Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Clin Pharmacol ; 64(1): 125-136, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37656820

RESUMO

The immunogenicity of cemiplimab, a fully human immunoglobulin G4 monoclonal antibody directed against programmed cell death 1, was assessed in patients across multiple tumor types. The development of antidrug antibodies (ADAs) against cemiplimab was monitored using a validated bridging immunoassay. To identify ADA-positive samples in the assay, statistically determined cut points were established by analyzing baseline clinical study samples from a mixed population of different tumor types, and this validation cut point was used to assess immunogenicity in all subsequent studies. Regulatory guidance requires that ADA assay cut points be verified for appropriateness in different patient populations. Thus, for the cemiplimab ADA assay, we evaluated whether each new oncology population was comparable with the validation population used to set the cut point. Assay responses from 2393 individual serum samples from 8 different tumor types were compared with the validation population, using established statistical methods for cut-point determination and comparison, with no significant differences observed. Across tumor types, the immunogenicity of cemiplimab was low, with an overall treatment-emergent ADA incidence rate of 1.9% and 2.5% at intravenous dose regimens of 3 mg/kg every 2 weeks and 350 mg every 3 weeks, respectively. Moreover, no neutralizing antibodies to cemiplimab were detected in patients with ADA-positive samples, and there was no observed impact of cemiplimab ADAs on pharmacokinetics. Study-specific cut points may be required in some diseases, such as immune and inflammatory diseases; however, based on this analysis, in-study cut points are not required for each new oncology disease indication for cemiplimab.


Assuntos
Anticorpos Monoclonais Humanizados , Neoplasias , Humanos , Incidência , Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Neoplasias/tratamento farmacológico
2.
AAPS J ; 24(4): 76, 2022 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-35725847

RESUMO

A cell-based assay was developed to detect neutralizing anti-drug antibodies (NAbs) against odronextamab, a CD20xCD3 bispecific monoclonal antibody (mAb) under investigation for treatment of CD20+ B cell malignancies. In this assay, odronextamab bridges between two cell types, CD20-expressing HEK293 cells and CD3-expressing Jurkat T cells that generate a luciferase signal upon CD3 clustering. Patient samples containing NAbs directed to either arm of the bispecific drug block the odronextamab bridge formation between the cell lines thus preventing the generation of the luciferase signal. We determined that other anti-CD20 therapeutics also block bridge formation, resulting in false-positive results. In patient samples from odronextamab clinical trials, approximately 30% of baseline samples had a strong false-positive NAb signal that correlated with the presence of prior rituximab (anti-CD20) therapy. We determined that rituximab interference can be minimized by the addition of anti-rituximab antibodies in the NAb assay. Understanding and mitigating the impact of prior biologic exposure is increasingly important for implementing a successful bioanalytical strategy to support clinical drug development, especially in the immuno-oncology field. Odronextamab neutralizing antibody assay, interference, and mitigation. A Design of the odronextamab neutralizing antibody (NAb) assay where anti-CD20xCD3 drug bridges between CD20-expressing HEK293 cells and Jurkat T cells expressing an NFAT response element and luciferase reporter. True NAb prevents odronextamab from bridging between target and effector cells, thus preventing the expression of luciferase. B Interference with odronextamab from other anti-CD20 therapeutic antibodies (e.g., rituximab) from prior disease treatment generates a false-positive NAb result. Assay interference can be mitigated with an anti-idiotypic antibody against the interfering therapy.


Assuntos
Anticorpos Biespecíficos , Antineoplásicos , Anticorpos Biespecíficos/farmacologia , Anticorpos Biespecíficos/uso terapêutico , Anticorpos Monoclonais , Anticorpos Neutralizantes , Antígenos CD20 , Células HEK293 , Humanos , Rituximab
3.
Bioanalysis ; 14(10): 627-692, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35578974

RESUMO

The 15th edition of the Workshop on Recent Issues in Bioanalysis (15th WRIB) was held on 27 September to 1 October 2021. Even with a last-minute move from in-person to virtual, an overwhelmingly high number of nearly 900 professionals representing pharma and biotech companies, contract research organizations (CROs), and multiple regulatory agencies still eagerly convened to actively discuss the most current topics of interest in bioanalysis. The 15th WRIB included three Main Workshops and seven Specialized Workshops that together spanned 1 week in order to allow exhaustive and thorough coverage of all major issues in bioanalysis, biomarkers, immunogenicity, gene therapy, cell therapy and vaccines. Moreover, in-depth workshops on biomarker assay development and validation (BAV) (focused on clarifying the confusion created by the increased use of the term "context of use" [COU]); mass spectrometry of proteins (therapeutic, biomarker and transgene); state-of-the-art cytometry innovation and validation; and critical reagent and positive control generation were the special features of the 15th edition. This 2021 White Paper encompasses recommendations emerging from the extensive discussions held during the workshop, and is aimed to provide the bioanalytical community with key information and practical solutions on topics and issues addressed, in an effort to enable advances in scientific excellence, improved quality and better regulatory compliance. Due to its length, the 2021 edition of this comprehensive White Paper has been divided into three parts for editorial reasons. This publication (Part 2) covers the recommendations on ISR for Biomarkers, Liquid Biopsies, Spectral Cytometry, Inhalation/Oral & Multispecific Biotherapeutics, Accuracy/LLOQ for Flow Cytometry. Part 1A (Endogenous Compounds, Small Molecules, Complex Methods, Regulated Mass Spec of Large Molecules, Small Molecule, PoC), Part 1B (Regulatory Agencies' Inputs on Bioanalysis, Biomarkers, Immunogenicity, Gene & Cell Therapy and Vaccine) and Part 3 (TAb/NAb, Viral Vector CDx, Shedding Assays; CRISPR/Cas9 & CAR-T Immunogenicity; PCR & Vaccine Assay Performance; ADA Assay Comparability & Cut Point Appropriateness) are published in volume 14 of Bioanalysis, issues 9 and 11 (2022), respectively.


Assuntos
Citometria de Fluxo , Biomarcadores/análise , Citometria de Fluxo/métodos , Humanos , Indicadores e Reagentes , Biópsia Líquida , Espectrometria de Massas
4.
Bioanalysis ; 13(23): 1751-1760, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34758642

RESUMO

Aim: IL-33 is a potential therapeutic target but commercially available assays for the quantitation of systemic IL-33 have poor reliability. Results: In commercial IL-33 kits, interference from endogenous binding partners (e.g., soluble ST2) causes under-quantitation. Mitigating this required acid dissociation and addition of the detection reagent simultaneously with the capture step. This enabled detection of total, reduced (active) levels of IL-33 in human serum (LLOQ 6.25 pg/ml). Conclusion: Acid treatment of serum samples dissociates IL-33 from endogenous binding partners, increasing soluble ST2 tolerance to >1000 ng/ml. The modified method was specific for reduced endogenous IL-33. Analysis of over 300 samples from individuals with and without asthma and with different smoking status revealed no difference in serum IL-33.


Assuntos
Proteína 1 Semelhante a Receptor de Interleucina-1/química , Interleucina-33/sangue , Asma/sangue , Asma/patologia , Humanos , Imunoensaio , Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Interleucina-33/química , Interleucina-33/genética , Interleucina-33/metabolismo , Limite de Detecção , Oxirredução , Ligação Proteica , Kit de Reagentes para Diagnóstico , Proteínas Recombinantes/análise , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Fumar
5.
AAPS J ; 23(6): 109, 2021 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-34608545

RESUMO

Monoclonal antibodies (mAbs) are a leading class of biotherapeutics. In oncology, patients often fail on early lines of biologic therapy to a specific target. Some patients may then enroll in a new clinical trial with a mAb specific for the same target. Therefore, immunoassays designed to quantify the current mAb therapy or assess immunogenicity to the drug may be susceptible to cross-reactivity or interference with residual prior biologics. The impact of two approved anti-PD-1 mAbs, pembrolizumab and nivolumab, was tested in several immunoassays for cemiplimab, another approved anti-PD-1 mAb. The methods included a target-capture drug concentration assay, a bridging anti-drug antibody (ADA) assay and a competitive ligand-binding neutralizing antibody (NAb) assay. We also tested bioanalytical strategies to mitigate cross-reactivity or interference in these assays from other anti-PD-1 biologics. Both pembrolizumab and nivolumab cross-reacted in the cemiplimab drug concentration assay. This was mitigated by addition of antibodies specific to pembrolizumab or nivolumab. ADA specific for pembrolizumab and nivolumab did not interfere in the cemiplimab ADA assay. However, pembrolizumab and nivolumab generated a false-positive response in a target-capture NAb assay. Our results demonstrate that similar exogenous pre-existing anti-PD-1 mAbs (biotherapeutics) such as pembrolizumab and nivolumab are detected and accurately quantified in the cemiplimab drug concentration assay. However, once steady state is achieved for the new therapy, prior biologics would likely not be detected. Cross-reactivity and interference in immunoassays from previous treatment with class-specific biotherapeutic(s) pose significant bioanalytical challenges, especially in immuno-oncology.


Assuntos
Anticorpos Monoclonais Humanizados/imunologia , Antineoplásicos Imunológicos/imunologia , Nivolumabe/imunologia , Anticorpos Monoclonais Humanizados/sangue , Anticorpos Neutralizantes/imunologia , Antineoplásicos Imunológicos/sangue , Ligação Competitiva , Reações Cruzadas , Humanos , Inibidores de Checkpoint Imunológico/sangue , Inibidores de Checkpoint Imunológico/imunologia , Imunoensaio/métodos , Nivolumabe/sangue
6.
Environ Health Perspect ; 120(6): 840-7, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22398240

RESUMO

BACKGROUND: The incidence of asbestos-induced human cancers is increasing worldwide, and considerable evidence suggests that reactive oxygen species (ROS) are important mediators of these diseases. Our previous studies suggested that mitochondria might be involved in the initiation of oxidative stress in asbestos-exposed mammalian cells. OBJECTIVE: We investigated whether mitochondria are a potential cytoplasmic target of asbestos using a mitochondrial DNA-depleted (ρ(0)) human small airway epithelial (SAE) cell model: ρ(0) SAE cells lack the capacity to produce mitochondrial ROS. METHODS: We examined nuclear DNA damage, micronuclei (MN), intracellular ROS production, and the expression of inflammation-related nuclear genes in both parental and ρ(0) SAE cells in response to asbestos treatment. RESULTS: Asbestos induced a dose-dependent increase in nuclear DNA oxidative damage and MN in SAE cells. Furthermore, there was a significant increase in intracellular oxidant production and activation of genes involved in nuclear factor κB and proinflammatory signaling pathways in SAE cells. In contrast, the effects of asbestos were minimal in ρ(0) SAE cells. CONCLUSIONS: Mitochondria are a major cytoplasmic target of asbestos. Asbestos may initiate mitochondria-associated ROS, which mediate asbestos-induced nuclear mutagenic events and inflammatory signaling pathways in exposed cells. These data provide new insights into the molecular mechanisms of asbestos-induced genotoxicity.


Assuntos
Amianto/efeitos adversos , Mitocôndrias/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , 8-Hidroxi-2'-Desoxiguanosina , Análise de Variância , Dano ao DNA , Desoxiguanosina/análogos & derivados , Relação Dose-Resposta a Droga , Células Epiteliais/efeitos dos fármacos , Imunofluorescência , Perfilação da Expressão Gênica , Humanos , Testes para Micronúcleos , Microscopia de Fluorescência , Mitocôndrias/metabolismo , Sistema Respiratório/citologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Int J Oncol ; 39(4): 1001-9, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21701776

RESUMO

As an uncommon cancer, mesothelioma is very hard to treat with a low average survival rate owing to its usual late detection and being highly invasive. The link between asbestos exposure and the development of mesothelioma in humans is unequivocal. TGFBI, a secreted protein that is induced by transforming growth factor-ß in various human cell types, has been shown to be associated with tumorigenesis in various types of tumors. It has been demonstrated that TGFBI expression is markedly suppressed in asbestos-induced tumorigenic cells, while an ectopic expression of TGFBI significantly suppresses tumorigenicity and progression in human bronchial epithelial cells. In order to delineate a potential role of TGFBI in mediating the molecular events that occur in mesothelioma tumorigenesis, we generated stable TGFBI knockdown mutants from the mesothelium cell line Met-5A by using an shRNA approach, and secondly created ectopic TGFBI overexpression mutants from the mesothelioma cell line H28 in which TGFBI is absent. We observed that in the absence of TGFBI, the knockdown mesothelial and mesothelioma cell lines exhibited an elevated proliferation rate, enhanced plating efficiency, increased anchorage-independent growth, as well as an increased cellular protein synthesis rate as compared with their respective controls. Furthermore, cell cycle regulatory proteins c-myc/cyclin D1/phosphor-Rb were upregulated; a more active PI3K/Akt/mTOR signaling pathway was also detected in TGFBI-depleted cell lines. These findings suggest that TGFBI may repress mesothelioma tumorigenesis and progression via the PI3K/Akt signaling pathway.


Assuntos
Processos de Crescimento Celular/efeitos dos fármacos , Proteínas da Matriz Extracelular/metabolismo , Mesotelioma/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Amianto/toxicidade , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Ciclina D1/metabolismo , Proteínas de Ligação a DNA/metabolismo , Progressão da Doença , Fator de Crescimento Epidérmico/metabolismo , Proteínas da Matriz Extracelular/antagonistas & inibidores , Proteínas da Matriz Extracelular/genética , Técnicas de Silenciamento de Genes/métodos , Humanos , Mesotelioma/etiologia , Mesotelioma/genética , Mesotelioma/patologia , Mutação , Neoplasias Mesoteliais/genética , Neoplasias Mesoteliais/metabolismo , Neoplasias Mesoteliais/patologia , Proteínas Oncogênicas v-mos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/genética
8.
Cancer Lett ; 308(1): 23-32, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21561707

RESUMO

Controversy has arisen as to the role of transforming growth factor-ß-induced protein (TGFBI) in the regulation of tumor metastasis. Using lung and breast cancer cell lines (H522 and MCF-7, respectively), we established that TGFBI induced cell adhesion to extracellular matrix proteins by activating adhesion-associated signaling and subsequent structure reformation, ultimately leading to cells less motile; whereas TGFBI reduced abilities of colony formation in soft agar, penetration through matrix gel, and activation of matrix metalloproteinases 2 and 9. Furthermore, injection of TGFBI-expressing cells into immuno-deficient mice resulted in a significant reduction in tumor metastasis in vivo. Taken together, these data suggest that TGFBI moderates the metastatic potential of cancer cells.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Fator de Crescimento Transformador beta1/biossíntese , Animais , Neoplasias da Mama/genética , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Citoesqueleto/metabolismo , Citoesqueleto/patologia , Modelos Animais de Doenças , Feminino , Humanos , Neoplasias Pulmonares/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Metástase Neoplásica , Transdução de Sinais , Transfecção , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
9.
J Cell Biochem ; 112(2): 463-75, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21268068

RESUMO

Melanoma is the most lethal form of human skin cancer. However, only limited chemotherapy is currently available for the metastatic stage of the disease. Since chemotherapy, radiation and sodium arsenite treatment operate mainly through induction of the intrinsic mitochondrial pathway, a strongly decreased mitochondrial function in metastatic melanoma cells, could be responsible for low efficacy of the conventional therapy of melanoma. Another feature of metastatic melanoma cells is their proinflammatory phenotype, linked to endogenous expression of the inflammatory cytokines, such as TNFα IL6 and IL8, their receptors, and constitutive NF-κB- and STAT3-dependent gene expression, including cyclooxygenase-2 (PTGS2/COX2). In the present study, we treated melanoma cells with immunological (monoclonal antibody against TNFα or IL6), pharmacological (small molecular inhibitors of IKKß-NF-κB and JAK2-STAT3) or genetic (specific RNAi for COX-2) agents that suppressed the inflammatory response in combination with induction of apoptosis via TRAIL. As a result of these combined treatments, exogenous TRAIL via interactions with TRAIL-R2/R1 strongly increased levels of apoptosis in resistant melanoma cells. The present study provides new understanding of the regulation of TRAIL-mediated apoptosis in melanoma and will serve as the foundation for the potential development of a novel approach for a therapy of resistant melanomas.


Assuntos
Apoptose/efeitos dos fármacos , Melanoma/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Western Blotting , Linhagem Celular , Ciclo-Oxigenase 2/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , NF-kappa B/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Reação em Cadeia da Polimerase , Fator de Transcrição STAT3/metabolismo
10.
Int J Radiat Biol ; 86(4): 321-8, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20353341

RESUMO

PURPOSE: After a radiological 'dirty bomb' incident in a major metropolitan center, substantial numbers of people may be exposed to radiation. However, only a fraction of those individuals will need urgent medical attention. Consequently, a rapid screening test is needed to identify those people who require immediate treatment. MATERIAL AND METHODS: Ten normal human cell lines were screened by enzyme-linked immunosorbent assay (ELISA) for the expression of a dozen secreted cytokines that have been reported to have changes in protein or mRNA levels at 1, 2, and 3 days after 0-10 Gy irradiation using (137)Cs gamma rays at 0.82 Gy min(-1). After this systematic in vitro screen, we measured changes in the level of a subset of these candidate proteins in plasma from irradiated C57BL/6 mice (n = 3 per group), comparing shams with a single radiation dose (5 Gy X-rays) at 3.7 Gy min(-1) at 6 h after irradiation. RESULTS: We identified four cytokine molecules that had altered levels after radiation exposure, one of which, Interleukin (IL) 6, was consistently elevated after irradiation in vitro and in vivo. CONCLUSIONS: Our findings underscore the potential for IL6 as a marker for an immunoassay-based, rapid, high-throughput biodosimeter.


Assuntos
Anticorpos/imunologia , Biomarcadores/metabolismo , Raios gama , Ensaios de Triagem em Larga Escala/métodos , Radiometria/métodos , Animais , Linhagem Celular Tumoral , Citocinas/sangue , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , Interleucina-6/sangue , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/metabolismo , Fatores de Tempo , Raios X
11.
Cancer Res ; 69(8): 3510-9, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19351839

RESUMO

The aim of the present study was to elucidate the effects of ataxia telangiectasia mutated (ATM) kinase on the regulation of the extrinsic tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor 2/DR5-mediated death pathway in human melanoma cells. We revealed that total ATM protein levels were high in some human melanoma lines compared with normal cells. The basal levels of active form ATM phospho-Ser(1981) were also detectable in many melanoma lines and could be further up-regulated by gamma-irradiation. Pretreatment of several melanoma lines just before gamma-irradiation with the inhibitor of ATM kinase KU-55933 suppressed p53 and nuclear factor-kappaB (NF-kappaB) activation but notably increased radiation-induced DR5 surface expression, down-regulated cFLIP (caspase-8 inhibitor) levels, and substantially enhanced exogenous TRAIL-induced apoptosis. Furthermore, gamma-irradiation in the presence of KU-55933 rendered TRAIL-resistant HHMSX melanoma cells susceptible to TRAIL-mediated apoptosis. In addition, suppression of ATM expression by the specific short hairpin RNA also resulted in down-regulation of cFLIP levels, up-regulation of surface DR5 expression, and TRAIL-mediated apoptosis in melanoma cells. Besides p53 and NF-kappaB, crucial regulators of DR5 expression, transcription factor STAT3 is known to negatively regulate DR5 expression. Suppression of Ser(727) and Tyr(705) phosphorylation of STAT3 by KU-55933 reduced STAT3 transacting activity accompanied by elevation in DR5 expression. Dominant-negative STAT3beta also efficiently up-regulated the DR5 surface expression and down-regulated cFLIP levels in melanoma cells in culture and in vivo. Taken together, our data show the existence of an ATM-dependent STAT3-mediated antiapoptotic pathway, which on suppression sensitizes human melanoma cells to TRAIL-mediated apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ligação a DNA/antagonistas & inibidores , Melanoma/tratamento farmacológico , Melanoma/enzimologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Proteínas Supressoras de Tumor/antagonistas & inibidores , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Humanos , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Morfolinas/farmacologia , NF-kappa B/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Pironas/farmacologia , RNA Interferente Pequeno/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/biossíntese , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Fator de Transcrição STAT3/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/genética , Ensaios Antitumorais Modelo de Xenoenxerto
12.
J Toxicol Environ Health A ; 72(5): 301-4, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19184745

RESUMO

One of the long-term objectives of the research in our laboratory was to determine whether mitochondrial DNA (mtDNA) mutations were generated in cell lines exposed to a variety of known mutagens. Many of these mutagens are known to increase oxidative stress in the cell, and one potential outcome of this would be an increased incidence of point mutations in mtDNA. Recently, there has been some controversy regarding the validity of point mutations in the regulatory region of mtDNA as a predictive or causative marker for carcinogenesis. Studies were undertaken to assess whether nuclear mutagens such as arsenic (As), asbestos, and ultraviolet (UV) and gamma-radiation, induced both heteroplasmic and homoplasmic point mutations in mtDNA. A direct sequencing approach was used to reduce the occurrence of experimental errors and cross-checked all base changes with databases of known polymorphisms. Our results showed that, while base changes did occur, there was no marked difference between the number of changes in treated and untreated cells. Furthermore, in human lymphocyte samples from subjects exposed to As, most of these base changes were previously reported. Interestingly, there was an increase in the number of transversions (purine ( pyrimidine) in smokers from a human population study, but as with the findings in cell culture samples, there was no difference in the total number of base changes. Data suggest that only a change in the number of rare transversions would be indicative of an increase in point mutations in mtDNA after exposure to mutagens.


Assuntos
DNA Mitocondrial/efeitos dos fármacos , DNA Mitocondrial/efeitos da radiação , Poluentes Ambientais/toxicidade , Raios gama , Regulação da Expressão Gênica/efeitos dos fármacos , Mutagênicos/toxicidade , Raios Ultravioleta , Animais , Arsênio/toxicidade , Amianto/toxicidade , Células CHO , Carcinógenos/toxicidade , Linhagem Celular , Cricetinae , Cricetulus , Dano ao DNA , Exposição Ambiental/efeitos adversos , Humanos , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Linfócitos/efeitos da radiação , Mutação/genética , Espécies Reativas de Oxigênio , Fumar/genética
13.
Sci Total Environ ; 407(3): 1174-81, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18977016

RESUMO

Inorganic arsenic (arsenate and arsenite) are well known human carcinogens. Apoptosis is a normal biological process that is involved in regulating cell development and differentiation, and is an important protective response to cell injury. The aim of this study was to determine the long term arsenic effect on human small airway epithelial cells (SAEC) by analyzing two distinct apoptosis-inducing agents, Fas ligand (Fas L), which evokes death receptor-mediated apoptosis, and hydrogen peroxide H2O2, which induces apoptosis mediated by reactive oxygen species (ROS). The SAEC were continuously exposed to 0.5 microg/mL arsenic for 28 weeks, and apoptosis was examined after 24 h treatment with either Fas L or H2O2. SAEC displayed decreased cell viability and increased apoptosis after treatment with Fas L and H2O2, compared to non-arsenic treated control cells. Furthermore, treatment of these arsenic-exposed SAEC with Fas L or H2O2 induced cleavage of the DNA damage recognition protein, poly (ADP-ribose) polymerase (PARP), and the 'effector' caspase, Caspase-3, both canonical indicators of apoptosis. We observed increased phosphorylation of p38, a member of the MAP kinase family, following treatment with Fas L or H2O2. To confirm the involvement of p38 in the regulation of apoptosis we pretreated cells with the p38 kinase inhibitor, SB 203580 and observed a significant decrease in apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Arseniatos/toxicidade , Mucosa Respiratória/citologia , Mucosa Respiratória/fisiologia , Carcinógenos/toxicidade , Caspase 3/efeitos dos fármacos , Caspase 3/metabolismo , Sobrevivência Celular/efeitos dos fármacos , DNA/efeitos dos fármacos , DNA/metabolismo , Dano ao DNA , Proteína Ligante Fas/efeitos dos fármacos , Proteína Ligante Fas/fisiologia , Citometria de Fluxo , Genes Reporter , Humanos , Peróxido de Hidrogênio/toxicidade , Microscopia Confocal , Microscopia Eletrônica de Varredura , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/ultraestrutura
14.
Exp Cell Res ; 314(5): 1163-76, 2008 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-18222423

RESUMO

Although many human melanomas express the death receptors TRAIL-R2/DR5 or TRAIL-R1/DR4 on cell surface, they often exhibit resistance to exogenous TRAIL. One of the main contributors to TRAIL-resistance of melanoma cells is upregulation of transcription factors STAT3 and NF-kappaB that control the expression of antiapoptotic genes, including cFLIP and Bcl-xL. On the other hand, the JNK-cJun pathway is involved in the negative regulation of cFLIP (a caspase-8 inhibitor) expression. Our observations indicated that resveratrol, a polyphenolic phytoalexin, decreased STAT3 and NF-kappaB activation, while activating JNK-cJun that finally suppressed expression of cFLIP and Bcl-xL proteins and increased sensitivity to exogenous TRAIL in DR5-positive melanomas. Interestingly, resveratrol did not increase surface expression of DR5 in human melanomas, while gamma-irradiation or sodium arsenite treatment substantially upregulated DR5 expression. Hence, an initial increase in DR5 surface expression (either by gamma-irradiation or arsenite), and subsequent downregulation of antiapoptotic cFLIP and Bcl-xL (by resveratrol), appear to constitute an efficient approach to reactivate apoptotic death pathways in TRAIL-resistant human melanomas. In spite of partial suppression of mitochondrial function and the mitochondrial death pathway, melanoma cells still retain the potential to undergo the DR5-mediated, caspase-8-dependent death pathway that could be accelerated by either an increase in DR5 surface expression or suppression of cFLIP. Taken together, these results suggest that resveratrol, in combination with TRAIL, may have a significant efficacy in the treatment of human melanomas.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Melanoma/patologia , Estilbenos/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/efeitos dos fármacos , Antineoplásicos Fitogênicos , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Linhagem Celular Tumoral , Regulação da Expressão Gênica/imunologia , Humanos , Melanoma/tratamento farmacológico , NF-kappa B/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Resveratrol , Fator de Transcrição STAT3/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/análise
15.
Mol Med ; 14(1-2): 2-10, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18037969

RESUMO

Human small airway epithelial cells (SAECs) previously immortalized with human telomerase reverse transcriptase (h-TERT) were continuously treated with sodium arsenite at a dose of 0.5 microg/mL in culture for up to 6 months. Arsenic-treated cells progressively displayed an increase in transformed phenotype including enhanced growth saturation density, plating efficiency, and anchorage-independent growth and invasion capability compared with their nontreated control cells. To determine whether arsenic-induced cell transformation was associated with genomic instability, treated and control cells were also analyzed for micronuclei formation. A 4.8-fold increase in micronuclei incidence in arsenic-treated cells was detected in conjunction with increased N-phosphonacetyl-l-aspartate (PALA)-resistant characteristics. In addition, arsenic-treated cells showed an increase in c-H-ras, c-myc, and c-fos protein expression relative to controls. The change in oncoprotein expression correlated with a decrease in wild-type p53 expression and hyperphosphorylated retinoblastoma. Taken together, these results strongly suggest that h-TERT immortalized human small airway epithelial cells underwent step-wise transformation after inorganic arsenic treatment.


Assuntos
Arsenitos/toxicidade , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/patologia , Células Epiteliais/efeitos dos fármacos , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/patologia , Compostos de Sódio/toxicidade , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Transformada , Proliferação de Células/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Micronúcleos com Defeito Cromossômico/induzido quimicamente , Proteínas Proto-Oncogênicas c-fos/biossíntese , Proteínas Proto-Oncogênicas c-myc/biossíntese , Proteínas Proto-Oncogênicas p21(ras)/biossíntese , Mucosa Respiratória/metabolismo , Telomerase , Proteína Supressora de Tumor p53/biossíntese
16.
DNA Seq ; 18(5): 341-6, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17654009

RESUMO

Mammalian mitochondria contain their own approximately 16.5 kb circular genome. Mitochondrial DNA (mtDNA) encodes for a subset of the proteins involved in the electron transport chain and depletion or mutation of the sequence is implicated in a number of human disease processes. The recent finding is that mitochondrial damage mediates genotoxicity after exposure to chemical carcinogens has focused attention on the role of mtDNA mutations in the development of cancer. Although the entire genome has been sequenced for a number of mammals, only a small fraction of the mtDNA sequence is available for hamsters. We have obtained here the entire 16,284 bp sequence of the Chinese hamster mitochondrial genome, which will enable detailed analysis of mtDNA mutations caused by exposure to mutagens in hamster-derived cell lines.


Assuntos
DNA Mitocondrial/química , DNA Mitocondrial/genética , Animais , Sequência de Bases , Células CHO , Cricetinae , Cricetulus , Genoma , Dados de Sequência Molecular , Análise de Sequência de DNA
17.
Cancer Res ; 67(11): 5239-47, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17545603

RESUMO

Arsenic is a well-established human carcinogen that is chronically consumed in drinking water by millions of people worldwide. Recent evidence has suggested that arsenic is a genotoxic carcinogen. Furthermore, we have shown that mitochondria mediate the mutagenic effects of arsenic in mammalian cells, as arsenic did not induce nuclear mutations in mitochondrial DNA (mtDNA)-depleted cells. Using the human-hamster hybrid A(L) cells, we show here that arsenic alters mitochondrial function by decreasing cytochrome c oxidase function and oxygen consumption but increasing citrate synthase function. These alterations correlated with depletion in mtDNA copy number and increase in large heteroplasmic mtDNA deletions. In addition, mtDNA isolated periodically from cultures treated continuously with arsenic did not consistently display the same deletion pattern, indicating that the mitochondrial genome was subjected to repeated and continuous damage. These data support the theory that the mitochondria, and particularly mtDNA, are important targets of the mutagenic effects of arsenic in mammalian cells.


Assuntos
Arsênio/toxicidade , Dano ao DNA , DNA Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Animais , Cricetinae , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Relação Dose-Resposta a Droga , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Humanos , Células Híbridas , Mitocôndrias/genética , Mitocôndrias/metabolismo , Testes de Mutagenicidade , Oxirredução , Consumo de Oxigênio/efeitos dos fármacos
18.
Mol Cell Biol ; 26(12): 4399-409, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16738308

RESUMO

Src kinase is a crucial mediator of adhesion-related signaling and motility. Src binds to focal adhesion kinase (FAK) through its SH2 domain and subsequently activates it for phosphorylation of downstream substrates. In addition to this binding function, data suggested that the SH2 domain might also perform an important role in targeting Src to focal adhesions (FAs) to enable further substrate phosphorylations. To examine this, we engineered an R175L mutation in cSrc to prevent the interaction with FAK pY397. This constitutively open Src kinase mediated up-regulated substrate phosphorylation in SYF cells but was unable to promote malignant transformation. Significantly, SrcR175L cells also had a profound motility defect and an impaired FA generation capacity. Importantly, we were able to recapitulate wild-type motile behavior and FA formation by directing the kinase to FAs, clearly implicating the SH2 domain in recruitment to FAK and indicating that this targeting capacity, and not simply Src-FAK scaffolding, was critical for normal Src function.


Assuntos
Movimento Celular/fisiologia , Proteína-Tirosina Quinases de Adesão Focal/fisiologia , Adesões Focais/fisiologia , Quinases da Família src/química , Quinases da Família src/fisiologia , Substituição de Aminoácidos , Animais , Arginina/química , Linhagem Celular , Galinhas , Proteína-Tirosina Quinases de Adesão Focal/química , Proteína-Tirosina Quinases de Adesão Focal/genética , Genes src , Humanos , Técnicas In Vitro , Mutagênese Sítio-Dirigida , Fosforilação , Domínios de Homologia de src , Quinases da Família src/genética
19.
Nat Cell Biol ; 7(6): 581-90, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15895076

RESUMO

Imaging studies implicate microtubule targeting of focal adhesions in focal adhesion disassembly, although the molecular mechanism is unknown. Here, we develop a model system of focal adhesion disassembly based on the finding that microtubule regrowth after nocodazole washout induces disassembly of focal adhesions, and that this disassembly occurs independently of Rho and Rac, but depends on focal adhesion kinase (FAK) and dynamin. During disassembly, dynamin interacts with FAK and colocalizes with focal adhesions. Inhibition of dynamin prevents migration of cells with a focal adhesion phenotype. Our results show that focal adhesion disassembly involves microtubules, dynamin and FAK, and is not simply the reversal of focal adhesion formation.


Assuntos
Adesão Celular/fisiologia , Dinaminas/metabolismo , Adesões Focais/metabolismo , Microtúbulos/metabolismo , Proteínas Tirosina Quinases/metabolismo , Animais , Antineoplásicos/farmacologia , Movimento Celular/fisiologia , Dinaminas/ultraestrutura , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Adesões Focais/ultraestrutura , Proteínas de Fluorescência Verde , Integrinas/metabolismo , Camundongos , Microtúbulos/ultraestrutura , Células NIH 3T3 , Nocodazol/farmacologia , Proteínas Tirosina Quinases/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA