Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 11(1): 17052, 2021 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-34426596

RESUMO

Activation of a telomere length maintenance mechanism (TMM), including telomerase and alternative lengthening of telomeres (ALT), is essential for replicative immortality of tumor cells, although its regulatory mechanisms are incompletely understood. We conducted a microRNA (miRNA) microarray analysis on isogenic telomerase positive (TEP) and ALT cancer cell lines. Amongst nine miRNAs that showed difference in their expression in TEP and ALT cancer cells in array analysis, miR-708 was selected for further analysis since it was consistently highly expressed in a large panel of ALT cells. miR-708 in TEP and ALT cancer cells was not correlated with C-circle levels, an established feature of ALT cells. Its overexpression induced suppression of cell migration, invasion, and angiogenesis in both TEP and ALT cells, although cell proliferation was inhibited only in TEP cells suggesting that ALT cells may have acquired the ability to escape inhibition of cell proliferation by sustained miR-708 overexpression. Further, cell proliferation regulation in TEP cells by miR708 appears to be through the CARF-p53 pathway. We demonstrate here that miR-708 (i) is the first miRNA shown to be differentially regulated in TEP and ALT cancer cells, (ii) possesses tumor suppressor function, and (iii) deregulates CARF and p21WAF1-mediated signaling to limit proliferation in TEP cells.


Assuntos
MicroRNAs/metabolismo , Neoplasias/metabolismo , Telomerase/genética , Células A549 , Movimento Celular , Proliferação de Células , Células HEK293 , Humanos , MicroRNAs/genética , Telomerase/deficiência
2.
Sci Rep ; 8(1): 375, 2018 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-29321561

RESUMO

microRNAs (miRs) have recently emerged as small non-coding regulators of gene expression. We performed a loss-of-function screening by recruiting retrovirus mediated arbitrary manipulation of genome coupled with escape of cells from 5-Aza-2'-deoxycytidine (5-Aza-dC)-induced senescence. miRNA pool from cells that emerged from 5-Aza-dC-induced senescence was subjected to miR-microarray analysis with respect to the untreated control. We identified miR-451 as one of the upregulated miRs and characterized its functional relevance to drug resistance, cell growth, tumor suppressor proteins p53 and pRb, and stress response. We report that miR-451 caused growth arrest in cells leading to their resistance to 5-Aza-dC-induced senescence. Decrease in cyclin D1, CDK4 and phosphorylated pRB supported the growth arrest in miR-451 transfected cells. We demonstrate that Collaborator of ARF (CARF) protein is a new target of miR-451 that intermediates its function in tumor suppressor and stress signaling.


Assuntos
Azacitidina/análogos & derivados , Perfilação da Expressão Gênica/métodos , MicroRNAs/genética , Neoplasias/genética , Fatores de Transcrição/genética , Regulação para Cima , Células A549 , Animais , Azacitidina/farmacologia , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Proteínas de Ligação a DNA , Decitabina , Resistencia a Medicamentos Antineoplásicos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7 , Camundongos , Transplante de Neoplasias , Neoplasias/patologia , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Transdução de Sinais
3.
Cell Death Dis ; 8(4): e2755, 2017 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-28425984

RESUMO

Maintenance of telomere length is the most consistent attribute of cancer cells. Tightly connected to their capacity to overcome replicative mortality, it is achieved either by activation of telomerase or an Alternative mechanism of Lengthening of Telomeres (ALT). Disruption of either of these mechanisms has been shown to induce DNA damage signalling leading to senescence or apoptosis. Telomerase inhibitors are considered as potential anticancer drugs but are ineffective for ALT cancers (~15% of all cancers). Withaferin-A (Wi-A), a major constituent of the medicinal plant, Withania somnifera (Ashwagandha), has been shown to exert anti-tumour activity. However, its effect on either telomerase or ALT mechanisms has not been investigated. Here, by using isogenic cancer cells with/without telomerase, we found that Wi-A caused stronger cytotoxicity to ALT cells. It was associated with inhibition of ALT-associated promyelocytic leukemia nuclear bodies, an established marker of ALT. Comparative analyses of telomerase positive and ALT cells revealed that Wi-A caused stronger telomere dysfunction and upregulation of DNA damage response in ALT cells. Molecular computational and experimental analyses revealed that Wi-A led to Myc-Mad mediated transcriptional suppression of NBS-1, an MRN complex protein that is an essential component of the ALT mechanism. The results suggest that Wi-A could be a new candidate drug for ALT cancers.


Assuntos
Modelos Moleculares , Neoplasias/enzimologia , Neoplasias/patologia , Telomerase/metabolismo , Vitanolídeos/química , Vitanolídeos/farmacologia , Sítios de Ligação , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , DNA/metabolismo , Regulação para Baixo/efeitos dos fármacos , Humanos , Simulação de Dinâmica Molecular , Complexos Multiproteicos/metabolismo , Fenótipo , Ligação Proteica/efeitos dos fármacos , Telômero/metabolismo , Homeostase do Telômero/efeitos dos fármacos
4.
Genes Chromosomes Cancer ; 56(8): 617-631, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28398700

RESUMO

Cancer cells require telomere maintenance to enable uncontrolled growth. Most often telomerase is activated, although a subset of human cancers are telomerase-negative and depend on recombination-based mechanisms known as ALT (Alternative Lengthening of Telomeres). ALT depends on proteins that are essential for homologous recombination, including BLM and the MRN complex, to extend telomeres. This study surveyed the requirement for requisite homologous recombination proteins, yet to be studied in human ALT cell lines, by protein depletion using RNA interference. Effects on ALT were evaluated by measuring C-circle abundance, a marker of ALT. Surprisingly, several proteins essential for homologous recombination, BARD1, BRCA2, and WRN, were dispensable for C-circle production, while PALB2 had varying effects on C-circles among ALT cell lines. Depletion of homologous recombination proteins BRCA1 and BLM, which have been previously studied in ALT, decreased C-circles in all ALT cell lines. Depletion of the non-homologous end joining proteins 53BP1 and LIG4 had no effect on C-circles in any ALT cell line. Proteins such as chromatin modifiers that recruit double-strand break proteins, RNF8 and RNF168, and other proteins loosely grouped into excision DNA repair processes, XPA, MSH2, and MPG, reduced C-circles in some ALT cell lines. MSH2 depletion also reduced recombination at telomeres as measured by intertelomeric exchanges. Collectively, the requirement for DNA repair proteins varied between the ALT cell lines compared. In sum, our study suggests that ALT proceeds by multiple mechanisms that differ between cell lines and that some of these depend on DNA repair proteins not associated with homologous recombination pathways.


Assuntos
Enzimas Reparadoras do DNA/genética , Neoplasias/genética , Homeostase do Telômero , Reparo do DNA , Enzimas Reparadoras do DNA/metabolismo , Células HeLa , Humanos
5.
Sci Rep ; 6: 30185, 2016 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-27457128

RESUMO

Significance of microRNAs (miRs), small non-coding molecules, has been implicated in a variety of biological processes. Here, we recruited retroviral insertional mutagenesis to obtain induction of an arbitrary noncoding RNAs, and coupled it with a cell based loss-of-function (5-Aza-2'-deoxycytidine (5Aza-dC)-induced senescence bypass) screening system. Cells that escaped 5-Aza-dC-induced senescence were subjected to miR-microarray analysis with respect to the untreated control. We identified miR-335 as one of the upregulated miRs. In order to characterize the functional significance, we overexpressed miR-335 in human cancer cells and found that it caused growth suppression. We demonstrate that the latter accounted for inhibition of 5-Aza-dC incorporation into the cell genome, enabling them to escape from induction of senescence. We also report that CARF (Collaborator of ARF) is a new target of miR-335 that regulates its growth suppressor function by complex crosstalk with other proteins including p16(INK4A), pRB, HDM2 and p21(WAF1).


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Genes Supressores de Tumor , MicroRNAs/fisiologia , Proteínas de Ligação a RNA/metabolismo , Animais , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Linhagem Celular Tumoral , Senescência Celular/efeitos dos fármacos , Decitabina , Genes p16 , Xenoenxertos , Humanos , Camundongos , Camundongos Nus , MicroRNAs/genética , Proteína do Retinoblastoma/metabolismo
6.
Cancer Res ; 76(9): 2754-2765, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26960973

RESUMO

Mortalin/mthsp70 (HSPA9) is a stress chaperone enriched in many cancers that has been implicated in carcinogenesis by promoting cell proliferation and survival. In the present study, we examined the clinical relevance of mortalin upregulation in carcinogenesis. Consistent with high mortalin expression in various human tumors and cell lines, we found that mortalin overexpression increased the migration and invasiveness of breast cancer cells. Expression analyses revealed that proteins involved in focal adhesion, PI3K-Akt and JAK-STAT signaling, all known to play key roles in cell migration and epithelial-to-mesenchymal transition (EMT), were upregulated in mortalin-expressing cancer cells. We further determined that expression levels of the mesenchymal markers vimentin (VIM), fibronectin (FN1), ß-catenin (CTNNB1), CK14 (KRT14) and hnRNP-K were also increased upon mortalin overexpression, whereas the epithelial markers E-cadherin (CDH1), CK8 (KRT8), and CK18 (KRT18) were downregulated. Furthermore, shRNA-mediated and pharmacological inhibition of mortalin suppressed the migration and invasive capacity of cancer cells and was associated with a diminished EMT gene signature. Taken together, these findings support a role for mortalin in the induction of EMT, prompting further investigation of its therapeutic value in metastatic disease models.

7.
J Gerontol A Biol Sci Med Sci ; 70(6): 701-13, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24747666

RESUMO

The heat shock 70 family protein, mortalin, has pancytoplasmic distribution pattern in normal and perinuclear in cancer human cells. Cancer cells when induced to senesce by either chemicals or stress showed shift in mortalin staining pattern from perinuclear to pancytoplasmic type. Using such shift in mortalin staining as a reporter, we screened human shRNA library and identified nine senescence-inducing siRNA candidates. An independent Comparative Genomic Hybridization analysis of 35 breast cancer cell lines revealed that five (NBS1, BRCA1, TIN2, MRE11A, and KPNA2) of the nine genes located on chromosome regions identified as the gain of locus in more than 80% cell lines. By gene-specific PCR, these five genes were found to be frequently amplified in cancer cell lines. Bioinformatics revealed that the identified targets were connected to MRN (MRE11-RAD50-NBS1) complex, the DNA damage-sensing complex. We demonstrate that the identified shRNAs triggered DNA damage response and induced the expression of tumor suppressor protein p16(INK4A) causing growth arrest of cancer cells. Furthermore, cells showed decreased migration, mediated by decrease in matrix metalloproteases. Taken together, we demonstrate that the MRN complex is a potential target of cancer cell proliferation and migration, and staining pattern of mortalin could serve as an assay to identify senescence-inducing/anticancer reagents.


Assuntos
Movimento Celular , Senescência Celular , Dano ao DNA , Proteínas de Choque Térmico HSP70/metabolismo , Neoplasias/patologia , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Proteínas de Ligação a DNA/genética , Biblioteca Gênica , Humanos , Proteína Homóloga a MRE11 , Neoplasias/genética , Proteínas Nucleares , RNA de Cadeia Dupla , Proteínas de Ligação a Telômeros , Ubiquitina-Proteína Ligases , alfa Carioferinas
8.
PLoS One ; 9(8): e103819, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25084169

RESUMO

Fifteen percent of tumors utilize recombination-based alternative lengthening of telomeres (ALT) to maintain telomeres. The mechanisms underlying ALT are unclear but involve several proteins involved in homologous recombination including the BLM helicase, mutated in Bloom's syndrome, and the BRCA1 tumor suppressor. Cells deficient in either BLM or BRCA1 have phenotypes consistent with telomere dysfunction. Although BLM associates with numerous DNA damage repair proteins including BRCA1 during DNA repair, the functional consequences of BLM-BRCA1 association in telomere maintenance are not completely understood. Our earlier work showed the involvement of BRCA1 in different mechanisms of ALT, and telomere shortening upon loss of BLM in ALT cells. In order to delineate their roles in telomere maintenance, we studied their association in telomere metabolism in cells using ALT. This work shows that BLM and BRCA1 co-localize with RAD50 at telomeres during S- and G2-phases of the cell cycle in immortalized human cells using ALT but not in cells using telomerase to maintain telomeres. Co-immunoprecipitation of BRCA1 and BLM is enhanced in ALT cells at G2. Furthermore, BRCA1 and BLM interact with RAD50 predominantly in S- and G2-phases, respectively. Biochemical assays demonstrate that full-length BRCA1 increases the unwinding rate of BLM three-fold in assays using a DNA substrate that models a forked structure composed of telomeric repeats. Our results suggest that BRCA1 participates in ALT through its interactions with RAD50 and BLM.


Assuntos
Proteína BRCA1/metabolismo , RecQ Helicases/metabolismo , Homeostase do Telômero/fisiologia , Proteína BRCA1/genética , Western Blotting , Ciclo Celular/genética , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Reparo do DNA/genética , Reparo do DNA/fisiologia , Imunofluorescência , Células HeLa , Humanos , Imunoprecipitação , RNA Interferente Pequeno , RecQ Helicases/genética , Homeostase do Telômero/genética
9.
J Biol Chem ; 289(36): 24832-44, 2014 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-25012652

RESUMO

The Hsp70 family protein mortalin is an essential chaperone that is frequently enriched in cancer cells and exists in various subcellular sites, including the mitochondrion, plasma membrane, endoplasmic reticulum, and cytosol. Although the molecular mechanisms underlying its multiple subcellular localizations are not yet clear, their functional significance has been revealed by several studies. In this study, we examined the nuclear fractions of human cells and found that the malignantly transformed cells have more mortalin than the normal cells. We then generated a mortalin mutant that lacked a mitochondrial targeting signal peptide. It was largely localized in the nucleus, and, hence, is called nuclear mortalin (mot-N). Functional characterization of mot-N revealed that it efficiently protects cancer cells against endogenous and exogenous oxidative stress. Furthermore, compared with the full-length mortalin overexpressing cancer cells, mot-N derivatives showed increased malignant properties, including higher proliferation rate, colony forming efficacy, motility, and tumor forming capacity both in in vitro and in vivo assays. We demonstrate that mot-N promotes carcinogenesis and cancer cell metastasis by inactivation of tumor suppressor protein p53 functions and by interaction and functional activation of telomerase and heterogeneous ribonucleoprotein K (hnRNP-K) proteins.


Assuntos
Núcleo Celular/metabolismo , Transformação Celular Neoplásica/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Neoplasias/metabolismo , Animais , Western Blotting , Linhagem Celular , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Células HCT116 , Proteínas de Choque Térmico HSP70/genética , Células HeLa , Ribonucleoproteínas Nucleares Heterogêneas Grupo K/metabolismo , Humanos , Imuno-Histoquímica , Células MCF-7 , Camundongos Endogâmicos BALB C , Camundongos Nus , Mutação , Metástase Neoplásica , Neoplasias/genética , Neoplasias/patologia , Estresse Oxidativo , Telomerase/genética , Telomerase/metabolismo , Transplante Heterólogo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
10.
Exp Cell Res ; 322(2): 324-34, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24485912

RESUMO

CARF is an ARF-binding protein that has been shown to regulate the p53-p21-HDM2 pathway. CARF overexpression was shown to cause growth arrest of human cancer cells and premature senescence of normal cells through activation of the p53 pathway. Because replicative senescence involves permanent withdrawal from the cell cycle in response to DNA damage response-mediated signaling, in the present study we investigated the relationship between CARF and the cell cycle and whether it is involved in the DNA damage response. We demonstrate that the half-life of CARF protein is less than 60 min, and that in cycling cells CARF levels are highest in G2 and early prophase. Serially passaged normal human skin and stromal fibroblasts showed upregulation of CARF during replicative senescence. Induction of G1 growth arrest and senescence by a variety of drugs was associated with increase in CARF expression at the transcriptional and translational level and was seen to correlate with increase in DNA damage response and checkpoint proteins, ATM, ATR, CHK1, CHK2, γH2AX, p53 and p21. Induction of growth arrest by oncogenic RAS and shRNA-mediated knockdown of TRF2 in cancer cells also caused upregulation of CARF. We conclude that CARF is associated with DNA damage response and checkpoint signaling pathways.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Pontos de Checagem do Ciclo Celular/fisiologia , Dano ao DNA/fisiologia , Fibroblastos/metabolismo , Proteínas de Ligação a RNA/metabolismo , Pele/metabolismo , Células Estromais/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/genética , Western Blotting , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Senescência Celular/fisiologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Imunofluorescência , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Pele/citologia , Pele/efeitos dos fármacos , Células Estromais/citologia , Células Estromais/efeitos dos fármacos , Proteína 2 de Ligação a Repetições Teloméricas/antagonistas & inibidores , Proteína 2 de Ligação a Repetições Teloméricas/genética
11.
Development ; 140(10): 2149-59, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23578927

RESUMO

The anaphase inhibitor securin plays a crucial role in regulating the timing of sister chromatid separation during mitosis. When sister chromatid pairs become bioriented, the E3 ligase anaphase promoting complex/cyclosome (APC/C) ubiquitylates securin for proteolysis, triggering sister chromatid separation. Securin is also implicated in regulating meiotic progression. Securin protein levels change sharply during cell cycle progression, enabling its timely action. To understand the mechanism underlying the tightly regulated dynamics of securin, we analyzed the subcellular localization of the securin IFY-1 during C. elegans development. IFY-1 was highly expressed in the cytoplasm of germ cells. The cytoplasmic level of IFY-1 declined immediately following meiosis I division and remained low during meiosis II and following mitoses. We identified a C. elegans homolog of another type of E3 ligase, UBE3C, designated ETC-1, as a regulator of the cytoplasmic IFY-1 level. RNAi-mediated depletion of ETC-1 stabilized IFY-1 and CYB-1 (cyclin B1) in post-meiosis I embryos. ETC-1 knockdown in a reduced APC function background caused an embryonic lethal phenotype. In vitro, ETC-1 ubiquitylates IFY-1 and CYB-1 in the presence of the E2 enzyme UBC-18, which functions in pharyngeal development. Genetic analysis revealed that UBC-18 plays a distinct role together with ETC-1 in regulating the cytoplasmic level of IFY-1 during meiosis. Our study reports a novel mechanism, mediated by ETC-1, that co-operates with APC/C to maintain the meiotic arrest required for proper cell cycle timing during reproduction.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/citologia , Proteínas de Transporte/metabolismo , Ciclina B1/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Meiose/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Alelos , Anáfase , Animais , Proteínas de Caenorhabditis elegans/química , Proteínas de Transporte/química , Proteínas de Transporte/fisiologia , Citoplasma/metabolismo , Imunoprecipitação , Espectrometria de Massas , Mitose , Interferência de RNA , Ubiquitina/metabolismo
12.
EMBO Rep ; 13(1): 52-9, 2011 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-22157895

RESUMO

Replicative senescence is accompanied by a telomere-specific DNA damage response (DDR). We found that DDR+ telomeres occur spontaneously in early-passage normal human cells and increase in number with increasing cumulative cell divisions. DDR+ telomeres at replicative senescence retain TRF2 and RAP1 proteins, are not associated with end-to-end fusions and mostly result from strand-independent, postreplicative dysfunction. On the basis of the calculated number of DDR+ telomeres in G1-phase cells just before senescence and after bypassing senescence by inactivation of wild-type p53 function, we conclude that the accrual of five telomeres in G1 that are DDR+ but nonfusogenic is associated with p53-dependent senescence.


Assuntos
Senescência Celular/genética , Telômero/metabolismo , Células Cultivadas , Dano ao DNA , Humanos , Homeostase do Telômero
13.
Nucleic Acids Res ; 39(18): 8078-91, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21724611

RESUMO

MicroRNAs (miRNAs) are a class of noncoding small RNAs that act as negative regulators of gene expression. To identify miRNAs that may regulate human cell immortalization and carcinogenesis, we performed comparative miRNA array profiling of human normal and SV40-T antigen immortalized cells. We found that miR-296 was upregulated in immortalized cells that also had activation of telomerase. By an independent experiment on genomic analysis of cancer cells we found that chromosome region (20q13.32), where miR-296 is located, was amplified in 28/36 cell lines, and most of these showed enriched miR-296 expression. Overexpression of miR-296 in human cancer cells, with and without telomerase activity, had no effect on their telomerase function. Instead, it suppressed p53 function that is frequently downregulated during human cell immortalization and carcinogenesis. By monitoring the activity of a luciferase reporter connected to p53 and p21(WAF1) (p21) untranslated regions (UTRs), we demonstrate that miR-296 interacts with the p21-3'UTR, and the Hu binding site of p21-3'UTR was identified as a potential miR-296 target site. We demonstrate for the first time that miR-296 is frequently upregulated during immortalization of human cells and contributes to carcinogenesis by downregulation of p53-p21(WAF1) pathway.


Assuntos
Regiões 3' não Traduzidas , Inibidor de Quinase Dependente de Ciclina p21/genética , Regulação para Baixo , MicroRNAs/metabolismo , Neoplasias/genética , Linhagem Celular , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Inibidor de Quinase Dependente de Ciclina p21/antagonistas & inibidores , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Humanos , RNA Mensageiro/metabolismo , Proteína Supressora de Tumor p53/antagonistas & inibidores , Regulação para Cima
14.
J Tissue Eng Regen Med ; 5(6): 437-43, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20799242

RESUMO

The purpose of this study was to track mesenchymal stem cells (MSCs) labelled with internalizing quantum dots (i-QDs) in the reparative tissues, following the allogeneic transplantation of three-dimensional (3D) cartilaginous aggregates into the osteochondral defects of rabbits. QDs were conjugated with a unique internalizing antibody against a heat shock protein-70 (hsp70) family stress chaperone, mortalin, which is upregulated and expressed on the surface of dividing cells. The i-QDs were added to the culture medium for 24 h. Scaffold-free cartilaginous aggregates formed from i-QD-labelled MSCs (i-MSCs), using a 3D culture system with chondrogenic supplements for 1 week, were transplanted into osteochondral defects of rabbits. At 4, 8 and 26 weeks after the transplantation, the reparative tissues were evaluated macroscopically, histologically and fluoroscopically. At as early as 4 weeks, the defects were covered with a white tissue resembling articular cartilage. In histological appearance, the reparative tissues resembled hyaline cartilage on safranin-O staining throughout the 26 weeks. In the deeper portion, subchondral bone and bone marrow were well remodelled. On fluoroscopic evaluation, QDs were tracked mainly in bone marrow stromata, with some signals detected in cartilage and the subchondral bone layer. We showed that the labelling of rabbit MSCs with anti-mortalin antibody-conjugated i-QDs is a tolerable procedure and provides a stable fluorescence signal during the cartilage repair process for up to 26 weeks after transplantation. The results suggest that i-MSCs did not inhibit, and indeed contributed to, the regeneration of osteochondral defects.


Assuntos
Células da Medula Óssea/citologia , Osso e Ossos/patologia , Cartilagem Articular/citologia , Condrócitos/patologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Animais , Células da Medula Óssea/metabolismo , Osso e Ossos/metabolismo , Condrócitos/metabolismo , Imuno-Histoquímica , Células-Tronco Mesenquimais/metabolismo , Microscopia de Fluorescência , Pontos Quânticos , Coelhos , Transplante Homólogo
15.
Nat Struct Mol Biol ; 16(12): 1244-51, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19935685

RESUMO

Telomere dysfunction is typically studied under conditions in which a component of the six-subunit shelterin complex that protects chromosome ends is disrupted. The nature of spontaneous telomere dysfunction is less well understood. Here we report that immortalized human cell lines lacking wild-type p53 function spontaneously show many telomeres with a DNA damage response (DDR), commonly affecting only one sister chromatid and not associated with increased chromosome end-joining. DDR(+) telomeres represent an intermediate configuration between the fully capped and uncapped (fusogenic) states. In telomerase activity-positive (TA(+)) cells, DDR is associated with low TA and short telomeres. In cells using the alternative lengthening of telomeres mechanism (ALT(+)), DDR is partly independent of telomere length, mostly affects leading strand-replicated telomeres, and can be partly suppressed by TRF2 overexpression. In ALT(+) (but not TA(+)) cells, DDR(+) telomeres preferentially associate with large foci of extrachromosomal telomeric DNA and recombination proteins. DDR(+) telomeres therefore arise through different mechanisms in TA(+) and ALT(+) cells and have different consequences.


Assuntos
Cromossomos Humanos/metabolismo , Dano ao DNA , Telômero/metabolismo , Linhagem Celular , Humanos , Modelos Biológicos , Recombinases/metabolismo , Telomerase/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo
16.
Hum Gene Ther ; 20(3): 217-24, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19257853

RESUMO

Progress in stem cell research has prioritized the refinement of cell-labeling techniques for in vitro and in vivo basic and therapeutic studies. Although quantum dots, because of their optical properties, are emerging as favorable nanoparticles for bioimaging, substantial refinements or modifications that would improve their biocompatibility are still required. We report here that internalizing quantum dots (i-QDs) generated by their conjugation with an internalizing antibody against a heat shock protein-70 family stress chaperone, mortalin, offered an efficient, genetically noninvasive, nontoxic, and functionally inert way to label mesenchymal stem cells (MSCs). The i-QD-labeled MSCs underwent normal adipocyte, osteocyte, and chondrocyte differentiation in vitro and in vivo, suggesting the potential application of i-QDs in in vivo diagnostics, regenerative and therapeutic medicine.


Assuntos
Células-Tronco Mesenquimais/citologia , Pontos Quânticos , Coloração e Rotulagem/métodos , Adipogenia , Animais , Técnicas de Cultura de Células , Condrogênese , Proteínas de Choque Térmico HSP70/imunologia , Haplorrinos , Humanos , Imuno-Histoquímica , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/química , Células-Tronco Mesenquimais/metabolismo , Osteogênese , Coelhos , Ratos
17.
J Biol Chem ; 284(3): 1664-72, 2009 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-19001376

RESUMO

The tumor suppressor protein, p53, is central to the pathways that monitor the stress, DNA damage repair, cell cycle, aging, and cancer. Highly complex p53 networks involving its upstream sensors and regulators, downstream effectors and regulatory feedback loops have been identified. CARF (Collaborator of ARF) was shown to enhance ARF-dependent and -independent wild-type p53 function. Here we report that (i) CARF overexpression causes premature senescence of human fibroblasts, (ii) it is vital for replicative and stress-induced senescence, and (iii) the lack of CARF function causes aneuploidy and apoptosis. We provide evidence that CARF plays a dual role in regulating p53-mediated senescence and apoptosis, the two major tumor suppressor mechanisms.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose/fisiologia , Senescência Celular/fisiologia , Fibroblastos/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Dano ao DNA/fisiologia , Reparo do DNA/fisiologia , Fibroblastos/citologia , Células HeLa , Humanos , Proteína Supressora de Tumor p53/genética
18.
Biogerontology ; 9(4): 269-82, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18344010

RESUMO

Aging is associated with accumulation of toxic intracellular and extracellular protein aggregates. Cells manage "aged" proteins by mobilizing their molecular chaperones or heat shock proteins that are also considered as determinants of lifespan in diverse species. In this study, we tested whether an exogenous addition of the non-toxic chemical chaperone 'glycerol' could elicit stress and geronto-protective activities. We found that glycerol enhanced chaperoning of heat-denatured proteins. In addition to stimulating proteasome activity, glycerol led to an increased expression of the stress chaperone 'mortalin' and decreased p53 function in human cells. Glycerol-fed worms exhibited thermo-tolerance and lower level of age-associated auto-fluorescence. Through the combined stimulation of the proteasome and chaperoning activities of mortalin, in particular, glycerol treatment resulted in increased survival and fitness against oxidative- and heat-stress. These results may have significant implications in the use of glycerol as a candidate geronto-modulator in development of practical interventions for "healthy aging".


Assuntos
Caenorhabditis elegans/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Glicerol/farmacologia , Temperatura Alta , Chaperonas Moleculares/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Caenorhabditis elegans/enzimologia , Caenorhabditis elegans/crescimento & desenvolvimento , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Estabilidade Enzimática , Fibroblastos/enzimologia , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Longevidade/efeitos dos fármacos , Desnaturação Proteica , Dobramento de Proteína , Proteína Supressora de Tumor p53/metabolismo
19.
Hum Gene Ther ; 18(11): 1153-60, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17937579

RESUMO

Gene therapy, a potential solution to hereditary and nonhereditary diseases, faces the challenges of safe and specific gene delivery. Cationic carrier molecules (e.g., liposome and polymers) that form noncovalent complexes with negatively charged DNA have been in use as nonviral gene delivery vectors. Although they tend to be relatively less efficient than viral systems, they have inherent advantages of flexibility and safety. Their derivatives, in conjugation with functional molecules such as peptides, proteins, growth factors, and antibodies, have been focused on to generate nanocarriers with low toxicity, high stability, high efficiency, and cell-specific targeting features. Here we describe internalizing polyclonal and monoclonal antibodies against a stress chaperone, mortalin/mtHsp70. We demonstrate that these internalizing anti-mortalin antibodies (i-mot Ab) could be employed for (1) internalization of nanoparticles (quantum dots, Qdots) and the generation of illuminating cells and (2) gene delivery. By using cancer and normal human cells in parallel, we further demonstrate that gene delivery can be specifically enhanced in human cancer cells if cationic polymer polyethylenimine (PEI) and i-mot Ab complex are used and may provide a novel cancer-targeting nanocarrier.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Marcação de Genes/métodos , Técnicas de Transferência de Genes , Proteínas de Choque Térmico HSP70/imunologia , Neoplasias/terapia , Polietilenoimina , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/farmacocinética , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Neoplasias/imunologia , Neoplasias/metabolismo
20.
Ann N Y Acad Sci ; 1100: 368-72, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17460200

RESUMO

Quantum dots (QDs) are fluorescent nanocrystals that are emerging as fine alternatives to the conventional organic dyes. They have several advantages including greater photostability and a wider range of excitation-emission wavelengths. By using mortalin staining as a model, we initially demonstrated that the QDs are more stable and provide better resolution in protein imaging in fixed cells. With the help of an internalizing antibody, we generated internalizing QD (i-QD) and demonstrated its inertness to cell replication, structure, and viability. Based on the superior resolution, stability and inertness, we propose the use of QD staining of mortalin as a cell-based visual assay to screen for senescence-inducing drugs, proteins, and siRNAs.


Assuntos
Senescência Celular , Ensaios de Seleção de Medicamentos Antitumorais , Proteínas de Choque Térmico HSP70/biossíntese , Microscopia de Fluorescência/métodos , Neoplasias/metabolismo , Neoplasias/patologia , Pontos Quânticos , Antineoplásicos/farmacologia , Transporte Biológico , Linhagem Celular Transformada , Linhagem Celular Tumoral , Sobrevivência Celular , Corantes Fluorescentes/farmacologia , Humanos , Concentração Inibidora 50 , RNA Interferente Pequeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...