Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cell Cycle ; 19(18): 2260-2269, 2020 09.
Article in English | MEDLINE | ID: mdl-32787501

ABSTRACT

The E2F1 transcription factor and RB tumor suppressor are best known for their roles in regulating the expression of genes important for cell cycle progression but, they also have transcription-independent functions that facilitate DNA repair at sites of damage. Depending on the type of DNA damage, E2F1 can recruit either the GCN5 or p300/CBP histone acetyltransferases to deposit different histone acetylation marks in flanking chromatin. At DNA double-strand breaks, E2F1 also recruits RB and the BRG1 ATPase to remodel chromatin and promote loading of the MRE11-RAD50-NBS1 complex. Knock-in mouse models demonstrate important roles for E2F1 post-translational modifications in regulating DNA repair and physiological responses to DNA damage. This review highlights how E2F1 moonlights in DNA repair, thus revealing E2F1 as a versatile protein that recruits many of the same chromatin-modifying enzymes to sites of DNA damage to promote repair that it recruits to gene promoters to regulate transcription.


Subject(s)
DNA Damage , DNA Repair , E2F1 Transcription Factor/metabolism , Retinoblastoma Protein/metabolism , Acetylation , Animals , Chromatin Assembly and Disassembly , E2F1 Transcription Factor/genetics , Histones/metabolism , Humans , Retinoblastoma Protein/genetics , p300-CBP Transcription Factors/metabolism
2.
Nat Rev Cancer ; 20(5): 274-284, 2020 05.
Article in English | MEDLINE | ID: mdl-32235902

ABSTRACT

Tumours reprogram host physiology, metabolism and immune responses during cancer progression. The release of soluble factors, exosomes and metabolites from tumours leads to systemic changes in distant organs, where cancer cells metastasize and grow. These tumour-derived circulating factors also profoundly impact tissues that are rarely inhabited by metastatic cancer cells such as skeletal muscle and adipose tissue. In fact, the majority of patients with metastatic cancer develop a debilitating muscle-wasting syndrome, known as cachexia, that is associated with decreased tolerance to antineoplastic therapy, poor prognosis and accelerated death, with no approved treatments. In this Perspective, we discuss the development of cachexia in the context of metastatic progression. We briefly discuss how circulating factors either directly or indirectly promote cachexia development and examine how signals from the metastatic process can trigger and amplify this process. Finally, we highlight promising therapeutic opportunities for targeting cachexia in the context of metastatic cancers.


Subject(s)
Cachexia/etiology , Neoplasms/complications , Animals , Cachexia/pathology , Disease Progression , Humans , Neoplasm Metastasis , Neoplasms/pathology
3.
Article in English | MEDLINE | ID: mdl-31615873

ABSTRACT

Metastasis arises when cancer cells disseminate from their site of origin and invade distant organs. While cancer cells rarely colonize muscle, they often induce a debilitating muscle-wasting condition known as cachexia that compromises feeding, breathing, and cardiac function in metastatic cancer patients. In fact, nearly 80% of metastatic cancer patients experience a spectrum of muscle-wasting states, which deteriorates the quality of life and overall survival of cancer patients. Muscle wasting in cancer results from increased muscle catabolism induced by circulating tumor factors and a systemic metabolic dysfunction. In addition, muscle loss can be exacerbated by the exposure to antineoplastic therapies and the process of aging. With no approved therapies to alleviate cachexia, muscle health, therefore, becomes a key determinant of prognosis, treatment response, and survival in metastatic cancer patients. This review will discuss the current understanding of cancer-associated cachexia and highlight promising therapeutic strategies to treat muscle wasting in the context of metastatic cancers.


Subject(s)
Cachexia/physiopathology , Muscular Atrophy/physiopathology , Neoplasm Metastasis/pathology , Aging , Antineoplastic Agents/adverse effects , Cachexia/etiology , Humans , Muscle, Skeletal/metabolism , Muscular Atrophy/etiology , Neoplasm Metastasis/drug therapy , Quality of Life
4.
Nat Commun ; 10(1): 4951, 2019 10 30.
Article in English | MEDLINE | ID: mdl-31666529

ABSTRACT

E2F1 and retinoblastoma (RB) tumor-suppressor protein not only regulate the periodic expression of genes important for cell proliferation, but also localize to DNA double-strand breaks (DSBs) to promote repair. E2F1 is acetylated in response to DNA damage but the role this plays in DNA repair is unknown. Here we demonstrate that E2F1 acetylation creates a binding motif for the bromodomains of the p300/KAT3B and CBP/KAT3A acetyltransferases and that this interaction is required for the recruitment of p300 and CBP to DSBs and the induction of histone acetylation at sites of damage. A knock-in mutation that blocks E2F1 acetylation abolishes the recruitment of p300 and CBP to DSBs and also the accumulation of other chromatin modifying activities and repair factors, including Tip60, BRG1 and NBS1, and renders mice hypersensitive to ionizing radiation (IR). These findings reveal an important role for E2F1 acetylation in orchestrating the remodeling of chromatin structure at DSBs to facilitate repair.


Subject(s)
CREB-Binding Protein/metabolism , DNA Breaks, Double-Stranded , E1A-Associated p300 Protein/metabolism , E2F1 Transcription Factor/metabolism , Histones/metabolism , Acetylation , Animals , Cell Cycle Proteins/metabolism , DNA Helicases/metabolism , DNA Repair/genetics , DNA-Binding Proteins/metabolism , E2F1 Transcription Factor/genetics , Gene Knock-In Techniques , Histone Acetyltransferases , Lysine Acetyltransferase 5/metabolism , Mice , Nuclear Proteins/metabolism , Protein Interaction Domains and Motifs , Radiation, Ionizing , Trans-Activators/metabolism , Transcription Factors/metabolism , p300-CBP Transcription Factors/metabolism
5.
Nat Med ; 24(6): 770-781, 2018 06.
Article in English | MEDLINE | ID: mdl-29875463

ABSTRACT

Patients with metastatic cancer experience a severe loss of skeletal muscle mass and function known as cachexia. Cachexia is associated with poor prognosis and accelerated death in patients with cancer, yet its underlying mechanisms remain poorly understood. Here, we identify the metal-ion transporter ZRT- and IRT-like protein 14 (ZIP14) as a critical mediator of cancer-induced cachexia. ZIP14 is upregulated in cachectic muscles of mice and in patients with metastatic cancer and can be induced by TNF-α and TGF-ß cytokines. Strikingly, germline ablation or muscle-specific depletion of Zip14 markedly reduces muscle atrophy in metastatic cancer models. We find that ZIP14-mediated zinc uptake in muscle progenitor cells represses the expression of MyoD and Mef2c and blocks muscle-cell differentiation. Importantly, ZIP14-mediated zinc accumulation in differentiated muscle cells induces myosin heavy chain loss. These results highlight a previously unrecognized role for altered zinc homeostasis in metastatic cancer-induced muscle wasting and implicate ZIP14 as a therapeutic target for its treatment.


Subject(s)
Cachexia/metabolism , Cachexia/pathology , Cation Transport Proteins/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Neoplasms/metabolism , Neoplasms/pathology , Up-Regulation , Animals , Cell Differentiation , Cell Line , Cytokines/metabolism , Disease Models, Animal , Humans , Mice, Inbred C57BL , Myosin Heavy Chains/metabolism , Neoplasm Metastasis , Transforming Growth Factor beta/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Zinc/metabolism
6.
Genes Dev ; 30(22): 2500-2512, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27940962

ABSTRACT

The retinoblastoma (RB) tumor suppressor is recognized as a master regulator that controls entry into the S phase of the cell cycle. Its loss leads to uncontrolled cell proliferation and is a hallmark of cancer. RB works by binding to members of the E2F family of transcription factors and recruiting chromatin modifiers to the promoters of E2F target genes. Here we show that RB also localizes to DNA double-strand breaks (DSBs) dependent on E2F1 and ATM kinase activity and promotes DSB repair through homologous recombination (HR), and its loss results in genome instability. RB is necessary for the recruitment of the BRG1 ATPase to DSBs, which stimulates DNA end resection and HR. A knock-in mutation of the ATM phosphorylation site on E2F1 (S29A) prevents the interaction between E2F1 and TopBP1 and recruitment of RB, E2F1, and BRG1 to DSBs. This knock-in mutation also impairs DNA repair, increases genomic instability, and renders mice hypersensitive to IR. Importantly, depletion of RB in osteosarcoma and breast cancer cell lines results in sensitivity to DNA-damaging drugs, which is further exacerbated by poly-ADP ribose polymerase (PARP) inhibitors. We uncovered a novel, nontranscriptional function for RB in HR, which could contribute to genome instability associated with RB loss.


Subject(s)
DNA Breaks, Double-Stranded , DNA Helicases/metabolism , Homologous Recombination/genetics , Nuclear Proteins/metabolism , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Cell Line, Tumor , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Helicases/genetics , DNA Repair/genetics , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Gamma Rays , Gene Knock-In Techniques , Genomic Instability/genetics , Humans , Male , Mice , Mutagens/pharmacology , Mutation , Nuclear Proteins/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein Transport/genetics , Retinoblastoma Protein/genetics , Transcription Factors/genetics , Whole-Body Irradiation/mortality
7.
Cancer Res ; 72(1): 13-7, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22180494

ABSTRACT

E2F is a family of transcription factors that regulate the expression of genes involved in a wide range of cellular processes, including cell-cycle progression, DNA replication, DNA repair, differentiation, and apoptosis. E2F1, the founding member of the family, undergoes posttranslational modifications in response to DNA damage, resulting in E2F1 stabilization. In some cases, E2F1 is important for DNA damage-induced apoptosis through the transcriptional activation of p73 and perhaps other proapoptotic target genes. However, in other contexts, E2F1 can stimulate DNA repair and promote survival in response to DNA damage. The E2F1 protein accumulates at sites of both DNA double-strand breaks and UV radiation-induced damage, indicating that E2F1 has a nontranscriptional function at sites of damage. This review summarizes recent progress made in understanding the role of E2F1 in the DNA damage response, including transcription-independent activities that facilitate DNA repair in the context of chromatin.


Subject(s)
DNA Damage , E2F1 Transcription Factor/physiology , Transcription, Genetic , DNA Repair , Humans , Protein Processing, Post-Translational
8.
Cell Cycle ; 10(8): 1287-94, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21512314

ABSTRACT

The E2F1 transcription factor is post-translationally modified and stabilized in response to various forms of DNA damage to regulate the expression of cell cycle and pro-apoptotic genes. E2F1 also forms foci at DNA double-strand breaks (DSBs) but the function of E2F1 at sites of damage is unknown. Here we demonstrate that the absence of E2F1 leads to spontaneous DNA breaks and impaired recovery following exposure to ionizing radiation. E2F1 deficiency results in defective NBS1 phosphorylation and foci formation in response to DSBs but does not affect NBS1 expression levels. Moreover, an increased association between NBS1 and E2F1 is observed in response to DNA damage, suggesting that E2F1 may promote NBS1 foci formation through a direct or indirect interaction at sites of DNA breaks. E2F1 deficiency also impairs RPA and Rad51 foci formation indicating that E2F1 is important for DNA end resection and the formation of single-stranded DNA at DSBs. These findings establish new roles for E2F1 in the DNA damage response, which may directly contribute to DNA repair and genome maintenance.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Repair , DNA/metabolism , E2F1 Transcription Factor , Nuclear Proteins/metabolism , Animals , Cell Culture Techniques , Cell Cycle , Cell Cycle Proteins/genetics , Cell Line, Tumor , Comet Assay , DNA Breaks, Double-Stranded/radiation effects , DNA Damage/radiation effects , DNA Repair/radiation effects , DNA-Binding Proteins , E2F1 Transcription Factor/deficiency , E2F1 Transcription Factor/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Gene Silencing , Humans , Mice , Nuclear Proteins/genetics , Phosphorylation/radiation effects , RNA, Small Interfering/metabolism , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Radiation, Ionizing , Replication Protein A/genetics , Replication Protein A/metabolism
9.
J Biol Chem ; 285(25): 19308-15, 2010 Jun 18.
Article in English | MEDLINE | ID: mdl-20413589

ABSTRACT

The E2F1 transcription factor is a well known regulator of cell proliferation and apoptosis, but its role in the DNA damage response is less clear. Using a local UV irradiation technique and immunofluorescence staining, E2F1 is shown to accumulate at sites of DNA damage. Localization of E2F1 to UV-damaged DNA requires the ATM and Rad3-related (ATR) kinase and serine 31 of E2F1 but not an intact DNA binding domain. E2F1 deficiency does not appear to affect the expression of nucleotide excision repair (NER) factors, such as XPC and XPA. However, E2F1 depletion does impair the recruitment of NER factors to sites of damage and reduces the efficiency of DNA repair. E2F1 mutants unable to bind DNA or activate transcription retain the ability to stimulate NER. These findings demonstrate that E2F1 has a direct, non-transcriptional role in DNA repair involving increased recruitment of NER factors to sites of damage.


Subject(s)
DNA Damage , E2F1 Transcription Factor/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Chromatin/chemistry , DNA Repair , E2F1 Transcription Factor/chemistry , Fibroblasts/metabolism , HeLa Cells , Humans , Microscopy, Fluorescence/methods , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Retinoblastoma Protein/metabolism , Serine/chemistry , Transcriptional Activation , Ultraviolet Rays
10.
FEMS Microbiol Lett ; 261(1): 12-8, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16842352

ABSTRACT

DNA replication takes place at five different stages during the life cycle of Plasmodium falciparum including the human and mosquito hosts. DNA replication initiation, the rate-determining step is poorly understood in Plasmodium. Here we show that PfMCM4 and PfORC1, two members of prereplication initiation complex are expressed specifically in the nucleus during the trophozoite and schizont stages of the asexual parasitic life cycle where maximum amount of DNA replication takes place. Further, we show that these proteins are also expressed in gametocytes, where DNA replication also occurs. These results expand our knowledge on these proteins and resolves discrepancies arising from previous studies with respect to the expression pattern of replication initiation proteins during the parasite's life cycle.


Subject(s)
DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Erythrocytes/parasitology , Origin Recognition Complex/metabolism , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Trans-Activators/metabolism , Animals , Culicidae/parasitology , DNA Replication , Humans , Origin Recognition Complex/analysis , Origin Recognition Complex/physiology , Plasmodium falciparum/genetics , Protozoan Proteins/analysis , Protozoan Proteins/physiology , Reproduction , Reproduction, Asexual
11.
Biochem Biophys Res Commun ; 337(3): 955-66, 2005 Nov 25.
Article in English | MEDLINE | ID: mdl-16216221

ABSTRACT

In eukaryotes, the origin recognition complex (ORC) is essential for the initiation of DNA replication. The largest subunit of this complex (ORC1) has a regulatory role in origin activation. Here we report the cloning and functional characterization of Plasmodium falciparum ORC1 homolog. Using immunofluorescence and immunoelectron microscopy, we show here that PfORC1 is expressed in the nucleus during the late trophozoite and schizont stages where maximum amount of DNA replication takes place. Homology modelling of the carboxy terminal region of PfORC1 (781-1033) using Saccharomyces pombe Cdc6/Cdc18 homolog as a template reveals the presence of a similar AAA+ type nucleotide-binding fold. This region shows ATPase activity in vitro that is important for the origin activity. To our knowledge, this is the first evidence of an individual ORC subunit that shows ATPase activity. These observations strongly suggest that PfORC1 might be involved in DNA replication initiation during the blood stage of the parasitic life cycle.


Subject(s)
Erythrocytes/parasitology , Gene Expression Regulation, Developmental/physiology , Models, Molecular , Origin Recognition Complex/chemistry , Origin Recognition Complex/metabolism , Plasmodium falciparum/metabolism , Protein Engineering/methods , Amino Acid Sequence , Animals , Computer Simulation , Humans , Malaria, Falciparum/parasitology , Models, Chemical , Molecular Sequence Data , Plasmodium falciparum/pathogenicity , Protein Subunits , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...