Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
BBA Adv ; 2: 100035, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-37082595

RESUMO

Cellular protein homeostasis (proteostasis) requires an accurate balance between protein biosynthesis, folding, and degradation, and its instability is causally related to human diseases and cancers. Here, we created numerous engineered cancer cell lines targeting APP (amyloid ß precursor protein) and/or PRNP (cellular prion) genes and we showed that APP knocking-down impaired PRNP mRNA level and vice versa, suggesting a link between their gene regulation. PRNPKD, APPKD and PRNPKD/APPKD HeLa cells encountered major difficulties to grow in a 3D tissue-like environment. Unexpectedly, we found a cytoplasmic accumulation of the PrPc protein without PRNP gene up regulation, in both APPKD and APPKO HeLa cells. Interestingly, APP and/or PRNP gene ablation enhanced the chaperone/serine protease HTRA2 gene expression, which is a protein processing quality factor involved in Alzheimer's disease. Importantly, HTRA2 gene silencing decreased PRNP mRNA level and lowered PrPc protein amounts, and conversely, HTRA2 overexpression increased PRNP gene regulation and enhanced membrane-anchored and cytoplasmic PrPc fractions. PrPc, APP and HTRA2 destabilized membrane-associated CD24 protein, suggesting changes in the lipid raft structure. Our data show for the first time that APP and the dual chaperone/serine protease HTRA2 protein could modulate PrPc proteostasis hampering cancer cell behavior.

2.
Front Cell Neurosci ; 14: 14, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32116560

RESUMO

Human brain organoids (mini-brains) consist of self-organized three-dimensional (3D) neural tissue which can be derived from reprogrammed adult cells and maintained for months in culture. These 3D structures manifest substantial potential for the modeling of neurodegenerative diseases and pave the way for personalized medicine. However, as these 3D brain models can express the whole human genetic complexity, it is critical to have access to isogenic mini-brains that only differ in specific and controlled genetic variables. Genetic engineering based on retroviral vectors is incompatible with the long-term modeling needed here and implies a risk of random integration while methods using CRISPR-Cas9 are still too complex to adapt to stem cells. We demonstrate in this study that our strategy which relies on an episomal plasmid vector derived from the Epstein-Barr virus (EBV) offers a simple and robust approach, avoiding the remaining caveats of mini-brain models. For this proof-of-concept, we used a normal tau protein with a fluorescent tag and a mutant genetic form (P301S) leading to Fronto-Temporal Dementia. Isogenic cell lines were obtained which were stable for more than 30 passages expressing either form. We show that the presence of the plasmid in the cells does not interfere with the mini-brain differentiation protocol and obtain the development of a pathologically relevant phenotype in cerebral organoids, with pathological hyperphosphorylation of the tau protein. Such a simple and versatile genetic strategy opens up the full potential of human organoids to contribute to disease modeling, personalized medicine and testing of therapeutics.

3.
PLoS Genet ; 11(7): e1005384, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26181065

RESUMO

Genome stability is jeopardized by imbalances of the dNTP pool; such imbalances affect the rate of fork progression. For example, cytidine deaminase (CDA) deficiency leads to an excess of dCTP, slowing the replication fork. We describe here a novel mechanism by which pyrimidine pool disequilibrium compromises the completion of replication and chromosome segregation: the intracellular accumulation of dCTP inhibits PARP-1 activity. CDA deficiency results in incomplete DNA replication when cells enter mitosis, leading to the formation of ultrafine anaphase bridges between sister-chromatids at "difficult-to-replicate" sites such as centromeres and fragile sites. Using molecular combing, electron microscopy and a sensitive assay involving cell imaging to quantify steady-state PAR levels, we found that DNA replication was unsuccessful due to the partial inhibition of basal PARP-1 activity, rather than slower fork speed. The stimulation of PARP-1 activity in CDA-deficient cells restores replication and, thus, chromosome segregation. Moreover, increasing intracellular dCTP levels generates under-replication-induced sister-chromatid bridges as efficiently as PARP-1 knockdown. These results have direct implications for Bloom syndrome (BS), a rare genetic disease combining susceptibility to cancer and genomic instability. BS results from mutation of the BLM gene, encoding BLM, a RecQ 3'-5' DNA helicase, a deficiency of which leads to CDA downregulation. BS cells thus have a CDA defect, resulting in a high frequency of ultrafine anaphase bridges due entirely to dCTP-dependent PARP-1 inhibition and independent of BLM status. Our study describes previously unknown pathological consequences of the distortion of dNTP pools and reveals an unexpected role for PARP-1 in preventing DNA under-replication and chromosome segregation defects.


Assuntos
Síndrome de Bloom/genética , Citidina Desaminase/genética , Poli(ADP-Ribose) Polimerases/genética , Pirimidinas/metabolismo , Síndrome de Bloom/patologia , Linhagem Celular , Centrômero/genética , Sítios Frágeis do Cromossomo/genética , Segregação de Cromossomos/genética , Citidina Desaminase/deficiência , Replicação do DNA/genética , Instabilidade Genômica , Humanos , Mitose/genética , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/biossíntese , RecQ Helicases/genética , Troca de Cromátide Irmã/genética
4.
Proc Natl Acad Sci U S A ; 112(22): E2910-9, 2015 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-26038566

RESUMO

UV-sensitive syndrome (UV(S)S) and Cockayne syndrome (CS) are human disorders caused by CSA or CSB gene mutations; both conditions cause defective transcription-coupled repair and photosensitivity. Patients with CS also display neurological and developmental abnormalities and dramatic premature aging, and their cells are hypersensitive to oxidative stress. We report CSA/CSB-dependent depletion of the mitochondrial DNA polymerase-γ catalytic subunit (POLG1), due to HTRA3 serine protease accumulation in CS, but not in UV(s)S or control fibroblasts. Inhibition of serine proteases restored physiological POLG1 levels in either CS fibroblasts and in CSB-silenced cells. Moreover, patient-derived CS cells displayed greater nitroso-redox imbalance than UV(S)S cells. Scavengers of reactive oxygen species and peroxynitrite normalized HTRA3 and POLG1 levels in CS cells, and notably, increased mitochondrial oxidative phosphorylation, which was altered in CS cells. These data reveal critical deregulation of proteases potentially linked to progeroid phenotypes in CS, and our results suggest rescue strategies as a therapeutic option.


Assuntos
Síndrome de Cockayne/tratamento farmacológico , DNA Helicases/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Doenças Mitocondriais/tratamento farmacológico , Progéria/patologia , Inibidores de Serina Proteinase/farmacologia , Western Blotting , Células Cultivadas , Síndrome de Cockayne/patologia , DNA Polimerase gama , DNA Mitocondrial/metabolismo , DNA Polimerase Dirigida por DNA/metabolismo , Imunofluorescência , Células HeLa , Humanos , Doenças Mitocondriais/patologia , Ácido Peroxinitroso/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Inibidores de Serina Proteinase/metabolismo
5.
Free Radic Biol Med ; 53(11): 2171-7, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23010470

RESUMO

Although oxidatively damaged DNA is repaired primarily via the base excision repair (BER) pathway, it is now evident that multiple subpathways are needed. Yet, their relative contributions and coordination are still unclear. Here, mouse embryo fibroblasts (MEFs) from selected nucleotide excision repair (NER) and/or BER mouse mutants with severe (Csb(m/m)/Xpa(-/-) and Csb(m/m)/Xpc(-/-)), mild (Csb(m/m)), or no progeria (Xpa(-/-), Xpc(-/-), Ogg1(-/-), Csb(m/m)/Ogg1(-/-)) or wild-type phenotype were exposed to an oxidizing agent, potassium bromate, and genomic 8-oxo-7,8-dihydroguanine (8-oxoGua) levels were measured by HPLC-ED. The same oxidized DNA base was measured in NER/BER-defective human cell lines obtained after transfection with replicative plasmids encoding siRNA targeting DNA repair genes. We show that both BER and NER factors contribute to the repair of 8-oxoGua, although to different extents, and that the repair profiles are similar in human compared to mouse cells. The BER DNA glycosylase OGG1 dominates 8-oxoGua repair, whereas NER (XPC, XPA) and transcription-coupled repair proteins (CSB and CSA) are similar, but minor contributors. The comparison of DNA oxidation levels in double versus single defective MEFs indicates increased oxidatively damaged DNA only when both CSB and XPC/XPA are defective, indicating that these proteins operate in different pathways. Moreover, we provide the first evidence of an involvement of XPA in the control of oxidatively damaged DNA in human primary cells.


Assuntos
Reparo do DNA , Guanina/análogos & derivados , Animais , Sobrevivência Celular , Células Cultivadas , Dano ao DNA , DNA Glicosilases/metabolismo , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , Enzimas Reparadoras do DNA/fisiologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Guanina/metabolismo , Humanos , Cinética , Camundongos , Camundongos Knockout , Oxirredução , Proteínas de Ligação a Poli-ADP-Ribose , Especificidade da Espécie , Proteína de Xeroderma Pigmentoso Grupo A/genética , Proteína de Xeroderma Pigmentoso Grupo A/metabolismo , Proteína de Xeroderma Pigmentoso Grupo A/fisiologia
6.
Mol Cell Biol ; 29(12): 3344-54, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19380493

RESUMO

Human DNA polymerase eta (Pol eta) modulates susceptibility to skin cancer by promoting translesion DNA synthesis (TLS) past sunlight-induced cyclobutane pyrimidine dimers. Despite its well-established role in TLS synthesis, the role of Pol eta in maintaining genome stability in the absence of external DNA damage has not been well explored. We show here that short hairpin RNA-mediated depletion of Pol eta from undamaged human cells affects cell cycle progression and the rate of cell proliferation and results in increased spontaneous chromosome breaks and common fragile site expression with the activation of ATM-mediated DNA damage checkpoint signaling. These phenotypes were also observed in association with modified replication factory dynamics during S phase. In contrast to that seen in Pol eta-depleted cells, none of these cellular or karyotypic defects were observed in cells depleted for Pol iota, the closest relative of Pol eta. Our results identify a new role for Pol eta in maintaining genomic stability during unperturbed S phase and challenge the idea that the sole functional role of Pol eta in human cells is in TLS DNA damage tolerance and/or repair pathways following exogenous DNA damage.


Assuntos
Sítios Frágeis do Cromossomo/fisiologia , Replicação do DNA/fisiologia , DNA Polimerase Dirigida por DNA/metabolismo , Sequência de Bases , Ciclo Celular , Linhagem Celular , Proliferação de Células , Quebra Cromossômica , Dano ao DNA , DNA Polimerase Dirigida por DNA/genética , Instabilidade Genômica/fisiologia , Humanos , Hibridização in Situ Fluorescente , Mutagênese Sítio-Dirigida , Inibidores da Síntese de Ácido Nucleico , RNA Interferente Pequeno/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais
7.
PLoS One ; 4(3): e5018, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19325708

RESUMO

Cervical cancer is frequently associated with HPV infection. The expression of E6 and E7 HPV oncoproteins is a key factor in its carcinogenicity and might also influence its virulence, including metastatic conversion. The cellular mechanisms involved in metastatic spread remain elusive, but pro-adhesive receptors and their ligands, such as SDF-1alpha and CXCR4 are implicated. In the present study, we assessed the possible relationship between SDF-1alpha/CXCR4 signaling, E6/E7 status and the metastatic process. We found that SDF-1alpha stimulated the invasion of E6/E7-positive cancer cell lines (HeLa and TC-1) in Matrigel though CXCR4 and subsequent Rho/ROCK activation. In pulmonary metastatic foci generated by TC-1 cells IV injection a high proportion of cells expressed membrane-associated CXCR4. In both cases models (in vitro and in vivo) cell adhesion and invasion was abrogated by CXCR4 immunological blockade supporting a contribution of SDF-1alpha/CXCR4 to the metastatic process. E6 and E7 silencing using stable knock-down and the approved anti-viral agent, Cidofovir decreased CXCR4 gene expression as well as both, constitutive and SDF-1alpha-induced cell invasion. In addition, Cidofovir inhibited lung metastasis (both adhesion and invasion) supporting contribution of E6 and E7 oncoproteins to the metastatic process. Finally, potential signals activated downstream SDF-1alpha/CXCR4 and involved in lung homing of E6/E7-expressing tumor cells were investigated. The contribution of the Rho/ROCK pathway was suggested by the inhibitory effect triggered by Cidofovir and further confirmed using Y-27632 (a small molecule ROCK inhibitor). These data suggest a novel and highly translatable therapeutic approach to cervix cancer, by inhibition of adhesion and invasion of circulating HPV-positive tumor cells, using Cidofovir and/or ROCK inhibition.


Assuntos
Citosina/análogos & derivados , Metástase Neoplásica/tratamento farmacológico , Proteínas Oncogênicas Virais/antagonistas & inibidores , Organofosfonatos/farmacologia , Receptores CXCR4/antagonistas & inibidores , Proteínas rho de Ligação ao GTP/metabolismo , Quinases Associadas a rho/metabolismo , Alphapapillomavirus , Linhagem Celular Tumoral , Quimiocina CXCL12/metabolismo , Cidofovir , Citosina/farmacologia , Humanos , Proteínas E7 de Papillomavirus/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos
8.
Mol Carcinog ; 48(4): 369-78, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19117014

RESUMO

Accurate DNA replication during S-phase is fundamental to maintain genome integrity. During this critical process, replication forks frequently encounter obstacles that impede their progression. While the regulatory pathways which act in response to exogenous replication stress are beginning to emerge, the mechanisms by which fork integrity is maintained at naturally occurring endogenous replication-impeding sequences remains obscure. Notably, little is known about how cells replicate through special chromosomal regions containing structured non-B DNA, for example, G4 quartets, known to hamper fork progression or trigger chromosomal rearrangements. Here, we have investigated the role in this process of the human translesion synthesis (TLS) DNA polymerases of the Y-family (pol eta, pol iota, and pol kappa), specialized enzymes known to synthesize DNA through DNA damage. We show that depletion by RNA interference of expression of the genes for Pol eta or Pol kappa, but not Pol iota, sensitizes U2OS cells treated with the G4-tetraplex interactive compound telomestatin and triggers double-strand breaks in HeLa cells harboring multiple copies of a G-rich sequence from the promoter region of the human c-MYC gene, chromosomally integrated as a transgene. Moreover, we found that downregulation of Pol kappa only raises the level of DSB in HeLa cells containing either one of two breakage hotspot structured DNA sequences in the chromosome, the major break region (Mbr) of BCL-2 gene and the GA rich region from the far right-hand end of the genome of the Kaposi Sarcoma associated Herpesvirus. These data suggest that naturally occurring DNA structures are physiological substrates of both pol eta and pol kappa. We discuss these data in the light of their downregulation in human cancers.


Assuntos
Neoplasias Colorretais/genética , Replicação do DNA , DNA Polimerase Dirigida por DNA/fisiologia , Quadruplex G , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/secundário , Western Blotting , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Ensaio de Unidades Formadoras de Colônias , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Dano ao DNA , Citometria de Fluxo , Inativação Gênica , Genes myc/genética , Células HeLa , Histonas/metabolismo , Humanos , Inibidores da Síntese de Ácido Nucleico , Osteossarcoma/genética , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Oxazóis/farmacologia , Regiões Promotoras Genéticas/genética , DNA Polimerase iota
9.
DNA Repair (Amst) ; 7(10): 1757-64, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18678285

RESUMO

In the childhood tumor neuroblastoma, high expression of the TrkA neurotrophin receptor is associated with a favorable prognosis and a lack of structural chromosomal changes, whereas TrkB is expressed in aggressive neuroblastomas demonstrating high genomic instability. The ability to repair DNA double-strand breaks (DSBs) is considered a central determinant of chromosomal stability with nonhomologous end joining (NHEJ) being the major pathway of DSB repair in vertebrates. Here, we used the SH-SY5Y human neuroblastoma cell line ectopically expressing either TrkA or TrkB as a model system to analyze the impact of Trk receptor expression on NHEJ-mediated DSB repair. In a cell-free NHEJ assay, SY5Y-TrkA cells displayed a significantly higher efficiency for NHEJ compared to SY5Y-TrkB cells. To detect possible underlying mechanisms, gene expression data (Affymetrix U95A microarray chips) obtained from the same SY5Y-TrkA/TrkB model system were reanalyzed focussing on genes involved in DNA repair. Expression of XRCC4, a central component of NHEJ, was significantly upregulated in SY5Y-TrkA compared to SY5Y-TrkB cells. Expression data were confirmed using real-time PCR and western blotting. Additionally, XRCC4 expression was enhanced in most primary neuroblastomas with high TrkA expression. The TrkA-induced increase in NHEJ activity could be reverted by XRCC4 knock-down, confirming the induction of XRCC4 by TrkA to be essential for the observed phenotype. Our data provide the first evidence for a functional relationship between tyrosine kinase receptor signaling and NHEJ-mediated DSB repair in cancer cells, potentially contributing to their genomic stability.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , Neuroblastoma/enzimologia , Neuroblastoma/patologia , Receptor trkA/metabolismo , Receptor trkB/metabolismo , Linhagem Celular Tumoral , Sistema Livre de Células , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Cinética , Neuroblastoma/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Recombinação Genética , Regulação para Cima
10.
Cancer Res ; 67(6): 2526-34, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17363570

RESUMO

To study the relationships between different DNA repair pathways, we established a set of clones in which one specific DNA repair gene was silenced using long-term RNA interference in HeLa cell line. We focus here on genes involved in either nucleotide excision repair (XPA and XPC) or nonhomologous end joining (NHEJ; DNA-PKcs and XRCC4). As expected, XPA(KD) (knock down) and XPC(KD) cells were highly sensitive to UVC. DNA-PKcs(KD) and XRCC4(KD) cells presented an increased sensitivity to various inducers of double-strand breaks (DSBs) and a 70% to 80% reduction of in vitro NHEJ activity. Long-term silencing of XPC gene expression led to an increased sensitivity to etoposide, a topoisomerase II inhibitor that creates DSBs through the progression of DNA replication forks. XPC(KD) cells also showed intolerance toward acute gamma-ray irradiation. We showed that XPC(KD) cells exhibited an altered spectrum of NHEJ products with decreased levels of intramolecular joined products. Moreover, in both XPC(KD) and DNA-PKcs(KD) cells, XRCC4 and ligase IV proteins were mobilized on damaged nuclear structures at lower doses of DSB inducer. In XPC-proficient cells, XPC protein was released from nuclear structures after induction of DSBs. By contrast, silencing of XPA gene expression did not have any effect on sensitivity to DSB or NHEJ. Our results suggest that XPC deficiency, certainly in combination with other genetic defects, may contribute to impair DSB repair.


Assuntos
Dano ao DNA , Reparo do DNA/fisiologia , Proteínas de Ligação a DNA/genética , Inativação Gênica , DNA Ligase Dependente de ATP , DNA Ligases/metabolismo , Reparo do DNA/genética , Reparo do DNA/efeitos da radiação , Proteína Quinase Ativada por DNA/biossíntese , Proteína Quinase Ativada por DNA/genética , Proteína Quinase Ativada por DNA/metabolismo , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/metabolismo , Etoposídeo/farmacologia , Raios gama , Células HeLa , Humanos , Interferência de RNA
11.
Mol Cancer Res ; 3(9): 519-29, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16179499

RESUMO

We developed and characterized replicative small interfering RNA (siRNA) vectors for efficient, specific, and long-term gene silencing in human cells. We created stable XPA(KD) and XPC(KD) (knockdown) syngeneic cell lines to mimic human cancer-prone syndromes. We also silenced (HSA)KIN17. Several clones displaying undetectable protein levels of XPA, XPC, or (HSA)kin17 were grown for more than 300 days. This stability of gene silencing over several months of culture allows us to assess the specific involvement of these proteins in UVC sensitivity in syngeneic cells. Unlike XPA, (HSA)KIN17, and XPC gene silencing dramatically impeded HeLa cell growth for several weeks after transfection. As expected, XPA(KD) and XPC(KD) HeLa cells were highly UVC sensitive. They presented an impaired unscheduled DNA synthesis after UVC irradiation. Interestingly, XPC(KD) HeLa clones were more sensitive to UVC than their XPA(KD) or KIN17(KD) counterparts. Hygromycin B withdrawal led to the total disappearance of EBV vectors and the resumption of normal XPA or XPC protein levels. Whereas reverted XPA(KD) cells recovered a normal UVC sensitivity, XPC(KD) cells remained highly sensitive, suggestive of irreversible damage following long-term XPC silencing. Our results show that in HeLa cells, (HSA)kin17 participates indirectly in early events following UVC irradiation, and XPC deficiency strongly affects cell physiology and contributes to UVC sensitivity to a greater extent than does XPA. EBV-based siRNA vectors improve the interest of siRNA by permitting long-term gene silencing without the safety concerns inherent in viral-based siRNA vehicles.


Assuntos
Reparo do DNA , Inativação Gênica , Vetores Genéticos/fisiologia , Herpesvirus Humano 4/genética , RNA Interferente Pequeno/genética , Raios Ultravioleta , Southern Blotting , Western Blotting , Ciclo Celular/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Proteínas de Ligação a DNA/genética , Citometria de Fluxo , Células HeLa/fisiologia , Células HeLa/efeitos da radiação , Humanos , Proteínas Nucleares/genética , Proteínas de Ligação a RNA , Ensaio Tumoral de Célula-Tronco , Proteína de Xeroderma Pigmentoso Grupo A
12.
Mol Cell Biol ; 25(9): 3814-30, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15831485

RESUMO

The human stress-activated protein kin17 accumulates in the nuclei of proliferating cells with predominant colocalization with sites of active DNA replication. The distribution of kin17 protein is in equilibrium between chromatin-DNA and the nuclear matrix. An increased association with nonchromatin nuclear structure is observed in S-phase cells. We demonstrated here that kin17 protein strongly associates in vivo with DNA fragments containing replication origins in both human HeLa and monkey CV-1 cells. This association was 10-fold higher than that observed with nonorigin control DNA fragments in exponentially growing cells. In addition, the association of kin17 protein to DNA fragments containing replication origins was also analyzed as a function of the cell cycle. High binding of kin17 protein was found at the G(1)/S border and throughout the S phase and was negligible in both G(0) and M phases. Specific monoclonal antibodies against kin17 protein induced a threefold inhibition of in vitro DNA replication of a plasmid containing a minimal replication origin that could be partially restored by the addition of recombinant kin17 protein. Immunoelectron microscopy confirmed the colocalization of kin17 protein with replication proteins like RPA, PCNA, and DNA polymerase alpha. A two-step chromatographic fractionation of nuclear extracts from HeLa cells revealed that kin17 protein localized in vivo in distinct protein complexes of high molecular weight. We found that kin17 protein purified within an approximately 600-kDa protein complex able to support in vitro DNA replication by means of two different biochemical methods designed to isolate replication complexes. In addition, the reduced in vitro DNA replication activity of the multiprotein replication complex after immunodepletion for kin17 protein highlighted for a direct role in DNA replication at the origins.


Assuntos
Replicação do DNA/fisiologia , Proteínas de Ligação a DNA/fisiologia , Proteínas Nucleares/fisiologia , Origem de Replicação/fisiologia , Animais , Anticorpos Monoclonais/farmacologia , Ciclo Celular/fisiologia , Núcleo Celular/química , Cromatina/metabolismo , DNA Polimerase I/análise , DNA Polimerase I/metabolismo , Replicação do DNA/efeitos dos fármacos , Replicação do DNA/genética , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/metabolismo , Haplorrinos , Humanos , Microscopia Imunoeletrônica , Complexos Multiproteicos/fisiologia , Complexos Multiproteicos/ultraestrutura , Proteínas Nucleares/análise , Proteínas Nucleares/antagonistas & inibidores , Antígeno Nuclear de Célula em Proliferação/análise , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteínas de Ligação a RNA , Proteína de Replicação A
13.
Nucleic Acids Res ; 31(14): 4162-75, 2003 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12853634

RESUMO

Several proteins involved in DNA synthesis are part of the so-called 'replication factories' that are anchored on non-chromatin nuclear structures. We report here that human kin17, a nuclear stress-activated protein, associates with both chromatin and non-chromatin nuclear structures in a cell cycle- and DNA damage-dependent manner. After L-mimosine block and withdrawal we observed that kin17 protein was recruited in the nucleus during re-entry and progression through S phase. These results are consistent with a role of kin17 protein in DNA replication. About 50% of the total amount of kin17 protein was detected on nuclear structures and could not be released by detergents. Furthermore, the amount of kin17 protein greatly increased in both G(1)/S and S phase-arrested cells in fractions containing proteins anchored to nuclear structures. The detection of kin17 protein showed for the first time its preferential assembly within non-chromatin nuclear structures in G(1)/S and S phase-arrested cells, while the association with these structures was found to be less stable in the G(2)/M phase, as judged by fractionation of human cells and immunostaining. In asynchronous growing cells, kin17 protein interacted with both chromatin DNA and non-chromatin nuclear structures, while in S phase-arrested cells it interacted mostly with non-chromatin nuclear structures, as judged by DNase I treatment and in vivo UV cross-linking. In the presence of DNA damage in S phase cells, the distribution of kin17 protein became mainly associated with chromosomal DNA, as judged by limited formaldehyde cross-linking of living cells. The physical interaction of kin17 protein with components of the nuclear matrix was confirmed and visualized by indirect immunofluorescence and immunoelectron microscopy. Our results indicate that, during S phase, a fraction of the human kin17 protein preferentially associates with the nuclear matrix, a fundamentally non-chromatin higher order nuclear structure, and to chromatin DNA in the presence of DNA damage.


Assuntos
Ciclo Celular/fisiologia , Cromatina/metabolismo , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Matriz Nuclear/metabolismo , Proteínas Nucleares , Cromatina/genética , DNA/genética , DNA/metabolismo , Replicação do DNA , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Immunoblotting , Microscopia Imunoeletrônica , Matriz Nuclear/ultraestrutura , Ligação Proteica , Proteínas de Ligação a RNA , Proteína de Replicação A , Fase S/fisiologia , Células Tumorais Cultivadas
14.
Mol Cancer Res ; 1(7): 519-31, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12754299

RESUMO

The Homo sapiens kin17 ((HSA)kin17) protein is a chromatin-associated protein conserved during evolution and overproduced in certain human tumor cell lines. For the first time, immunoelectron microscopy analysis of endogenous (HSA)kin17 protein revealed an ultrastructural co-localization of (HSA)kin17 and bromodeoxyuridine (BrdUrd) at sites of DNA replication after either short (15 min) or long (120 min) pulses of BrdUrd labeling. After hydroxyurea (HU) or L-mimosine (Mimo) block and withdrawal, we observed that (HSA)kin17 was recruited onto the chromatin during the re-entry and the progression in the S phase. These results are consistent with a major role of (HSA)kin17 protein in DNA replication factories. Other treatments hampering replication fork progression and/or inducing double-strand breaks also triggered an accumulation and a concentration of the chromatin-bound (HSA)kin17 protein into large intranuclear foci 24 h post-treatment. Moreover, HU- and Mimo-induced (HSA)kin17 foci were retained in the nucleus after detergent extraction, suggesting a strong association with nuclear structures. Gel filtration analyses of cellular extracts showed that endogenous (HSA)kin17 protein co-eluted with both replication proteins RPA32 and RPA70 in a fraction containing complexes of M(r) 600,000. Interestingly, HU-induced G(1)-S arrest triggered an increase in the molecular weight of complexes containing (HSA)kin17 protein. Hence, treatments interfering with either initiation and/or elongation of DNA replication also recruited chromatin-bound (HSA)kin17 protein. We hypothesize that in the presence of unrepaired DNA damage, (HSA)kin17 protein concentrated into high molecular weight complexes probably to create a bridge that contributes to the harmonization of DNA replication and repair.


Assuntos
Replicação do DNA/fisiologia , Proteínas de Ligação a DNA/fisiologia , Proteínas Nucleares/fisiologia , Animais , Antineoplásicos/toxicidade , Bromodesoxiuridina , Carcinoma Pulmonar de Células não Pequenas , Divisão Celular , Neoplasias do Colo , Replicação do DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/análise , Citometria de Fluxo , Humanos , Hidroxiureia/toxicidade , Neoplasias Pulmonares , Microscopia Imunoeletrônica , Mimosina/toxicidade , Proteínas Nucleares/análise , Proteínas de Ligação a RNA , Células Tumorais Cultivadas , Dedos de Zinco
15.
Cancer Res ; 62(19): 5425-35, 2002 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-12359749

RESUMO

Kin17 is an evolutionarily conserved DNA-binding protein, which forms intranuclear foci in proliferating cells. Recent data have suggested that human kin17 protein is associated with cell proliferation and unrepaired DNA lesions. Herein, we show that human fibroblasts (MRC5-V2 and CHSV4) immortalized with SV40 overexpress endogenous kin17 protein, as compared with normal diploid human fibroblasts. We observed that certain carcinoma cell lines also up-regulated kin17 protein, suggesting that increased kin17 protein levels may be a consequence of the immortalized phenotype. We report here that the endogenous kin17 protein is located in nucleoplasmic foci and colocalizes with SV40 large T antigen. Purification of human kin17 protein allowed analysis of the physical interaction with T antigen by several in vitro and in vivo assays. Large T antigen and human kin17 protein are part of the same high molecular weight multiprotein complex in human cells. Furthermore, human kin17 protein interacts with T antigen bound to the SV40 DNA origin of replication. Strikingly, the overexpression of human kin17 protein in vivo and the introduction of increased amounts of human kin17 protein in an in vitro assay reduced T-antigen-dependent DNA replication, suggesting that kin17 protein may be involved in the DNA replication process in human cells.


Assuntos
Antígenos Virais de Tumores/metabolismo , Replicação do DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , DNA/antagonistas & inibidores , Proteínas Nucleares , Animais , Baculoviridae/genética , Linhagem Celular Transformada , Núcleo Celular/metabolismo , Cromatina/metabolismo , DNA/biossíntese , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Fibroblastos/metabolismo , Histidina/metabolismo , Humanos , Testes de Precipitina , Proteínas de Ligação a RNA , Spodoptera/metabolismo , Spodoptera/virologia , Transfecção , Células Tumorais Cultivadas , Regulação para Cima
16.
J Biol Chem ; 277(21): 19156-65, 2002 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-11880372

RESUMO

The human DNA-binding (HSA)kin17 protein cross-reacts with antibodies raised against the stress-activated Escherichia coli RecA protein. We show here that (HSA)kin17 protein is directly associated with chromosomal DNA as judged by cross-linking experiments on living cells. We detected increased amounts of DNA-bound (HSA)kin17 protein 24 h after gamma irradiation, with 2.6-fold more (HSA)kin17 molecules after 6 Gy of irradiation (46,000-117,000 molecules). At this time we observed that highly proliferating RKO cells displayed the concentration and co-localization of (HSA)kin17 and replication protein A in nucleoplasmic foci. Our results suggest that 24 h post-irradiation (HSA)kin17 protein may localize at the sites of unrepaired DNA damages. RKO clones expressing an (HSA)KIN17 antisense transcript (RASK.5 and RASK.13 cells) revealed that reduced (HSA)kin17 protein levels are correlated with a decrease in clonogenic cell growth and cell proliferation, as well as an accumulation of cells in early and mid-S phase. Taken together our observations support the idea that (HSA)kin17 protein is a DNA maintenance protein involved in the cellular response to the presence of DNA damage and suggest that it helps to overcome the perturbation of DNA replication produced by unrepaired lesions.


Assuntos
Cromatina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares , DNA/metabolismo , Proteínas de Ligação a DNA/genética , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Ligação Proteica , RNA Mensageiro/genética , Proteínas de Ligação a RNA , Fase S , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA