , Microscopia de Fluorescência , Microtúbulos/química , Dados de Sequência Molecular
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa