Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Am Soc Nephrol ; 33(11): 1966-1979, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36041790

RESUMO

Anemia is a common complication of chronic kidney disease; it is mainly treated with erythropoiesis-stimulating agents (ESAs) and iron. Experimental studies extensively investigated the mechanisms involved in the body's response to hypoxia and led to the discovery of the hypoxia-inducible factor (HIF) pathway and the enzymes regulating its function. HIF-prolyl-hydroxyl domain (PHD) inhibitors are a new class of oral drugs developed to treat anemia in chronic kidney disease. By inhibiting the function of PHD enzymes, they mimic the exposure to moderate hypoxia and stimulate the production of endogenous erythropoietin and very likely increase iron availability. Some data also suggest that their efficacy and, consequently, dose needs are less influenced by inflammation than ESAs. Overall, data from phases 2 and 3 clinical development showed efficacy in anemia correction and maintenance for all of the class molecules compared with placebo (superiority) or erythropoiesis-stimulating agents (noninferiority). Three molecules, roxadustat, vadadustat, and daprodustat, underwent extensive clinical investigation to assess their safety on hard cardiovascular end points, mortality, and special interest events (including cancer and thrombosis). Aside from vadadustat in the nondialysis population, at the prespecified primary analyses, all three molecules met the noninferiority margin for the risk of major cardiovascular events compared with erythropoiesis-stimulating agents or placebo. The reason for this discrepancy is difficult to explain. Other safety signals came from secondary analyses of some of the other randomized clinical trials, including a higher incidence of thrombosis. A more extensive clinical experience with post-marketing data on hard safety issues is needed to define better when and how to use HIF-PHD inhibitors compared with already available ESAs.


Assuntos
Anemia , Eritropoetina , Hematínicos , Insuficiência Renal Crônica , Humanos , Hematínicos/efeitos adversos , Prolina Dioxigenases do Fator Induzível por Hipóxia , Eritropoetina/uso terapêutico , Anemia/etiologia , Anemia/complicações , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/tratamento farmacológico , Ferro/uso terapêutico , Hipóxia
2.
Bioorg Med Chem ; 73: 117039, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36198217

RESUMO

Hypoxia-inducible factor (HIF) activators aid the treatment of renal anemia and ischemia. Recently, PyrzA (5-(1-acetyl-5-phenylpyrazolidin-3-ylidene)-1,3-dimethylbarbituric acid), a HIF activator by PHD inhibition without a 2-oxoglutarate moiety was reported. However, PyrzA has low lipophilicity, and it was necessary to improve its solubility by synthesizing derivatives. In this study, we synthesized and evaluated a higher lipophilic derivative of PyrzA and found that it exhibited higher HIF activity and stabilizing ability at low concentrations compared to Roxadustat, a commercially available HIF activator.


Assuntos
Hipóxia , Ácidos Cetoglutáricos , Humanos , Barbitúricos , Subunidade alfa do Fator 1 Induzível por Hipóxia , Prolina Dioxigenases do Fator Induzível por Hipóxia
3.
Nephrol Dial Transplant ; 36(8): 1369-1377, 2021 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-32206785

RESUMO

Erythropoiesis-stimulating agents (ESAs) are effective drugs to correct and maintain haemoglobin (Hb) levels, however, their use at doses to reach high Hb targets has been associated with an increased risk of cardiovascular adverse events, mortality and cancer. Presently used ESAs have a common mechanism of action but different pharmacokinetic and pharmacodynamic characteristics. Accordingly, the mode of activation of the erythropoietin (EPO) receptor can exert marked differences in downstream events. It is unknown whether the various ESA molecules have different efficacy/safety profiles. The relative mortality and morbidity risks associated with the use of different types of ESAs remains poorly evaluated. Recently an observational study and a randomized clinical trial provided conflicting results regarding this matter. However, these two studies displayed several differences in patient characteristics and ESA molecules used. More importantly, by definition, randomized clinical trials avoid bias by indication and suffer less from confounding factors. Therefore they bring a higher degree of evidence. The scenario becomes even more complex when considering the new class of ESAs, called prolyl-hydroxylase domain (PHD) inhibitors. They are oral drugs that mimic exposure to hypoxia and stabilize hypoxia-inducible factor α. They profoundly differ from presently used ESAs, as they have multiple targets of action, including the stimulation of endogenous EPO synthesis, direct mobilization/absorption of iron and a higher reduction of hepcidin. Accordingly, they have the potential to be more effective in inflamed patients with functional iron deficiency, i.e. the setting of patients who are at higher risk of cardiovascular events and mortality in response to present ESA use. As for ESAs, individual PHD inhibitors differ in molecular structure and degree of selectivity for the three main PHD isoforms; their efficacy and safety profiles may therefore be different from that of presently available ESAs.


Assuntos
Anemia , Eritropoetina , Hematínicos , Inibidores de Prolil-Hidrolase , Anemia/tratamento farmacológico , Anemia/etiologia , Epoetina alfa , Eritropoese , Hematínicos/efeitos adversos , Humanos , Estudos Observacionais como Assunto , Ensaios Clínicos Controlados Aleatórios como Assunto
4.
Nephrol Dial Transplant ; 35(6): 926-932, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30844045

RESUMO

Prolyl-hydroxylase (PHD) inhibitors (PHD-I) are the most appealing drugs undergoing clinical development for the treatment of anaemia in patients with chronic kidney disease. PHD inhibition mimics the exposure of the body to hypoxia and activates the hypoxia-inducible factor system. Among many other pathways, this activation promotes the production of endogenous erythropoietin (EPO) and the absorption and mobilization of iron. PHD-I are given orally and, differing from erythropoiesis-stimulating agents (ESAs), they correct and maintain haemoglobin levels by stimulating endogenous EPO production. Their efficacy and safety are supported by several Phases I and II studies with relatively short follow-up. This class of drugs has the potential to have a better safety profile than ESAs and there may be additional advantages for cardiovascular disease (CVD), osteoporosis and metabolism. However, possible adverse outcomes are feared. These span from the worsening or occurrence of new cancer, to eye complications or pulmonary hypertension. The data from the ongoing Phase III studies are awaited to better clarify the long-term safety and possible advantages of PHD-I.


Assuntos
Anemia/tratamento farmacológico , Eritropoetina/metabolismo , Hematínicos/uso terapêutico , Inibidores de Prolil-Hidrolase/uso terapêutico , Insuficiência Renal Crônica/complicações , Anemia/etiologia , Anemia/patologia , Humanos , Prognóstico
5.
Bioorg Med Chem Lett ; 28(10): 1725-1730, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29681433

RESUMO

Prolyl hydroxylase domain-containing protein (PHD) inhibitors are useful as orally administered agents for the treatment of renal anemia. Based on the common structures of known PHD inhibitors, we found novel PHD inhibitor 1 with a 2-[(4-hydroxy-6-oxo-2,3-dihydro-1H-pyridine-5-carbonyl)amino]acetic acid motif. The PHD2-inhibitory activity, lipophilicity (CLogP), and PK profiles (hepatocyte metabolism, protein binding, and/or elimination half-life) of this inhibitor were used as the evaluation index to optimize the structure and eventually discovered clinical candidate 42 as the suitable compound. Compound 42 was demonstrated to promote the production of erythropoietin (EPO) following oral administration in mice and rats. The predicted half-life of this compound in humans was 1.3-5.6 h, therefore, this drug may be expected to administer once daily with few adverse effects caused by excessive EPO production.


Assuntos
Ácido Acético/farmacologia , Anemia/tratamento farmacológico , Descoberta de Drogas , Prolina Dioxigenases do Fator Induzível por Hipóxia/antagonistas & inibidores , Inibidores de Prolil-Hidrolase/farmacologia , Insuficiência Renal Crônica/tratamento farmacológico , Ácido Acético/administração & dosagem , Ácido Acético/química , Administração Oral , Anemia/metabolismo , Animais , Cães , Relação Dose-Resposta a Droga , Humanos , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Camundongos , Estrutura Molecular , Inibidores de Prolil-Hidrolase/administração & dosagem , Inibidores de Prolil-Hidrolase/química , Ratos , Insuficiência Renal Crônica/metabolismo , Relação Estrutura-Atividade
6.
Int Endod J ; 51 Suppl 2: e146-e156, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28656722

RESUMO

AIM: To evaluate the impact of hypoxia and hypoxia mimetic agents (HMA) on the formation and activity of spheroids by dental pulp cells (DPC). METHODOLOGY: DPC on agarose-coated plates were treated with hypoxia and the HMA dimethyloxallyl glycine (DMOG), desferrioxamine (DFO) and L-mimosine (L-MIM). Images of spheroids were taken directly after seeding and at 6 h and 24 h. Spheroid sizes were quantified by area measurement with ImageJ software. Viability was assessed with Live-Dead staining, MTT and resazurin-based toxicity assay. Production of VEGF, IL-8 and SDF-1 was evaluated using immunoassays. Data were analysed using Kruskal-Wallis test and post hoc Mann-Whitney U-test. RESULTS: DPC formed spheroids in the presence of hypoxia, HMA and combined treatment with hypoxia and HMA. No pronounced difference in spheroid size was found in the groups treated with hypoxia, DMOG, DFO, L-MIM and the combination of hypoxia and the HMA relative to their normoxic controls (P > 0.05). Spheroids appeared vital in Live-Dead and MTT staining and the resazurin-based toxicity assay. Evaluation of protein production with immunoassays revealed significantly enhanced levels of VEGF and IL-8 (P < 0.05), but there was no significant effect on SDF-1 production (P > 0.05). Treatment with a combination of hypoxia and HMA did not further boost VEGF and IL-8 production (P > 0.05). CONCLUSIONS: Pre-conditioning with hypoxia and HMA increased the pro-angiogenic capacity of spheroids whilst not interfering with their formation. Pre-clinical studies will reveal whether pre-conditioning of spheroids with hypoxia and HMA can effectively improve the efficiency of cell transplantation approaches for regenerative endodontics.


Assuntos
Polpa Dentária/citologia , Esferoides Celulares/citologia , Quimiocina CXCL12/metabolismo , Desferroxamina/farmacologia , Polpa Dentária/efeitos dos fármacos , Polpa Dentária/fisiologia , Ensaio de Imunoadsorção Enzimática , Glicina/análogos & derivados , Glicina/farmacologia , Humanos , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Interleucina-8/metabolismo , Mimosina/farmacologia , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Curr Hypertens Rep ; 18(3): 23, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26894597

RESUMO

Anemia of chronic kidney disease (CKD) is common and is associated with diminished quality of life, cognitive impairment, cardiovascular morbidity, hospitalizations, and mortality. As the prevalence of end-stage renal disease continues to rise, the management of anemia represents a growing economic burden. Erythropoiesis-stimulating agents (ESA) are the mainstay of anemia management but their use is limited due to the associated cardiovascular adverse events. Prolyl hydroxylase domain enzyme (PHD) inhibitors are a new class of drugs that stabilize the hypoxia-inducible factors and are under clinical investigation for the treatment of renal anemia. The advantages of PHD inhibitors include the oral route of administration, improved iron profile, restoration of diurnal rhythm of erythropoietin secretion, and endogenous erythropoietin production near physiological range. Emerging but limited data indicates a small blood pressure lowering effect of PHD inhibitors. The effect of PHD inhibitors on cardiovascular endpoints and the potential risks of CKD progression and pulmonary hypertension remains to be addressed in the ongoing clinical trials.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Pressão Sanguínea , Hemoglobinas/metabolismo , Fator 1 Induzível por Hipóxia/metabolismo , Animais , Homeostase , Humanos , Hipertensão/tratamento farmacológico , Insuficiência Renal Crônica/complicações
8.
ChemMedChem ; : e202400504, 2024 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-39291299

RESUMO

Prolyl hydroxylase domain-containing proteins 1-3 (PHD1-3) are 2-oxoglutarate (2OG)-dependent oxygenases catalysing C-4 hydroxylation of prolyl residues in α-subunits of the heterodimeric transcription factor hypoxia-inducible factor (HIF), modifications that promote HIF-α degradation via the ubiquitin-proteasome pathway. Pharmacological inhibition of the PHDs induces HIF-α stabilisation, so promoting HIF target gene transcription. PHD inhibitors are used to treat anaemia caused by chronic kidney disease (CKD) due to their ability to stimulate erythropoietin (EPO) production. We report studies on the effects of the approved PHD inhibitors Desidustat and Enarodustat, and the clinical candidate TP0463518, on activities of a representative set of isolated recombinant human 2OG oxygenases. The three molecules manifest selectivity for PHD inhibition over that of the other 2OG oxygenases evaluated. We obtained crystal structures of Desidustat and Enarodustat in complex with the human 2OG oxygenase factor inhibiting hypoxia-inducible factor-α (FIH), which, together with modelling studies, inform on the binding modes of Desidustat and Enarodustat to active site Fe(II) in 2OG oxygenases, including PHD1-3. The results will help in the design of selective inhibitors of both the PHDs and other 2OG oxygenases, which are of medicinal interest due to their involvement inter alia in metabolic regulation, epigenetic signalling, DNA-damage repair, and agrochemical resistance.

9.
Bioorg Med Chem ; 21(21): 6349-58, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24055079

RESUMO

We have discovered a novel complex crystal structure of the PHD2 enzyme with its inhibitor, the 2,8-diazaspiro[4.5]decan-1-one analogue 4b. The widely reported salt bridge between Arg383 of the enzyme and its inhibitors in all complex structures published thus far was not observed in our case. In our complex structure compound 4b forms several novel interactions with the enzyme, which include a hydrogen bond with Arg322, a π-cation interaction with Arg322, a π-π stacking with Trp389, and a π-π stacking with His313. Guided by the structural information, SAR studies were performed on the 2,8-diazaspiro[4.5]decan-1-one series leading to the discovery of compound 9p with high potency and good oral pharmacokinetic profile in mice.


Assuntos
Compostos Aza/química , Prolina Dioxigenases do Fator Induzível por Hipóxia/antagonistas & inibidores , Inibidores de Prolil-Hidrolase/química , Piridinas/química , Administração Oral , Animais , Sítios de Ligação , Cristalografia por Raios X , Meia-Vida , Humanos , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular , Inibidores de Prolil-Hidrolase/síntese química , Inibidores de Prolil-Hidrolase/farmacocinética , Ligação Proteica , Estrutura Terciária de Proteína , Piridinas/síntese química , Piridinas/farmacocinética , Compostos de Espiro/síntese química , Compostos de Espiro/química , Compostos de Espiro/farmacocinética , Relação Estrutura-Atividade
10.
Genes (Basel) ; 13(3)2022 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-35327980

RESUMO

MAPK-organizer 1 (MORG1) is a molecular scaffold for prolyl-hydroxylase-3 containing a domain (PHD3) protein linking MORG1 to mechanisms of adaptation in hypoxic conditions. In this paper, we report the cloning of the promoter region of the murine and human MORG1 gene. Among other transcriptional factors binding sites, we identified that both (mouse and human) promoter regions contained several putative hypoxia-inducible factor binding motifs. Analyses of the human MORG1 promoter by reporter assays revealed that hypoxia and pharmacological inhibitors of prolyl-hydroxylases under in vitro conditions in HEK 293 cells differentially regulate the MORG1 promoter reporter activity. The exposure of the cells to 10% hypoxia showed inhibition of MORG1 promotor activity at 6 and 12 h, but stimulation after 24 h while treated with prolyl-hydroxylase inhibitors led to a time-independent MORG1 promoter activation. Mutational analyses of the individual HIF binding sites on human MORG1 promoter suggest that the binding sites work in a complex corporation because single mutations were not sufficient to abolish completely the MORG1 reporter activation by PHD inhibitors. Our data provide the first evidence that not only MORG1 regulate HIF stabilization through a PHD complex, but also that, vice versa, HIFs control MORG1 expression directly or indirectly by a complex regulatory mechanism.


Assuntos
Inibidores de Prolil-Hidrolase , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Clonagem Molecular , Células HEK293 , Humanos , Hipóxia/genética , Camundongos , Inibidores de Prolil-Hidrolase/farmacologia , Regiões Promotoras Genéticas
11.
ChemMedChem ; 17(1): e202100398, 2022 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-34581506

RESUMO

Studies on the inhibition of the human 2-oxoglutarate dependent oxygenase JMJD6, which is a cancer target, by 2-oxoglutarate mimics / competitors, including human drugs, drug candidates, and metabolites relevant to cancer are described. JMJD6 assays employed NMR to monitor inhibitor binding and use of mass spectrometry to monitor JMJD6-catalysed lysine hydroxylation. Notably, some clinically applied prolyl hydroxylase inhibitors also inhibit JMJD6. The results will help enable the development of inhibitors selective for human oxygenases, including JMJD6.


Assuntos
Histona Desmetilases com o Domínio Jumonji/antagonistas & inibidores , Ácidos Cetoglutáricos/farmacologia , Inibidores de Prolil-Hidrolase/farmacologia , Relação Dose-Resposta a Droga , Humanos , Histona Desmetilases com o Domínio Jumonji/metabolismo , Ácidos Cetoglutáricos/química , Estrutura Molecular , Inibidores de Prolil-Hidrolase/química , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Relação Estrutura-Atividade
12.
Cardiol Clin ; 39(3): 319-333, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34247747

RESUMO

Erythropoiesis-stimulating agents (ESAs) have improved the quality of life and reduced the need for transfusions in patients with chronic kidney disease. However, randomized trials showed no benefit but possible safety issues following high doses of ESAs given to reach normal hemoglobin levels. Iron therapy is used together with ESA; when given proactively, it may reduce the risk of mortality and cardiovascular events in hemodialysis patients. Recent trials also showed benefits of intravenous iron therapy in patients with heart failure. New drugs for correcting anemia may retain the present efficacy of ESAs as antianemic drugs and reduce cardiovascular risks.


Assuntos
Anemia , Insuficiência Cardíaca , Hematínicos , Insuficiência Renal Crônica , Anemia/tratamento farmacológico , Anemia/etiologia , Insuficiência Cardíaca/complicações , Hematínicos/uso terapêutico , Humanos , Qualidade de Vida , Insuficiência Renal Crônica/complicações
13.
J Clin Med ; 10(4)2021 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-33670704

RESUMO

Anemia is a well-known consequence of chronic kidney disease (CKD); it is mainly due to a relative insufficiency of erythropoietin synthesis by the failing kidneys. Over the years, the combination of erythropoiesis stimulating agents (ESA) and iron has become the standard of care of anemia. All ESAs effectively increase hemoglobin (Hb) levels in a substantial percentage of patients. However, in the last decade, their use has been surrounded by safety issues in increased cardiovascular risk, especially when used at high doses in inflamed and hyporesponsive patients. This has led to the definition of a more cautious Hb target. Iron deficiency is very frequent in CKD patients, with a higher frequency in non-dialysis patients. Traditionally, iron supplementation is mostly used as supportive therapy for anemia control. However, the concept is growing that intravenous iron therapy per se could be beneficial in the presence of heart failure. A new class of drugs, prolyl hydroxylase domain (PHD) inhibitors (PHD inhibitors) is becoming available for the treatment of anemia in CKD patients. Theoretically, these agents have a number of advantages, the main ones being that of stimulating the synthesis of endogenous erythropoietin and increasing iron availability. The impact of their future use in clinical practice is still to be defined. Another possible strategy could be targeting serum hepcidin and its related pathways. This possibility is fascinating from the scientific point of view, but at present its development phase is still far from clinical application.

14.
Intern Emerg Med ; 15(6): 911-915, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32617906

RESUMO

The 2019 Nobel Prize for Medicine or Physiology was assigned to three prestigious physician-scientists, Gregg L. Semenza, William G. Kaelin, and Peter J. Ratcliffe, who clarified the molecular mechanisms of hypoxia adaptation. This viewpoint traces their fundamental findings, which have paved the way for the development of innovative drugs for a wide range of common diseases, including cancer and anemia.


Assuntos
Hipóxia/fisiopatologia , Medicina/tendências , Prêmio Nobel , Fisiologia/tendências , Aniversários e Eventos Especiais , Pesquisa Biomédica/história , História do Século XX , História do Século XXI , Humanos , Medicina/instrumentação , Fisiologia/instrumentação
15.
Clin Kidney J ; 13(4): 494-499, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32905208

RESUMO

Coronavirus disease 2019 (COVID-19) is a pandemic of unprecedented severity affecting millions of people around the world and causing several hundred thousands of deaths. The presentation of the disease ranges from asymptomatic manifestations through to acute respiratory distress syndrome with the necessity of mechanical ventilation. Cytokine storm and maladaptive responses to the viral spread in the body could be responsible for the severity of disease. Many patients develop acute kidney injury (AKI) during the course of their disease, especially in more severe cases. Many factors could cause kidney damage during infection from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. It is still unclear whether direct viral damage or the overexpression of cytokines and inflammatory factors are preeminent. According to autoptic studies, in most of the cases, AKI is due proximal tubular damage. However, cases of collapsing focal segmental glomerulosclerosis were reported as well in the absence of signs of direct viral infection of the kidney. Considering that severe hypoxia is a hallmark of severe SARS-CoV-2 infection, the involvement of the hypoxia-inducible factor (HIF) system is very likely, possibly influencing the inflammatory response and outcome in both the lungs and kidneys. Several bodies of evidence have shown a possible role of the HIF pathway during AKI in various kidney disease models. Similar observations were made in the setting of acute lung injury. In both organs, HIF activation by means of inhibition of the prolyl-hydroxylases domain (PHD) could be protective. Considering these promising experimental data, we hypothesize that PHD inhibitors could be considered as a possible new therapy against severe SARS-CoV-2 infection.

16.
Kidney360 ; 1(6): 447-457, 2020 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-35368589

RESUMO

Background: Mannose-binding lectin (MBL) is an important component of innate immune defense. MBL undergoes oligomerization to generate high mol weight (HMW) forms which act as pattern recognition molecules to detect and opsonize various microorganisms. Several post-translational modifications including prolyl hydroxylation are known to affect the oligomerization of MBL. Yet, the enzyme(s) which hydroxylate proline in the collagen-like domain residues have not been identified and the significance of prolyl hydroxylation is incompletely understood. Methods: To investigate post-translational modifications of MBL, we stably expressed Myc-DDK tagged MBL in HEK293S cells. We used pharmacologic and genetic inhibition of 2-oxoglutarate-dependent dioxygenases (2OGDD) to identify the enzyme required for prolyl hydroxylation of MBL. We performed mass spectrometry to determine the effects of various inhibitors on MBL modifications. Results: Secretion of HMW MBL was impaired by inhibitors of the superfamily of 2OGDD, and was dependent on prolyl-4-hydroxylase subunit α1. Roxadustat and vadadustat, but not molidustat, led to significant suppression of hydroxylation and secretion of HMW forms of MBL. Conclusions: These data suggest that prolyl hydroxylation in the collagen-like domain of MBL is mediated by collagen prolyl-4-hydroxylase. Reduced MBL activity is likely to be an off-target effect of some, but not all, prolyl hydroxylase domain (PHD) inhibitors. There may be advantages in selective PHD inhibitors that would not interfere with MBL production.


Assuntos
Inibidores de Prolil-Hidrolase , Colágeno , Prolina Dioxigenases do Fator Induzível por Hipóxia , Pró-Colágeno-Prolina Dioxigenase , Prolil Hidroxilases , Inibidores de Prolil-Hidrolase/química
17.
ChemMedChem ; 14(1): 94-99, 2019 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-30380199

RESUMO

Prolyl hydroxylation domain (PHD) enzymes catalyze the hydroxylation of the transcription factor hypoxia-inducible factor (HIF) and serve as cellular oxygen sensors. HIF and the PHD enzymes regulate numerous potentially tissue-protective target genes which can adapt cells to metabolic and ischemic stress. We describe a fluorescent PHD inhibitor (1-chloro-4-hydroxybenzo[g]isoquinoline-3-carbonyl)glycine which is suited to fluorescence-based detection assays and for monitoring PHD inhibitors in biological systems. In cell-based assays, application of the fluorescent PHD inhibitor allowed co-localization with a cellular PHD enzyme and led to live cell imaging of processes involved in cellular oxygen sensing.


Assuntos
Benzilisoquinolinas/farmacologia , Corantes Fluorescentes/farmacologia , Imagem Molecular/métodos , Imagem Óptica/métodos , Prolil Hidroxilases/metabolismo , Inibidores de Prolil-Hidrolase/farmacologia , Benzilisoquinolinas/síntese química , Benzilisoquinolinas/química , Biocatálise/efeitos dos fármacos , Relação Dose-Resposta a Droga , Corantes Fluorescentes/síntese química , Corantes Fluorescentes/química , Células HeLa , Humanos , Estrutura Molecular , Inibidores de Prolil-Hidrolase/síntese química , Inibidores de Prolil-Hidrolase/química , Relação Estrutura-Atividade
19.
ACS Med Chem Lett ; 1(9): 526-9, 2010 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-24900242

RESUMO

HIF prolyl 4-hydroxylases (PHD) are a family of enzymes that mediate key physiological responses to hypoxia by modulating the levels of hypoxia inducible factor 1-α (HIF1α). Certain benzimidazole-2-pyrazole carboxylates were discovered to be PHD2 inhibitors using ligand- and structure-based methods and found to be potent, orally efficacious stimulators of erythropoietin secretion in vivo.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA