Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 48(4): 1652-1668, 2020 02 28.
Article in English | MEDLINE | ID: mdl-31930303

ABSTRACT

The excision of mutagenic DNA adducts by the nucleotide excision repair (NER) pathway is essential for genome stability, which is key to avoiding genetic diseases, premature aging, cancer and neurologic disorders. Due to the need to process an extraordinarily high damage density embedded in the nucleosome landscape of chromatin, NER activity provides a unique functional caliper to understand how histone modifiers modulate DNA damage responses. At least three distinct lysine methyltransferases (KMTs) targeting histones have been shown to facilitate the detection of ultraviolet (UV) light-induced DNA lesions in the difficult to access DNA wrapped around histones in nucleosomes. By methylating core histones, these KMTs generate docking sites for DNA damage recognition factors before the chromatin structure is ultimately relaxed and the offending lesions are effectively excised. In view of their function in priming nucleosomes for DNA repair, mutations of genes coding for these KMTs are expected to cause the accumulation of DNA damage promoting cancer and other chronic diseases. Research on the question of how KMTs modulate DNA repair might pave the way to the development of pharmacologic agents for novel therapeutic strategies.


Subject(s)
Chromatin/genetics , DNA Damage/genetics , Histone Methyltransferases/genetics , Histones/genetics , Chromatin/radiation effects , DNA Damage/radiation effects , DNA Repair/genetics , DNA Repair/radiation effects , Genomic Instability/genetics , Genomic Instability/radiation effects , Histone Methyltransferases/chemistry , Methylation/radiation effects , Nucleosomes/genetics , Nucleosomes/radiation effects , Saccharomyces cerevisiae/genetics , Signal Transduction/radiation effects , Ultraviolet Rays
2.
Oncotarget ; 9(47): 28666-28690, 2018 Jun 19.
Article in English | MEDLINE | ID: mdl-29983888

ABSTRACT

Breast cancer is one of the most common malignancies among women which is often treated with hormone therapy and chemotherapy. Despite the improvements in detection and treatment of breast cancer, the vast majority of breast cancer patients are diagnosed with metastatic disease either at the beginning of the disease or later during treatment. Still, the molecular mechanisms causing a therapy resistant metastatic breast cancer are still elusive. In the present study we addressed the function of the transcriptional activator ZRF1 during breast cancer progression. We provide evidence that ZRF1 plays an essential role for the early metastatic events in vitro and acts like a tumor suppressor protein during the progression of breast invasive ductal carcinoma into a more advanced stage. Hence, depletion of ZRF1 results in the acquisition of metastatic behavior by facilitating the initiation of the metastatic cascade, notably for cell adhesion, migration and invasion. Furthermore absence of ZRF1 provokes endocrine resistance via misregulation of cell death and cell survival related pathways. Taken together, we have identified ZRF1 as an important regulator of breast cancer progression that holds the potential to be explored for new treatment strategies in the future.

3.
FEBS J ; 285(13): 2367-2376, 2018 07.
Article in English | MEDLINE | ID: mdl-29630766

ABSTRACT

Autophagy is a ubiquitous catabolic process, which causes cellular bulk degradation through vesicular engulfment of obsolete, damaged or harmful cytoplasmic components. While autophagy regulates cellular homeostasis during development and in youth, there is mounting evidence that autophagy becomes increasingly dysfunctional with age. Recent work in Caenorhabditis elegans even suggests that late-life dysfunctional autophagy exhibits detrimental effects that drive the ageing process. Other studies link elevated autophagy closely to increased health and longevity. This review aims to put these apparently opposing views into perspective and define our current understanding of the role of autophagy during ageing.


Subject(s)
Aging/physiology , Autophagy/physiology , Longevity/physiology , Signal Transduction , Animals , Caenorhabditis elegans/physiology , Drosophila melanogaster/physiology , Humans , Saccharomyces cerevisiae/physiology
4.
Nucleus ; 9(1): 212-215, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29482435

ABSTRACT

The integrity of the genome is maintained by specific DNA repair pathways. The main pathway removing DNA lesions induced by exposure to UV light is nucleotide excision repair (NER). The DNA damage response at chromatin is accompanied by the recruitment of DNA repair factors to the lesion site and the deposition of specific histone marks. The function of these histone marks in NER stays for the most part elusive. We have recently reported that the methyltransferase MMSET catalyzes the dimethylation of histone H4 at lysine 20 (H4K20me2) at the lesion site. The deposition of H4K20me2 at DNA damage sites elicits the recruitment of the NER factor XPA providing evidence for an H4K20me2-dependent DNA repair factor recruitment mechanism during lesion recognition in the global-genomic branch of NER. Here we discuss how H4K20me2 might impact on the chromatin conformation and the DNA damage response.


Subject(s)
DNA Repair , DNA/metabolism , Histones/metabolism , Lysine/metabolism , DNA Damage , Humans , Methyltransferases/metabolism , Nucleotides/metabolism
5.
J Cell Biol ; 217(2): 527-540, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29233865

ABSTRACT

Ultraviolet (UV) irradiation triggers the recruitment of DNA repair factors to the lesion sites and the deposition of histone marks as part of the DNA damage response. The major DNA repair pathway removing DNA lesions caused by exposure to UV light is nucleotide excision repair (NER). We have previously demonstrated that the endoribonuclease DICER facilitates chromatin decondensation during lesion recognition in the global-genomic branch of NER. Here, we report that DICER mediates the recruitment of the methyltransferase MMSET to the DNA damage site. We show that MMSET is required for efficient NER and that it catalyzes the dimethylation of histone H4 at lysine 20 (H4K20me2). H4K20me2 at DNA damage sites facilitates the recruitment of the NER factor XPA. Our work thus provides evidence for an H4K20me2-dependent mechanism of XPA recruitment during lesion recognition in the global-genomic branch of NER.


Subject(s)
DEAD-box RNA Helicases/metabolism , DNA Damage , DNA Repair , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Lysine/metabolism , Repressor Proteins/metabolism , Ribonuclease III/metabolism , Xeroderma Pigmentosum Group A Protein/metabolism , Biocatalysis , Cell Line, Tumor , HEK293 Cells , Humans , Ultraviolet Rays
7.
Genes Dev ; 31(15): 1561-1572, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28882853

ABSTRACT

Autophagy is a ubiquitous catabolic process that causes cellular bulk degradation of cytoplasmic components and is generally associated with positive effects on health and longevity. Inactivation of autophagy has been linked with detrimental effects on cells and organisms. The antagonistic pleiotropy theory postulates that some fitness-promoting genes during youth are harmful during aging. On this basis, we examined genes mediating post-reproductive longevity using an RNAi screen. From this screen, we identified 30 novel regulators of post-reproductive longevity, including pha-4 Through downstream analysis of pha-4, we identified that the inactivation of genes governing the early stages of autophagy up until the stage of vesicle nucleation, such as bec-1, strongly extend both life span and health span. Furthermore, our data demonstrate that the improvements in health and longevity are mediated through the neurons, resulting in reduced neurodegeneration and sarcopenia. We propose that autophagy switches from advantageous to harmful in the context of an age-associated dysfunction.


Subject(s)
Autophagy/physiology , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/physiology , Cytoplasm/metabolism , Longevity , Neurons/metabolism , Aging/physiology , Animals , Caenorhabditis elegans Proteins/genetics , Gene Silencing/physiology , Genetic Pleiotropy , RNA Interference/physiology , Reproduction , Signal Transduction , Trans-Activators/genetics , Trans-Activators/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
8.
Int J Mol Sci ; 18(8)2017 Aug 05.
Article in English | MEDLINE | ID: mdl-28783053

ABSTRACT

Cellular DNA is constantly challenged by damage-inducing factors derived from exogenous or endogenous sources. In order to maintain genome stability and integrity, cells have evolved a wide variety of DNA repair pathways which counteract different types of DNA lesions, also referred to as the DNA damage response (DDR). However, DNA in eukaryotes is highly organized and compacted into chromatin representing major constraints for all cellular pathways, including DNA repair pathways, which require DNA as their substrate. Therefore, the chromatin configuration surrounding the lesion site undergoes dramatic remodeling to facilitate access of DNA repair factors and subsequent removal of the DNA lesion. In this review, we focus on the question of how the cellular DNA repair pathways overcome the chromatin barrier, how the chromatin environment is rearranged to facilitate efficient DNA repair, which proteins mediate this re-organization process and, consequently, how the altered chromatin landscape is involved in the regulation of DNA damage responses.


Subject(s)
Chromatin/metabolism , DNA Damage , DNA Repair , Animals , Humans , Models, Biological , Phosphorylation , Ubiquitination
9.
Oncotarget ; 8(25): 39937-39938, 2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28537916
10.
Nucleic Acids Res ; 45(10): 5901-5912, 2017 Jun 02.
Article in English | MEDLINE | ID: mdl-28402505

ABSTRACT

Repair of damaged DNA relies on the recruitment of DNA repair factors in a well orchestrated manner. As a prerequisite, the chromatin needs to be decondensed by chromatin remodelers to allow for binding of repair factors and for DNA repair to occur. Recent studies have implicated members of the SWI/SNF and INO80 families as well as PARP1 in nucleotide excision repair (NER). In this study, we report that the endonuclease DICER is implicated in chromatin decondensation during NER. In response to UV irradiation, DICER is recruited to chromatin in a ZRF1-mediated manner. The H2A-ubiquitin binding protein ZRF1 and DICER together impact on the chromatin conformation via PARP1. Moreover, DICER-mediated chromatin decondensation is independent of its catalytic activity. Taken together, we describe a novel function of DICER at chromatin and its interaction with the ubiquitin signalling cascade during GG-NER.


Subject(s)
Caenorhabditis elegans/genetics , Chromatin/chemistry , DEAD-box RNA Helicases/genetics , DNA Repair , DNA-Binding Proteins/genetics , Oncogene Proteins/genetics , Ribonuclease III/genetics , Animals , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/radiation effects , Cell Line , Cell Line, Tumor , Chromatin/metabolism , Chromatin Assembly and Disassembly , DEAD-box RNA Helicases/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/radiation effects , HEK293 Cells , Histones/genetics , Histones/metabolism , Humans , Molecular Chaperones , Oncogene Proteins/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Osteoblasts/radiation effects , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , RNA-Binding Proteins , Ribonuclease III/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism , Ultraviolet Rays
11.
Oncotarget ; 8(19): 30870-30887, 2017 May 09.
Article in English | MEDLINE | ID: mdl-28416769

ABSTRACT

One of the major cellular DNA repair pathways is nucleotide excision repair (NER). It is the primary pathway for repair of various DNA lesions caused by exposure to ultraviolet (UV) light, such as cyclobutane pyrimidine dimers (CPDs) and 6-4 photoproducts. Although lesion-containing DNA associates with the nuclear matrix after UV irradiation it is still not understood how nuclear organization affects NER. Analyzing unscheduled DNA synthesis (UDS) indicates that NER preferentially occurs in specific nuclear areas, viz the nucleolus. Upon inducing localized damage, we observe migration of damaged DNA towards the nucleolus. Employing a LacR-based tethering system we demonstrate that H2A-ubiquitylation via the UV-RING1B complex localizes chromatin close to the nucleolus. We further show that the H2A-ubiquitin binding protein ZRF1 resides in the nucleolus, and that it anchors ubiquitylated chromatin along with XPC. Our data thus provide insight into the sub-nuclear organization of NER and reveal a novel role for histone H2A-ubiquitylation.


Subject(s)
DNA Repair , Histones/metabolism , Polycomb Repressive Complex 1/metabolism , Cell Line , Cell Nucleolus/genetics , Cell Nucleolus/metabolism , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin/genetics , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Humans , Molecular Chaperones , Oncogene Proteins/metabolism , RNA-Binding Proteins , Ubiquitination , Ultraviolet Rays
12.
Cell Cycle ; 16(2): 163-171, 2017 Jan 17.
Article in English | MEDLINE | ID: mdl-27929739

ABSTRACT

Damaged DNA is repaired by specialized repair factors that are recruited in a well-orchestrated manner to the damage site. The DNA damage response at UV inflicted DNA lesions is accompanied by posttranslational modifications of DNA repair factors and the chromatin environment sourrounding the lesion. In particular, mono- and poly-ubiquitylation events are an integral part of the DNA damage signaling. Whereas ubiquitin signaling at DNA doublestrand breaks has been subject to intensive studies comparatively little is known about the intricacies of ubiquitylation events occurring during nucleotide excision repair (NER), the major pathway to remove bulky helix lesions. Both, the global genomic (GG-NER) and the transcription-coupled (TC-NER) branches of NER are subject to ubiquitylation and deubiquitylation processes.Here we summarize our current knowledge of the ubiquitylation network that drives DNA repair in the NER pathway and we discuss the crosstalk of ubiquitin signaling with other prominent post-translational modfications that might be essential to time the DNA damage recognition step.


Subject(s)
DNA Damage , DNA Repair , Signal Transduction , Ubiquitin/metabolism , Animals , Humans , Time Factors , Ubiquitination
13.
Cell Cycle ; 15(23): 3306-3317, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27754813

ABSTRACT

In the present study we addressed the function of the transcriptional activator Zrf1 in the generation of the 3 germ layers during in vitro development. Currently, Zrf1 is rather regarded as a factor that drives the expression of neuronal genes. Here, we have employed mouse embryonic stem cells and P19 cells to understand the role of Zrf1 in the generation of mesoderm-derived tissues like adipocytes, cartilage and heart. Our data shows that Zrf1 is essential for the transcriptional activation of genes that give rise to mesoderm and in particular heart development. In both, the mESC and P19 systems, we provide evidence that Zrf1 contributes to the generation of functional cardiomyocytes. We further demonstrate that Zrf1 binds to the transcription start sites (TSSs) of heart tissue-specific genes from the first and second heart field where it drives their temporal expression during differentiation. Taken together, we have identified Zrf1 as a novel regulator of the mesodermal lineage that might facilitate spatiotemporal expression of genes.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , HSP40 Heat-Shock Proteins/metabolism , Mesoderm/cytology , Myocytes, Cardiac/cytology , Animals , DNA-Binding Proteins , Embryoid Bodies/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , HEK293 Cells , Humans , Mice , Molecular Chaperones , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Organogenesis/genetics , Phenotype , RNA-Binding Proteins , Time Factors , Transcriptional Activation/genetics
14.
Bioessays ; 38(11): 1130-1140, 2016 11.
Article in English | MEDLINE | ID: mdl-27599465

ABSTRACT

In this review, we discuss a novel on-site remodeling function that is mediated by the H2A-ubiquitin binding protein ZRF1. ZRF1 facilitates the remodeling of multiprotein complexes at chromatin and lies at the heart of signaling processes that occur at DNA damage sites and during transcriptional activation. In nucleotide excision repair ZRF1 remodels E3 ubiquitin ligase complexes at the damage site. During embryonic stem cell differentiation, it contributes to retinoic acid-mediated gene activation by altering the subunit composition of the Mediator complex. We postulate that ZRF1 operates in conjunction with cellular remodeling machines and suggest that on-site remodeling might be a hallmark of many chromatin-associated signaling pathways. We discuss yet unexplored functions of ZRF1-mediated remodeling in replication and double strand break repair. In conclusion, we postulate that on-site remodeling of multiprotein complexes is essential for the timing of chromatin signaling processes.


Subject(s)
Chromatin Assembly and Disassembly , DNA Repair , DNA-Binding Proteins/metabolism , Oncogene Proteins/metabolism , Transcriptional Activation , Animals , Humans , Molecular Chaperones , Multiprotein Complexes/metabolism , RNA-Binding Proteins
15.
Cell Cycle ; 15(11): 1479-93, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27096886

ABSTRACT

Mediator is considered an enhancer of RNA-Polymerase II dependent transcription but its function and regulation in pluripotent mouse embryonic stem cells (mESCs) remains unresolved. One means of controlling the function of Mediator is provided by the binding of the Cdk8 module (Med12, Cdk8, Ccnc and Med13) to the core Mediator. Here we report that Med12 operates together with PRC1 to silence key developmental genes in pluripotency. At the molecular level, while PRC1 represses genes it is also required to assemble ncRNA containing Med12-Mediator complexes. In the course of cellular differentiation the H2A ubiquitin binding protein Zrf1 abrogates PRC1-Med12 binding and facilitates the association of Cdk8 with Mediator. This remodeling of Mediator-associated protein complexes converts Mediator from a transcriptional repressor to a transcriptional enhancer, which then mediates ncRNA-dependent activation of Polycomb target genes. Altogether, our data reveal how the interplay of PRC1, ncRNA and Mediator complexes controls pluripotency and cellular differentiation.


Subject(s)
Mediator Complex/genetics , Mouse Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Polycomb-Group Proteins/genetics , RNA, Untranslated/genetics , Transcriptional Activation , Animals , Cell Differentiation , Cell Line , Cyclin C/genetics , Cyclin C/metabolism , Cyclin-Dependent Kinase 8/genetics , Cyclin-Dependent Kinase 8/metabolism , DNA-Binding Proteins , Gene Expression Profiling , HEK293 Cells , HSP40 Heat-Shock Proteins/genetics , HSP40 Heat-Shock Proteins/metabolism , Humans , Mediator Complex/metabolism , Mice , Molecular Chaperones , Mouse Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Polycomb-Group Proteins/metabolism , Protein Binding , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA, Untranslated/metabolism , RNA-Binding Proteins , Signal Transduction
16.
J Cell Biol ; 213(2): 185-200, 2016 04 25.
Article in English | MEDLINE | ID: mdl-27091446

ABSTRACT

Faithful DNA repair is essential to maintain genome integrity. Ultraviolet (UV) irradiation elicits both the recruitment of DNA repair factors and the deposition of histone marks such as monoubiquitylation of histone H2A at lesion sites. Here, we report how a ubiquitin E3 ligase complex specific to DNA repair is remodeled at lesion sites in the global genome nucleotide excision repair (GG-NER) pathway. Monoubiquitylation of histone H2A (H2A-ubiquitin) is catalyzed predominantly by a novel E3 ligase complex consisting of DDB2, DDB1, CUL4B, and RING1B (UV-RING1B complex) that acts early during lesion recognition. The H2A-ubiquitin binding protein ZRF1 mediates remodeling of this E3 ligase complex directly at the DNA lesion site, causing the assembly of the UV-DDB-CUL4A E3 ligase complex (DDB1-DDB2-CUL4A-RBX1). ZRF1 is an essential factor in GG-NER, and its function at damaged chromatin sites is linked to damage recognition factor XPC. Overall, the results shed light on the interplay between epigenetic and DNA repair recognition factors at DNA lesion sites.


Subject(s)
DNA Repair/physiology , DNA-Binding Proteins/physiology , Oncogene Proteins/physiology , Ubiquitin-Protein Ligases/metabolism , Cullin Proteins/metabolism , DNA Damage , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HEK293 Cells , Histones/metabolism , Humans , Molecular Chaperones , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 1/physiology , RNA-Binding Proteins , Ubiquitination , Ultraviolet Rays
17.
Cell Cycle ; 10(5): 745-50, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21311219

ABSTRACT

We have recently reported that the protein ZRF1 specifically binds to monoubiquitinated histone H2A and derepresses Polycomb target genes at the onset of cellular differentiation. Our results suggest that ZRF1 exerts its function in a two-step mechanism, by initially displacing the Polycomb-repressive complex 1 (PRC1) from chromatin and subsequently acting together with histone H2A-specific deubiquitinases to facilitate transcriptional activation of its target genes. These findings demonstrate an ambiguity of the epigenetic monoubiquitin mark at histone H2A. Once considered to be a hallmark of gene silencing, it is now clear that this mark can also be utilized as a recruitment platform for proteins engaged in gene activation. Genome-wide analyses demonstrate that ZRF1 is recruited to typical Polycomb target genes, thereby putting it in a position to have an impact on differentiation and animal development. This molecular mechanism for ZRF1 may represent one of the first steps in switching silenced genes to a transcriptionally active state. We discuss here our recent findings in the light of progress made in understanding Polycomb-mediated silencing.


Subject(s)
DNA-Binding Proteins/physiology , Histones/physiology , Oncogene Proteins/physiology , Transcriptional Activation , Cell Cycle Proteins/metabolism , Cell Differentiation , Chromatin/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Gene Silencing , Histones/genetics , Histones/metabolism , Humans , Molecular Chaperones , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , RNA-Binding Proteins , S Phase , Ubiquitination
18.
Nature ; 468(7327): 1124-8, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-21179169

ABSTRACT

Covalent modification of histones is fundamental in orchestrating chromatin dynamics and transcription. One example of such an epigenetic mark is the mono-ubiquitination of histones, which mainly occurs at histone H2A and H2B. Ubiquitination of histone H2A has been implicated in polycomb-mediated transcriptional silencing. However, the precise role of the ubiquitin mark during silencing is still elusive. Here we show in human cell lines that ZRF1 (zuotin-related factor 1) is specifically recruited to histone H2A when it is ubiquitinated at Lys 119 by means of a novel ubiquitin-interacting domain that is located in the evolutionarily conserved zuotin domain. At the onset of differentiation, ZRF1 specifically displaces polycomb-repressive complex 1 (PRC1) from chromatin and facilitates transcriptional activation. A genome-wide mapping of ZRF1, RING1B and H2A-ubiquitin targets revealed its involvement in the regulation of a large set of polycomb target genes, emphasizing the key role ZRF1 has in cell fate decisions. We provide here a model of the molecular mechanism of switching polycomb-repressed genes to an active state.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Silencing , Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Transcriptional Activation , Cell Line, Tumor , Chromatin/metabolism , Chromosome Mapping , Gene Expression Regulation , HEK293 Cells , Histones/metabolism , Humans , Models, Biological , Molecular Chaperones , Polycomb-Group Proteins , RNA-Binding Proteins , U937 Cells , Ubiquitins/metabolism
19.
Bioessays ; 32(8): 669-79, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20658705

ABSTRACT

Epigenetic modifications, such as DNA methylation and post-translation modifications of histones, have been shown to play an important role in chromatin structure, promoter activity, and cellular reprogramming. Large protein complexes, such as Polycomb and trithorax, often harbor multiple activities which affect histone tail modification. Nevertheless, the mechanisms underlying the deposition of these marks, their propagation during cell replication, and the alteration on their distribution during transformation still require further study. Here we review recent data on those processes in both normal and cancer cells, and we propose that the unscheduled expression of oncogenic transcription factors causes reprogramming of normal cells into cancer stem cells.


Subject(s)
Epigenesis, Genetic/genetics , Neoplasms/genetics , Animals , Cell Transformation, Neoplastic/genetics , Chromatin/genetics , Histones/genetics , Humans , Neoplastic Stem Cells/metabolism
20.
Cell ; 120(1): 73-84, 2005 Jan 14.
Article in English | MEDLINE | ID: mdl-15652483

ABSTRACT

Protein degradation in eukaryotes usually requires multiubiquitylation and subsequent delivery of the tagged substrates to the proteasome. Recent studies suggest the involvement of the AAA ATPase CDC48, its cofactors, and other ubiquitin binding factors in protein degradation, but how these proteins work together is unclear. Here we show that these factors cooperate sequentially through protein-protein interactions and thereby escort ubiquitin-protein conjugates to the proteasome. Central to this pathway is the chaperone CDC48/p97, which coordinates substrate recruitment, E4-catalyzed multiubiquitin chain assembly, and proteasomal targeting. Concomitantly, CDC48 prevents the formation of excessive multiubiquitin chain sizes that are surplus to requirements for degradation. In yeast, this escort pathway guides a transcription factor from its activation in the cytosol to its final degradation and also mediates proteolysis at the endoplasmic reticulum by the ERAD pathway.


Subject(s)
Cell Cycle Proteins/metabolism , Multienzyme Complexes/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitins/metabolism , Adenosine Triphosphatases , Animals , Endoplasmic Reticulum/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Protein Binding , Protein Folding , Ubiquitin-Conjugating Enzymes/metabolism , Valosin Containing Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...