Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Odontology ; 112(1): 91-99, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37166745

RESUMO

Periodontal ligament (PDL) cells play an important role in mechanosensing and secretion of signaling molecules during bone remodeling. However, the regulatory mechanism is unknown. The aim of the present study is to investigate the expression pattern of periostin and sclerostin in response to orthodontic forces in periodontal ligament cells in vitro. PDL cells were isolated from extracted teeth and treated with compressive forces of 25 gr/cm2 or equiaxial tension forces at frequency 1 Hz for 0, 24, 48, and 72 h. qRT-PCR was applied to evaluate the gene expressions. The secretion of sclerostin and periostin was assessed using ELISA. DAPI staining was used to evaluate apoptosis. The expression of sclerostin elevated significantly at protein and gene levels under compression forces after 24 h, while the application of tensile forces induced the expression of periostin and its upstream regulator RUNX2 (p < 0.05). Gene expression up-regulation was significant for POSTN and RUNX2 after 48 and 72 h tensile forces. Also, the gene expression of sclerostin reduced in a time-dependent manner after application of tensile force. The compression forces enhanced apoptosis to 7.5 ± 3.5% and induced gene expression of apoptotic markers of CASP9, and BCL2 within 72 h of exposure. Periostin and sclerostin play an important role in orthodontic loads and their expressions are affected oppositely by compressive and tensile forces that might be suggested as a biomarker for assessment of bone remodeling during orthodontic treatment.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core , Ligamento Periodontal , Humanos , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Remodelação Óssea , Biomarcadores , Pressão , Estresse Mecânico , Técnicas de Movimentação Dentária , Células Cultivadas , Moléculas de Adesão Celular/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo
2.
Lasers Med Sci ; 39(1): 9, 2023 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-38110767

RESUMO

An in vitro study was designed to evaluate the effects of photobiomodulation (PBM) with 915-nm diode laser on human gingival fibroblast (HGF) cells under hyperglycemic condition. The HGF cells were cultured in Dulbecco's modified eagle medium (DMEM) medium containing 30 mM glucose concentration for 48 h to mimic the hyperglycemic condition. Subsequently, the cells received three sessions of PBM (915 nm, continuous emission mode, 200 mW, energy density values of 3.2, 6, and 9.2 J/cm2). Twenty-four hours post-irradiation, cell proliferation, expression of interleukin 6 (IL-6), and vascular endothelial growth factor (VEGF) were assessed with MTT and real-time polymerase chain reaction (PCR) tests, respectively. Also, reactive oxygen species (ROS) production was measured using CM-H2DCFDA fluorimetry. No changes were detected in the cell proliferation rate between the high glucose control group and laser-treated cells, while VEGF and IL-6 gene expression levels increased significantly after PBM in the high glucose-treated cells group. ROS level was significantly decreased in the irradiated cells in high-glucose medium compared with the high glucose control group. Our study revealed the inductive role of 915-nm-mediated PBM on VEGF and the inflammatory response while concurrently reducing reactive oxygen species production in HGF cells in hyperglycemic conditions.


Assuntos
Interleucina-6 , Terapia com Luz de Baixa Intensidade , Humanos , Interleucina-6/genética , Fator A de Crescimento do Endotélio Vascular/genética , Espécies Reativas de Oxigênio/metabolismo , Glicemia , Fibroblastos/efeitos da radiação , Células Cultivadas
3.
Artigo em Inglês | MEDLINE | ID: mdl-35649677

RESUMO

Graphene oxide (GO), an engineered nanomaterial, has a two-dimensional structure with carbon atoms arranged in a hexagonal array. While it has been widely used in many industries, such as biomedicine, electronics, and biosensors, there are still concerns over its safety. Recently, many studies have focused on the potential toxicity of GO. Epigenetic toxicity is an important aspect of a material's toxicological profile, since changes in gene expression have been associated with carcinogenicity and disease progression. In this review, we focus on the epigenetic alterations caused by GO, including DNA methylation, histone modification, and altered expression of non-coding RNAs. GO can affect DNA methyltransferase activity and disrupt the methylation of cytosine bases in DNA strands, leading to alteration of genome expression. Modulation of histones by GO, targeting histone deacetylase and demethylase, as well as dysregulation of miRNA and lncRNA expression have been reported. Further studies are required to determine the mechanisms of GO-induced epigenetic alterations.


Assuntos
Epigênese Genética , Grafite , Metilação de DNA , Grafite/toxicidade , Histonas/genética , Histonas/metabolismo
4.
Arch Oral Biol ; 133: 105296, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34735927

RESUMO

OBJECTIVE: The present in vitro study aims to investigate the potential use of epigenetic inhibitors as treatment modalities in tongue squamous cell carcinoma. DESIGN: The human tongue squamous cell carcinoma cell line (CAL-27) was cultured and exposed to varying concentrations of 5-Azacitidine (5-Aza) or Trichostatin A (TSA) in the culture medium. The cell apoptosis was evaluated using Annexin V/PI by flow cytometry. To evaluate DNA damage response, γH2AX foci analysis was performed using immunofluorescence. Single cell gel electrophoresis (SCGE) was applied to measure DNA strand breaks. Gene expression was assessed by quantitative real-time PCR. RESULTS: The results showed that 5-Aza and TSA had apoptotic effects on the SCC cell line at concentrations of 50-200 µM and 0.5-5 µM, respectively. Immunofluorescence analysis showed increased expression of γH2AX, the marker of DNA damage response after treatment of 5-Aza and TSA that was associated with increased DNA strand breaks. The expressions of urokinase plasminogen activator, its receptor and matrix metalloproteinase-2, were significantly reduced in TSA- and 5-Aza-treated cells. CONCLUSIONS: Our results showed that 5-Aza and TSA increase apoptotic and DNA damage response in squamous cell carcinoma cell line while reducing the expression of tumor invasion genes that further indicating the potential therapeutic value of two epigenetic modifiers in squamous cell carcinoma.


Assuntos
Carcinoma de Células Escamosas , Neoplasias da Língua , Azacitidina/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/genética , Dano ao DNA , Decitabina , Inibidores de Histona Desacetilases/farmacologia , Humanos , Ácidos Hidroxâmicos , Metaloproteinase 2 da Matriz , Língua , Neoplasias da Língua/tratamento farmacológico , Neoplasias da Língua/genética
5.
Iran J Pharm Res ; 20(4): 165-177, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35194437

RESUMO

Exposure to certain environmental toxins has been shown to be associated with cellular senescence mainly through induction of oxidative stress and impact on cellular systems. Acrylamide (ACR) has raised worldwide concerns regarding the high risk of human dietary exposure to its hazardous effect. Although there is ample evidence about the carcinogenicity of ACR, limited studies have focused on its impact on cellular aging. The levels of ß-galactosidase (SA-ß-gal) activity, cell cycle distribution, and the expression of the senescence-associated gene and inflammatory markers were evaluated following exposure of embryonic fibroblast cells to ACR. A significant elevation in SA-ß-gal activity after exposure to different concentrations of ACR was accompanied by a considerably increased level of reactive oxygen species and lipid peroxidation. ACR-treated cells showed a noticeable decline in the total antioxidant capacity and thiol molecules. Moreover, the expression of cellular senescence-related genes including p38, p53, and p21 significantly upregulated at the high concentration of 5 mM ACR. ACR also induced G0/G1 phase arrest in embryonic fibroblast cells. The current study results revealed that exposure to ACR could enhance senescence response, contributing to increased oxidative stress and cellular damage.

6.
Curr Mol Pharmacol ; 14(1): 60-67, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32394848

RESUMO

BACKGROUND: Head and neck squamous cell carcinoma (HNSCC) is one of the most fatal malignancies worldwide and despite using various therapeutic strategies for the treatment of HNSCC, the surveillance rate is low. Telomerase has been remarked as the primary target in cancer therapy. Considering the key regulatory role of epigenetic mechanisms in controlling genome expression, the present study aimed to investigate the effects of two epigenetic modulators, a DNA methylation inhibitor and a histone deacetylase inhibitor on cell migration, proliferation, hTERT gene expression, and telomerase activity in HNSCC cell lines. METHODS: Human HNSCC cell lines were treated with Azacitidine and Trichostatin A to investigate their effects on telomerase gene expression and activity. Cell viability, migration, hTERT gene expression, and telomerase activity were studied using MTT colorimetric assay, scratch wound assay, qRT-PCR, and TRAP assay, respectively. RESULTS: Azacitidine at concentrations of ≤1µM and Trichostatin A at 0.1 to 0.3nM concentrations significantly decreased FaDu and Cal-27 cells migration. The results showed that Azacitidine significantly decreased hTERT gene expression and telomerase activity in FaDu and Cal-27 cell lines. However, there were no significant changes in hTERT gene expression at different concentrations of Trichostatin A in both cell lines. Trichostatin A treatment affected telomerase activity at the high dose of 0.3 nM Trichostatin A. CONCLUSION: The findings revealed that unlike histone deacetylase inhibitor, Azacitidine as an inhibitor of DNA methylation decreases telomerase expression in HNSCC cells. This might suggest the potential role of DNA methyltransferase inhibitors in telomerase-based therapeutic approaches in squamous cell carcinoma.


Assuntos
Antineoplásicos/química , Azacitidina/química , Metilases de Modificação do DNA/antagonistas & inibidores , Inibidores Enzimáticos/química , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Inibidores de Histona Desacetilases/química , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico , Telomerase/metabolismo , Antineoplásicos/farmacologia , Apoptose , Azacitidina/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular , Metilação de DNA , Desenvolvimento de Medicamentos , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Epigênese Genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Histonas/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Fragmentos de Peptídeos/genética , Transdução de Sinais
7.
Asian Pac J Cancer Prev ; 20(6): 1649-1654, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-31244283

RESUMO

Objective: Oral squamous cell carcinoma (OSCC) accounts for over 90% of oral neoplasms. Finding molecular markers for predicting prognosis is a high priority. The core transcription factors, OCT4, SOX2, and NANOG that regulate embryonic stem cell pluripotency have been implicated in progression of various malignancies. The predictive value of these markers and their role in the development of OSCC is still controversial. In this study, we therefore evaluated their expression in OSCCs and adjacent non-tumor tissue. Methods: A total of 60 frozen tumor and adjacent non-tumor tissue samples from 30 patients with OSCC were examined using quantitative reverse transcription polymerase chain reaction (qRT-PCR). Clinical and pathological data of patients including tumor stage, lymph node metastasis and tumor grade were also recorded. Results: Expression of SOX2 was significantly higher in adjacent non-tumor as compared to tumor tissue (P=0.04). No statistically significant differences were found for expression of OCT4 (P=0.50) and NANOG (P=0.68). Also, there was no significant association between expression of OCT4, SOX2, and NANOG and clinical or pathological data (P>0.05), although slightly higher values were noted in patients without lymph node metastasis. Conclusion: Based on the present data, decreased expression of SOX2 is correlated with carcinogenesis in the oral cavity and development of OSCC.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/metabolismo , Neoplasias Bucais/metabolismo , Proteína Homeobox Nanog/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Biomarcadores Tumorais/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Estudos de Casos e Controles , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias Bucais/genética , Neoplasias Bucais/patologia , Proteína Homeobox Nanog/genética , Fator 3 de Transcrição de Octâmero/genética , Prognóstico , Fatores de Transcrição SOXB1/genética
8.
Environ Toxicol Pharmacol ; 64: 26-40, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30290328

RESUMO

Poisoning with aluminum phosphide (AlP) has been attributed to the high rate of mortality among many Asian countries. It affects several organs, mainly heart and kidney. Numerous literature demonstrated the valuable effect of minocycline in mitigating pathological symptoms of heart and kidney disease. The aim of the present study was to evaluate the probable protective effect of minocycline on cardiac hemodynamic parameters abnormalities and renal toxicity induced by AlP-poisoning in the rat model. AlP was administered by gavage at 12 mg/kg body weight followed by injection of minocycline for two interval times of 12 and 24 h, at 40, 80, 120 mg/kg body weight. Electrocardiographic (ECG) parameters were monitored, 30 min after AlP gavage for 6 h using an electronic cardiovascular monitoring device. Kidney tissue and serum were collected for the study of histology, mitochondrial complexes I, II, IV, lactate dehydrogenase (LDH) and myeloperoxidase (MPO) activity, ADP/ATP ratio, mitochondrial cytochrome c release, apoptosis, lactate, BUN, and Cr levels. The results demonstrated that AlP induces ECG abnormalities, and failure of heart rate and blood pressure, which improved significantly by minocycline. Minocycline treatment significantly improved complexes I, IV, MPO and LDH activities, and also reduced the ADP/ATP ratio, lactate level, release of cytochrome c, and apoptosis in the kidney following AlP-poisoning. Also, the histological results showed an improvement of kidney injury in minocycline treated groups. In conclusion, the findings of this study showed that minocycline could improve cardiac hemodynamic abnormalities and kidney injury following AlP-poisoning, suggesting minocycline might be a possible candidate for the treatment of AlP-poisoning.


Assuntos
Injúria Renal Aguda/tratamento farmacológico , Compostos de Alumínio/toxicidade , Frequência Cardíaca/efeitos dos fármacos , Minociclina/uso terapêutico , Fosfinas/toxicidade , Substâncias Protetoras/uso terapêutico , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Animais , Apoptose/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Citocromos c/metabolismo , Eletrocardiografia/efeitos dos fármacos , Coração/efeitos dos fármacos , Coração/fisiologia , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , L-Lactato Desidrogenase/metabolismo , Ácido Láctico/metabolismo , Masculino , Ratos Wistar
9.
Arch Toxicol ; 91(7): 2577-2597, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28516248

RESUMO

Epigenotoxicology is an emerging field of study that investigates the non-genotoxic epigenetic effects of environmental toxicants resulting in alteration of normal gene expression and disruption of cell function. Recent findings on the role of toxicant-induced epigenetic modifications in the development of degenerative diseases have opened up a promising research direction to explore epigenetic therapy approaches and related prognostic biomarkers. In this review, we presented comprehensive data on epigenetic alterations identified in various diseases, including cancer, autoimmune disorders, pulmonary conditions as well as cardiovascular, gastrointestinal and bone disease. Although data on abnormalities of DNA methylation and their role in the development of diseases are abundant, less is known about the impact of histone modifications and microRNA expressions. Further, we discussed the effects of selected common environmental toxicants on epigenetic modifications and their association with particular abnormalities. A number of different environmental toxicants have been identified for their role in aberrant DNA methylation, histone modifications, and microRNA expression. Such epigenetic effects were shown to be tissue-type specific and highly associated with the level and duration of exposure. Finally, we described present and future therapeutic strategies, including medicines and dietary compounds for combating the toxicant-induced epigenetic alterations. There are currently seven histone deacetylase inhibitors and two DNA methyltransferase inhibitors approved for clinical use and many other promising candidates are in preclinical and clinical testing. Dietary compounds are thought to be the effective and safe strategies for treating and prevention of epigenetic pathophysiological conditions. Still more concentrated epigenetic researches are required for evaluation of chemical toxicity and identifying the causal association between key epigenetic alteration and disease.


Assuntos
Exposição Ambiental/efeitos adversos , Poluentes Ambientais/toxicidade , Epigênese Genética/efeitos dos fármacos , Doenças Autoimunes/epidemiologia , Doenças Autoimunes/genética , Doenças Cardiovasculares/epidemiologia , Doenças Cardiovasculares/genética , Metilação de DNA/efeitos dos fármacos , Dieta , Inibidores de Histona Desacetilases/farmacologia , Histonas/genética , Histonas/metabolismo , Humanos , Neoplasias/epidemiologia , Neoplasias/genética
10.
In Vitro Cell Dev Biol Anim ; 52(10): 1060-1071, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27503516

RESUMO

This is the first report on differentiation of mouse amniotic membrane mesenchymal stem cells (AM-MSCs) into male germ cells (GCs). AM-MSCs have the multipotent differentiation capacity and can be differentiated into various cell types. In the present study, AM-MSCs were induced for differentiation into GCs. AM-MSCs were isolated from mouse embryonic membrane by enzymatic digestion. AM-MSCs were characterized with osteogenic and adipogenic differentiation test and flow cytometric analysis of some CD-markers. AM-MSCs were induced to differentiate into GCs using a creative two-step method. Passage-3 AM-MSCs were firstly treated with 25 ng/ml bone morphogenetic protein 4 (BMP4) for 5 d and in continuing with 1 µM retinoic acid (RA) for 12 d (total treatment time was 17 d). At the end of the treatment period, real-time reverse transcription (RT)-PCR was performed to evaluate the expression of GC-specific markers-Itgb1, Dazl, Stra8, Piwil2, Mvh, Oct4, and c-Kit- in the cells. Moreover, flow cytometry and immunofluorescence staining were performed to evaluate the expression of Mvh and Dazl at protein level. Real-time RT-PCR showed that most of the tested markers were upregulated in the treated AM-MSCs. Furthermore, flow cytometric and immunofluorescence analyses both revealed that a considerable part of the treated cells expressed GC-specific markers. The percentage of positive cells for Mvh and Dazl was about 23 and 46%, respectively. Our results indicated that a number of AM-MSCs successfully differentiated into the GCs. Finally, it seems that AM-MSCs would be a potential source of adult pluripotent stem cells for in vitro generation of GCs and cell-based therapies for treatment of infertility.


Assuntos
Âmnio/citologia , Diferenciação Celular , Células Germinativas/citologia , Células-Tronco Mesenquimais/citologia , Animais , Biomarcadores/metabolismo , Separação Celular , Forma Celular , Células Cultivadas , Feminino , Citometria de Fluxo , Células Germinativas/metabolismo , Imuno-Histoquímica , Células-Tronco Mesenquimais/metabolismo , Camundongos , Reação em Cadeia da Polimerase em Tempo Real
11.
Food Chem Toxicol ; 86: 298-308, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26582272

RESUMO

Stem cells are an important target for environmental toxicants. As they are the main source for replenishing of organs in the body, any changes in their normal function could affect the regenerative potential of organs, leading to the appearance of age-related disease and acceleration of the aging process. Environmental toxicants could exert their adverse effect on stem cell function via multiple cellular and molecular mechanisms, resulting in changes in the stem cell differentiation fate and cell transformation, and reduced self-renewal capacity, as well as induction of stress-induced cellular senescence. The present review focuses on the effect of environmental toxicants on stem cell function associated with the aging process. We categorized environmental toxicants according to their preferred molecular mechanism of action on stem cells, including changes in genomic, epigenomic, and proteomic levels and enhancing oxidative stress. Pesticides, tobacco smoke, radiation and heavy metals are well-studied toxicants that cause stem cell dysfunction via induction of oxidative stress. Transgenerational epigenetic changes are the most important effects of a variety of toxicants on germ cells and embryos that are heritable and could affect health in the next several generations. A better understanding of the underlying mechanisms of toxicant-induced stem cell aging will help us to develop therapeutic intervention strategies against environmental aging. Meanwhile, more efforts are required to find the direct in vivo relationship between adverse effect of environmental toxicants and stem cell aging, leading to organismal aging.


Assuntos
Envelhecimento/efeitos dos fármacos , Envelhecimento/fisiologia , Poluentes Ambientais/toxicidade , Células-Tronco/efeitos dos fármacos , Células-Tronco/fisiologia , Animais
12.
PLoS One ; 9(7): e101529, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24987841

RESUMO

DNA damage induced by numerous exogenous or endogenous factors may have irreversible consequences on the cell leading to cell cycle arrest, senescence and cell death. The DNA damage response (DDR) is powerful signaling machinery triggered in response to DNA damage, to provide DNA damage recognition, signaling and repair. Most anticancer drugs induce DNA damage, and DNA repair in turn attenuates therapeutic efficiency of those drugs. Approaches delaying DNA repair are often used to increase efficiency of treatment. Recent data show that ubiquitin-proteasome system is essential for signaling and repair of DNA damage. However, mechanisms providing regulation of proteasome intracellular localization, activity, and recruitment to DNA damage sites are elusive. Even less investigated are the roles of extranuclear signaling proteins in these processes. In this study, we report the involvement of the serine protease urokinase-type plasminogen activator receptor (uPAR) in DDR-associated regulation of proteasome. We show that in vascular smooth muscle cells (VSMC) uPAR activates DNA single strand break repair signaling pathway. We provide evidence that uPAR is essential for functional assembly of the 26S proteasome. We further demonstrate that uPAR mediates DNA damage-induced phosphorylation, nuclear import, and recruitment of the regulatory subunit PSMD6 to proteasome. We found that deficiency of uPAR and PSMD6 delays DNA repair and leads to decreased cell survival. These data may offer new therapeutic approaches for diseases such as cancer, cardiovascular and neurodegenerative disorders.


Assuntos
Quebras de DNA de Cadeia Simples , Reparo do DNA , Músculo Liso Vascular/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Transporte Ativo do Núcleo Celular , Animais , Linhagem Celular , Sobrevivência Celular , Células Cultivadas , Deleção de Genes , Humanos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/citologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo
13.
J Vasc Res ; 50(2): 109-23, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23172421

RESUMO

The anthracycline doxorubicin is a widely used effective anti-cancer drug. However, its application and dosage are severely limited due to its cardiotoxicity. The exact mechanisms of doxorubicin-induced cardiotoxic side effects remain poorly understood. Even less is known about the impact of doxorubicin treatment on vascular damage. We found that low doses of doxorubicin induced a senescent response in human primary vascular smooth muscle cells (VSMC). We observed that expression of urokinase receptor (uPAR) was upregulated in response to doxorubicin. Furthermore, the level of uPAR expression played a decisive role in developing doxorubicin-induced senescence. uPAR silencing in human VSMC by means of RNA interference as well as uPAR knockout in mouse VSMC resulted in abrogation of doxorubicin-induced cellular senescence. On the contrary, uPAR overexpression promoted VSMC senescence. We further found that proteasomal degradation of telomeric repeat binding factor 2 (TRF2) mediates doxorubicin-induced VSMC senescence. Our results demonstrate that uPAR controls the ubiquitin-proteasome system in VSMC and regulates doxorubicin-induced TRF2 ubiquitination and proteasomal degradation via this mechanism. Therefore, VSMC senescence induced by low doses of doxorubicin may contribute to vascular damage upon doxorubicin treatment. uPAR-mediated TRF2 ubiquitination and proteasomal degradation are further identified as a molecular mechanism underlying this process.


Assuntos
Senescência Celular/efeitos dos fármacos , Doxorrubicina/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/fisiologia , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Animais , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Doxorrubicina/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Miócitos de Músculo Liso/metabolismo , Cultura Primária de Células , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/deficiência , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Proteínas Recombinantes de Fusão/fisiologia , Telômero/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/antagonistas & inibidores , Proteína 2 de Ligação a Repetições Teloméricas/genética , Transfecção , Ubiquitinação/efeitos dos fármacos , Artérias Umbilicais/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA