Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Genet ; 50: 45-66, 2016 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-27893964

RESUMEN

Chromosomal instability (CIN), the persistent inability of a cell to faithfully segregate its genome, is a feature of many cancer cells. It stands to reason that CIN enables the acquisition of multiple cancer hallmarks; however, there is a growing body of evidence suggesting that CIN impairs cellular fitness and prevents neoplastic transformation. Here, we suggest a new perspective to reconcile this apparent paradox and share an unexpected link between aneuploidy and aging that was discovered through attempts to investigate the CIN-cancer relationship. Additionally, we provide a comprehensive overview of the function and regulation of the anaphase-promoting complex, an E3 ubiquitin ligase that mediates high-fidelity chromosome segregation, and describe the mechanisms that lead to whole-chromosome gain or loss. With this review, we aim to expand our understanding of the role of CIN in cancer and aging with the long-term objective of harnessing this information for the advancement of patient care.


Asunto(s)
Envejecimiento/genética , Aneuploidia , Neoplasias/genética , Ciclosoma-Complejo Promotor de la Anafase/genética , Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Cromátides , Inestabilidad Cromosómica , Humanos
2.
Nature ; 562(7728): 578-582, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30232451

RESUMEN

Cellular senescence, which is characterized by an irreversible cell-cycle arrest1 accompanied by a distinctive secretory phenotype2, can be induced through various intracellular and extracellular factors. Senescent cells that express the cell cycle inhibitory protein p16INK4A have been found to actively drive naturally occurring age-related tissue deterioration3,4 and contribute to several diseases associated with ageing, including atherosclerosis5 and osteoarthritis6. Various markers of senescence have been observed in patients with neurodegenerative diseases7-9; however, a role for senescent cells in the aetiology of these pathologies is unknown. Here we show a causal link between the accumulation of senescent cells and cognition-associated neuronal loss. We found that the MAPTP301SPS19 mouse model of tau-dependent neurodegenerative disease10 accumulates p16INK4A-positive senescent astrocytes and microglia. Clearance of these cells as they arise using INK-ATTAC transgenic mice prevents gliosis, hyperphosphorylation of both soluble and insoluble tau leading to neurofibrillary tangle deposition, and degeneration of cortical and hippocampal neurons, thus preserving cognitive function. Pharmacological intervention with a first-generation senolytic modulates tau aggregation. Collectively, these results show that senescent cells have a role in the initiation and progression of tau-mediated disease, and suggest that targeting senescent cells may provide a therapeutic avenue for the treatment of these pathologies.


Asunto(s)
Senescencia Celular , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Neuroglía/metabolismo , Neuroglía/patología , Proteínas tau/metabolismo , Compuestos de Anilina/farmacología , Animales , Astrocitos/metabolismo , Astrocitos/patología , Senescencia Celular/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Femenino , Gliosis/metabolismo , Humanos , Masculino , Ratones , Ratones Transgénicos , Ovillos Neurofibrilares/metabolismo , Fosforilación/efectos de los fármacos , Solubilidad , Sulfonamidas/farmacología , Transgenes , Proteínas tau/química
3.
Mol Cell ; 62(2): 272-283, 2016 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-27151442

RESUMEN

Expanded CAG repeats lead to debilitating neurodegenerative disorders characterized by aggregation of proteins with expanded polyglutamine (polyQ) tracts. The mechanism of aggregation involves primary and secondary nucleation steps. We show how a noncanonical member of the DNAJ-chaperone family, DNAJB6, inhibits the conversion of soluble polyQ peptides into amyloid fibrils, in particular by suppressing primary nucleation. This inhibition is mediated by a serine/threonine-rich region that provides an array of surface-exposed hydroxyl groups that bind to polyQ peptides and may disrupt the formation of the H bonds essential for the stability of amyloid fibrils. Early prevention of polyQ aggregation by DNAJB6 occurs also in cells and leads to delayed neurite retraction even before aggregates are visible. In a mouse model, brain-specific coexpression of DNAJB6 delays polyQ aggregation, relieves symptoms, and prolongs lifespan, pointing to DNAJB6 as a potential target for disease therapy and tool for unraveling early events in the onset of polyQ diseases.

4.
Psychosom Med ; 2023 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-37910129

RESUMEN

OBJECTIVE: Despite advances toward understanding the etiology of Alzheimer's disease (AD), it remains unclear which aspects of this disease are affected by environmental factors. Chronic life stress increases risk for aging-related diseases including AD. The impact of stress on tauopathies remains understudied. We examined the effects of stress elicited by social (chronic subordination stress, CSS) or psychological/physical (chronic restraint stress, CRS) factors - on the PS19 mouse model of tauopathy. METHODS: Male PS19 mice (average age 6.3 months) were randomized to receive CSS, CRS, or to remain as singly-housed controls. Behavioral tests were used to assess anxiety-like behaviors and cognitive functions. Immunofluorescence staining and western blotting analysis were used to measure levels of astrogliosis, microgliosis and tau burden. Immunohistochemistry was used to assess glucocorticoid receptor expression. RESULTS: PS19 mice exhibit neuroinflammation (GFAP, t-tests; p = 0.0297; Iba1, t-tests; p = 0.006) and tau hyperphosphorylation (t-test, p = 0.0446) in the hippocampus, reduced anxiety (post hoc, p = 0.046), and cognitive deficits, when compared to wild type mice. Surprisingly, CRS reduced hippocampal levels of both total tau and phospho-tauS404 (t-test, p = 0.0116), and attenuated some aspects of both astrogliosis and microgliosis in PS19 mice (t-tests, p = 0.068 to p = 0.0003); however, this was not associated with significant changes in neurodegeneration or cognitive function. Anxiety-like behaviors were increased by CRS (post hoc, p = 0.046). Conversely, CSS impaired spatial learning in Barnes Maze without impacting tau phosphorylation or neurodegeneration and having a minimal impact on gliosis. CONCLUSIONS: Our results demonstrate that social or psychological stress can differentially impact anxiety-like behavior, select cognitive functions, and some aspects of tau-dependent pathology in PS19 male mice, providing entry points for the development of experimental approaches designed to slow AD progression.

5.
Cell ; 133(1): 103-15, 2008 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-18394993

RESUMEN

RanBP2 is a nucleoporin with SUMO E3 ligase activity that functions in both nucleocytoplasmic transport and mitosis. However, the biological relevance of RanBP2 and the in vivo targets of its E3 ligase activity are unknown. Here we show that animals with low amounts of RanBP2 develop severe aneuploidy in the absence of overt transport defects. The main chromosome segregation defect in cells from these mice is anaphase-bridge formation. Topoisomerase IIalpha (Topo IIalpha), which decatenates sister centromeres prior to anaphase onset to prevent bridges, fails to accumulate at inner centromeres when RanBP2 levels are low. We find that RanBP2 sumoylates Topo IIalpha in mitosis and that this modification is required for its proper localization to inner centromeres. Furthermore, mice with low amounts of RanBP2 are highly sensitive to tumor formation. Together, these data identify RanBP2 as a chromosomal instability gene that regulates Topo IIalpha by sumoylation and suppresses tumorigenesis.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Centrómero/metabolismo , ADN-Topoisomerasas de Tipo II/metabolismo , Proteínas de Unión al ADN/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Anafase , Aneuploidia , Animales , Carcinógenos , Fibroblastos/citología , Fibroblastos/metabolismo , Ratones , Ratones Noqueados , Mitosis , Chaperonas Moleculares/genética , Mutación , Neoplasias/inducido químicamente , Neoplasias/metabolismo , Proteínas de Complejo Poro Nuclear/genética , Estructura Terciaria de Proteína , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo
6.
Mol Cell ; 60(1): 21-34, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26387737

RESUMEN

Mutations in the E3 ubiquitin ligase Parkin have been linked to familial Parkinson's disease. Parkin has also been implicated in mitosis through mechanisms that are unclear. Here we show that Parkin interacts with anaphase promoting complex/cyclosome (APC/C) coactivators Cdc20 and Cdh1 to mediate the degradation of several key mitotic regulators independent of APC/C. We demonstrate that ordered progression through mitosis is orchestrated by two distinct E3 ligases through the shared use of Cdc20 and Cdh1. Furthermore, Parkin is phosphorylated and activated by polo-like kinase 1 (Plk1) during mitosis. Parkin deficiency results in overexpression of its substrates, mitotic defects, genomic instability, and tumorigenesis. These results suggest that the Parkin-Cdc20/Cdh1 complex is an important regulator of mitosis.


Asunto(s)
Cadherinas/metabolismo , Proteínas Cdc20/metabolismo , Inestabilidad Genómica , Mitosis , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Carcinogénesis/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Embrión de Mamíferos/citología , Fibroblastos/citología , Fibroblastos/metabolismo , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Ratones , Mutación , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Quinasa Tipo Polo 1
7.
Hepatology ; 73(1): 303-317, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32259305

RESUMEN

BACKGROUND AND AIMS: Up-regulation of the E2F-dependent transcriptional network has been identified in nearly every human malignancy and is an important driver of tumorigenesis. Two members of the E2F family, E2F7 and E2F8, are potent repressors of E2F-dependent transcription. They are atypical in that they do not bind to dimerization partner proteins and are not controlled by retinoblastoma protein. The physiological relevance of E2F7 and E2F8 remains incompletely understood, largely because tools to manipulate their activity in vivo have been lacking. APPROACH AND RESULTS: Here, we generated transgenic mice with doxycycline-controlled transcriptional activation of E2f7 and E2f8 and induced their expression during postnatal development, in adulthood, and in the context of cancer. Systemic induction of E2f7 and, to lesser extent, E2f8 transgenes in juvenile mice impaired cell proliferation, caused replication stress, DNA damage, and apoptosis, and inhibited animal growth. In adult mice, however, E2F7 and E2F8 induction was well tolerated, yet profoundly interfered with DNA replication, DNA integrity, and cell proliferation in diethylnitrosamine-induced liver tumors. CONCLUSION: Collectively, our findings demonstrate that atypical E2Fs can override cell-cycle entry and progression governed by other E2F family members and suggest that this property can be exploited to inhibit proliferation of neoplastic hepatocytes when growth and development have subsided during adulthood.


Asunto(s)
Proliferación Celular , Factor de Transcripción E2F7/fisiología , Hepatocitos/metabolismo , Neoplasias Hepáticas/patología , Proteínas Represoras/fisiología , Animales , Apoptosis/fisiología , Ciclo Celular/fisiología , Daño del ADN , Factor de Transcripción E2F7/deficiencia , Factor de Transcripción E2F7/genética , Células HeLa , Humanos , Neoplasias Hepáticas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Activación Transcripcional
8.
Nature ; 530(7589): 184-9, 2016 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-26840489

RESUMEN

Cellular senescence, a stress-induced irreversible growth arrest often characterized by expression of p16(Ink4a) (encoded by the Ink4a/Arf locus, also known as Cdkn2a) and a distinctive secretory phenotype, prevents the proliferation of preneoplastic cells and has beneficial roles in tissue remodelling during embryogenesis and wound healing. Senescent cells accumulate in various tissues and organs over time, and have been speculated to have a role in ageing. To explore the physiological relevance and consequences of naturally occurring senescent cells, here we use a previously established transgene, INK-ATTAC, to induce apoptosis in p16(Ink4a)-expressing cells of wild-type mice by injection of AP20187 twice a week starting at one year of age. We show that compared to vehicle alone, AP20187 treatment extended median lifespan in both male and female mice of two distinct genetic backgrounds. The clearance of p16(Ink4a)-positive cells delayed tumorigenesis and attenuated age-related deterioration of several organs without apparent side effects, including kidney, heart and fat, where clearance preserved the functionality of glomeruli, cardio-protective KATP channels and adipocytes, respectively. Thus, p16(Ink4a)-positive cells that accumulate during adulthood negatively influence lifespan and promote age-dependent changes in several organs, and their therapeutic removal may be an attractive approach to extend healthy lifespan.


Asunto(s)
Envejecimiento/patología , Envejecimiento/fisiología , Senescencia Celular/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Salud , Longevidad/fisiología , Adipocitos/citología , Adipocitos/patología , Adipocitos/fisiología , Animales , Apoptosis , Separación Celular , Transformación Celular Neoplásica/patología , Células Epiteliales/citología , Células Epiteliales/patología , Femenino , Riñón/citología , Riñón/patología , Riñón/fisiología , Riñón/fisiopatología , Lipodistrofia/patología , Masculino , Ratones , Miocardio/citología , Miocardio/metabolismo , Miocardio/patología , Especificidad de Órganos , Células Madre/citología , Células Madre/patología , Factores de Tiempo
9.
Proc Natl Acad Sci U S A ; 116(27): 13311-13319, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31209047

RESUMEN

Cellular senescence defines an irreversible cell growth arrest state linked to loss of tissue function and aging in mammals. This transition from proliferation to senescence is typically characterized by increased expression of the cell-cycle inhibitor p16INK4a and formation of senescence-associated heterochromatin foci (SAHF). SAHF formation depends on HIRA-mediated nucleosome assembly of histone H3.3, which is regulated by the serine/threonine protein kinase Pak2. However, it is unknown if Pak2 contributes to cellular senescence. Here, we show that depletion of Pak2 delayed oncogene-induced senescence in IMR90 human fibroblasts and oxidative stress-induced senescence of mouse embryonic fibroblasts (MEFs), whereas overexpression of Pak2 accelerated senescence of IMR90 cells. Importantly, depletion of Pak2 in BubR1 progeroid mice attenuated the onset of aging-associated phenotypes and extended life span. Pak2 is required for expression of genes involved in cellular senescence and regulated the deposition of newly synthesized H3.3 onto chromatin in senescent cells. Together, our results demonstrate that Pak2 is an important regulator of cellular senescence and organismal aging, in part through the regulation of gene expression and H3.3 nucleosome assembly.


Asunto(s)
Envejecimiento , Senescencia Celular , Quinasas p21 Activadas/fisiología , Envejecimiento/metabolismo , Animales , Línea Celular , Inmunoprecipitación de Cromatina , Regulación de la Expresión Génica , Histonas/metabolismo , Longevidad , Ratones Noqueados , Estrés Oxidativo , Reacción en Cadena en Tiempo Real de la Polimerasa , Quinasas p21 Activadas/metabolismo
10.
Hum Mol Genet ; 28(11): 1822-1836, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30668728

RESUMEN

BUB-related 1 (BubR1) encoded by Budding Uninhibited by Benzimidazole 1B (BUB1B) is a crucial mitotic checkpoint protein ensuring proper segregation of chromosomes during mitosis. Mutations of BUB1B are responsible for mosaic variegated aneuploidy (MVA), a human congenital disorder characterized by extensive abnormalities in chromosome number. Although microcephaly is a prominent feature of MVA carrying the BUB1B mutation, how BubR1 deficiency disturbs neural progenitor proliferation and neuronal output and leads to microcephaly is unknown. Here we show that conditional loss of BubR1 in mouse cerebral cortex recapitulates microcephaly. BubR1-deficient cortex includes a strikingly reduced number of late-born, but not of early-born, neurons, although BubR1 expression is substantially reduced from an early stage. Importantly, absence of BubR1 decreases the proportion of neural progenitors in mitosis, specifically in metaphase, suggesting shortened mitosis owing to premature chromosome segregation. In the BubR1 mutant, massive apoptotic cell death, which is likely due to the compromised genomic integrity that results from aberrant mitosis, depletes progenitors and neurons during neurogenesis. There is no apparent alteration in centrosome number, spindle formation or primary cilia, suggesting that the major effect of BubR1 deficiency on neural progenitors is to impair the mitotic checkpoint. This finding highlights the importance of the mitotic checkpoint in the pathogenesis of microcephaly. Furthermore, the ependymal cell layer does not form in the conditional knockout, revealing an unrecognized role of BubR1 in assuring the integrity of the ventricular system, which may account for the presence of hydrocephalus in some patients.


Asunto(s)
Proteínas de Ciclo Celular/genética , Microcefalia/genética , Mitosis/genética , Neurogénesis/genética , Proteínas Serina-Treonina Quinasas/genética , Alelos , Animales , Apoptosis/genética , Proteínas de Ciclo Celular/deficiencia , Proliferación Celular/genética , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Trastornos de los Cromosomas/genética , Trastornos de los Cromosomas/fisiopatología , Segregación Cromosómica/genética , Modelos Animales de Enfermedad , Humanos , Ratones , Microcefalia/metabolismo , Microcefalia/fisiopatología , Mosaicismo , Mutación/genética , Neuronas/metabolismo , Neuronas/patología , Proteínas Serina-Treonina Quinasas/deficiencia , Huso Acromático/genética , Huso Acromático/patología
11.
Gastroenterology ; 157(1): 210-226.e12, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30878468

RESUMEN

BACKGROUND & AIMS: The CCNE1 locus, which encodes cyclin E1, is amplified in many types of cancer cells and is activated in hepatocellular carcinomas (HCCs) from patients infected with hepatitis B virus or adeno-associated virus type 2, due to integration of the virus nearby. We investigated cell-cycle and oncogenic effects of cyclin E1 overexpression in tissues of mice. METHODS: We generated mice with doxycycline-inducible expression of Ccne1 (Ccne1T mice) and activated overexpression of cyclin E1 from age 3 weeks onward. At 14 months of age, livers were collected from mice that overexpress cyclin E1 and nontransgenic mice (controls) and analyzed for tumor burden and by histology. Mouse embryonic fibroblasts (MEFs) and hepatocytes from Ccne1T and control mice were analyzed to determine the extent to which cyclin E1 overexpression perturbs S-phase entry, DNA replication, and numbers and structures of chromosomes. Tissues from 4-month-old Ccne1T and control mice (at that age were free of tumors) were analyzed for chromosome alterations, to investigate the mechanisms by which cyclin E1 predisposes hepatocytes to transformation. RESULTS: Ccne1T mice developed more hepatocellular adenomas and HCCs than control mice. Tumors developed only in livers of Ccne1T mice, despite high levels of cyclin E1 in other tissues. Ccne1T MEFs had defects that promoted chromosome missegregation and aneuploidy, including incomplete replication of DNA, centrosome amplification, and formation of nonperpendicular mitotic spindles. Whereas Ccne1T mice accumulated near-diploid aneuploid cells in multiple tissues and organs, polyploidization was observed only in hepatocytes, with losses and gains of whole chromosomes, DNA damage, and oxidative stress. CONCLUSIONS: Livers, but not other tissues of mice with inducible overexpression of cyclin E1, develop tumors. More hepatocytes from the cyclin E1-overexpressing mice were polyploid than from control mice, and had losses or gains of whole chromosomes, DNA damage, and oxidative stress; all of these have been observed in human HCC cells. The increased risk of HCC in patients with hepatitis B virus or adeno-associated virus type 2 infection might involve activation of cyclin E1 and its effects on chromosomes and genomes of liver cells.


Asunto(s)
Adenoma de Células Hepáticas/genética , Carcinoma Hepatocelular/genética , Inestabilidad Cromosómica/genética , Ciclina E/genética , Neoplasias Hepáticas/genética , Hígado/metabolismo , Proteínas Oncogénicas/genética , Adenoma de Células Hepáticas/patología , Adenoma de Células Hepáticas/virología , Animales , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/virología , Estructuras Cromosómicas , Daño del ADN/genética , Replicación del ADN , Dependovirus , Fibroblastos , Hepatitis B Crónica , Hepatocitos , Hígado/patología , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/patología , Ratones , Estrés Oxidativo/genética , Infecciones por Parvoviridae , Parvovirinae , Poliploidía , Puntos de Control de la Fase S del Ciclo Celular
12.
Nature ; 509(7501): 439-46, 2014 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-24848057

RESUMEN

Cellular senescence has historically been viewed as an irreversible cell-cycle arrest mechanism that acts to protect against cancer, but recent discoveries have extended its known role to complex biological processes such as development, tissue repair, ageing and age-related disorders. New insights indicate that, unlike a static endpoint, senescence represents a series of progressive and phenotypically diverse cellular states acquired after the initial growth arrest. A deeper understanding of the molecular mechanisms underlying the multi-step progression of senescence and the development and function of acute versus chronic senescent cells may lead to new therapeutic strategies for age-related pathologies and extend healthy lifespan.


Asunto(s)
Envejecimiento/patología , Senescencia Celular/fisiología , Animales , Enfermedad , Humanos , Longevidad , Mitosis , Modelos Biológicos
13.
Curr Opin Lipidol ; 30(3): 177-185, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30913069

RESUMEN

PURPOSE OF REVIEW: Senescent cells have recently been identified as key players in the development of metabolic dysfunction. In this review, we will highlight recent developments in this field and discuss the concept of targeting these cells to prevent or treat cardiometabolic diseases. RECENT FINDINGS: Evidence is accumulating that cellular senescence contributes to adipose tissue dysfunction, presumably through induction of low-grade inflammation and inhibition of adipogenic differentiation leading to insulin resistance and dyslipidaemia. Senescent cells modulate their surroundings through their bioactive secretome and only a relatively small number of senescent cells is sufficient to cause persistent physical dysfunction even in young mice. Proof-of-principle studies showed that selective elimination of senescent cells can prevent or delay the development of cardiometabolic diseases in mice. SUMMARY: The metabolic consequences of senescent cell accumulation in various tissues are now unravelling and point to new therapeutic opportunities for the treatment of cardiometabolic diseases.


Asunto(s)
Senescencia Celular , Enfermedades Metabólicas/patología , Miocardio/metabolismo , Miocardio/patología , Tejido Adiposo/patología , Animales , Humanos , Enfermedades Metabólicas/complicaciones , Enfermedades Metabólicas/metabolismo
14.
Nucleic Acids Res ; 45(8): 4564-4576, 2017 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-28199696

RESUMEN

Germline mutations in SPRTN cause Ruijs-Aalfs syndrome (RJALS), a disorder characterized by genome instability, progeria and early onset hepatocellular carcinoma. Spartan, the protein encoded by SPRTN, is a nuclear metalloprotease that is involved in the repair of DNA-protein crosslinks (DPCs). Although Sprtn hypomorphic mice recapitulate key progeroid phenotypes of RJALS, whether this model expressing low amounts of Spartan is prone to DPC repair defects and spontaneous tumors is unknown. Here, we showed that the livers of Sprtn hypomorphic mice accumulate DPCs containing Topoisomerase 1 covalently linked to DNA. Furthermore, these mice exhibited DNA damage, aneuploidy and spontaneous tumorigenesis in the liver. Collectively, these findings provide evidence that partial loss of Spartan impairs DPC repair and tumor suppression.


Asunto(s)
Carcinogénesis/genética , Carcinoma Hepatocelular/genética , Proteínas Cromosómicas no Histona/deficiencia , ADN-Topoisomerasas de Tipo I/genética , Neoplasias Hepáticas/genética , Progeria/genética , Aneuploidia , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Proteínas Cromosómicas no Histona/genética , Aductos de ADN/genética , Aductos de ADN/metabolismo , ADN-Topoisomerasas de Tipo I/metabolismo , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Humanos , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Noqueados , Progeria/metabolismo , Progeria/patología , Proteolisis , Síndrome
15.
EMBO J ; 33(13): 1438-53, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24825348

RESUMEN

Mice overexpressing the mitotic checkpoint kinase gene BubR1 live longer, whereas mice hypomorphic for BubR1 (BubR1(H/H)) live shorter and show signs of accelerated aging. As wild-type mice age, BubR1 levels decline in many tissues, a process that is proposed to underlie normal aging and age-related diseases. Understanding why BubR1 declines with age and how to slow this process is therefore of considerable interest. The sirtuins (SIRT1-7) are a family of NAD(+)-dependent deacetylases that can delay age-related diseases. Here, we show that the loss of BubR1 levels with age is due to a decline in NAD(+) and the ability of SIRT2 to maintain lysine-668 of BubR1 in a deacetylated state, which is counteracted by the acetyltransferase CBP. Overexpression of SIRT2 or treatment of mice with the NAD(+) precursor nicotinamide mononucleotide (NMN) increases BubR1 abundance in vivo. Overexpression of SIRT2 in BubR1(H/H) animals increases median lifespan, with a greater effect in male mice. Together, these data indicate that further exploration of the potential of SIRT2 and NAD(+) to delay diseases of aging in mammals is warranted.


Asunto(s)
Longevidad/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Sirtuina 2/metabolismo , Animales , Proteínas de Ciclo Celular , Inducción Enzimática/fisiología , Células HeLa , Humanos , Masculino , Ratones , Ratones Noqueados , NAD/genética , NAD/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Sirtuina 2/genética
16.
Gastroenterology ; 152(5): 1126-1138.e6, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28065787

RESUMEN

BACKGROUND & AIMS: The role of the intestine in the maintenance of cholesterol homeostasis increasingly is recognized. Fecal excretion of cholesterol is the last step in the atheroprotective reverse cholesterol transport pathway, to which biliary and transintestinal cholesterol excretion (TICE) contribute. The mechanisms controlling the flux of cholesterol through the TICE pathway, however, are poorly understood. We aimed to identify mechanisms that regulate and stimulate TICE. METHODS: We performed studies with C57Bl/6J mice, as well as with mice with intestine-specific knockout of the farnesoid X receptor (FXR), mice that express an FXR transgene specifically in the intestine, and ABCG8-knockout mice. Mice were fed a control diet or a diet supplemented with the FXR agonist PX20606, with or without the cholesterol absorption inhibitor ezetimibe. Some mice with intestine-specific knockout of FXR were given daily injections of fibroblast growth factor (FGF)19. To determine fractional cholesterol absorption, mice were given intravenous injections of cholesterol D5 and oral cholesterol D7. Mice were given 13C-acetate in drinking water for measurement of cholesterol synthesis. Bile cannulations were performed and biliary cholesterol secretion rates were assessed. In a separate set of experiments, bile ducts of male Wistar rats were exteriorized, allowing replacement of endogenous bile by a model bile. RESULTS: In mice, we found TICE to be regulated by intestinal FXR via induction of its target gene Fgf15 (FGF19 in rats and human beings). Stimulation of this pathway caused mice to excrete up to 60% of their total cholesterol content each day. PX20606 and FGF19 each increased the ratio of muricholate:cholate in bile, inducing a more hydrophilic bile salt pool. The altered bile salt pool stimulated robust secretion of cholesterol into the intestinal lumen via the sterol-exporting heterodimer adenosine triphosphate binding cassette subfamily G member 5/8 (ABCG5/G8). Of note, the increase in TICE induced by PX20606 was independent of changes in cholesterol absorption. CONCLUSIONS: Hydrophilicity of the bile salt pool, controlled by FXR and FGF15/19, is an important determinant of cholesterol removal via TICE. Strategies that alter bile salt pool composition might be developed for the prevention of cardiovascular disease. Transcript profiling: http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?token=irsrayeohfcntqx&acc=GSE74101.


Asunto(s)
Transportador de Casete de Unión a ATP, Subfamilia G, Miembro 8/genética , Ácidos y Sales Biliares/metabolismo , Colesterol/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Eliminación Intestinal/genética , Mucosa Intestinal/metabolismo , Lipoproteínas/genética , Receptores Citoplasmáticos y Nucleares/genética , Transportador de Casete de Unión a ATP, Subfamilia G, Miembro 8/metabolismo , Animales , Anticolesterolemiantes/farmacología , Benzoatos/farmacología , Conductos Biliares , Ezetimiba/farmacología , Eliminación Intestinal/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Intestinos/efectos de los fármacos , Isoxazoles/farmacología , Lipoproteínas/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Ratas , Ratas Wistar , Receptores Citoplasmáticos y Nucleares/agonistas
17.
Chromosome Res ; 24(1): 67-76, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26615533

RESUMEN

Centrosomes are microtubule-organizing centers that duplicate in S phase to form bipolar spindles that separate duplicated chromosomes faithfully into two daughter cells during cell division. Recent studies show that proper timing of centrosome dynamics, the disjunction and movement of centrosomes, is tightly linked to spindle symmetry, correct microtubule-kinetochore attachment, and chromosome segregation. Here, we review mechanisms that regulate centrosome dynamics, with emphasis on the roles of key mitotic kinases in the proper timing of centrosome dynamics and how aberrancies in these processes may cause chromosomal instability and cancer.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Centrosoma/metabolismo , Inestabilidad Cromosómica , Cromosomas Humanos/metabolismo , Neoplasias/metabolismo , Proteínas Quinasas/metabolismo , Fase S , Animales , Proteínas de Ciclo Celular/genética , Cromosomas Humanos/genética , Humanos , Cinetocoros/metabolismo , Neoplasias/genética , Neoplasias/patología , Proteínas Quinasas/genética
18.
Nature ; 479(7372): 232-6, 2011 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-22048312

RESUMEN

Advanced age is the main risk factor for most chronic diseases and functional deficits in humans, but the fundamental mechanisms that drive ageing remain largely unknown, impeding the development of interventions that might delay or prevent age-related disorders and maximize healthy lifespan. Cellular senescence, which halts the proliferation of damaged or dysfunctional cells, is an important mechanism to constrain the malignant progression of tumour cells. Senescent cells accumulate in various tissues and organs with ageing and have been hypothesized to disrupt tissue structure and function because of the components they secrete. However, whether senescent cells are causally implicated in age-related dysfunction and whether their removal is beneficial has remained unknown. To address these fundamental questions, we made use of a biomarker for senescence, p16(Ink4a), to design a novel transgene, INK-ATTAC, for inducible elimination of p16(Ink4a)-positive senescent cells upon administration of a drug. Here we show that in the BubR1 progeroid mouse background, INK-ATTAC removes p16(Ink4a)-positive senescent cells upon drug treatment. In tissues--such as adipose tissue, skeletal muscle and eye--in which p16(Ink4a) contributes to the acquisition of age-related pathologies, life-long removal of p16(Ink4a)-expressing cells delayed onset of these phenotypes. Furthermore, late-life clearance attenuated progression of already established age-related disorders. These data indicate that cellular senescence is causally implicated in generating age-related phenotypes and that removal of senescent cells can prevent or delay tissue dysfunction and extend healthspan.


Asunto(s)
Envejecimiento/fisiología , Senescencia Celular/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Tejido Adiposo/citología , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/patología , Envejecimiento/efectos de los fármacos , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Recuento de Células , Proteínas de Ciclo Celular , Células Cultivadas , Senescencia Celular/efectos de los fármacos , Ojo/citología , Ojo/efectos de los fármacos , Ojo/patología , Femenino , Expresión Génica , Genotipo , Longevidad/efectos de los fármacos , Longevidad/fisiología , Masculino , Ratones , Ratones Transgénicos , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Fenotipo , Progeria/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Tacrolimus/análogos & derivados , Tacrolimus/farmacología , Factores de Tiempo , Destete
19.
Stroke ; 47(4): 1068-77, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26883501

RESUMEN

BACKGROUND AND PURPOSE: Age-related changes in the cerebrovasculature, including blood-brain barrier (BBB) disruption, are emerging as potential risks for diverse neurological conditions. Because the accumulation of senescent cells in tissues is increasingly recognized as a critical step leading to age-related organ dysfunction, we evaluated whether senescent vascular cells are associated with compromised BBB integrity. METHODS: Effects of vascular cell senescence on tight junction and barrier integrity were studied using an in vitro BBB model, composed of endothelial cells, pericytes, and astrocytes. In addition, tight junction coverage in microvessels and BBB integrity in BubR1 hypomorphic (BubR1(H/H)) mice, which display senescence cell-dependent phenotypes, were examined. RESULTS: When an in vitro BBB model was constructed with senescent endothelial cells and pericytes, tight junction structure and barrier integrity (evaluated by transendothelial electric resistance and tracer efflux assay using sodium fluorescein and Evans blue-albumin were significantly impaired. Endothelial cells and pericytes from BubR1(H/H) mice had increased senescent-associated ß-galactosidase activity and p16(INK4a) expression, demonstrating an exacerbation of senescence. The coverage by tight junction proteins in the cortical microvessels were reduced in BubR1(H/H) mice, consistent with a compromised BBB integrity from permeability assays. Importantly, the coverage of microvessels by end-feet of aquaporin 4-immunoreactive astrocytes was not altered in the cortex of the BubR1(H/H) mice. CONCLUSIONS: Our results indicate that accumulation of senescent vascular cells is associated with compromised BBB integrity, providing insights into the mechanism of BBB disruption related to biological aging.


Asunto(s)
Envejecimiento/patología , Barrera Hematoencefálica/patología , Senescencia Celular/fisiología , Células Endoteliales/patología , Pericitos/patología , Envejecimiento/metabolismo , Animales , Acuaporina 4/metabolismo , Astrocitos/metabolismo , Astrocitos/patología , Barrera Hematoencefálica/metabolismo , Encéfalo/irrigación sanguínea , Permeabilidad Capilar , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Células Endoteliales/metabolismo , Ratones , Pericitos/metabolismo , Uniones Estrechas/metabolismo , beta-Galactosidasa/metabolismo
20.
Hepatology ; 61(4): 1269-83, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25503294

RESUMEN

UNLABELLED: In vitro studies have proposed a tumor suppressor role for sulfatase 1 (SULF1) in hepatocellular carcinoma (HCC); however, high expression in human HCC has been associated with poor prognosis. The reason underlying this paradoxical observation remains to be explored. Using a transgenic (Tg) mouse model overexpressing Sulf1 (Sulf1-Tg), we assessed the effects of SULF1 on the diethylnitrosamine model of liver carcinogenesis. Sulf1-Tg mice show a higher incidence of large and multifocal tumors with diethylnitrosamine injection compared to wild-type mice. Lung metastases were found in 75% of Sulf1-Tg mice but not in wild-type mice. Immunohistochemistry, immunoblotting, and reporter assays all show a significant activation of the transforming growth factor-ß (TGF-ß)/SMAD transcriptional pathway by SULF1 both in vitro and in vivo. This effect of SULF1 on the TGF-ß/SMAD pathway is functional; overexpression of SULF1 promotes TGF-ß-induced gene expression and epithelial-mesenchymal transition and enhances cell migration/invasiveness. Mechanistic analyses demonstrate that inactivating mutation of the catalytic site of SULF1 impairs the above actions of SULF1 and diminishes the release of TGF-ß from the cell surface. We also show that SULF1 expression decreases the interaction between TGF-ß1 and its heparan sulfate proteoglycan sequestration receptor, TGFßR3. Finally, using gene expression from human HCCs, we show that patients with high SULF1 expression have poorer recurrence-free survival (hazard ratio 4.1, 95% confidence interval 1.9-8.3; P = 0.002) compared to patients with low SULF1. We also found strong correlations of SULF1 expression with TGF-ß expression and with several TGF-ß-related epithelial-mesenchymal transition genes in human HCC. CONCLUSION: Our study proposes a novel role of SULF1 in HCC tumor progression through augmentation of the TGF-ß pathway, thus defining SULF1 as a potential biomarker for tumor progression and a novel target for drug development for HCC.


Asunto(s)
Carcinoma Hepatocelular/etiología , Neoplasias Hepáticas/etiología , Proteínas Smad/fisiología , Sulfotransferasas/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Masculino , Ratones , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA