Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
2.
EMBO J ; 39(11): e101573, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32323871

ABSTRACT

High expression of 2',5'-oligoadenylate synthetase 1 (OAS1), which adds AMP residues in 2',5' linkage to a variety of substrates, is observed in many cancers as a part of the interferon-related DNA damage resistance signature (IRDS). Poly(ADP-ribose) (PAR) is rapidly synthesized from NAD+ at sites of DNA damage to facilitate repair, but excessive PAR synthesis due to extensive DNA damage results in cell death by energy depletion and/or activation of PAR-dependent programmed cell death pathways. We find that OAS1 adds AMP residues in 2',5' linkage to PAR, inhibiting its synthesis in vitro and reducing its accumulation in cells. Increased OAS1 expression substantially improves cell viability following DNA-damaging treatments that stimulate PAR synthesis during DNA repair. We conclude that high expression of OAS1 in cancer cells promotes their ability to survive DNA damage by attenuating PAR synthesis and thus preventing cell death.


Subject(s)
2',5'-Oligoadenylate Synthetase/biosynthesis , DNA Damage , Gene Expression Regulation, Enzymologic , Poly ADP Ribosylation , 2',5'-Oligoadenylate Synthetase/genetics , Adenosine Monophosphate/genetics , Adenosine Monophosphate/metabolism , Cell Death , Cell Line, Transformed , Humans
3.
Cell Rep ; 21(8): 2223-2235, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-29166612

ABSTRACT

Naturally stalled replication forks are considered to cause structurally abnormal chromosomes in tumor cells. However, underlying mechanisms remain speculative, as capturing naturally stalled forks has been a challenge. Here, we captured naturally stalled forks in tumor cells and delineated molecular processes underlying the structural evolution of circular mini-chromosomes (double-minute chromosomes; DMs). Replication forks stalled on the DM by the co-directional collision with the transcription machinery for long non-coding RNA. RPA, BRCA2, and DNA polymerase eta (Polη) were recruited to the stalled forks. The recruitment of Polη was critical for replication to continue, as Polη knockdown resulted in DM loss. Rescued stalled forks were error-prone and switched replication templates repeatedly to create complex fusions of multiple short genomic segments. In mice, such complex fusions circularized the genomic region surrounding MYC to create a DM during tumorigenesis. Our results define a molecular path that guides stalled replication forks to complex chromosomal rearrangements.


Subject(s)
BRCA2 Protein/metabolism , Chromosome Aberrations , DNA Repair/genetics , DNA Replication/genetics , RNA, Long Noncoding/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Acid Anhydride Hydrolases , Animals , DNA-Binding Proteins/metabolism , Mice , RNA, Long Noncoding/genetics , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism
4.
Nucleic Acids Res ; 43(5): 2678-90, 2015 Mar 11.
Article in English | MEDLINE | ID: mdl-25672394

ABSTRACT

Gene amplification is a phenotype-causing form of chromosome instability and is initiated by DNA double-strand breaks (DSBs). Cells with mutant p53 lose G1/S checkpoint and are permissive to gene amplification. In this study we show that mammalian cells become proficient for spontaneous gene amplification when the function of the DSB repair protein complex MRN (Mre11/Rad50/Nbs1) is impaired. Cells with impaired MRN complex experienced severe replication stress and gained substrates for gene amplification during replication, as evidenced by the increase of replication-associated single-stranded breaks that were converted to DSBs most likely through replication fork reversal. Impaired MRN complex directly compromised ATM/ATR-mediated checkpoints and allowed cells to progress through cell cycle in the presence of DSBs. Such compromised intra-S phase checkpoints promoted gene amplification independently from mutant p53. Finally, cells adapted to endogenous replication stress by globally suppressing genes for DNA replication and cell cycle progression. Our results indicate that the MRN complex suppresses gene amplification by stabilizing replication forks and by securing DNA damage response to replication-associated DSBs.


Subject(s)
DNA Repair , DNA Replication/genetics , Gene Amplification , S Phase Cell Cycle Checkpoints/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Blotting, Western , CHO Cells , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cricetinae , Cricetulus , DNA Breaks, Double-Stranded , DNA Breaks, Single-Stranded , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Flow Cytometry , Gene Expression Profiling , HEK293 Cells , Humans , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
5.
Aging (Albany NY) ; 6(8): 675-89, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25239872

ABSTRACT

The mTOR signaling pathway modulates metabolic processes with respect to nutrient availability and other growth-related cues. According to the existing paradigm, mTOR complex 1 (mTORC1) activityin vivo is induced by food and gradually decreases during fasting. We found that mTORC1 activity is controlled by an internal clock mechanism different from the known light-entrainable circadian clock. We observed 24-hr rhythms in phosphorylation of mTORC1 downstream targets, which were entrained by food, persisted during fasting and could be uncoupled from oscillating expression of the canonical circadian clock genes. Furthermore, these rhythms were present in tissues of mice with disrupted light-entrainable circadian clock. We propose tissue-specific rhythms in the expression of tor and its negative regulator deptor as the molecular mechanism of the mTORC1 activity oscillation. Our data demonstrate the existence of at least two independent molecular circadian clocks: one providing metabolic adaptation to periodic light/darkness and the other - to feeding.


Subject(s)
Biological Clocks/physiology , Feeding Behavior/physiology , Signal Transduction/physiology , TOR Serine-Threonine Kinases/metabolism , Animals , Liver/metabolism , Mice , Phosphorylation/physiology
7.
Aging (Albany NY) ; 6(1): 48-57, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24481314

ABSTRACT

The circadian clock, an internal time-keeping system, has been linked with control of aging, but molecular mechanisms of regulation are not known. BMAL1 is a transcriptional factor and core component of the circadian clock; BMAL1 deficiency is associated with premature aging and reduced lifespan. Here we report that activity of mammalian Target of Rapamycin Complex 1 (mTORC1) is increased upon BMAL1 deficiency both in vivo and in cell culture. Increased mTOR signaling is associated with accelerated aging; in accordance with that, treatment with the mTORC1 inhibitor rapamycin increased lifespan of Bmal1-/- mice by 50%. Our data suggest that BMAL1 is a negative regulator of mTORC1 signaling. We propose that the circadian clock controls the activity of the mTOR pathway through BMAL1-dependent mechanisms and this regulation is important for control of aging and metabolism.


Subject(s)
ARNTL Transcription Factors/metabolism , Aging/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , ARNTL Transcription Factors/deficiency , ARNTL Transcription Factors/genetics , Aging/genetics , Animals , Cell Proliferation , Cells, Cultured , Circadian Rhythm , Enzyme Inhibitors/pharmacology , Fibroblasts/enzymology , Genotype , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lung/enzymology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Multiprotein Complexes/metabolism , Phenotype , Phosphorylation , Signal Transduction/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Time Factors
8.
Nucleic Acids Res ; 41(21): 9732-40, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23975201

ABSTRACT

Breakage-fusion-bridge (BFB) cycle is a series of chromosome breaks and duplications that could lead to the increased copy number of a genomic segment (gene amplification). A critical step of BFB cycles leading to gene amplification is a palindromic fusion of sister chromatids following the rupture of a dicentric chromosome during mitosis. It is currently unknown how sister chromatid fusion is produced from a mitotic break. To delineate the process, we took an integrated genomic, cytogenetic and molecular approach for the recurrent MCL1 amplicon at chromosome 1 in human tumor cells. A newly developed next-generation sequencing-based approach identified a cluster of palindromic fusions within the amplicon at ∼50-kb intervals, indicating a series of breaks and fusions by BFB cycles. The physical location of the amplicon (at the end of a broken chromosome) further indicated BFB cycles as underlying processes. Three palindromic fusions were mediated by the homologies between two nearby inverted Alu repeats, whereas the other two fusions exhibited microhomology-mediated events. Such breakpoint sequences indicate that homology-mediated fold-back capping of broken ends followed by DNA replication is an underlying mechanism of sister chromatid fusion. Our results elucidate nucleotide-level events during BFB cycles and end processing for naturally occurring mitotic breaks.


Subject(s)
Chromatids/genetics , Chromosome Breakage , Cell Line , Cell Line, Tumor , Chromosome Breakpoints , Gene Amplification , Genomics , Humans , Inverted Repeat Sequences
9.
J Immunol ; 190(2): 832-41, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23241877

ABSTRACT

During growth in the host, tumor cells are subjected to the stresses of innate and adaptive immunity (immunoediting), which provoke epigenetic changes in the tumor and increase tumor resistance to these immune responses. Our recent studies in methylcholanthrene-induced fibrosarcomas have indicated the appearance and rapid growth of tumor variants deficient in producing the T cell chemoattractant chemokine CXCL9/Mig, an important component of antitumor immunity. In the current report, we demonstrate that highly tumorigenic Mig-deficient tumor variants arise in both cutaneous fibrosarcoma and melanoma as a result of immune stress imposed by IFN-γ and T cells. The consequence of the loss of tumor-derived Mig expression is the increased resistance of Mig-deficient tumors to T cell-mediated immunity, which promotes the accelerated growth of these tumor variants. Remarkably, the ability of Mig-deficient tumor cells to express another CXCR3 ligand, CXCL10/IFN-γ-inducible protein, does not compensate for the absent antitumor functions of Mig, suggesting a nonredundant role for this chemokine in the suppression of tumor growth. To our knowledge, these studies report for the first time that IFN-γ-mediated stress leads to the loss of specific chemokine expression by tumor cells, which in turn promotes tumor growth and evasion of the immune response.


Subject(s)
Chemokine CXCL9/metabolism , Interferon-gamma/pharmacology , Skin Neoplasms/immunology , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Chemokine CXCL9/deficiency , Chemokine CXCL9/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Interferon-gamma/genetics , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Mice , Mice, Knockout , Skin Neoplasms/genetics
10.
Breast Cancer Res ; 14(6): R150, 2012 Nov 26.
Article in English | MEDLINE | ID: mdl-23181561

ABSTRACT

INTRODUCTION: Segmental duplications (low-copy repeats) are the recently duplicated genomic segments in the human genome that display nearly identical (> 90%) sequences and account for about 5% of euchromatic regions. In germline, duplicated segments mediate nonallelic homologous recombination and thus cause both non-disease-causing copy-number variants and genomic disorders. To what extent duplicated segments play a role in somatic DNA rearrangements in cancer remains elusive. Duplicated segments often cluster and form genomic blocks enriched with both direct and inverted repeats (complex genomic regions). Such complex regions could be fragile and play a mechanistic role in the amplification of the ERBB2 gene in breast tumors, because repeated sequences are known to initiate gene amplification in model systems. METHODS: We conducted polymerase chain reaction (PCR)-based assays for primary breast tumors and analyzed publically available array-comparative genomic hybridization data to map a common copy-number breakpoint in ERBB2-amplified primary breast tumors. We further used molecular, bioinformatics, and population-genetics approaches to define duplication contents, structural variants, and haplotypes within the common breakpoint. RESULTS: We found a large (> 300-kb) block of duplicated segments that was colocalized with a common-copy number breakpoint for ERBB2 amplification. The breakpoint that potentially initiated ERBB2 amplification localized in a region 1.5 megabases (Mb) on the telomeric side of ERBB2. The region is very complex, with extensive duplications of KRTAP genes, structural variants, and, as a result, a paucity of single-nucleotide polymorphism (SNP) markers. Duplicated segments are varied in size and degree of sequence homology, indicating that duplications have occurred recurrently during genome evolution. CONCLUSIONS: Amplification of the ERBB2 gene in breast tumors is potentially initiated by a complex region that has unusual genomic features and thus requires rigorous, labor-intensive investigation. The haplotypes we provide could be useful to identify the potential association between the complex region and ERBB2 amplification.


Subject(s)
Breast Neoplasms/genetics , Chromosome Breakpoints , DNA Copy Number Variations , Receptor, ErbB-2/genetics , Segmental Duplications, Genomic/genetics , Base Sequence , Chromosomes, Human, Pair 17/genetics , Comparative Genomic Hybridization , Female , Gene Amplification/genetics , Gene Dosage , Genome, Human , Haplotypes/genetics , Humans , Keratins, Hair-Specific/genetics , Polymorphism, Single Nucleotide , Sequence Deletion/genetics
11.
Nat Rev Neurosci ; 13(5): 325-35, 2012 Mar 07.
Article in English | MEDLINE | ID: mdl-22395806

ABSTRACT

Ageing leads to a functional deterioration of many brain systems, including the circadian clock--an internal time-keeping system that generates ∼24-hour rhythms in physiology and behaviour. Numerous clinical studies have established a direct correlation between abnormal circadian clock functions and the severity of neurodegenerative and sleep disorders. Latest data from experiments in model organisms, gene expression studies and clinical trials imply that dysfunctions of the circadian clock contribute to ageing and age-associated pathologies, thereby suggesting a functional link between the circadian clock and age-associated decline of brain functions. Potential molecular mechanisms underlying this link include the circadian control of physiological processes such as brain metabolism, reactive oxygen species homeostasis, hormone secretion, autophagy and stem cell proliferation.


Subject(s)
Aging/pathology , Brain/pathology , Circadian Clocks/physiology , Circadian Rhythm/physiology , Aging/physiology , Animals , Brain/physiology , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Humans
12.
Cell Cycle ; 10(23): 4162-9, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22101268

ABSTRACT

Deficiency of the circadian clock transcriptional factor BMAL1 results in the development of premature aging in mice. In agreement with the accelerated aging phenotype, we observed an increase in the number of senescent cells in different tissues (lungs, liver and spleen) of Bmal1(-/-) mice, which suggests the important role of BMAL1 in the control of senescence in vivo. However, no difference in the rate of proliferation and senescence between primary fibroblasts isolated from wild-type and Bmal1(-/-) mice has been detected, suggesting that BMAL1 does not play a significant role in replicative senescence in vitro. BMAL1 deficient fibroblasts had an increased sensitivity to hydrogen peroxide treatment, and reduced sensitivity to DNA damaging anticancer drugs etoposide and daunorubicin. Increased sensitivity of Bmal1(-/-) cells to oxidative stress was p53 independent and correlated with the disrupted regulation of reactive oxygen species (ROS) homeostasis in BMAL1 deficient cells: indeed, circadian oscillations of ROS level can be induced in wild-type but not in Bmal1(-/-) cells. We propose that BMAL1 is important for the regulation of oxidative stress and DNA damage responses, while deregulation of these processes upon BMAL1 deficiency leads to development of stress induced senescence in vivo.


Subject(s)
ARNTL Transcription Factors/metabolism , Cellular Senescence , Circadian Clocks , DNA Damage , Oxidative Stress , ARNTL Transcription Factors/genetics , Animals , Antineoplastic Agents/pharmacology , Cell Death , Culture Media, Serum-Free/metabolism , Daunorubicin/pharmacology , Drug Screening Assays, Antitumor , Etoposide/pharmacology , Fibroblasts/drug effects , Gene Expression Regulation, Neoplastic , Homeostasis , Hydrogen Peroxide/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Primary Cell Culture , Reactive Oxygen Species , Time Factors
13.
Aging (Albany NY) ; 2(5): 285-97, 2010 May.
Article in English | MEDLINE | ID: mdl-20519775

ABSTRACT

Deficiency of the transcription factor BMAL1, a core component of the circadian clock, results in an accelerated aging phenotype in mice. The circadian clock regulates many physiological processes and was recently implicated in control of brain-based activities, such as memory formation and the regulation of emotions. Aging is accompanied by the decline in brain physiology, particularly decline in the response and adaptation to novelty. We investigated the role of the circadian clock in exploratory behavior and habituation to novelty using the open field paradigm. We found that mice with a deficiency of the circadian transcription factor BMAL1 display hyperactivity in novel environments and impaired intra- and intersession habituation, indicative of defects in short- and long-term memory formation. In contrast, mice double-deficient for the circadian proteins CRY1 and CRY2 (repressors of the BMAL1-mediated transcription) demonstrate reduced activity and accelerated habituation when compared to wild type mice. Mice with mutation in theClock gene (encoding the BMAL1 transcription partner) show normal locomotion, but increased rearing activity and impaired intersession habituation. BMAL1 is highly expressed in the neurons of the hippocampus - a brain region associated with spatial memory formation; BMAL1 deficiency disrupts circadian oscillation in gene expression and reactive oxygen species homeostasis in the brain, which may be among the possible mechanisms involved. Thus, we suggest that the BMAL1:CLOCK activity is critical for the proper exploratory and habituation behavior, and that the circadian clock prepares organism for a new round of everyday activities through optimization of behavioral learning.


Subject(s)
ARNTL Transcription Factors/genetics , CLOCK Proteins/genetics , Cryptochromes/genetics , Exploratory Behavior/physiology , Memory/physiology , ARNTL Transcription Factors/metabolism , Aging/physiology , Animals , Biological Clocks/genetics , CLOCK Proteins/metabolism , Cerebral Cortex/metabolism , Circadian Rhythm/genetics , Cryptochromes/metabolism , Habituation, Psychophysiologic/genetics , Hippocampus/metabolism , Hyperkinesis/genetics , Mice , Mice, Knockout , Motor Activity/genetics , Reactive Oxygen Species/metabolism
14.
Aging (Albany NY) ; 1(12): 979-87, 2009 Dec 30.
Article in English | MEDLINE | ID: mdl-20157581

ABSTRACT

Deficiency of the circadian clock protein BMAL1 leads to premature aging and increased levels of reactivate oxygen species in several tissues of mice. In order to investigate the role of oxidative stress in accelerated aging and development of age-related pathologies, we continuously administered the antioxidant N-acetyl-L-cysteine toBmal1-deficient mice through their entire lifespan by supplementing drinking water. We found that the life long treatment with antioxidant significantly increased average and maximal lifespan and reduced the rate of age-dependent weight loss and development of cataracts. At the same time, it had no effect on time of onset and severity of other age-related pathologies characteristic of Bmal1-/- mice, such as joint ossification, reduced hair regrowth and sarcopenia. We conclude that chronic oxidative stress affects longevity and contributes to the development of at least some age-associated pathology, although ROS-independent mechanisms may also play a role. Our bioinformatics analysis identified the presence of a conservative E box element in the promoter regions of several genes encoding major antioxidant enzymes. We speculate that BMAL1 controls antioxidant defense by regulating the expression of major antioxidant enzymes.


Subject(s)
ARNTL Transcription Factors/physiology , Acetylcysteine/pharmacology , Aging, Premature/drug therapy , Antioxidants/pharmacology , Longevity/drug effects , ARNTL Transcription Factors/genetics , Aging, Premature/genetics , Animals , Arthritis/genetics , Arthritis/prevention & control , Body Weight/drug effects , Catalase/genetics , Cataract/genetics , Cataract/prevention & control , E-Box Elements/physiology , Glutathione Peroxidase/genetics , Humans , Longevity/genetics , Macaca mulatta , Male , Mice , Ossification, Heterotopic/genetics , Ossification, Heterotopic/prevention & control , Oxidative Stress/drug effects , Pan troglodytes , Peroxiredoxins/genetics , Rats , Reactive Oxygen Species/metabolism , Superoxide Dismutase/genetics
15.
Cell Cycle ; 7(9): 1197-204, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18418054

ABSTRACT

The mammalian circadian system has been implicated in the regulation of various biological processes including those involved in genotoxic stress responses and tumor suppression. Here we report that mice with the functional deficiency in circadian transcription factor CLOCK (Clock/Clock mutant mice) do not display predisposition to tumor formation both during their normal lifespan or when challenged by gamma- radiation. This phenotype is consistent with high apoptotic and low proliferation rate in lymphoid tissues of Clock mutant mice and is supported by the gene expression profiling of a number of apoptosis and cell cycle-related genes, as well as by growth inhibition of cells with CLOCK downregulation. At the same time, Clock mutant mice respond to low-dose irradiation by accelerating their aging program, and develop phenotypes that are reminiscent of those in Bmal1-deficient mice. Taken together, our results demonstrate the dichotomy in biological consequences of the disruption of the circadian clock with respect to ageing and cancer. They also highlight the existence of a complex interconnection between ageing, carcinogenesis and individual components of the circadian clock machinery.


Subject(s)
Aging/genetics , Cell Transformation, Neoplastic/genetics , Chronobiology Disorders/genetics , Circadian Rhythm/genetics , Mutation/genetics , Trans-Activators/genetics , Aging/metabolism , Aging/radiation effects , Aging, Premature/genetics , Aging, Premature/metabolism , Animals , Apoptosis/genetics , Apoptosis/radiation effects , CLOCK Proteins , Cell Proliferation/radiation effects , Cell Transformation, Neoplastic/metabolism , Chronobiology Disorders/metabolism , Chronobiology Disorders/physiopathology , Down-Regulation/genetics , Down-Regulation/radiation effects , Female , Gamma Rays/adverse effects , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/radiation effects , Genetic Predisposition to Disease/genetics , Immunity, Innate/genetics , Immunity, Innate/radiation effects , Lymphoid Tissue/metabolism , Lymphoid Tissue/radiation effects , Male , Mice , Phenotype
16.
DNA Cell Biol ; 27(4): 191-8, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18067462

ABSTRACT

Flavonoid quercetin and its derivative, methylquercetin, inhibit the replication of poliovirus in several cell lines. Here, we show that replication of poliovirus is inhibited by quercetin and that the extent of this inhibition depends on the intracellular content of pirin, a quercetinase. HeLa cells contain higher content of pirin protein than normal kidney human epithelial (NKE) or 293 cells do. Poliovirus replication in HeLa cells is significantly more resistant to quercetin than its replication in NKE and 293 cells. Overexpression of pirin reduced antiviral inhibitory effect of quercetin, while siRNA-induced suppression of pirin level made poliovirus replication more sensitive to the flavonoid. The results suggest that quercetinase activity of pirin determines the resistance of poliovirus infection to quercetin.


Subject(s)
Antioxidants/pharmacology , Carrier Proteins/metabolism , Drug Resistance, Viral , Nuclear Proteins/metabolism , Poliovirus/drug effects , Poliovirus/physiology , Quercetin/pharmacology , Virus Replication/drug effects , Androstadienes/pharmacology , Cell Line , Dioxygenases/metabolism , Drug Resistance, Viral/drug effects , HeLa Cells , Humans , Protein Kinase Inhibitors/pharmacology , Wortmannin
17.
Genes Dev ; 20(14): 1868-73, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16847346

ABSTRACT

Mice deficient in the circadian transcription factor BMAL1 (brain and muscle ARNT-like protein) have impaired circadian behavior and demonstrate loss of rhythmicity in the expression of target genes. Here we report that Bmal1(-/-) mice have reduced lifespans and display various symptoms of premature aging including sarcopenia, cataracts, less subcutaneous fat, organ shrinkage, and others. The early aging phenotype correlates with increased levels of reactive oxygen species in some tissues of the Bmal1(-/- )animals. These findings, together with data on CLOCK/BMAL1-dependent control of stress responses, may provide a mechanistic explanation for the early onset of age-related pathologies in the absence of BMAL1.


Subject(s)
Aging, Premature/genetics , Aging/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Circadian Rhythm/genetics , ARNTL Transcription Factors , Adipose Tissue/growth & development , Adipose Tissue/pathology , Aging, Premature/physiopathology , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/metabolism , Body Weight , Female , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/pathology , Organ Size/genetics , Reactive Oxygen Species/metabolism
18.
Cell Cycle ; 5(8): 890-5, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16628007

ABSTRACT

Mammalian CLOCK(NPAS2), BMAL1 and CRYPTOCHROMEs are core components of the circadian oscillatory mechanism. The active CLOCK/BMAL1 or NPAS2/BMAL1 complexes regulate expression of numerous genes including two Cryptochromes. The products of these genes, CRY1 and CRY2, in turn repress CLOCK/BMAL1 transcriptional activity by an unknown mechanism. We have examined the effect of CRYPTOCHROMEs on posttranslational modifications and intracellular distribution of endogenous and ectopically expressed CLOCK(NPAS2) and BMAL1 proteins. We found that ectopic coexpression with CRY led to stabilization and nuclear accumulation of unphosphorylated forms of the proteins, which directly correlated with the inhibition of their transcriptional activity. This effect was CRY-specific, as other known repressors of CLOCK/BMAL1 and NPAS2/ BMAL1 transcriptional activity were not able to induce similar effects. CRYs had no effect on CLOCK(NPAS2)/BMAL1 complex formation or its ability to bind DNA. Altogether, these results demonstrate that CRYs regulate the functional activity of circadian transcriptional complex at the posttranslational level. Importantly, the posttranslational modifications and intracellular distribution of CLOCK and BMAL1 proteins were critically impaired in the tissues of mice with targeted disruption of both Cry genes, thus confirming the suggested role of CRY in clock function in vivo. Based on these findings we propose a modified model of the circadian transcriptional control, which implies CRY-mediated periodic rotation of transcriptionally active and inactive forms of CLOCK/BMAL1 on the promoter. This model provides mechanistic explanation for previously reported dual functional activity of CLOCK/BMAL1 and highlights the involvement of the circadian system in modulating the organism's response to various types of genotoxic stress, including chemotherapy and radiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/physiology , Flavoproteins/metabolism , Nerve Tissue Proteins/physiology , Protein Processing, Post-Translational , ARNTL Transcription Factors , Animals , Biological Clocks , Circadian Rhythm , Cryptochromes , Humans , Mice , Mice, Inbred C57BL
19.
FASEB J ; 20(3): 530-2, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16507766

ABSTRACT

The basic helix-loop-helix (bHLH) -PAS domain containing transcription factors CLOCK and BMAL1 are two major components of the circadian molecular oscillator. It is known that the CLOCK/BMAL1 complex positively regulates the activity of E-box containing promoters. Here we demonstrate that the CLOCK/BMAL1 complex can also suppress the activity of some promoters upon its interaction with CRYPTOCHROME (CRY). Such a dual function of the circadian transcriptional complex provides a mechanistic explanation for the unpredicted pattern of circadian gene expression in the tissues of Bmal1 null mice. We speculate that the switch from transcriptional activation to transcriptional repression may provide a highly efficient mechanism for circadian control of gene expression. We also show that CLOCK/BMAL1 can interfere with promoter regulation by other, non-circadian, transcription factors including N-MYC and ETS, leading to attenuation or abrogation of transcription of CLOCK/BMAL1-controlled stress-induced genes. We propose that, based upon these results, both circadian repression and activation of the transcription of different target genes are required for circadian responses to various external stimuli, including genotoxic stress induced by anticancer treatment.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Circadian Rhythm/physiology , Flavoproteins/physiology , Gene Expression Regulation/physiology , Trans-Activators/physiology , Transcriptional Activation/physiology , ARNTL Transcription Factors , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , CLOCK Proteins , Cell Cycle Proteins , Circadian Rhythm/genetics , Crosses, Genetic , Cryptochromes , Flavoproteins/genetics , Gene Expression Profiling , Gene Expression Regulation/genetics , Genes, Reporter , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Genetic , Multiprotein Complexes , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Period Circadian Proteins , Promoter Regions, Genetic , Proto-Oncogene Proteins c-ets/physiology , Proto-Oncogene Proteins c-myc/physiology , Recombinant Fusion Proteins/physiology , Stress, Physiological/genetics , Stress, Physiological/metabolism , Trans-Activators/deficiency , Trans-Activators/genetics , Transcriptional Activation/genetics
20.
Cell Cycle ; 4(10): 1403-10, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16138011

ABSTRACT

Many viruses encode anti-apoptotic proteins that have been used as valuable tools for identification and analysis of key cellular regulators of programmed cell death. Here we demonstrate that the poliovirus protein 3A, previously shown to exhibit anti-apoptotic activity, binds and inactivates LIS1, a component of the dynein/dynactin motor complex, encoded by the gene mutated in patients with type I lissencephaly ("smooth brain"), thereby causing deregulation of endoplasmatic reticilum-to-Golgi vesicular transport, resulting in rapid disappearance of short-living receptors from the plasma membrane and loss of cell sensitivity to TNF and interferon. Truncated derivatives of LIS1, acting in a dominant negative manner, cause similar effects. However, 3A, being an endoplasmic reticulum-bound protein, locks Golgi-targeted YFP in the endoplasmatic reticilum, while expression of LIS1 mutants results in a dispersed cytoplasmic localization of the reporter protein. LIS1 dysfunction caused by ectopic expressing 3A or LIS1 mutants, as well as by overexpression of wild type LIS1, leads to cell blocking at the postmitotic stage associated with inability to undergo cytokinesis. Thus, the use of poliovirus protein as a research tool allowed us to reveal the role of cellular protein LIS1 in membrane protein trafficking, maintenance of Golgi integrity, surface presentation of unstable receptors, cell sensitivity to TNF-induced apoptosis and cell cycle progression.


Subject(s)
Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Viral Core Proteins/metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase , Animals , Cell Division , Cell Line , Cell Membrane/metabolism , Gene Expression Regulation , Golgi Apparatus/metabolism , Humans , Mice , Microtubule-Associated Proteins/genetics , Mutation/genetics , Poliovirus , Protein Binding , Protein Transport , Viral Core Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...