Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros











Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(48)2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34819364

RESUMEN

Mitotic errors can activate cyclic GMP-AMP synthase (cGAS) and induce type I interferon (IFN) signaling. Current models propose that chromosome segregation errors generate micronuclei whose rupture activates cGAS. We used a panel of antimitotic drugs to perturb mitosis in human fibroblasts and measured abnormal nuclear morphologies, cGAS localization, and IFN signaling in the subsequent interphase. Micronuclei consistently recruited cGAS without activating it. Instead, IFN signaling correlated with formation of cGAS-coated chromatin bridges that were selectively generated by microtubule stabilizers and MPS1 inhibitors. cGAS activation by chromatin bridges was suppressed by drugs that prevented cytokinesis. We confirmed cGAS activation by chromatin bridges in cancer lines that are unable to secrete IFN by measuring paracrine transfer of 2'3'-cGAMP to fibroblasts, and in mouse cells. We propose that cGAS is selectively activated by self-chromatin when it is stretched in chromatin bridges. Immunosurveillance of cells that fail mitosis, and antitumor actions of taxanes and MPS1 inhibitors, may depend on this effect.


Asunto(s)
Cromatina/fisiología , Mitosis/fisiología , Nucleotidiltransferasas/metabolismo , Línea Celular Tumoral , Cromatina/genética , Humanos , Interferón Tipo I/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/genética , Micronúcleo Germinal/genética , Micronúcleo Germinal/fisiología , Mitosis/efectos de los fármacos , Mitosis/genética , Neoplasias/metabolismo , Nucleótidos Cíclicos/metabolismo , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/fisiología , Transducción de Señal
2.
Cancer Cell ; 39(9): 1214-1226.e10, 2021 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-34375612

RESUMEN

PARP7 is a monoPARP that catalyzes the transfer of single units of ADP-ribose onto substrates to change their function. Here, we identify PARP7 as a negative regulator of nucleic acid sensing in tumor cells. Inhibition of PARP7 restores type I interferon (IFN) signaling responses to nucleic acids in tumor models. Restored signaling can directly inhibit cell proliferation and activate the immune system, both of which contribute to tumor regression. Oral dosing of the PARP7 small-molecule inhibitor, RBN-2397, results in complete tumor regression in a lung cancer xenograft and induces tumor-specific adaptive immune memory in an immunocompetent mouse cancer model, dependent on inducing type I IFN signaling in tumor cells. PARP7 is a therapeutic target whose inhibition induces both cancer cell-autonomous and immune stimulatory effects via enhanced IFN signaling. These data support the targeting of a monoPARP in cancer and introduce a potent and selective PARP7 inhibitor to enter clinical development.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Interferón Tipo I/metabolismo , Neoplasias/tratamiento farmacológico , Proteínas de Transporte de Nucleósidos/genética , Proteínas de Transporte de Nucleósidos/metabolismo , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Inmunidad Adaptativa/efectos de los fármacos , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células HEK293 , Células HeLa , Humanos , Ratones , Neoplasias/genética , Neoplasias/metabolismo , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Escape del Tumor/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
3.
BMC Biol ; 19(1): 149, 2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34325682

RESUMEN

Natural killer (NK) cells participate in cancer immunosurveillance and cancer immunotherapy. Live cell imaging of cancer cells targeted by NK cells, published today in BMC Biology by Zhu et al., reveals a remarkable diversity of programmed cell death pathways induced in individual cells. Pathway choice depends on the state of the target cell actin cytoskeleton and a novel death pathway, granzyme-induced necroptosis, could be of broad importance in cancer immunotherapy.


Asunto(s)
Células Asesinas Naturales , Neoplasias , Apoptosis , Granzimas , Humanos , Inmunoterapia , Neoplasias/terapia
4.
Nat Metab ; 3(4): 513-522, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33846641

RESUMEN

Colchicine has served as a traditional medicine for millennia and remains widely used to treat inflammatory and other disorders. Colchicine binds tubulin and depolymerizes microtubules, but it remains unclear how this mechanism blocks myeloid cell recruitment to inflamed tissues. Here we show that colchicine inhibits myeloid cell activation via an indirect mechanism involving the release of hepatokines. We find that a safe dose of colchicine depolymerizes microtubules selectively in hepatocytes but not in circulating myeloid cells. Mechanistically, colchicine triggers Nrf2 activation in hepatocytes, leading to secretion of anti-inflammatory hepatokines, including growth differentiation factor 15 (GDF15). Nrf2 and GDF15 are required for the anti-inflammatory action of colchicine in vivo. Plasma from colchicine-treated mice inhibits inflammatory signalling in myeloid cells in a GDF15-dependent manner, by positive regulation of SHP-1 (PTPN6) phosphatase, although the precise molecular identities of colchicine-induced GDF15 and its receptor require further characterization. Our work shows that the efficacy and safety of colchicine depend on its selective action on hepatocytes, and reveals a new axis of liver-myeloid cell communication. Plasma GDF15 levels and myeloid cell SHP-1 activity may be useful pharmacodynamic biomarkers of colchicine action.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Colchicina/farmacología , Citocinas/fisiología , Hígado/efectos de los fármacos , Hígado/metabolismo , Células Mieloides/efectos de los fármacos , Animales , Antiinflamatorios no Esteroideos/farmacocinética , Antioxidantes/farmacología , Colchicina/farmacocinética , Simulación por Computador , Citocinas/biosíntesis , Factor 15 de Diferenciación de Crecimiento/genética , Hepatocitos/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Peritonitis/inducido químicamente , Peritonitis/prevención & control , Proteína Tirosina Fosfatasa no Receptora Tipo 6/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
5.
Elife ; 102021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33554860

RESUMEN

Individual cancers rely on distinct essential genes for their survival. The Cancer Dependency Map (DepMap) is an ongoing project to uncover these gene dependencies in hundreds of cancer cell lines. To make this drug discovery resource more accessible to the scientific community, we built an easy-to-use browser, shinyDepMap (https://labsyspharm.shinyapps.io/depmap). shinyDepMap combines CRISPR and shRNA data to determine, for each gene, the growth reduction caused by knockout/knockdown and the selectivity of this effect across cell lines. The tool also clusters genes with similar dependencies, revealing functional relationships. shinyDepMap can be used to (1) predict the efficacy and selectivity of drugs targeting particular genes; (2) identify maximally sensitive cell lines for testing a drug; (3) target hop, that is, navigate from an undruggable protein with the desired selectivity profile, such as an activated oncogene, to more druggable targets with a similar profile; and (4) identify novel pathways driving cancer cell growth and survival.


Asunto(s)
Biología Computacional/métodos , Neoplasias/genética , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Genes Esenciales , Humanos , Internet , Neoplasias/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Programas Informáticos
6.
Elife ; 92020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-33284105

RESUMEN

How bulk cytoplasm generates forces to separate post-anaphase microtubule (MT) asters in Xenopus laevis and other large eggs remains unclear. Previous models proposed that dynein-based, inward organelle transport generates length-dependent pulling forces that move centrosomes and MTs outwards, while other components of cytoplasm are static. We imaged aster movement by dynein and actomyosin forces in Xenopus egg extracts and observed outward co-movement of MTs, endoplasmic reticulum (ER), mitochondria, acidic organelles, F-actin, keratin, and soluble fluorescein. Organelles exhibited a burst of dynein-dependent inward movement at the growing aster periphery, then mostly halted inside the aster, while dynein-coated beads moved to the aster center at a constant rate, suggesting organelle movement is limited by brake proteins or other sources of drag. These observations call for new models in which all components of the cytoplasm comprise a mechanically integrated aster gel that moves collectively in response to dynein and actomyosin forces.


Asunto(s)
Actinas/metabolismo , Actomiosina/metabolismo , Citoplasma/metabolismo , Dineínas/metabolismo , Microtúbulos/metabolismo , Orgánulos/metabolismo , Animales , Citocinesis , Femenino , Oocitos , Xenopus laevis
7.
Nat Commun ; 11(1): 3521, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32665556

RESUMEN

Microtubules (MTs) mediate mitosis, directional signaling, and are therapeutic targets in cancer. Yet in vivo analysis of cancer cell MT behavior within the tumor microenvironment remains challenging. Here we developed an imaging pipeline using plus-end tip tracking and intravital microscopy to quantify MT dynamics in live xenograft tumor models. Among analyzed features, cancer cells in vivo displayed higher coherent orientation of MT dynamics along their cell major axes compared with 2D in vitro cultures, and distinct from 3D collagen gel cultures. This in vivo MT phenotype was reproduced in vitro when cells were co-cultured with IL4-polarized MΦ. MΦ depletion, MT disruption, targeted kinase inhibition, and altered MΦ polarization via IL10R blockade all reduced MT coherence and/or tumor cell elongation. We show that MT coherence is a defining feature for in vivo tumor cell dynamics and migration, modulated by local signaling from pro-tumor macrophages.


Asunto(s)
Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Animales , Ciclo Celular/genética , Ciclo Celular/fisiología , Movimiento Celular/genética , Movimiento Celular/fisiología , Femenino , Humanos , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos/genética , Mitosis/genética , Mitosis/fisiología , Análisis de Componente Principal , Células RAW 264.7
8.
Cells ; 9(5)2020 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-32349222

RESUMEN

Dysregulation of microtubules and tubulin homeostasis has been linked to developmental disorders, neurodegenerative diseases, and cancer. In general, both microtubule-stabilizing and destabilizing agents have been powerful tools for studies of microtubule cytoskeleton and as clinical agents in oncology. However, many cancers develop resistance to these agents, limiting their utility. We sought to address this by developing a different kind of agent: tubulin-targeted small molecule degraders. Degraders (also known as proteolysis-targeting chimeras (PROTACs)) are compounds that recruit endogenous E3 ligases to a target of interest, resulting in the target's degradation. We developed and examined several series of α- and ß-tubulin degraders, based on microtubule-destabilizing agents. Our results indicate, that although previously reported covalent tubulin binders led to tubulin degradation, in our hands, cereblon-recruiting PROTACs were not efficient. In summary, while we consider tubulin degraders to be valuable tools for studying the biology of tubulin homeostasis, it remains to be seen whether the PROTAC strategy can be applied to this target of high clinical relevance.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteolisis , Tubulina (Proteína) , Ubiquitina-Proteína Ligasas , Humanos , Proteínas Adaptadoras Transductoras de Señales/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Línea Celular Tumoral , Ingeniería de Proteínas/métodos , Bibliotecas de Moléculas Pequeñas , Tubulina (Proteína)/metabolismo , Tubulina (Proteína)/fisiología , Ubiquitina-Proteína Ligasas/efectos de los fármacos , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
9.
Science ; 367(6473): 100-104, 2020 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-31727855

RESUMEN

Tubulins play crucial roles in cell division, intracellular traffic, and cell shape. Tubulin concentration is autoregulated by feedback control of messenger RNA (mRNA) degradation via an unknown mechanism. We identified tetratricopeptide protein 5 (TTC5) as a tubulin-specific ribosome-associating factor that triggers cotranslational degradation of tubulin mRNAs in response to excess soluble tubulin. Structural analysis revealed that TTC5 binds near the ribosome exit tunnel and engages the amino terminus of nascent tubulins. TTC5 mutants incapable of ribosome or nascent tubulin interaction abolished tubulin autoregulation and showed chromosome segregation defects during mitosis. Our findings show how a subset of mRNAs can be targeted for coordinated degradation by a specificity factor that recognizes the nascent polypeptides they encode.


Asunto(s)
Retroalimentación Fisiológica , Estabilidad del ARN , ARN Mensajero/química , Ribosomas/metabolismo , Factores de Transcripción/fisiología , Tubulina (Proteína)/metabolismo , Células HEK293 , Humanos , Mutación , Biosíntesis de Proteínas , Factores de Transcripción/genética , Tubulina (Proteína)/genética
10.
Mol Cell Oncol ; 6(6): e1648739, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31692941

RESUMEN

We have discovered an organoid culture approach that recapitulates morphology and coordinated development of a benign breast tumor. This system may be useful to groups investigating normal mammary gland biology and coordination of collective cell behavior in the mammary gland.

11.
Proc Natl Acad Sci U S A ; 116(23): 11444-11453, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31101720

RESUMEN

As 3D culture has become central to investigation of tissue biology, mammary epithelial organoids have emerged as powerful tools for investigation of epithelial cell polarization and carcinogenesis. However, most current protocols start from single cells suspended in Matrigel, which can also restrict cell differentiation and behavior. Here, we show that the noncancerous mammary cell line HMT-3522 S1, when allowed to spontaneously form cell aggregates ("spheroids") in medium without Matrigel, switches to a collective growth mode that recapitulates many attributes of "usual ductal hyperplasia" (UDH), a common benign mammary lesion. Interestingly, these spheroids undergo a complex maturation process reminiscent of embryonic development: solid-cell cords form their own basement membrane, grow on the surface of initially homogeneous cell aggregates, and form asymmetric lumina lined by two distinct cell types that express basal and luminal cytokeratins. This sequence of events provides a cellular mechanism that explains how the characteristic crescent-shaped, asymmetrical lumina form in UDH. Our results suggest that HMT-3522 S1 spheroids are useful as an in vitro model system to study UDH biology, glandular lumen formation, and stem cell biology of the mammary gland.


Asunto(s)
Neoplasias de la Mama/patología , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Glándulas Mamarias Humanas/patología , Organoides/patología , Membrana Basal/patología , Línea Celular Tumoral , Células Epiteliales/patología , Femenino , Humanos
12.
Mol Syst Biol ; 15(2): e8636, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30782979

RESUMEN

The liver and kidney in mammals play central roles in protecting the organism from xenobiotics and are at high risk of xenobiotic-induced injury. Xenobiotic-induced tissue injury has been extensively studied from both classical histopathological and biochemical perspectives. Here, we introduce a machine-learning approach to analyze toxicological response. Unsupervised characterization of physiological and histological changes in a large toxicogenomic dataset revealed nine discrete toxin-induced disease states, some of which correspond to known pathology, but others were novel. Analysis of dynamics revealed transitions between disease states at constant toxin exposure, mostly toward decreased pathology, implying induction of tolerance. Tolerance correlated with induction of known xenobiotic defense genes and decrease of novel ferroptosis sensitivity biomarkers, suggesting ferroptosis as a druggable driver of tissue pathophysiology. Lastly, mechanism of body weight decrease, a known primary marker for xenobiotic toxicity, was investigated. Combined analysis of food consumption, body weight, and molecular biomarkers indicated that organ injury promotes cachexia by whole-body signaling through Gdf15 and Igf1, suggesting strategies for therapeutic intervention that may be broadly relevant to human disease.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/diagnóstico , Riñón/efectos de los fármacos , Hígado/efectos de los fármacos , Xenobióticos/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/fisiopatología , Factor 15 de Diferenciación de Crecimiento/genética , Humanos , Riñón/patología , Hígado/patología , Transducción de Señal/efectos de los fármacos , Fenómenos Toxicológicos/genética , Aprendizaje Automático no Supervisado
13.
Proc Natl Acad Sci U S A ; 115(48): E11406-E11414, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30429313

RESUMEN

Drug receptor site occupancy is a central pharmacology parameter that quantitatively relates the biochemistry of drug binding to the biology of drug action. Taxanes and epothilones bind to overlapping sites in microtubules (MTs) and stabilize them. They are used to treat cancer and are under investigation for neurodegeneration. In cells, they cause concentration-dependent inhibition of MT dynamics and perturbation of mitosis, but the degree of site occupancy required to trigger different effects has not been measured. We report a live cell assay for taxane-site occupancy, and relationships between site occupancy and biological effects across four drugs and two cell lines. By normalizing to site occupancy, we were able to quantitatively compare drug activities and cell sensitivities independent of differences in drug affinity and uptake/efflux kinetics. Across all drugs and cells tested, we found that inhibition of MT dynamics, postmitotic micronucleation, and mitotic arrest required successively higher site occupancy. We also found interesting differences between cells and drugs, for example, insensitivity of the spindle assembly checkpoint to site occupancy. By extending our assay to a mouse xenograft tumor model, we estimated the initial site occupancy required for paclitaxel to completely prevent tumor growth as 80%. The most important cellular action of taxanes for cancer treatment may be formation of micronuclei, which occurs over a broad range of site occupancies.


Asunto(s)
Antineoplásicos/metabolismo , Hidrocarburos Aromáticos con Puentes/metabolismo , Taxoides/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacología , Transporte Biológico , Hidrocarburos Aromáticos con Puentes/química , Hidrocarburos Aromáticos con Puentes/farmacología , Línea Celular Tumoral , Epotilonas/química , Epotilonas/metabolismo , Epotilonas/farmacología , Humanos , Cinética , Microscopía , Microtúbulos/química , Microtúbulos/metabolismo , Taxoides/química , Taxoides/farmacología
14.
Nat Commun ; 9(1): 3116, 2018 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-30082792

RESUMEN

Glioblastoma multiforme (GBM) is an aggressive primary brain cancer that includes focal amplification of PDGFRα and for which there are no effective therapies. Herein, we report the development of a genetically engineered mouse model of GBM based on autocrine, chronic stimulation of overexpressed PDGFRα, and the analysis of GBM signaling pathways using proteomics. We discover the tubulin-binding protein Stathmin1 (STMN1) as a PDGFRα phospho-regulated target, and that this mis-regulation confers sensitivity to vinblastine (VB) cytotoxicity. Treatment of PDGFRα-positive mouse and a patient-derived xenograft (PDX) GBMs with VB in mice prolongs survival and is dependent on STMN1. Our work reveals a previously unconsidered link between PDGFRα activity and STMN1, and highlight an STMN1-dependent cytotoxic effect of VB in GBM.


Asunto(s)
Neoplasias de la Mama/metabolismo , Resistencia a Antineoplásicos , Glioblastoma/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Estatmina/metabolismo , Vinblastina/farmacología , Animales , Antineoplásicos/farmacología , Apoptosis , Ciclo Celular , Supervivencia Celular , Células Cultivadas , Biología Computacional , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Imagen por Resonancia Magnética , Masculino , Ratones , Trasplante de Neoplasias , Fosforilación , Proteómica , Transducción de Señal
15.
Cell Metab ; 27(5): 1067-1080.e5, 2018 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-29685734

RESUMEN

The redox cofactor nicotinamide adenine dinucleotide (NAD) plays a central role in metabolism and is a substrate for signaling enzymes including poly-ADP-ribose-polymerases (PARPs) and sirtuins. NAD concentration falls during aging, which has triggered intense interest in strategies to boost NAD levels. A limitation in understanding NAD metabolism has been reliance on concentration measurements. Here, we present isotope-tracer methods for NAD flux quantitation. In cell lines, NAD was made from nicotinamide and consumed largely by PARPs and sirtuins. In vivo, NAD was made from tryptophan selectively in the liver, which then excreted nicotinamide. NAD fluxes varied widely across tissues, with high flux in the small intestine and spleen and low flux in the skeletal muscle. Intravenous administration of nicotinamide riboside or mononucleotide delivered intact molecules to multiple tissues, but the same agents given orally were metabolized to nicotinamide in the liver. Thus, flux analysis can reveal tissue-specific NAD metabolism.


Asunto(s)
Hígado/metabolismo , NAD/análisis , NAD/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Sirtuinas/metabolismo , Triptófano/metabolismo , Animales , Femenino , Células HCT116 , Células Hep G2 , Humanos , Intestino Delgado/metabolismo , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , NAD/biosíntesis , Niacinamida/administración & dosificación , Niacinamida/farmacocinética , Bazo/metabolismo
17.
Mol Biol Cell ; 28(14): 1975-1983, 2017 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-28468979

RESUMEN

The spindle is a dynamic structure that changes its architecture and size in response to biochemical and physical cues. For example, a simple physical change, cell confinement, can trigger centrosome separation and increase spindle steady-state length at metaphase. How this occurs is not understood, and is the question we pose here. We find that metaphase and anaphase spindles elongate at the same rate when confined, suggesting that similar elongation forces can be generated independent of biochemical and spindle structural differences. Furthermore, this elongation does not require bipolar spindle architecture or dynamic microtubules. Rather, confinement increases numbers of astral microtubules laterally contacting the cortex, shifting contact geometry from "end-on" to "side-on." Astral microtubules engage cortically anchored motors along their length, as demonstrated by outward sliding and buckling after ablation-mediated release from the centrosome. We show that dynein is required for confinement-induced spindle elongation, and both chemical and physical centrosome removal demonstrate that astral microtubules are required for such spindle elongation and its maintenance. Together the data suggest that promoting lateral cortex-microtubule contacts increases dynein-mediated force generation and is sufficient to drive spindle elongation. More broadly, changes in microtubule-to-cortex contact geometry could offer a mechanism for translating changes in cell shape into dramatic intracellular remodeling.


Asunto(s)
Microtúbulos/fisiología , Huso Acromático/fisiología , Anafase/fisiología , Animales , Caenorhabditis elegans/fisiología , Técnicas de Cultivo de Célula , Ciclo Celular/fisiología , Forma de la Célula/fisiología , Centrosoma/fisiología , Dineínas/fisiología , Cinesinas/fisiología , Mamíferos , Metafase/fisiología , Proteínas Asociadas a Microtúbulos/fisiología , Microtúbulos/metabolismo , Huso Acromático/metabolismo
18.
Mol Biol Cell ; 28(11): 1444-1456, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28404751

RESUMEN

The chromosomal passenger complex (CPC) is a conserved, essential regulator of cell division. As such, significant anti-cancer drug development efforts have been focused on targeting it, most notably by inhibiting its AURKB kinase subunit. The CPC is activated by AURKB-catalyzed autophosphorylation on multiple subunits, but how this regulates CPC interactions with other mitotic proteins remains unclear. We investigated the hydrodynamic behavior of the CPC in Xenopus laevis egg cytosol using sucrose gradient sedimentation and in HeLa cells using fluorescence correlation spectroscopy. We found that autophosphorylation of the CPC decreases its sedimentation coefficient in egg cytosol and increases its diffusion coefficient in live cells, indicating a decrease in mass. Using immunoprecipitation coupled with mass spectrometry and immunoblots, we discovered that inactive, unphosphorylated CPC interacts with nucleophosmin/nucleoplasmin proteins, which are known to oligomerize into pentamers and decamers. Autophosphorylation of the CPC causes it to dissociate from nucleophosmin/nucleoplasmin. We propose that nucleophosmin/nucleoplasmin complexes serve as chaperones that negatively regulate the CPC and/or stabilize its inactive form, preventing CPC autophosphorylation and recruitment to chromatin and microtubules in mitosis.


Asunto(s)
Proteínas Nucleares/metabolismo , Nucleoplasminas/metabolismo , Animales , División Celular , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Cromosomas , Células HeLa/metabolismo , Humanos , Hidrodinámica , Microtúbulos/metabolismo , Mitosis , Chaperonas Moleculares/metabolismo , Nucleofosmina , Huso Acromático/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo
19.
Endocr Relat Cancer ; 24(9): T83-T96, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28249963

RESUMEN

Anti-mitotic cancer drugs include classic microtubule-targeting drugs, such as taxanes and vinca alkaloids, and the newer spindle-targeting drugs, such as inhibitors of the motor protein; Kinesin-5 (aka KSP, Eg5, KIF11); and Aurora-A, Aurora-B and Polo-like kinases. Microtubule-targeting drugs are among the first line of chemotherapies for a wide spectrum of cancers, but patient responses vary greatly. We still lack understanding of how these drugs achieve a favorable therapeutic index, and why individual patient responses vary. Spindle-targeting drugs have so far shown disappointing results in the clinic, but it is possible that certain patients could benefit if we understand their mechanism of action better. Pre-clinical data from both cell culture and mouse tumor models showed that the cell death response is the most variable point of the drug action. Hence, in this review we focus on current mechanistic understanding of the cell death response to anti-mitotics. We first draw on extensive results from cell culture studies, and then cross-examine them with the more limited data from animal tumor models and the clinic. We end by discussing how cell type variation in cell death response might be harnessed to improve anti-mitotic chemotherapy by better patient stratification, new drug combinations and identification of novel targets for drug development.


Asunto(s)
Antimitóticos/farmacología , Antimitóticos/uso terapéutico , Muerte Celular/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Animales , Daño del ADN , Modelos Animales de Enfermedad , Humanos , Ratones , Microtúbulos , Neoplasias/genética , Huso Acromático , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética
20.
Elife ; 62017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-28244368

RESUMEN

Purified microtubules have been shown to align along the static magnetic field (SMF) in vitro because of their diamagnetic anisotropy. However, whether mitotic spindle in cells can be aligned by magnetic field has not been experimentally proved. In particular, the biological effects of SMF of above 20 T (Tesla) have never been reported. Here we found that in both CNE-2Z and RPE1 human cells spindle orients in 27 T SMF. The direction of spindle alignment depended on the extent to which chromosomes were aligned to form a planar metaphase plate. Our results show that the magnetic torque acts on both microtubules and chromosomes, and the preferred direction of spindle alignment relative to the field depends more on chromosome alignment than microtubules. In addition, spindle morphology was also perturbed by 27 T SMF. This is the first reported study that investigated the cellular responses to ultra-high magnetic field of above 20 T. Our study not only found that ultra-high magnetic field can change the orientation and morphology of mitotic spindles, but also provided a tool to probe the role of spindle orientation and perturbation in developmental and cancer biology.


Asunto(s)
Campos Magnéticos , Mitosis/efectos de la radiación , Huso Acromático/efectos de la radiación , Línea Celular , Cromosomas Humanos/efectos de la radiación , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA