Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 178
Filter
Add more filters

Uruguay Oncology Collection
Publication year range
1.
Genes Dev ; 35(15-16): 1079-1092, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34266888

ABSTRACT

Chromosome gains and losses are a frequent feature of human cancers. However, how these aberrations can outweigh the detrimental effects of aneuploidy remains unclear. An initial comparison of existing chromosomal instability (CIN) mouse models suggests that aneuploidy accumulates to low levels in these animals. We therefore developed a novel mouse model that enables unprecedented levels of chromosome missegregation in the adult animal. At the earliest stages of T-cell development, cells with random chromosome gains and/or losses are selected against, but CIN eventually results in the expansion of progenitors with clonal chromosomal imbalances. Clonal selection leads to the development of T-cell lymphomas with stereotypic karyotypes in which chromosome 15, containing the Myc oncogene, is gained with high prevalence. Expressing human MYC from chromosome 6 (MYCChr6) is sufficient to change the karyotype of these lymphomas to include universal chromosome 6 gains. Interestingly, while chromosome 15 is still gained in MYCChr6 tumors after genetic ablation of the endogenous Myc locus, this chromosome is not efficiently gained after deletion of one copy of Rad21, suggesting a synergistic effect of both MYC and RAD21 in driving chromosome 15 gains. Our results show that the initial detrimental effects of random missegregation are outbalanced by clonal selection, which is dictated by the chromosomal location and nature of certain genes and is sufficient to drive cancer with high prevalence.


Subject(s)
Aneuploidy , Chromosomal Instability , Animals , Cell Transformation, Neoplastic/genetics , Chromosomal Instability/genetics , Chromosome Aberrations , Karyotype , Mice , Prevalence , Stem Cells
2.
Annu Rev Genet ; 50: 45-66, 2016 Nov 23.
Article in English | MEDLINE | ID: mdl-27893964

ABSTRACT

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.


Subject(s)
Aging/genetics , Aneuploidy , Neoplasms/genetics , Anaphase-Promoting Complex-Cyclosome/genetics , Anaphase-Promoting Complex-Cyclosome/metabolism , Chromatids , Chromosomal Instability , Humans
3.
Nature ; 562(7728): 578-582, 2018 10.
Article in English | MEDLINE | ID: mdl-30232451

ABSTRACT

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.


Subject(s)
Cellular Senescence , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Neuroglia/metabolism , Neuroglia/pathology , tau Proteins/metabolism , Aniline Compounds/pharmacology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Female , Gliosis/metabolism , Humans , Male , Mice , Mice, Transgenic , Neurofibrillary Tangles/metabolism , Phosphorylation/drug effects , Solubility , Sulfonamides/pharmacology , Transgenes , tau Proteins/chemistry
4.
Mol Cell ; 62(2): 272-283, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27151442

ABSTRACT

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.

5.
Psychosom Med ; 2023 Oct 03.
Article in English | MEDLINE | ID: mdl-37910129

ABSTRACT

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.

6.
Cell ; 133(1): 103-15, 2008 Apr 04.
Article in English | MEDLINE | ID: mdl-18394993

ABSTRACT

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.


Subject(s)
Antigens, Neoplasm/metabolism , Centromere/metabolism , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Molecular Chaperones/metabolism , Nuclear Pore Complex Proteins/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Anaphase , Aneuploidy , Animals , Carcinogens , Fibroblasts/cytology , Fibroblasts/metabolism , Mice , Mice, Knockout , Mitosis , Molecular Chaperones/genetics , Mutation , Neoplasms/chemically induced , Neoplasms/metabolism , Nuclear Pore Complex Proteins/genetics , Protein Structure, Tertiary , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism
7.
Mol Cell ; 60(1): 21-34, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26387737

ABSTRACT

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.


Subject(s)
Cadherins/metabolism , Cdc20 Proteins/metabolism , Genomic Instability , Mitosis , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Animals , Carcinogenesis/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Knockout Techniques , HEK293 Cells , Humans , Mice , Mutation , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Polo-Like Kinase 1
8.
Hepatology ; 73(1): 303-317, 2021 01.
Article in English | MEDLINE | ID: mdl-32259305

ABSTRACT

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.


Subject(s)
Cell Proliferation , E2F7 Transcription Factor/physiology , Hepatocytes/metabolism , Liver Neoplasms/pathology , Repressor Proteins/physiology , Animals , Apoptosis/physiology , Cell Cycle/physiology , DNA Damage , E2F7 Transcription Factor/deficiency , E2F7 Transcription Factor/genetics , HeLa Cells , Humans , Liver Neoplasms/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Repressor Proteins/deficiency , Repressor Proteins/genetics , Transcriptional Activation
9.
Nature ; 530(7589): 184-9, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26840489

ABSTRACT

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.


Subject(s)
Aging/pathology , Aging/physiology , Cellular Senescence/physiology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Health , Longevity/physiology , Adipocytes/cytology , Adipocytes/pathology , Adipocytes/physiology , Animals , Apoptosis , Cell Separation , Cell Transformation, Neoplastic/pathology , Epithelial Cells/cytology , Epithelial Cells/pathology , Female , Kidney/cytology , Kidney/pathology , Kidney/physiology , Kidney/physiopathology , Lipodystrophy/pathology , Male , Mice , Myocardium/cytology , Myocardium/metabolism , Myocardium/pathology , Organ Specificity , Stem Cells/cytology , Stem Cells/pathology , Time Factors
10.
Proc Natl Acad Sci U S A ; 116(27): 13311-13319, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31209047

ABSTRACT

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.


Subject(s)
Aging , Cellular Senescence , p21-Activated Kinases/physiology , Aging/metabolism , Animals , Cell Line , Chromatin Immunoprecipitation , Gene Expression Regulation , Histones/metabolism , Longevity , Mice, Knockout , Oxidative Stress , Real-Time Polymerase Chain Reaction , p21-Activated Kinases/metabolism
11.
Hum Mol Genet ; 28(11): 1822-1836, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30668728

ABSTRACT

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.


Subject(s)
Cell Cycle Proteins/genetics , Microcephaly/genetics , Mitosis/genetics , Neurogenesis/genetics , Protein Serine-Threonine Kinases/genetics , Alleles , Animals , Apoptosis/genetics , Cell Cycle Proteins/deficiency , Cell Proliferation/genetics , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Chromosome Disorders/genetics , Chromosome Disorders/physiopathology , Chromosome Segregation/genetics , Disease Models, Animal , Humans , Mice , Microcephaly/metabolism , Microcephaly/physiopathology , Mosaicism , Mutation/genetics , Neurons/metabolism , Neurons/pathology , Protein Serine-Threonine Kinases/deficiency , Spindle Apparatus/genetics , Spindle Apparatus/pathology
12.
Gastroenterology ; 157(1): 210-226.e12, 2019 07.
Article in English | MEDLINE | ID: mdl-30878468

ABSTRACT

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.


Subject(s)
Adenoma, Liver Cell/genetics , Carcinoma, Hepatocellular/genetics , Chromosomal Instability/genetics , Cyclin E/genetics , Liver Neoplasms/genetics , Liver/metabolism , Oncogene Proteins/genetics , Adenoma, Liver Cell/pathology , Adenoma, Liver Cell/virology , Animals , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Chromosome Structures , DNA Damage/genetics , DNA Replication , Dependovirus , Fibroblasts , Hepatitis B, Chronic , Hepatocytes , Liver/pathology , Liver Neoplasms/pathology , Liver Neoplasms/virology , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Mice , Oxidative Stress/genetics , Parvoviridae Infections , Parvovirinae , Polyploidy , S Phase Cell Cycle Checkpoints
13.
Circ Res ; 122(12): 1648-1660, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29545368

ABSTRACT

RATIONALE: COMMD (copper metabolism MURR1 domain)-containing proteins are a part of the CCC (COMMD-CCDC22 [coiled-coil domain containing 22]-CCDC93 [coiled-coil domain containing 93]) complex facilitating endosomal trafficking of cell surface receptors. Hepatic COMMD1 inactivation decreases CCDC22 and CCDC93 protein levels, impairs the recycling of the LDLR (low-density lipoprotein receptor), and increases plasma low-density lipoprotein cholesterol levels in mice. However, whether any of the other COMMD members function similarly as COMMD1 and whether perturbation in the CCC complex promotes atherogenesis remain unclear. OBJECTIVE: The main aim of this study is to unravel the contribution of evolutionarily conserved COMMD proteins to plasma lipoprotein levels and atherogenesis. METHODS AND RESULTS: Using liver-specific Commd1, Commd6, or Commd9 knockout mice, we investigated the relation between the COMMD proteins in the regulation of plasma cholesterol levels. Combining biochemical and quantitative targeted proteomic approaches, we found that hepatic COMMD1, COMMD6, or COMMD9 deficiency resulted in massive reduction in the protein levels of all 10 COMMDs. This decrease in COMMD protein levels coincided with destabilizing of the core (CCDC22, CCDC93, and chromosome 16 open reading frame 62 [C16orf62]) of the CCC complex, reduced cell surface levels of LDLR and LRP1 (LDLR-related protein 1), followed by increased plasma low-density lipoprotein cholesterol levels. To assess the direct contribution of the CCC core in the regulation of plasma cholesterol levels, Ccdc22 was deleted in mouse livers via CRISPR/Cas9-mediated somatic gene editing. CCDC22 deficiency also destabilized the complete CCC complex and resulted in elevated plasma low-density lipoprotein cholesterol levels. Finally, we found that hepatic disruption of the CCC complex exacerbates dyslipidemia and atherosclerosis in ApoE3*Leiden mice. CONCLUSIONS: Collectively, these findings demonstrate a strong interrelationship between COMMD proteins and the core of the CCC complex in endosomal LDLR trafficking. Hepatic disruption of either of these CCC components causes hypercholesterolemia and exacerbates atherosclerosis. Our results indicate that not only COMMD1 but all other COMMDs and CCC components may be potential targets for modulating plasma lipid levels in humans.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Atherosclerosis/prevention & control , Cholesterol, LDL/blood , Cytoskeletal Proteins/metabolism , Endosomes/metabolism , Receptors, LDL/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Atherosclerosis/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cholesterol/analysis , Chromatography, High Pressure Liquid , Cytoskeletal Proteins/genetics , Gene Deletion , Gene Expression , HEK293 Cells , Hep G2 Cells , Humans , Liver/chemistry , Liver/metabolism , Low Density Lipoprotein Receptor-Related Protein-1 , Male , Mice , Mice, Knockout , Protein Transport , Triglycerides/analysis , Tumor Suppressor Proteins/metabolism
14.
Nature ; 509(7501): 439-46, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24848057

ABSTRACT

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.


Subject(s)
Aging/pathology , Cellular Senescence/physiology , Animals , Disease , Humans , Longevity , Mitosis , Models, Biological
16.
Proc Natl Acad Sci U S A ; 114(15): 3933-3938, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28348210

ABSTRACT

Focal adhesion kinase (FAK) is a nonreceptor tyrosine kinase involved in development and human disease, including cancer. It is currently thought that the four-point one, ezrin, radixin, moesin (FERM)-kinase domain linker, which contains autophosphorylation site tyrosine (Y) 397, is not required for in vivo FAK function until late midgestation. Here, we directly tested this hypothesis by generating mice with FAK Y397-to-phenylalanine (F) mutations in the germline. We found that Y397F embryos exhibited reduced mesodermal fibronectin (FN) and osteopontin expression and died during mesoderm development akin to FAK kinase-dead mice. We identified myosin-1E (MYO1E), an actin-dependent molecular motor, to interact directly with the FAK FERM-kinase linker and induce FAK kinase activity and Y397 phosphorylation. Active FAK in turn accumulated in the nucleus where it led to the expression of osteopontin and other FN-type matrix in both mouse embryonic fibroblasts and human melanoma. Our data support a model in which FAK Y397 autophosphorylation is required for FAK function in vivo and is positively regulated by MYO1E.


Subject(s)
Focal Adhesion Kinase 1/metabolism , Melanoma/metabolism , Myosins/metabolism , Skin Neoplasms/metabolism , Animals , Embryo Loss/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Female , Fibroblasts/metabolism , Fibronectins/metabolism , Focal Adhesion Kinase 1/chemistry , Focal Adhesion Kinase 1/genetics , Humans , Melanoma/pathology , Mesoderm/embryology , Mice, Mutant Strains , Myosin Type I , Myosins/chemistry , Myosins/genetics , Osteopontin/genetics , Osteopontin/metabolism , Phosphorylation , Pregnancy , Protein Domains , Skin Neoplasms/pathology , Tyrosine/metabolism
17.
Curr Opin Lipidol ; 30(3): 177-185, 2019 06.
Article in English | MEDLINE | ID: mdl-30913069

ABSTRACT

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.


Subject(s)
Cellular Senescence , Metabolic Diseases/pathology , Myocardium/metabolism , Myocardium/pathology , Adipose Tissue/pathology , Animals , Humans , Metabolic Diseases/complications , Metabolic Diseases/metabolism
18.
J Lipid Res ; 60(9): 1547-1561, 2019 09.
Article in English | MEDLINE | ID: mdl-31253637

ABSTRACT

The bile acid-activated nuclear receptor, FXR (NR1H4), has been implicated in the control of lipid and energy metabolism, but its role in fat tissue, where it is moderately expressed, is not understood. In view of the recent development of FXR-targeting therapeutics for treatment of human metabolic diseases, understanding the tissue-specific actions of FXR is essential. Transgenic mice expressing human FXR in adipose tissue (aP2-hFXR mice) at three to five times higher levels than endogenous Fxr, i.e., much lower than its expression in liver and intestine, have markedly enlarged adipocytes and show extensive extracellular matrix remodeling. Ageing and exposure to obesogenic conditions revealed a strongly limited capacity for adipose expansion and development of fibrosis in adipose tissues of aP2-hFXR transgenic mice. This was associated with impaired lipid storage capacity, leading to elevated plasma free fatty acids and ectopic fat deposition in liver and muscle as well as whole-body insulin resistance. These studies establish that adipose FXR is a determinant of adipose tissue architecture and contributes to whole-body lipid homeostasis.


Subject(s)
Adipose Tissue/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Adipocytes/metabolism , Aging/physiology , Animals , Body Weight/physiology , Diet, High-Fat/adverse effects , Extracellular Matrix/metabolism , Humans , Insulin Resistance/physiology , Lipid Metabolism/physiology , Metabolic Diseases/metabolism , Mice , Mice, Transgenic , Tissue Culture Techniques
19.
Gastroenterology ; 154(8): 2209-2221.e14, 2018 06.
Article in English | MEDLINE | ID: mdl-29454793

ABSTRACT

BACKGROUND & AIMS: Hepatic stellate cells (HSCs) contribute to desmoplasia and stiffness of liver metastases by differentiating into matrix-producing myofibroblasts. We investigated whether stiffness due to the presence of tumors increases activation of HSCs into myofibroblasts and their tumor-promoting effects, as well as the role of E1A binding protein p300, a histone acetyltransferase that regulates transcription, in these processes. METHODS: HSCs were isolated from liver tissues of patients, mice in which the p300 gene was flanked by 2 loxP sites (p300F/F mice), and p300+/+ mice (controls). The HSCs were placed on polyacrylamide gels with precisely defined stiffness, and their activation (differentiation into myofibroblasts) was assessed by immunofluorescence and immunoblot analyses for alpha-smooth muscle actin. In HSCs from mice, the p300 gene was disrupted by cre recombinase. In human HSCs, levels of p300 were knocked down with small hairpin RNAs or a mutant form of p300 that is not phosphorylated by AKT (p300S1834A) was overexpressed. Human HSCs were also cultured with inhibitors of p300 (C646), PI3K signaling to AKT (LY294002), or RHOA (C3 transferase) and effects on stiffness-induced activation were measured. RNA sequencing and chromatin immunoprecipitation-quantitative polymerase chain reaction were used to identify HSC genes that changed expression levels in response to stiffness. We measured effects of HSC-conditioned media on proliferation of HT29 colon cancer cells and growth of tumors following subcutaneous injection of these cells into mice. MC38 colon cancer cells were injected into portal veins of p300F/Fcre and control mice, and liver metastases were measured. p300F/Fcre and control mice were given intraperitoneal injections of CCl4 to induce liver fibrosis. Liver tissues were collected and analyzed by immunofluorescence, immunoblot, and histology. RESULTS: Substrate stiffness was sufficient to activate HSCs, leading to nuclear accumulation of p300. Disrupting p300 level or activity blocked stiffness-induced activation of HSCs. In HSCs, substrate stiffness activated AKT signaling via RHOA to induce phosphorylation of p300 at serine 1834; this caused p300 to translocate to the nucleus, where it up-regulated transcription of genes that increase activation of HSCs and metastasis, including CXCL12. MC38 cells, injected into portal veins, formed fewer metastases in livers of p300F/Fcre mice than control mice. Expression of p300 was increased in livers of mice following injection of CCl4; HSC activation and collagen deposition were reduced in livers of p300F/Fcre mice compared with control mice. CONCLUSIONS: In studies of mice, we found liver stiffness to activate HSC differentiation into myofibroblasts, which required nuclear accumulation of p300. p300 increases HSC expression of genes that promote metastasis.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/metabolism , E1A-Associated p300 Protein/metabolism , Hepatic Stellate Cells/pathology , Liver Neoplasms/pathology , Myofibroblasts/pathology , Animals , Benzoates/pharmacology , Carbon Tetrachloride/toxicity , Cell Nucleus/metabolism , Cell Transdifferentiation , E1A-Associated p300 Protein/genetics , Gene Expression Profiling , Gene Knockdown Techniques , HT29 Cells , Hepatic Stellate Cells/metabolism , Humans , Liver/cytology , Liver/metabolism , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Mice , Mice, Knockout , Mice, SCID , Myofibroblasts/metabolism , Nitrobenzenes , Phosphorylation , Primary Cell Culture , Pyrazoles/pharmacology , Pyrazolones , RNA, Small Interfering/antagonists & inhibitors , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , rhoA GTP-Binding Protein/metabolism
20.
Nucleic Acids Res ; 45(8): 4564-4576, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28199696

ABSTRACT

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.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Hepatocellular/genetics , Chromosomal Proteins, Non-Histone/deficiency , DNA Topoisomerases, Type I/genetics , Liver Neoplasms/genetics , Progeria/genetics , Aneuploidy , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Chromosomal Proteins, Non-Histone/genetics , DNA Adducts/genetics , DNA Adducts/metabolism , DNA Topoisomerases, Type I/metabolism , DNA-Binding Proteins , Disease Models, Animal , Female , Gene Expression , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Knockout , Progeria/metabolism , Progeria/pathology , Proteolysis , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL