Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Métodos Terapêuticos e Terapias MTCI
Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 116(49): 24819-24829, 2019 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-31740620

RESUMO

Chronic gut inflammatory diseases are associated with disruption of intestinal epithelial barriers and impaired mucosal immunity. HIV-1 (HIV) causes depletion of mucosal CD4+ T cells early in infection and disruption of gut epithelium, resulting in chronic inflammation and immunodeficiency. Although antiretroviral therapy (ART) is effective in suppressing viral replication, it is incapable of restoring the "leaky gut," which poses an impediment for HIV cure efforts. Strategies are needed for rapid repair of the epithelium to protect intestinal microenvironments and immunity in inflamed gut. Using an in vivo nonhuman primate intestinal loop model of HIV/AIDS, we identified the pathogenic mechanism underlying sustained disruption of gut epithelium and explored rapid repair of gut epithelium at the intersection of microbial metabolism. Molecular, immunological, and metabolomic analyses revealed marked loss of peroxisomal proliferator-activated receptor-α (PPARα) signaling, predominant impairment of mitochondrial function, and epithelial disruption both in vivo and in vitro. To elucidate pathways regulating intestinal epithelial integrity, we introduced probiotic Lactobacillus plantarum into Simian immunodeficiency virus (SIV)-inflamed intestinal lumen. Rapid recovery of the epithelium occurred within 5 h of L. plantarum administration, independent of mucosal CD4+ T cell recovery, and in the absence of ART. This intestinal barrier repair was driven by L. plantarum-induced PPARα activation and restoration of mitochondrial structure and fatty acid ß-oxidation. Our data highlight the critical role of PPARα at the intersection between microbial metabolism and epithelial repair in virally inflamed gut and as a potential mitochondrial target for restoring gut barriers in other infectious or gut inflammatory diseases.


Assuntos
Metabolismo Energético/fisiologia , Microbioma Gastrointestinal/fisiologia , Intestinos/imunologia , Intestinos/microbiologia , Mitocôndrias/metabolismo , PPAR alfa/metabolismo , Animais , Terapia Antirretroviral de Alta Atividade , Linfócitos T CD4-Positivos/imunologia , Modelos Animais de Doenças , Metabolismo Energético/efeitos dos fármacos , Epitélio/imunologia , Infecções por HIV , Humanos , Imunidade nas Mucosas , Interleucina-1beta/metabolismo , Intestinos/patologia , Lactobacillus plantarum/fisiologia , Macaca mulatta , Masculino , Metabolômica , Mitocôndrias/efeitos dos fármacos , Probióticos/administração & dosagem , Probióticos/uso terapêutico , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia
2.
PLoS One ; 14(10): e0223209, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31665133

RESUMO

Friedreich's ataxia (FA) is a neurodegenerative disease with no approved therapy that is the result of frataxin deficiency. The identification of human FA blood biomarkers related to disease severity and neuro-pathomechanism could support clinical trials of drug efficacy. To try to identify human biomarkers of neuro-pathomechanistic relevance, we compared the overlapping gene expression changes of primary blood and skin cells of FA patients with changes in the Dorsal Root Ganglion (DRG) of the KIKO FA mouse model. As DRG is the primary site of neurodegeneration in FA, our goal was to identify which changes in blood and skin of FA patients provide a 'window' into the FA neuropathomechanism inside the nervous system. In addition, gene expression in frataxin-deficient neuroglial cells and FA mouse hearts were compared for a total of 5 data sets. The overlap of these changes strongly supports mitochondrial changes, apoptosis and alterations of selenium metabolism. Consistent biomarkers were observed, including three genes of mitochondrial stress (MTIF2, ENO2), apoptosis (DDIT3/CHOP), oxidative stress (PREX1), and selenometabolism (SEPW1). These results prompted our investigation of the GPX1 activity as a marker of selenium and oxidative stress, in which we observed a significant change in FA patients. We believe these lead biomarkers that could be assayed in FA patient blood as indicators of disease severity and progression, and also support the involvement of mitochondria, apoptosis and selenium in the neurodegenerative process.


Assuntos
Biomarcadores/sangue , Ataxia de Friedreich/sangue , Gânglios Espinais/metabolismo , Estresse Oxidativo/genética , Animais , Antioxidantes/metabolismo , Apoptose/genética , Modelos Animais de Doenças , Fatores de Iniciação em Eucariotos/sangue , Ataxia de Friedreich/genética , Ataxia de Friedreich/patologia , Gânglios Espinais/patologia , Regulação da Expressão Gênica/genética , Fatores de Troca do Nucleotídeo Guanina/sangue , Humanos , Proteínas de Ligação ao Ferro/genética , Camundongos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/sangue , Miocárdio/metabolismo , Selênio/metabolismo , Fator de Transcrição CHOP/sangue , Frataxina
3.
Toxicology ; 393: 123-139, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29141199

RESUMO

Menadione, also known as vitamin K3, is a 2-methyl-1,4 naphthoquinone with a potent cytotoxic activity mainly resulting from its quinone redox-cycling with production of reactive oxygen species (ROS). Although increased ROS generation is considered a relevant mechanism in cancer cell death, it may not be sufficiently effective to kill cancer cells due to phenotypic adaptations. Therefore, combining ROS-generating agents with other molecules targeting important cancer cell phenotypes can be an effective therapeutic strategy. As mitochondrial dysfunction has been implicated in many human diseases, including cancer, we describe here the discovery of a mitochondrial-directed agent (MitoK3), which was developed by conjugating a TPP cation to the C3 position of the menadione's naphthoquinone ring, increasing its selective accumulation in mitochondria, as well as led to alterations of its redox properties and consequent biological outcome. MitoK3 disturbed the mitochondrial bioenergetic apparatus, with subsequent loss of mitochondrial ATP production. The combinatory strategy of MitoK3 with anticancer agent doxorubicin (DOX) resulted in a degree of cytotoxicity higher than those of the individual molecules, as the combination triggered tumour apoptotic cell death evident by caspase 3/9 activities, probably through mitochondrial destabilization or by interference with mitochondrial redox processes. The results of this investigation support the importance of drug discovery process in developing molecules that can be use as adjuvant therapy in patients with specific cancer subtypes.


Assuntos
Adjuvantes Farmacêuticos/farmacologia , Antineoplásicos/farmacologia , Doxorrubicina/farmacologia , Mitocôndrias/efeitos dos fármacos , Vitamina K 3/análogos & derivados , Vitamina K 3/farmacologia , Células A549 , Trifosfato de Adenosina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 9/metabolismo , Linhagem Celular , Respiração Celular/efeitos dos fármacos , Células Hep G2 , Humanos , Células MCF-7 , Masculino , Mitocôndrias/metabolismo , Oxirredução , Consumo de Oxigênio , Ratos
4.
Mitochondrion ; 30: 177-86, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27497748

RESUMO

Inherited mitochondrial complex I mutations cause blinding Leber's hereditary optic neuropathy (LHON), for which no curative therapy exists. A specific biochemical consequence of LHON mutations in the presence of trace rotenone was observed: deficient complex I-dependent ATP synthesis (CIDAS) and mitochondrial O2 consumption, proportional to the clinical severity of the three primary LHON mutations. We optimized a high-throughput assay of CIDAS to screen 1600 drugs to 2, papaverine and zolpidem, which protected CIDAS in LHON cells concentration-dependently. TSPO and cAMP were investigated as protective mechanisms, but a conclusive mechanism remains to be elucidated; next steps include testing in animal models.


Assuntos
Trifosfato de Adenosina/biossíntese , Complexo I de Transporte de Elétrons/metabolismo , Redes e Vias Metabólicas/efeitos dos fármacos , Atrofia Óptica Hereditária de Leber/tratamento farmacológico , Papaverina/metabolismo , Piridinas/metabolismo , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Humanos , Zolpidem
5.
Anal Biochem ; 434(1): 105-11, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23219563

RESUMO

The EPXH2 gene encodes soluble epoxide hydrolase (sEH), which has two distinct enzyme activities: epoxide hydrolase (Cterm-EH) and phosphatase (Nterm-phos). The Cterm-EH is involved in the metabolism of arachidonic acid epoxides that play important roles in blood pressure, cell growth, inflammation, and pain. While recent findings suggested complementary biological roles for Nterm-phos, research is limited by the lack of potent bioavailable inhibitors of this phosphatase activity. Also, a potent bioavailable inhibitor of this activity could be important in the development of therapy for cardiovascular diseases. We report herein the development of an HTS enzyme-based assay for Nterm-phos (Z'>0.9) using AttoPhos as the substrate. This assay was used to screen a wide variety of chemical entities, including a library of known drugs that have reached through clinical evaluation (Pharmakon 1600), as well as a library of pesticides and environmental toxins. We discovered that ebselen inhibits sEH phosphatase activity. Ebselen binds to the N-terminal domain of sEH (K(I)=550 nM) and chemically reacts with the enzyme to quickly and irreversibly inhibit Nterm-phos, and subsequently Cterm-EH, and thus represents a new class of sEH inhibitor.


Assuntos
Epóxido Hidrolases/análise , Ensaios de Triagem em Larga Escala , Bibliotecas de Moléculas Pequenas/química , Azóis/metabolismo , Epóxido Hidrolases/genética , Epóxido Hidrolases/metabolismo , Humanos , Isoindóis , Cinética , Compostos Organosselênicos/metabolismo , Praguicidas/metabolismo , Ligação Proteica , Proteínas Recombinantes/análise , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Especificidade por Substrato , Toxinas Biológicas/metabolismo
6.
Hum Mol Genet ; 16(8): 929-41, 2007 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17331979

RESUMO

The neurodegenerative disorder Friedreich's ataxia (FRDA) is caused by mutations in frataxin, a mitochondrial protein whose function remains controversial. Using co-immunoprecipitation and mass spectrometry we identified multiple interactors of mitochondrial frataxin in mammalian cells. One interactor was mortalin/GRP75, a homolog of the yeast ssq1 chaperone that integrates iron-sulfur clusters into imported mitochondrial proteins. Another interactor was ISD11, recently identified as a component of the eukaryotic complex Nfs1/ISCU, an essential component of iron-sulfur cluster biogenesis. Interactions between frataxin and ISD11, and frataxin and GRP75 were confirmed by co-immunoprecipitation experiments in both directions. Immunofluorescence analysis demonstrated that ISD11 co-localized with both frataxin and with mitochondria. The point mutations I154F and W155R in frataxin cause FRDA and are clustered to one surface of the protein, and these mutations decrease the interaction of frataxin with ISD11. The frataxin/ISD11 interaction was also decreased by the chelator EDTA, and was increased by supplementation with nickel but not other metal ions. Nickel supplementation rescued the defective interaction of mutant frataxin I154F and W155R with ISD11. Upon ISD11 depletion by siRNA in HEK293T cells, the amount of the Nfs1/ISCU protein complex declined, as did the activity of the iron-sulfur cluster enzyme aconitase, while the cellular iron content was increased, as seen in tissues from FRDA patients. Furthermore, ISD11 mRNA levels were decreased in FRDA patient cells. These data suggest that frataxin binds the iron-sulfur biogenesis Nfs1/ISCU complex through ISD11, that the interaction is nickel-dependent, and that multiple consequences of frataxin deficiency are duplicated by ISD11 deficiency.


Assuntos
Liases de Carbono-Enxofre/metabolismo , Proteínas de Ligação ao Ferro/metabolismo , Proteínas Ferro-Enxofre/metabolismo , Proteínas Mitocondriais/metabolismo , Chaperonas Moleculares/metabolismo , Aconitato Hidratase/genética , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Ataxia de Friedreich/genética , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Ferro/metabolismo , Proteínas de Ligação ao Ferro/genética , Proteínas de Membrana/metabolismo , Modelos Biológicos , Complexos Multiproteicos/metabolismo , Níquel/fisiologia , Ligação Proteica , Transporte Proteico , Frataxina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA