Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Biomed Pharmacother ; 173: 116342, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38430635

RESUMO

BACKGROUND: Neuroinflammation is responsible for neuropsychiatric dysfunction following acute brain injury and neurodegenerative diseases. This study describes how a hypoxia-inducible factor prolyl hydroxylase (HIF-PHD) inhibitor FG-4592 prevents the lipopolysaccharide (LPS)-induced acute neuroinflammation in microglia. METHODS: The distribution of FG-4592 in mouse brain tissues was determined by collision-induced dissociation tandem mass spectrometry. Microglial activation in the hippocampus was analyzed by immunofluorescence. Moreover, we determined the activation of HIF-1 and nuclear factor-κB (NF-κB) signaling pathways, proinflammatory responses using molecular biological techniques. Transcriptome sequencing and BNIP3 silencing were conducted to explore signaling pathway and molecular mechanisms underlying FG-4592 anti-inflammatory activity. RESULTS: FG-4592 was transported into the brain tissues and LPS increased its transportation. FG-4592 promoted the expression of HIF-1α and induced the downstream gene transcription in the hippocampus. Administration with FG-4592 significantly inhibited microglial hyperactivation and decreased proinflammatory cytokine levels following LPS treatment in the hippocampus. The LPS-induced inflammatory responses and the NF-κB signaling pathway were also downregulated by FG-4592 pretreatment in microglial cells. Mechanistically, Venn diagram analysis of transcriptomic changes of BV2 cells identified that BNIP3 was a shared and common differentially expressed gene among different treatment groups. FG-4592 markedly upregulated the protein levels of BNIP3 in microglia. Importantly, BNIP3 knockdown aggravated the LPS-stimulated inflammatory responses and partially reversed the protection of FG-4592 against microglial inflammatory signaling and microglial activation in the mouse hippocampus. CONCLUSIONS: FG-4592 alleviates neuroinflammation through facilitating microglial HIF-1/BNIP3 signaling pathway in mice. Targeting HIF-PHD/HIF-1/BNIP3 axis is a promising strategy for the development of anti-neuroinflammation drugs.


Assuntos
NF-kappa B , Inibidores de Prolil-Hidrolase , Camundongos , Animais , NF-kappa B/metabolismo , Microglia/metabolismo , Inibidores de Prolil-Hidrolase/metabolismo , Doenças Neuroinflamatórias , Lipopolissacarídeos/toxicidade , Lipopolissacarídeos/metabolismo , Transdução de Sinais , Fator 1 Induzível por Hipóxia/metabolismo
2.
Gut Microbes ; 13(1): 1938380, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34190032

RESUMO

The gut microbiota is essential for human health. Microbial supply of short-chain fatty acids (SCFAs), particularly butyrate, is a well-established contributor to gut homeostasis and disease resistance. Reaching millimolar luminal concentrations, butyrate is sequestered and utilized in the colon as the favored energy source for intestinal epithelia. Given the steep oxygen gradient across the anoxic lumen and the highly oxygenated lamina propria, the colon provides a particularly interesting environment to study oxygen sensing. Previous studies have shown that the transcription factor hypoxia-inducible factor (HIF) is stabilized in healthy colonic epithelia. Here we show that butyrate directly inhibits HIF prolyl hydroxylases (PHDs) to stabilize HIF. We find that butyrate stabilizes HIF in vitro despite eliminating ß-oxidation and resultant oxygen consumption. Using recombinant PHD protein in combination with nuclear magnetic resonance and enzymatic biochemical assays, we identify butyrate to bind and function as a unique, noncompetitive inhibitor of PHDs relative to other SCFAs. Butyrate inhibited PHD with a noncompetitive Ki of 5.3 ± 0.5 mM, a physiologically relevant concentration. We also confirm that microbiota-derived butyrate is necessary to stabilize HIF in mice colonic tissue through antibiotic-induced butyrate depletion and reconstitution experiments. Our results suggest that the co-evolution of mammals and mutualistic microbiota has selected for butyrate to impact a critical gene regulation pathway that can be extended beyond the mammalian gut. As PHDs are a major target for drug development in the stabilization of HIF, butyrate holds great potential as a well-tolerated endogenous inhibitor with far-reaching therapeutic impact.


Assuntos
Bactérias/metabolismo , Butiratos/química , Colo/microbiologia , Microbioma Gastrointestinal , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia/química , Inibidores de Prolil-Hidrolase/química , Animais , Bactérias/classificação , Bactérias/genética , Butiratos/metabolismo , Colo/enzimologia , Colo/metabolismo , Ácidos Graxos Voláteis/metabolismo , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Mucosa Intestinal/enzimologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oxirredução , Oxigênio/metabolismo , Inibidores de Prolil-Hidrolase/metabolismo
3.
J Photochem Photobiol B ; 210: 111980, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32745950

RESUMO

The three hypoxia-inducible factor (HIF) prolyl-4-hydroxylase domain (PHD) 1-3 enzymes confer oxygen sensitivity to the HIF pathway and are novel therapeutic targets for treatment of renal anemia. Inhibition of the PHDs may further be beneficial in other hypoxia-associated diseases, including ischemia and chronic inflammation. Several pharmacologic PHD inhibitors (PHIs) are available, but our understanding of their selectivity and its chemical basis is limited. We here report that the PHI JNJ-42041935 (JNJ-1935) is structurally similar to the firefly luciferase substrate D-luciferin. Our results demonstrate that JNJ-1935 is a novel inhibitor of firefly luciferase enzymatic activity. In contrast, the PHIs FG-4592 (roxadustat) and FG-2216 (ICA, BIQ, IOX3, YM 311) did not affect firefly luciferase. The JNJ-1935 mode of inhibition is competitive with a Ki of 1.36 µM. D-luciferin did not inhibit the PHDs, despite its structural similarity to JNJ-1935. This study provides insights into a previously unknown JNJ-1935 off-target effect as well as into the chemical requirements for firefly luciferase and PHD inhibitors and may inform the development of novel compounds targeting these enzymes.


Assuntos
Luciferases de Vaga-Lume/metabolismo , Inibidores de Prolil-Hidrolase/química , Animais , Benzotiazóis/metabolismo , Ligação Competitiva , Vaga-Lumes/enzimologia , Glicina/análogos & derivados , Glicina/química , Glicina/metabolismo , Isoquinolinas/química , Isoquinolinas/metabolismo , Cinética , Luciferases de Vaga-Lume/antagonistas & inibidores , Luciferases de Vaga-Lume/genética , Inibidores de Prolil-Hidrolase/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Renilla/enzimologia
4.
Mol Metab ; 41: 101039, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32534258

RESUMO

OBJECTIVE: Recent evidence indicates that inhibition of prolyl hydroxylase domain (PHD) proteins can exert beneficial effects to improve metabolic abnormalities in mice and humans. However, the underlying mechanisms are not clearly understood. This study was designed to address this question. METHODS: A pan-PHD inhibitor compound was injected into WT and liver-specific hypoxia-inducible factor (HIF)-2α KO mice, after onset of obesity and glucose intolerance, and changes in glucose and glucagon tolerance were measured. Tissue-specific changes in basal glucose flux and insulin sensitivity were also measured by hyperinsulinemic euglycemic clamp studies. Molecular and cellular mechanisms were assessed in normal and type 2 diabetic human hepatocytes, as well as in mouse hepatocytes. RESULTS: Administration of a PHD inhibitor compound (PHDi) after the onset of obesity and insulin resistance improved glycemic control by increasing insulin and decreasing glucagon sensitivity in mice, independent of body weight change. Hyperinsulinemic euglycemic clamp studies revealed that these effects of PHDi treatment were mainly due to decreased basal hepatic glucose output and increased liver insulin sensitivity. Hepatocyte-specific deletion of HIF-2α markedly attenuated these effects of PHDi treatment, showing PHDi effects are HIF-2α dependent. At the molecular level, HIF-2α induced increased Irs2 and cyclic AMP-specific phosphodiesterase gene expression, leading to increased and decreased insulin and glucagon signaling, respectively. These effects of PHDi treatment were conserved in human and mouse hepatocytes. CONCLUSIONS: Our results elucidate unknown mechanisms for how PHD inhibition improves glycemic control through HIF-2α-dependent regulation of hepatic insulin and glucagon sensitivity.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Insulina/metabolismo , Inibidores de Prolil-Hidrolase/farmacologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Glucagon/metabolismo , Glucose/metabolismo , Hepatócitos/metabolismo , Humanos , Resistência à Insulina/fisiologia , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/metabolismo , Prolil Hidroxilases/metabolismo , Inibidores de Prolil-Hidrolase/metabolismo , Transdução de Sinais
5.
Chemistry ; 25(8): 2019-2024, 2019 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-30427558

RESUMO

Human prolyl hydroxylases are involved in the modification of transcription factors, procollagen, and ribosomal proteins, and are current medicinal chemistry targets. To date, there are few reports on inhibitors selective for the different types of prolyl hydroxylases. We report a structurally informed template-based strategy for the development of inhibitors selective for the human ribosomal prolyl hydroxylase OGFOD1. These inhibitors did not target the other human oxygenases tested, including the structurally similar hypoxia-inducible transcription factor prolyl hydroxylase, PHD2.


Assuntos
Prolil Hidroxilases , Inibidores de Prolil-Hidrolase , Ribossomos/efeitos dos fármacos , Proteínas de Transporte/antagonistas & inibidores , Desenho de Fármacos , Humanos , Proteínas Nucleares/antagonistas & inibidores , Prolil Hidroxilases/metabolismo , Inibidores de Prolil-Hidrolase/química , Inibidores de Prolil-Hidrolase/metabolismo , Inibidores de Prolil-Hidrolase/farmacologia , Ribossomos/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato
6.
Nephrol Ther ; 13 Suppl 1: S29-S34, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28577740

RESUMO

Hypoxia is a common clinical problem that has profound effects on renal homeostasis. Prolyl-4-hydroxylases PHD1, 2 and 3 function as oxygen sensors and control the activity of hypoxia-inducible factor (HIF), an oxygen-sensitive transcription factor that regulates a multitude of hypoxia responses, which help cells and tissues to adapt to low oxygen environments. This review provides an overview of the molecular mechanisms that govern these hypoxia responses and discusses clinical experience with compounds that inhibit prolyl-4-hydroxylases to harness HIF responses for therapy in nephrology.


Assuntos
Homeostase/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Hipóxia/metabolismo , Rim/metabolismo , Oxigênio/metabolismo , Inibidores de Prolil-Hidrolase/metabolismo , Insuficiência Renal Crônica/enzimologia , Eritropoetina/biossíntese , Humanos , Insuficiência Renal Crônica/metabolismo
7.
J Infect Dis ; 215(9): 1459-1467, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28368464

RESUMO

Our understanding of how the course of opportunistic bacterial infection is influenced by the microenvironment is limited. We demonstrate that the pathogenicity of Pseudomonas aeruginosa strains derived from acute clinical infections is higher than that of strains derived from chronic infections, where tissues are hypoxic. Exposure to hypoxia attenuated the pathogenicity of strains from acute (but not chronic) infections, implicating a role for hypoxia in regulating bacterial virulence. Mass spectrometric analysis of the secretome of P. aeruginosa derived from an acute infection revealed hypoxia-induced repression of multiple virulence factors independent of altered bacterial growth. Pseudomonas aeruginosa lacking the Pseudomonas prolyl-hydroxylase domain-containing protein, which has been implicated in bacterial oxygen sensing, displays reduced virulence factor expression. Furthermore, pharmacological hydroxylase inhibition reduces virulence factor expression and pathogenicity in a murine model of pneumonia. We hypothesize that hypoxia reduces P. aeruginosa virulence at least in part through the regulation of bacterial hydroxylases.


Assuntos
Hipóxia Celular/fisiologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/patogenicidade , Fatores de Virulência/metabolismo , ADP Ribose Transferases/metabolismo , Doença Aguda , Animais , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Microambiente Celular/fisiologia , Doença Crônica , Exotoxinas/metabolismo , Camundongos , Oxigênio/farmacologia , Prolil Hidroxilases/metabolismo , Inibidores de Prolil-Hidrolase/metabolismo , Pseudomonas aeruginosa/metabolismo , Sideróforos/metabolismo , Fatores de Virulência/análise , Exotoxina A de Pseudomonas aeruginosa
8.
Am J Physiol Regul Integr Comp Physiol ; 310(11): R1226-43, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-26936783

RESUMO

Intrauterine growth restriction induced by placental restriction (PR) in sheep leads to chronic hypoxemia and reduced surfactant maturation. The underlying molecular mechanism involves altered regulation of hypoxia signaling by increased prolyl hydroxylase domain (PHD) expression. Here, we evaluated the effect of intratracheal administration of the PHD inhibitor dimethyloxalylglycine (DMOG) on functional, molecular, and structural determinants of lung maturation in the control and PR sheep fetus. There was no effect of DMOG on fetal blood pressure or fetal breathing movements. DMOG reduced lung expression of genes regulating hypoxia signaling (HIF-3α, ACE1), antioxidant defense (CAT), lung liquid reabsorption (SCNN1-A, ATP1-A1, AQP-1, AQP-5), and surfactant maturation (SFTP-A, SFTP-B, SFTP-C, PCYT1A, LPCAT, ABCA3, LAMP3) in control fetuses. There were very few effects of DMOG on gene expression in the PR fetal lung (reduced lung expression of angiogenic factor ADM, water channel AQP-5, and increased expression of glucose transporter SLC2A1). DMOG administration in controls reduced total lung lavage phosphatidylcholine to the same degree as in PR fetuses. These changes appear to be regulated at the molecular level as there was no effect of DMOG on the percent tissue, air space, or numerical density of SFTP-B positive cells in the control and PR lung. Hence, DMOG administration mimics the effects of PR in reducing surfactant maturation in the lung of control fetuses. The limited responsiveness of the PR fetal lung suggests a potential biochemical limit or reduced plasticity to respond to changes in regulation of hypoxia signaling following exposure to chronic hypoxemia in utero.


Assuntos
Retardo do Crescimento Fetal/enzimologia , Pulmão/enzimologia , Pulmão/crescimento & desenvolvimento , Prolil Hidroxilases/metabolismo , Inibidores de Prolil-Hidrolase/metabolismo , Animais , Feminino , Idade Gestacional , Pulmão/embriologia , Prolil Hidroxilases/química , Domínios Proteicos , Espécies Reativas de Oxigênio/metabolismo , Ovinos , Relação Estrutura-Atividade
9.
J Chromatogr B Analyt Technol Biomed Life Sci ; 1009-1010: 7-16, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26688343

RESUMO

GSK1278863 is an investigative drug under investigation for treatment of anemia associated with chronic kidney disease. Its metabolism is primarily metabolized by P450 enzymes where 19 unique metabolic species have been identified. These include multiple products of mono-, di-, and tri-oxygenation. Initially, two separate and complex ultra high performance liquid chromatography (UHPLC) reverse phase methodologies were developed, validated and applied to measure parent and various predominant and circulating metabolites in numerous clinical studies. However, 5 of the 6 oxidative metabolites may exist in different stereoisomeric forms, resulting in 14 separate species; therefore a chiral methodology was required to determine which stereoisomeric forms circulated in human. A variety of conventional approaches were explored, where in the end a supercritical fluid chromatography (SFC) method was required to separate this complex mixture of 14 stereoisomeric metabolites; data from these experiments provided important information on which species circulate in human. The details of these methodologies will be discussed herein.


Assuntos
Barbitúricos/sangue , Cromatografia Líquida de Alta Pressão/métodos , Cromatografia com Fluido Supercrítico/métodos , Glicina/análogos & derivados , Inibidores de Prolil-Hidrolase/sangue , Barbitúricos/isolamento & purificação , Barbitúricos/metabolismo , Cromatografia de Fase Reversa/métodos , Sistema Enzimático do Citocromo P-450/metabolismo , Glicina/sangue , Glicina/isolamento & purificação , Glicina/metabolismo , Humanos , Oxirredução , Inibidores de Prolil-Hidrolase/isolamento & purificação , Inibidores de Prolil-Hidrolase/metabolismo , Reprodutibilidade dos Testes , Estereoisomerismo
10.
J Endod ; 41(11): 1852-61, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26395911

RESUMO

INTRODUCTION: Proangiogenic prolyl hydroxylase (PHD) inhibitors represent a novel approach to stimulate tissue regeneration. Diabetes mellitus involves the accumulation of advanced glycation end products (AGEs). Here we evaluated the impact of AGEs on the response of human pulp tissue to the PHD inhibitor L-mimosine (L-MIM) in monolayer cultures of dental pulp-derived cells (DPCs) and tooth slice organ cultures. METHODS: In monolayer cultures, DPCs were incubated with L-MIM and AGEs. Viability was assessed based on formazan formation, live-dead staining, annexin V/propidium iodide, and trypan blue exclusion assay. Vascular endothelial growth factor (VEGF), interleukin (IL)-6, and IL-8 production was evaluated by quantitative polymerase chain reaction and immunoassays. Furthermore, expression levels of odontoblast markers were assessed, and alizarin red staining was performed. Tooth slice organ cultures were performed, and VEGF, IL-6, and IL8 levels in their supernatants were measured by immunoassays. Pulp tissue vitality and morphology were assessed by MTT assay and histology. RESULTS: In monolayer cultures of DPCs, L-MIM at nontoxic concentrations increased the production of VEGF and IL-8 in the presence of AGEs. Stimulation with L-MIM decreased alkaline phosphatase levels and matrix mineralization also in the presence of AGEs, whereas no significant changes in dentin matrix protein 1 and dentin sialophosphoprotein expression were observed. In tooth slice organ cultures, L-MIM increased VEGF but not IL-6 and IL-8 production in the presence of AGEs. The pulp tissue was vital, and no signs of apoptosis or necrosis were observed. CONCLUSIONS: Overall, in the presence of AGEs, L-MIM increases the proangiogenic capacity, but decreases alkaline phosphatase expression and matrix mineralization.


Assuntos
Polpa Dentária/efeitos dos fármacos , Produtos Finais de Glicação Avançada , Mimosina/metabolismo , Inibidores de Prolil-Hidrolase/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citocinas/biossíntese , Ensaio de Imunoadsorção Enzimática , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Modelos Biológicos , Técnicas de Cultura de Órgãos
11.
Chem Commun (Camb) ; 51(53): 10730-3, 2015 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-26051901

RESUMO

We report herein that Zn(II) selectively inhibits the hypoxia-inducible factor prolyl hydroxylase PHD3 over PHD2, and does not compete with Fe(II). Independent of the oligomer formation induced by Zn(II), inhibition of the activity of PHD3 by Zn(II) involves Cys42 and Cys52 residues distantly located from the active site.


Assuntos
Prolina Dioxigenases do Fator Induzível por Hipóxia/antagonistas & inibidores , Inibidores de Prolil-Hidrolase/química , Zinco/química , Sequência de Aminoácidos , Polarização de Fluorescência , Humanos , Prolina Dioxigenases do Fator Induzível por Hipóxia/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Inibidores de Prolil-Hidrolase/metabolismo , Alinhamento de Sequência , Zinco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA