Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Medicinas Complementares
Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Front Immunol ; 12: 748519, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34777360

RESUMO

Inherited defects that abrogate the function of the adenosine deaminase (ADA) enzyme and consequently lead to the accumulation of toxic purine metabolites cause profound lymphopenia and severe combined immune deficiency. Additionally, neutropenia and impaired neutrophil function have been reported among ADA-deficient patients. However, due to the rarity of the disorder, the neutrophil developmental abnormalities and the mechanisms contributing to them have not been characterized. Induced pluripotent stem cells (iPSC) generated from two unrelated ADA-deficient patients and from healthy controls were differentiated through embryoid bodies into neutrophils. ADA deficiency led to a significant reduction in the number of all early multipotent hematopoietic progenitors. At later stages of differentiation, ADA deficiency impeded the formation of granulocyte colonies in methylcellulose cultures, leading to a significant decrease in the number of neutrophils generated from ADA-deficient iPSCs. The viability and apoptosis of ADA-deficient neutrophils isolated from methylcellulose cultures were unaffected, suggesting that the abnormal purine homeostasis in this condition interferes with differentiation or proliferation. Additionally, there was a significant increase in the percentage of hyperlobular ADA-deficient neutrophils, and these neutrophils demonstrated significantly reduced ability to phagocytize fluorescent microspheres. Supplementing iPSCs and methylcellulose cultures with exogenous ADA, which can correct adenosine metabolism, reversed all abnormalities, cementing the critical role of ADA in neutrophil development. Moreover, chemical inhibition of the ribonucleotide reductase (RNR) enzyme, using hydroxyurea or a combination of nicotinamide and trichostatin A in iPSCs from healthy controls, led to abnormal neutrophil differentiation similar to that observed in ADA deficiency, implicating RNR inhibition as a potential mechanism for the neutrophil abnormalities. In conclusion, the findings presented here demonstrate the important role of ADA in the development and function of neutrophils while clarifying the mechanisms responsible for the neutrophil abnormalities in ADA-deficient patients.


Assuntos
Adenosina Desaminase/fisiologia , Agamaglobulinemia/imunologia , Células-Tronco Pluripotentes Induzidas/citologia , Neutrófilos/citologia , Imunodeficiência Combinada Severa/imunologia , Adenosina Desaminase/genética , Células Cultivadas , Corpos Embrioides/citologia , Fibroblastos/enzimologia , Granulócitos/citologia , Humanos , Ácidos Hidroxâmicos/farmacologia , Hidroxiureia/farmacologia , Lactente , Masculino , Mutação de Sentido Incorreto , Mielopoese , Niacinamida/farmacologia , Mutação Puntual , Ribonucleotídeo Redutases/antagonistas & inibidores
2.
Artigo em Inglês | MEDLINE | ID: mdl-28893786

RESUMO

We have previously reported that an erg11 mutation affecting ergosterol synthesis and a hem13 mutation in the heme synthesis pathway significantly sensitize the fission yeast Schizosaccharomyces pombe to hydroxyurea (HU) (1, 2). Here we show that treatment with inhibitors of Erg11 and heme biosynthesis phenocopies the two mutations in sensitizing wild-type cells to HU. Importantly, HU synergistically interacts with the heme biosynthesis inhibitor sampangine and several Erg11 inhibitors, the antifungal azoles, in causing cell lethality. Since the synergistic drug interactions are also observed in the phylogenetically divergent Saccharomyces cerevisiae and the opportunistic fungal pathogen Candida albicans, the synergism is likely conserved in eukaryotes. Interestingly, our genetic data for S. pombe has also led to the discovery of a robust synergism between sampangine and the azoles in C. albicans Thus, combinations of HU, sampangine, and the azoles can be further studied as a new method for the treatment of fungal infections.


Assuntos
Alcaloides/farmacologia , Antifúngicos/farmacologia , Azóis/farmacologia , Inibidores Enzimáticos/farmacologia , Hidroxiureia/farmacologia , Schizosaccharomyces/efeitos dos fármacos , Candida albicans/efeitos dos fármacos , Candidíase/tratamento farmacológico , Coproporfirinogênio Oxidase/genética , Sistema Enzimático do Citocromo P-450/genética , Citocinese/efeitos dos fármacos , Sinergismo Farmacológico , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Testes de Sensibilidade Microbiana , Naftiridinas , Ribonucleotídeo Redutases/antagonistas & inibidores , Saccharomyces cerevisiae/efeitos dos fármacos , Proteínas de Saccharomyces cerevisiae/antagonistas & inibidores , Proteínas de Saccharomyces cerevisiae/genética
3.
Indian J Dent Res ; 25(5): 580-5, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25511055

RESUMO

BACKGROUND: Heat shock proteins-47 (HSP47) is a collagen specific molecular chaperone, involved in the processing and/or secretion of procollagen. It seems to be regularly upregulated in various fibrotic or collagen disorders. Hence, this protein can be a potential target for the treatment of various fibrotic diseases including oral submucous fibrosis (OSF), which is a collagen metabolic disorder of oral cavity and whose etiopathogeneic mechanism and therapeutic protocols are still not well documented. AIM: The aim of this study is to identify the novel therapeutic agents using in-silico methods for the management of OSF. OBJECTIVES: The objectives of this study are to identify the binding sites of HSP47 on the collagen molecule and to identify the lead compound with anti-HSP47 activity from the library of natural compounds, using in-silico methodology. MATERIALS AND METHODS: The web-based and tool based in-silico analysis of the HSP47 and collagen molecules are used in this study. The crystal structure of collagen and HSP47 were retrieved from Protein Data Bank website. The binding site identification and the docking studies are done using Molegro Virtual Docker offline tool. RESULTS: Out of the 104 Natural compounds, six ligands are found to possess best binding affinity to the binding amino acid residues. Silymarin binds with the 4AU2A receptor and the energy value are found to be -178.193 with four Hbonds. The other best five natural compounds are hesperidin, ginkgolides, withanolides, resveratrol, and gingerol. Our findings provide the basis for the in-vitro validation of the above specified compounds, which can possibly act as "lead" molecules in designing the drugs for OSF. CONCLUSION: HSP47 can be a potential candidate to target, in order to control the production of abundance collagen in OSF. Hence, the binding sites of HSP47 with collagen are identified and some natural compounds with a potential to bind with these binding receptors are also recognized. These natural compounds might act as anti-HSP47 lead molecules in designing novel therapeutic agents for OSF, which are so far unavailable.


Assuntos
Simulação por Computador , Proteínas de Choque Térmico HSP47/química , Modelos Químicos , Fibrose Oral Submucosa/tratamento farmacológico , Marcadores de Afinidade/química , Antioxidantes/química , Sítios de Ligação , Catecóis/química , Colágeno/química , Cristalografia , Álcoois Graxos/química , Zingiber officinale/química , Ginkgolídeos/química , Hesperidina/química , Humanos , Ligantes , Ligação Proteica , Resveratrol , Ribonucleotídeo Redutases/antagonistas & inibidores , Silimarina/química , Estilbenos/química , Interface Usuário-Computador , Vitanolídeos/química
4.
PLoS One ; 9(11): e112619, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25402485

RESUMO

Acute Myeloid Leukemia (AML) is an aggressive malignancy which leads to marrow failure, and ultimately death. There is a desperate need for new therapeutics for these patients. Ribonucleotide reductase (RR) is the rate limiting enzyme in DNA synthesis. Didox (3,4-Dihydroxybenzohydroxamic acid) is a novel RR inhibitor noted to be more potent than hydroxyurea. In this report we detail the activity and toxicity of Didox in preclinical models of AML. RR was present in all AML cell lines and primary patient samples tested. Didox was active against all human and murine AML lines tested with IC50 values in the low micromolar range (mean IC50 37 µM [range 25.89-52.70 µM]). It was active against primary patient samples at concentrations that did not affect normal hematopoietic stem cells (HSCs). Didox exposure resulted in DNA damage and p53 induction culminating in apoptosis. In syngeneic, therapy-resistant AML models, single agent Didox treatment resulted in a significant reduction in leukemia burden and a survival benefit. Didox was well tolerated, as marrow from treated animals was morphologically indistinguishable from controls. Didox exposure at levels that impaired leukemia growth did not inhibit normal HSC engraftment. In summary, Didox was well tolerated and effective against preclinical models of AML.


Assuntos
Inibidores Enzimáticos/farmacologia , Ácidos Hidroxâmicos/farmacologia , Leucemia Mieloide Aguda/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Resistencia a Medicamentos Antineoplásicos , Inibidores Enzimáticos/administração & dosagem , Feminino , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Humanos , Ácidos Hidroxâmicos/administração & dosagem , Cariótipo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/mortalidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Ribonucleotídeo Redutases/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Ensaio Tumoral de Célula-Tronco , Proteína Supressora de Tumor p53/metabolismo
5.
Age (Dordr) ; 35(1): 69-81, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22083438

RESUMO

Resveratrol, a polyphenolic compound, has been shown to extend lifespan in different organisms. Emerging evidence suggests that the prolongevity effect of resveratrol depends on dietary composition. However, the mechanisms underlying the interaction of resveratrol and dietary nutrients in modulating lifespan remain elusive. Here, we investigated the effect of resveratrol on lifespan of Drosophila melanogaster fed diets differing in the concentrations of sugar, yeast extract, and palmitic acid representing carbohydrate, protein, and fat, respectively. Resveratrol at up to 200 µM in diets did not affect lifespan of wild-type female flies fed a standard, restricted or high sugar-low protein diet, but extended lifespan of females fed a low sugar-high protein diet. Resveratrol at 400 µM extended lifespan of females fed a high-fat diet. Lifespan extension by resveratrol was associated with downregulation of genes in aging-related pathways, including antioxidant peroxiredoxins, insulin-like peptides involved in insulin-like signaling and several downstream genes in Jun-kinase signaling involved in oxidative stress response. Furthermore, resveratrol increased lifespan of superoxide dismutase 1 (sod1) knockdown mutant females fed a standard or high-fat diet. No lifespan extension by resveratrol was observed in wild-type and sod1 knockdown males under the culture conditions in this study. Our results suggest that the gender-specific prolongevity effect of resveratrol is influenced by dietary composition and resveratrol promotes the survival of flies by modulating genetic pathways that can reduce cellular damage. This study reveals the context-dependent effect of resveratrol on lifespan and suggests the importance of dietary nutrients in implementation of effective aging interventions using dietary supplements.


Assuntos
Envelhecimento/efeitos dos fármacos , Restrição Calórica , Drosophila melanogaster/fisiologia , Longevidade/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Estilbenos/farmacologia , Envelhecimento/fisiologia , Animais , Antioxidantes/farmacologia , Dieta Hiperlipídica , Feminino , Masculino , Resveratrol , Ribonucleotídeo Redutases/antagonistas & inibidores , Transdução de Sinais
6.
Arch Oral Biol ; 58(3): 299-310, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22770624

RESUMO

OBJECTIVE: In dentistry, the use of metals in fillings, braces, implants, bridges and other prosthodontic restorations is a common practice. Previous studies revealed that zinc (Zn) and copper (Cu) released from gold alloys, and nickel (Ni) released from nickel-chromium alloys, have a highly cytotoxic effect on fibroblast cell cultures. Our working hypothesis is that oral fibroblasts are susceptible to damage from metals because they elevate reaction oxygen species (ROS). In this study, we investigated specific antioxidant (AO) combinations to determine if they counteract the effects of Cu, Ni and Zn on cultured oral fibroblast proliferation and oxidative damage. METHODS: Oral fibroblasts were pretreated with Cu, Ni and Zn for 60min. Thereafter, cells were treated with 10(-5)M combinations of bioactive AO resveratrol (R), ferulic acid (F), phloretin (P) and tetrahydrocurcuminoids (T) (RFT, PFR, PFT) for 24h. Cell viability and DNA synthesis were monitored by 3-[4,5-dimethylthiazol-2-yl]-5-[3-carboxymethoxyphenyl]-2-[4-sulfophenyl]-2H-tetrazolium (MTS) and 5-bromo-2-deoxyuridine (BrDU) assays. ROS was measured using the fluorescence response of dichlorodihydrofluorescein diacetate (DCF). RESULTS: AO compounds increased recovery of cells exposed to Cu and Zn. Moreover, AO treatment induced DNA synthesis in the presence of the metal stressors. Cu and Ni stimulated production of ROS. PFR treatment decreased ROS in the presence of Cu, Ni and Zn. SIGNIFICANCE: These data indicate that pure AOs counteracted the detrimental effects of Cu, Ni, Zn on oral fibroblasts in vitro by increasing cell viability, and DNA synthesis and decreasing ROS activity.


Assuntos
Antioxidantes/uso terapêutico , Materiais Dentários/toxicidade , Fibroblastos/efeitos dos fármacos , Gengiva/efeitos dos fármacos , Metais/toxicidade , Ligamento Periodontal/efeitos dos fármacos , Antioxidantes/administração & dosagem , Técnicas de Cultura de Células , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Cobre/toxicidade , Ácidos Cumáricos/administração & dosagem , Ácidos Cumáricos/uso terapêutico , Curcumina/administração & dosagem , Curcumina/análogos & derivados , Curcumina/uso terapêutico , DNA/biossíntese , DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Sequestradores de Radicais Livres/administração & dosagem , Sequestradores de Radicais Livres/uso terapêutico , Gengiva/citologia , Humanos , Níquel/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Ligamento Periodontal/citologia , Fenóis/administração & dosagem , Fenóis/uso terapêutico , Floretina/administração & dosagem , Floretina/uso terapêutico , Espécies Reativas de Oxigênio/antagonistas & inibidores , Resveratrol , Ribonucleotídeo Redutases/antagonistas & inibidores , Estilbenos/administração & dosagem , Estilbenos/uso terapêutico , Fatores de Tempo , Zinco/toxicidade
7.
Age (Dordr) ; 35(5): 1851-65, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23129026

RESUMO

Resveratrol is a polyphenol that is mainly found in grapes and red wine and has been reported to be a caloric restriction (CR) mimetic driven by Sirtuin 1 (SIRT1) activation. Resveratrol increases metabolic rate, insulin sensitivity, mitochondrial biogenesis and physical endurance, and reduces fat accumulation in mice. In addition, resveratrol may be a powerful agent to prevent age-associated neurodegeneration and to improve cognitive deficits in Alzheimer's disease (AD). Moreover, different findings support the view that longevity in mice could be promoted by CR. In this study, we examined the role of dietary resveratrol in SAMP8 mice, a model of age-related AD. We found that resveratrol supplements increased mean life expectancy and maximal life span in SAMP8 and in their control, the related strain SAMR1. In addition, we examined the resveratrol-mediated neuroprotective effects on several specific hallmarks of AD. We found that long-term dietary resveratrol activates AMPK pathways and pro-survival routes such as SIRT1 in vivo. It also reduces cognitive impairment and has a neuroprotective role, decreasing the amyloid burden and reducing tau hyperphosphorylation.


Assuntos
Envelhecimento , Doença de Alzheimer/dietoterapia , Biomarcadores/metabolismo , Restrição Calórica , Suplementos Nutricionais , Longevidade/efeitos dos fármacos , Estilbenos/administração & dosagem , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Animais , Western Blotting , Cognição/efeitos dos fármacos , Modelos Animais de Doenças , Inibidores Enzimáticos/administração & dosagem , Imuno-Histoquímica , Camundongos , Resveratrol , Ribonucleotídeo Redutases/antagonistas & inibidores
8.
PLoS One ; 7(6): e38465, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22768044

RESUMO

BACKGROUND: Persistence of γ-H2AX after ionizing radiation (IR) or drug therapy is a robust reporter of unrepaired DNA double strand breaks in treated cells. METHODS: DU-145 prostate cancer cells were treated with a chemical library ±IR and assayed for persistence of γ-H2AX using an automated 96-well immunocytochemistry assay at 4 hours after treatment. Hits that resulted in persistence of γ-H2AX foci were tested for effects on cell survival. The molecular targets of hits were validated by molecular, genetic and biochemical assays and in vivo activity was tested in a validated Drosophila cancer model. RESULTS: We identified 2 compounds, MS0019266 and MS0017509, which markedly increased persistence of γ-H2AX, apoptosis and radiosensitization in DU-145 cells. Chemical evaluation demonstrated that both compounds exhibited structurally similar and biochemical assays confirmed that these compounds inhibit ribonucleotide reductase. DNA microarray analysis and immunoblotting demonstrates that MS0019266 significantly decreased polo-like kinase 1 gene and protein expression. MS0019266 demonstrated in vivo antitumor activity without significant whole organism toxicity. CONCLUSIONS: MS0019266 and MS0017509 are promising compounds that may be candidates for further development as radiosensitizing compounds as inhibitors of ribonucleotide reductase.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Histonas/metabolismo , Radiossensibilizantes/análise , Radiossensibilizantes/farmacologia , Bibliotecas de Moléculas Pequenas/análise , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/efeitos da radiação , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , DNA/biossíntese , Dano ao DNA , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/efeitos da radiação , Modelos Animais de Doenças , Drosophila melanogaster/efeitos dos fármacos , Drosophila melanogaster/metabolismo , Olho/efeitos dos fármacos , Olho/patologia , Olho/efeitos da radiação , Olho/ultraestrutura , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Cinética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Radiação Ionizante , Radiossensibilizantes/administração & dosagem , Ribonucleotídeo Redutases/antagonistas & inibidores , Ribonucleotídeo Redutases/metabolismo , Bibliotecas de Moléculas Pequenas/administração & dosagem , Quinase 1 Polo-Like
9.
Ann Oncol ; 23(11): 2799-2805, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22771827

RESUMO

BACKGROUND: Sorafenib is an oral anticancer agent targeting Ras-dependent signaling and angiogenic pathways. A phase I trial demonstrated that the combination of gemcitabine and sorafenib was well tolerated and had activity in advanced pancreatic cancer (APC) patients. The BAYPAN study was a multicentric, placebo-controlled, double-blind, randomized phase III trial comparing gemcitabine/sorafenib and gemcitabine/placebo in the treatment of APC. PATIENTS AND METHODS: The patient eligibility criteria were locally advanced or metastatic pancreatic adenocarcinoma, no prior therapy for advanced disease and a performance status of zero to two. The primary end point was progression-free survival (PFS). The patients received gemcitabine 1000 mg/m(2) i.v., weekly seven times followed by 1 rest week, then weekly three times every 4 weeks plus sorafenib 200 mg or placebo, two tablets p.o., twice daily continuously. RESULTS: Between December 2006 and September 2009, 104 patients were enrolled on the study (52 pts in each arm) and 102 patients were treated. The median and the 6-month PFS were 5.7 months and 48% for gemcitabine/placebo and 3.8 months and 33% for gemcitabine/sorafenib (P = 0.902, stratified log-rank test), respectively. The median overall survivals were 9.2 and 8 months, respectively (P = 0.231, log-rank test). The overall response rates were similar (19 and 23%, respectively). CONCLUSION: The addition of sorafenib to gemcitabine does not improve PFS in APC patients.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Desoxicitidina/análogos & derivados , Niacinamida/análogos & derivados , Neoplasias Pancreáticas/tratamento farmacológico , Compostos de Fenilureia/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Antimetabólitos Antineoplásicos/administração & dosagem , Antimetabólitos Antineoplásicos/efeitos adversos , Antimetabólitos Antineoplásicos/uso terapêutico , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Desoxicitidina/administração & dosagem , Desoxicitidina/efeitos adversos , Desoxicitidina/uso terapêutico , Intervalo Livre de Doença , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Niacinamida/administração & dosagem , Niacinamida/efeitos adversos , Niacinamida/uso terapêutico , Compostos de Fenilureia/administração & dosagem , Compostos de Fenilureia/efeitos adversos , Placebos , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/uso terapêutico , Ribonucleotídeo Redutases/antagonistas & inibidores , Sorafenibe , Gencitabina
10.
Future Oncol ; 8(2): 145-50, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22335579

RESUMO

The anticancer agent 3-aminopyridine-2-carboxaldehyde thiosemicarbazone is a ribonucleotide reductase inhibitor. It inactivates ribonucleotide reductase by disrupting an iron-stabilized radical in ribonucleotide reductase's small subunits, M2 and M2b (p53R2). Unfortunately, 3-aminopyridine-2-carboxaldehyde thiosemicarbazone also alters iron II (Fe(2+)) in hemoglobin. This creates Fe(3+) methemoglobin that does not deliver oxygen. Fe(2+) in hemoglobin normally auto-oxidizes to inactive Fe(3+) methemoglobin at a rate of nearly 3% per day and this is counterbalanced by a reductase system that normally limits methemoglobin concentrations to less than 1% of hemoglobin. This balance may be perturbed by symptomatic toxicity levels during 3-aminopyridine-2-carboxaldehyde thiosemicarbazone therapy. Indications of 3-aminopyridine-2-carboxaldehyde thiosemicarbazone sequelae attributable to methemoglobinemia include resting dyspnea, headaches and altered cognition. Management of methemoglobinemia includes supplemental oxygen, ascorbate and, most importantly, intravenously administered methylene blue as a therapeutic antidote.


Assuntos
Antídotos/uso terapêutico , Antineoplásicos/efeitos adversos , Inibidores Enzimáticos/efeitos adversos , Metemoglobinemia/tratamento farmacológico , Azul de Metileno/uso terapêutico , Piridinas/efeitos adversos , Ribonucleotídeo Redutases/antagonistas & inibidores , Tiossemicarbazonas/efeitos adversos , Antineoplásicos/farmacocinética , Inibidores Enzimáticos/farmacocinética , Hemoglobinas/metabolismo , Humanos , Metemoglobina/metabolismo , Metemoglobinemia/induzido quimicamente , Metemoglobinemia/diagnóstico , Piridinas/farmacocinética , Tiossemicarbazonas/farmacocinética
11.
J Gerontol A Biol Sci Med Sci ; 66(7): 751-64, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21454355

RESUMO

This study analyzed the capacity of resveratrol, a naturally occurring polyphenol, to reduce aging-induced oxidative stress and protect against sarcopenia. Middle-aged (18 months) C57/BL6 mice were randomly assigned to receive either a control diet or a diet supplemented with 0.05% trans-resveratrol for 10 months. Young (6 months) and middle-aged (18 months) mice were used as controls. Resveratrol supplementation did not reduce the aging-associated loss of muscle mass or improve maximal isometric force production, but it appeared to preserve fast-twitch fiber contractile function. Resveratrol supplementation did not improve mitochondrial content, the subcellular localization of cytochrome c protein content, or PGC1 protein content. Resveratrol increased manganese superoxide dismutase (MnSOD), reduced hydrogen peroxide(,) and lipid peroxidation levels in muscle samples, but it was unable to significantly reduce protein carbonyl levels. The data suggest that resveratrol has a protective effect against aging-induced oxidative stress in skeletal muscle, likely through the upregulation of MnSOD activity, but sarcopenia was not attenuated by resveratrol.


Assuntos
Envelhecimento/fisiologia , Suplementos Nutricionais , Músculo Esquelético/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Sarcopenia/tratamento farmacológico , Estilbenos/administração & dosagem , Envelhecimento/efeitos dos fármacos , Animais , Antioxidantes/administração & dosagem , Modelos Animais de Doenças , Seguimentos , Immunoblotting , Contração Isométrica/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias Musculares/efeitos dos fármacos , Mitocôndrias Musculares/metabolismo , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Resveratrol , Ribonucleotídeo Redutases/antagonistas & inibidores , Sarcopenia/metabolismo , Sarcopenia/patologia , Fatores de Tempo
12.
Shock ; 34(5): 535-44, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20386495

RESUMO

Immature myeloid cells have been implicated as a source of postburn inflammation, and the appearance of these cells correlates with enhanced upregulation of hematopoiesis. The role of proliferative cells in postburn immune changes has not been directly tested. Gemcitabine, a ribonucleotide reductase inhibitor, has been shown to deplete proliferative immature myeloid cells in tumor models while sparing mature cells, leading to restored lymphocyte function and tumor regression. We treated burn mice at postburn day 6 (PBD6) with 120 mg/kg gemcitabine. On PBD8, splenocytes were taken and stimulated with LPS, peptidoglycan, or concanavalin A. The blood and spleen cell populations were enumerated by flow cytometry or automated cell counter. In addition, mice treated with gemcitabine were given LPS or infected with Pseudomonas aeruginosa at PBD8, and mortality was monitored. Gemcitabine depleted burn-induced polymorphonuclear leukocytes and inflammatory monocytes without affecting mature F4/80 macrophages. This was accompanied by reduced TNFα, IL-6, and IL-10 production by burn splenocytes. Burn splenocytes stimulated with mitogens exhibited increased nitric oxide production relative to sham mice. In vivo treatment of burn mice with gemcitabine blocked these burn-induced changes without damaging lymphocyte function. Treatment of burn mice with gemcitabine ameliorated burn-induced susceptibility to LPS and infiltration of polymorphonuclear leukocytes into the liver and lung. Finally, gemcitabine treatment blocked the protective effect of burn injury upon P. aeruginosa infection. Our report shows that proliferative cells are major drivers of postburn immune changes and provides evidence that implicates immature myeloid cells in these processes.


Assuntos
Queimaduras/imunologia , Desoxicitidina/análogos & derivados , Células Mieloides/efeitos dos fármacos , Ribonucleotídeo Redutases/antagonistas & inibidores , Animais , Queimaduras/tratamento farmacológico , Concanavalina A/farmacologia , Desoxicitidina/farmacologia , Desoxicitidina/uso terapêutico , Avaliação Pré-Clínica de Medicamentos , Interleucina-10/biossíntese , Interleucina-6/biossíntese , Contagem de Leucócitos , Lipopolissacarídeos/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/efeitos dos fármacos , Monócitos/fisiologia , Células Mieloides/imunologia , Células Mieloides/metabolismo , Óxido Nítrico/biossíntese , Peptidoglicano/farmacologia , Infecções por Pseudomonas/complicações , Baço/imunologia , Baço/patologia , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Gencitabina
13.
Mol Cancer Ther ; 8(4): 980-7, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19372571

RESUMO

A phase I dose-escalation study of telomerase-specific oncolytic adenovirus, OBP-301 (Telomelysin), is now under way in the United States to assess feasibility and to characterize its pharmacokinetics in patients with advanced solid tumors. The present preclinical study investigates whether OBP-301 and a chemotherapeutic agent that is commonly used for lung cancer treatment, gemcitabine, are able to enhance antitumor effects in vitro and in vivo. The antitumor effects of OBP-301 infection and gemcitabine were evaluated by 2,3-bis[2-methoxy-4-nitro-5-sulfophenyl]-2H-tetrazolium-5-carboxanilide inner salt assay. In vivo antitumor effects of intratumoral injection of OBP-301 in combination with systemic administration of gemcitabine were assessed on nu/nu mice s.c. xenografted with human lung tumors. OBP-301 infection combined with gemcitabine resulted in very potent synergistic cytotoxicity in human lung cancer cells. The three human lung cancer cell lines treated with OBP-301 for 24 hours tended to accumulate in S phase compared with controls. The proportion of cells in S phase increased from 43.85% to 56.41% in H460 cells, from 46.72% to 67.09% in H322 cells, and from 38.22% to 57.67% in H358 cells. Intratumoral injection of OBP-301 combined with systemic administration of gemcitabine showed therapeutic synergism in human lung tumor xenografts. Our data suggest that the combination of OBP-301 and gemcitabine enhances the antitumor effects against human lung cancer. We also found that the synergistic mechanism may be due to OBP-301-mediated cell cycle accumulation in S phase. These results have important implications for the treatment of human lung cancer.


Assuntos
Adenocarcinoma Bronquioloalveolar/terapia , Antimetabólitos Antineoplásicos/uso terapêutico , Carcinoma de Células Grandes/terapia , Desoxicitidina/análogos & derivados , Neoplasias Pulmonares/terapia , Terapia Viral Oncolítica , Telomerase/metabolismo , Adenocarcinoma Bronquioloalveolar/patologia , Adenocarcinoma Bronquioloalveolar/virologia , Proteínas E1A de Adenovirus/metabolismo , Animais , Western Blotting , Carcinoma de Células Grandes/patologia , Carcinoma de Células Grandes/virologia , Proliferação de Células/efeitos dos fármacos , Terapia Combinada , Desoxicitidina/uso terapêutico , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Técnicas Imunoenzimáticas , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ribonucleotídeo Redutases/antagonistas & inibidores , Fase S/efeitos dos fármacos , Fase S/fisiologia , Transdução de Sinais , Células Tumorais Cultivadas , Replicação Viral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
14.
Nitric Oxide ; 18(3): 216-22, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18230367

RESUMO

Radio-toxins are toxic metabolites produced by ionizing irradiation and have toxic effects similar to those caused by direct irradiation. We have investigated the effect of a quinoid radio-toxin (QRT) obtained from gamma-irradiated potato tuber on various organs in mice using ex vivo and in vivo EPR spectroscopy. Results indicate a decrease in the activity of ribonucleotide reductase enzyme in spleen of mice treated with 0.2mg QRT. A dose of 2mg QRT was fatal to mice within 45-60 min of treatment. Nitrosyl hemoglobin complexes alpha-(Fe(2+)-NO)alpha-(Fe(2+))beta-(Fe(2+))(2) were detected from spleen, blood, liver, kidney, heart, and lung tissue samples of mice treated with lethal doses of QRT. A significant decrease of pO(2) in liver and brain was observed after administration of QRT at the lethal dose. The time of the appearance of the nitrosyl hemoglobin complex and its intensity varied with the dose of QRT and the type of tissue. These results indicate that the effect of the QRT is more prominent in spleen and to a lesser extent in liver and blood. The QRT action at the lethal doses resulted in an increased hypoxia over time with disruption of compensatory adaptive response. The results indicate similar outcome of QRT as observed with gamma-irradiation.


Assuntos
Quinonas/toxicidade , Ribonucleotídeo Redutases/antagonistas & inibidores , Baço/efeitos dos fármacos , Baço/metabolismo , Toxinas Biológicas/toxicidade , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Relação Dose-Resposta a Droga , Espectroscopia de Ressonância de Spin Eletrônica/métodos , Ativação Enzimática/efeitos dos fármacos , Raios gama , Coração/efeitos dos fármacos , Hemoglobinas/biossíntese , Injeções Intraperitoneais , Rim/efeitos dos fármacos , Rim/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Oxigênio/antagonistas & inibidores , Oxigênio/metabolismo , Tubérculos/química , Tubérculos/efeitos da radiação , Quinonas/isolamento & purificação , Ribonucleotídeo Redutases/metabolismo , Solanum tuberosum/química , Solanum tuberosum/efeitos da radiação , Baço/enzimologia , Fatores de Tempo , Toxinas Biológicas/isolamento & purificação
15.
Cancer Lett ; 250(2): 323-8, 2007 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-17137710

RESUMO

Avemar (MSC) is a nontoxic fermented wheat germ extract demonstrated to significantly improve the survival rate in patients suffering from various malignancies. We investigated its effects in human HL-60 promyelocytic leukemia cells. After 24, 48, and 72 h of incubation, Avemar inhibited the growth of HL-60 cells with IC50 values of 400, 190, and 160 microg/ml, respectively. Incubation with MSC caused dose-dependent induction of apoptosis in up to 85% of tumor cells. In addition, Avemar attenuated the progression from G2-M to G0-G1 phase of the cell cycle and was also found to significantly reduce the in situ activity of ribonucleotide reductase, the key enzyme of de novo DNA synthesis. We conclude that Avemar exerts a number of beneficial effects which could support conventional chemotherapy of human malignancies.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Extratos Vegetais/farmacologia , Ribonucleotídeo Redutases/antagonistas & inibidores , Divisão Celular/efeitos dos fármacos , Citidina/metabolismo , DNA/metabolismo , Células HL-60 , Humanos
16.
Exp Hematol ; 34(10): 1377-84, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16982330

RESUMO

OBJECTIVE: Resveratrol (3,4',5,-trihydroxystilbene, RV), an ingredient of wine, is an inhibitor of the proliferation-linked enzyme ribonucleotide reductase (RR) and shows a broad spectrum of cytotoxic effects against human cancer cells. In order to enhance these effects, we introduced additional hydroxyl moieties into the molecule. In the present study, the activity of a novel RV analog, 3,3',4,4',5,5'-hexahydroxystilbene (M8), was investigated in HL-60 human promyelocytic leukemia cells. METHODS: Cytotoxicity of M8 alone or in combination with Ara-C was assessed employing growth inhibition assays. Effects of M8 on nucleoside triphosphates (NTPs) and deoxynucleoside triphosphates (dNTPs) were examined by HPLC. The apoptotic potential of M8 and RV was compared using a specific double-staining method and inhibition of TNF-alpha-induced activation of NF-kappaB was studied. Cell-cycle distribution was analyzed by FACS. RESULTS: Addition of ascorbic acid decreased the IC(50) value of M8 from 6.25 microM to 2 microM. M8 depleted dATP and dTTP pools to 41% and 21% of control values, whereas dCTP pools increased to 199% of untreated controls. In addition, TTP, ATP, CTP, and GTP concentrations were decreased while UTP concentrations increased. M8 induced apoptosis at concentrations significantly lower than RV and could remarkably inhibit the activation of NF-kappaB. M8 arrested cells in the S phase of the cell cycle while depleting cells in the G2-M phase and exhibited synergistic combination effects when applied simultaneously with Ara-C. CONCLUSION: Due to these promising results, this novel polyhydroxylated stilbene derivative might become an additional option for the treatment of leukemia and therefore deserves further preclinical and in vivo testing.


Assuntos
Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Leucemia Promielocítica Aguda/enzimologia , Pirogalol/análogos & derivados , Ribonucleotídeo Redutases/antagonistas & inibidores , Estilbenos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Citarabina/farmacologia , Desoxirribonucleotídeos/metabolismo , Avaliação Pré-Clínica de Medicamentos , Células HL-60 , Humanos , Leucemia Promielocítica Aguda/tratamento farmacológico , NF-kappa B/metabolismo , Pirogalol/farmacologia , Resveratrol , Ribonucleotídeos/metabolismo , Estilbenos/química , Fator de Necrose Tumoral alfa/farmacologia , Vinho
17.
Br J Haematol ; 135(1): 52-61, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16925573

RESUMO

Ribonucleotide reductase (RR) is the enzyme that catalyses the rate-limiting step in DNA synthesis, the production of deoxynucleotides. RR activity is markedly elevated in tumour tissue and is crucial for cell division. It is therefore an excellent target for cancer chemotherapy. This study examined the anti-myeloma activity of Didox (3,4-Dihydroxybenzohydroxamic acid), a novel RR inhibitor (RRI). Our data showed that Didox induced caspase-dependent multiple myeloma (MM) cell apoptosis. Didox, unlike other RRIs that mainly target the pyrimidine metabolism pathway, targets both purine and pyrimidine metabolism pathways in MM, as demonstrated by transcriptional profiling using the Affymetrix U133A 2.0 gene chip. Specifically, a >or=2-fold downregulation of genes in these anabolic pathways was shown as early as 12 h after exposure to Didox. Furthermore, apoptosis was accompanied by downregulation of bcl family proteins including bcl-2, bcl(xl), and XIAP. Importantly, RR M1 component transcript was also downregulated, associated with decreased protein expression. Genes involved in DNA repair mechanisms, specifically RAD 51 homologue, were also downregulated. As Didox acts on MM cells by inhibiting DNA synthesis and repair, combination studies with melphalan, an agent commonly used in MM, were performed. A strong in vitro synergism was shown, with combination indices of <0.7 as determined by the Chou-Talalay method. These studies therefore provide the preclinical rationale for evaluation of Didox, alone and in combination with DNA-damaging agents, to improve patient outcome in MM.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Ácidos Hidroxâmicos/farmacologia , Mieloma Múltiplo/patologia , Antineoplásicos Alquilantes/farmacologia , Caspases/fisiologia , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Reparo do DNA/genética , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Melfalan/farmacologia , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ribonucleotídeo Redutases/antagonistas & inibidores , Células Tumorais Cultivadas
18.
Oncology (Williston Park) ; 18(8 Suppl 5): 5-11, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15339053

RESUMO

Several new antimetabolites, administered alone or in combination, are changing the therapeutic landscape for thoracic cancer. Two-drug combinations involving these newer drugs are becoming the standard of care for non-small-cell lung cancer (NSCLC), largely due to improvements in survival rates, time to disease progression, and response rates as well as an improved safety profile. Gemcitabine (Gemzar) has elicited considerable interest in this disease, as a combination partner in chemotherapeutic regimens. Another promising agent is pemetrexed (Alimta), a folate-based inhibitor of thymidylate synthase. In preclinical development, pemetrexed both alone and in combination with other cytotoxic agents has exhibited activity across a broad range of tumor models, including NSCLC and mesothelioma. In clinical trials of patients with NSCLC, pemetrexed has been an effective, well-tolerated agent that can be used as monotherapy or in combination with other agents at full dose. In clinical trials of patients with mesothelioma, the combination of pemetrexed and cisplatin demonstrated a significant improvement in survival, response, and patient quality-of-life parameters. The principle toxicities of pemetrexed can be minimized by folate and vitamin B12 supplements.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Desoxicitidina/análogos & derivados , Guanina/análogos & derivados , Neoplasias Pulmonares/tratamento farmacológico , Mesotelioma/tratamento farmacológico , Idoso , Desoxicitidina/administração & dosagem , Glutamatos/administração & dosagem , Guanina/administração & dosagem , Humanos , Pemetrexede , Ribonucleotídeo Redutases/antagonistas & inibidores , Timidilato Sintase/antagonistas & inibidores , Gencitabina
19.
Biotechnol Lett ; 25(2): 143-7, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12882290

RESUMO

Cell division of the wild type strain Corynebacterium (formerly Brevibacterium) ammoniagenes ATCC 6872 which requires 1 microM Mn2+ for balanced growth was inhibited by addition of 20 mM hydroxyurea (HU) or 10 mM p-methoxyphenol (MP) to a Mn2+-supplemented fermentation medium at an appropriate time. Scanning electron microscopy (SEM) showed a restricted elongation characteristic of arrest of the cell cycle in coryneform bacteria. The cultures treated with HU or MP had, respectively, a fourfold or sixfold enhanced accumulation of NAD+ by a salvage biosynthetic pathway. An assay of nucleotide-permeable cells for ribonucleotide reductase activity using [3H-CDP] as substrate revealed a pre-early and complete decline of DNA precursor biosynthesis not found in the untreated control. Overproduction of NAD+ is an alternative to the conventional fermentation process using Mn2+ deficiency. A simple model is presented to discuss the metabolic regulation of the new process based on the presence of a manganese ribonucleotide reductase (Mn-RNR) in the producing strain.


Assuntos
Corynebacterium/crescimento & desenvolvimento , Magnésio/farmacologia , NAD/biossíntese , Ribonucleotídeo Redutases/antagonistas & inibidores , Anisóis/farmacologia , Ciclo Celular/fisiologia , Corynebacterium/efeitos dos fármacos , Corynebacterium/metabolismo , Corynebacterium/ultraestrutura , Homeostase/efeitos dos fármacos , Homeostase/fisiologia , Hidroxiureia/farmacologia , Ribonucleotídeo Redutases/metabolismo
20.
Clin Breast Cancer ; 3 Suppl 1: 12-6, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12057039

RESUMO

Overexpression of the HER2/neu oncogene and receptor protein has been reported in 20%-30% of patients with breast cancer and is associated with a poor prognosis. HER2/neu expression in breast cancer patients assessed by fluorescence in situ hybridization or immunohistochemistry is a predictor for response to trastuzumab, a humanized monoclonal antibody against the HER2/neu cell-surface protein. Data regarding HER2/neu expression in lung cancer are more limited, and there is little information regarding HER2/neu expression and response to trastuzumab alone or in combination with chemotherapeutic agents. Gemcitabine is an active agent against non-small-cell lung cancer (NSCLC) and has demonstrated activity in breast cancer as well. In vitro modified tetrazolium salt growth assays were performed to determine whether the combination of trastuzumab/gemcitabine produced synergistic or additive effects on breast and lung cancer cell lines. The effects of trastuzumab alone, gemcitabine alone, and the trastuzumab/gemcitabine combination was evaluated on 4 NSCLC cell lines, 1 small-cell lung cancer (SCLC) cell line, and 2 breast cancer cell lines. HER2/neu surface protein expression was assessed by fluorescence flow cytometry and immunohistochemistry. Fluorescence in situ hybridization analysis was used to study gene expression. Trastuzumab treatment alone resulted in growth inhibition in all cell lines expressing HER2/neu and the inhibitive effect correlated with the level of cell surface HER2/neu protein expression. Treatment with gemcitabine alone resulted in growth inhibition in both breast and NSCLC cell lines. A synergistic growth inhibition effect was seen with the trastuzumab/ gemcitabine combination as indicated by combination index values < 1. The degree of synergy observed did not directly correlate with the level of surface protein expression, as synergy was seen even in cancer cell lines expressing low levels of HER2/neu. No treatment effect was seen in the SCLC cell line, which did not express HER2/neu. These preclinical studies indicate a need to study the clinical synergistic effects of the gemcitabine/trastuzumab combination in breast cancer and NSCLC patients whose tumors overexpress HER2/ neu.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/metabolismo , Desoxicitidina/análogos & derivados , Neoplasias Pulmonares/metabolismo , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Antimetabólitos Antineoplásicos/farmacologia , Antimetabólitos Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma de Células Pequenas/tratamento farmacológico , Carcinoma de Células Pequenas/metabolismo , Desoxicitidina/farmacologia , Desoxicitidina/uso terapêutico , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Feminino , Regulação Neoplásica da Expressão Gênica , Genes erbB-2/genética , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Ribonucleotídeo Redutases/antagonistas & inibidores , Trastuzumab , Células Tumorais Cultivadas/efeitos dos fármacos , Gencitabina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA