Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 145(6): 863-74, 2011 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-21640374

RESUMO

Metabolites in the kynurenine pathway, generated by tryptophan degradation, are thought to play an important role in neurodegenerative disorders, including Alzheimer's and Huntington's diseases. In these disorders, glutamate receptor-mediated excitotoxicity and free radical formation have been correlated with decreased levels of the neuroprotective metabolite kynurenic acid. Here, we describe the synthesis and characterization of JM6, a small-molecule prodrug inhibitor of kynurenine 3-monooxygenase (KMO). Chronic oral administration of JM6 inhibits KMO in the blood, increasing kynurenic acid levels and reducing extracellular glutamate in the brain. In a transgenic mouse model of Alzheimer's disease, JM6 prevents spatial memory deficits, anxiety-related behavior, and synaptic loss. JM6 also extends life span, prevents synaptic loss, and decreases microglial activation in a mouse model of Huntington's disease. These findings support a critical link between tryptophan metabolism in the blood and neurodegeneration, and they provide a foundation for treatment of neurodegenerative diseases.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Huntington/tratamento farmacológico , Ácido Cinurênico/análise , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Sulfonamidas/uso terapêutico , Tiazóis/uso terapêutico , Administração Oral , Doença de Alzheimer/fisiopatologia , Animais , Química Encefálica , Modelos Animais de Doenças , Feminino , Humanos , Ácido Cinurênico/sangue , Masculino , Camundongos , Camundongos Transgênicos , Sulfonamidas/administração & dosagem , Tiazóis/administração & dosagem
2.
Molecules ; 27(1)2022 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-35011505

RESUMO

Under normal physiological conditions, the kynurenine pathway (KP) plays a critical role in generating cellular energy and catabolizing tryptophan. Under inflammatory conditions, however, there is an upregulation of the KP enzymes, particularly kynurenine 3-monooxygenase (KMO). KMO has garnered much attention due to its production of toxic metabolites that have been implicated in many diseases and disorders. With many of these illnesses having an inadequate or modest treatment, there exists a need to develop KMO inhibitors that reduce the production of these toxic metabolites. Though prior efforts to find an appropriate KMO inhibitor were unpromising, the development of a KMO crystal structure has provided the opportunity for a rational structure-based design in the development of inhibitors. Therefore, the purpose of this review is to describe the kynurenine pathway, the kynurenine 3-monooxygenase enzyme, and KMO inhibitors and their potential candidacy for clinical use.


Assuntos
Desenho de Fármacos , Inibidores Enzimáticos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Quinurenina 3-Mono-Oxigenase , Cinurenina , Animais , Inibidores Enzimáticos/química , Inibidores Enzimáticos/uso terapêutico , Humanos , Inflamação/tratamento farmacológico , Inflamação/enzimologia , Cinurenina/química , Cinurenina/metabolismo , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Quinurenina 3-Mono-Oxigenase/biossíntese , Quinurenina 3-Mono-Oxigenase/química , Relação Estrutura-Atividade
3.
Arch Biochem Biophys ; 697: 108702, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33275878

RESUMO

Kynurenine-3-monooxygenase (KMO) is an important therapeutic target for several brain disorders that has been extensively studied in recent years. Potent inhibitors towards KMO have been developed and tested within different disease models, showing great therapeutic potential, especially in models of neurodegenerative disease. The inhibition of KMO reduces the production of downstream toxic kynurenine pathway metabolites and shifts the flux to the formation of the neuroprotectant kynurenic acid. However, the efficacy of KMO inhibitors in neurodegenerative disease has been limited by their poor brain permeability. Combined with virtual screening and prodrug strategies, a novel brain penetrating KMO inhibitor has been developed which dramatically decreases neurotoxic metabolites. This review highlights the importance of KMO as a drug target in neurological disease and the benefits of brain permeable inhibitors in modulating kynurenine pathway metabolites in the central nervous system.


Assuntos
Encéfalo/metabolismo , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Doenças Neurodegenerativas/tratamento farmacológico , Animais , Encéfalo/efeitos dos fármacos , Descoberta de Drogas , Inibidores Enzimáticos/uso terapêutico , Humanos , Doenças Neurodegenerativas/enzimologia , Doenças Neurodegenerativas/metabolismo
4.
Bioorg Med Chem Lett ; 44: 128115, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34015507

RESUMO

Kynurenine monooxygenase (KMO) is expected to be a good drug target to treat Huntington's disease (HD). This study presents the structure-activity relationship of pyridazine derivatives to find novel KMO inhibitors. The most promising compound 14 resolved the problematic issues of lead compound 1, i.e., metabolic instability and reactive metabolite-derived side-effects. Compound 14 exhibited high brain permeability and a long-lasting pharmacokinetics profile in monkeys, and neuroprotective kynurenic acid was increased by a single administration of 14 in R6/2 mouse brain. These results demonstrated 14 may be a potential drug candidate to treat HD.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Animais , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Humanos , Quinurenina 3-Mono-Oxigenase/metabolismo , Camundongos , Estrutura Molecular , Ratos , Relação Estrutura-Atividade
5.
Bioorg Med Chem Lett ; 33: 127753, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33359168

RESUMO

Huntington's disease (HD) is one of the serious neurodegenerative diseases and no disease modifiers are available to date. The correction of unbalanced kynurenine pathway metabolites may be useful to treat disease progression and kynurenine monooxygenase (KMO) is considered an ideal drug target. A couple of KMO inhibitors have been reported, but their brain permeability was very poor. We found pyridazinylsulfonamide as a novel lead compound, and it was optimized to the brain-permeable and highly potent KMO inhibitor 12, which was equipotent with CHDI-340246 and superior to CHDI-340246 in terms of brain penetration. Compound 12 was effective in R6/2 mice (HD model mice), i.e. neuroprotective kynurenic acid was increased, whereas neurotoxic 3-hydroxykynurenine was suppressed. In addition, impaired cognitive function was improved. Therefore, the brain-permeable KMO inhibitor was considered to be a disease modifier for HD treatment.


Assuntos
Encéfalo/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Sulfonamidas/farmacologia , Administração Oral , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/química , Quinurenina 3-Mono-Oxigenase/metabolismo , Camundongos , Camundongos Transgênicos , Estrutura Molecular , Relação Estrutura-Atividade , Sulfonamidas/administração & dosagem , Sulfonamidas/química , Benzenossulfonamidas
6.
J Immunol ; 203(4): 899-910, 2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31285277

RESUMO

The kynurenine pathway (KP) is a key regulator of many important physiological processes and plays a harmful role in cancer, many neurologic conditions, and chronic viral infections. In HIV infection, KP activity is consistently associated with reduced CD4 T cell counts and elevated levels of T cell activation and viral load; it also independently predicts mortality and morbidity from non-AIDS events. Kynurenine 3-monooxygenase (KMO) is a therapeutically important target in the KP. Using the nonhuman primate model of SIV infection in rhesus macaques, we investigated whether KMO inhibition could slow the course of disease progression. We used a KMO inhibitor, CHDI-340246, to perturb the KP during early acute infection and followed the animals for 1 y to assess clinical outcomes and immune phenotype and function during pre-combination antiretroviral therapy acute infection and combination antiretroviral therapy-treated chronic infection. Inhibition of KMO in acute SIV infection disrupted the KP and prevented SIV-induced increases in downstream metabolites, improving clinical outcome as measured by both increased CD4+ T cell counts and body weight. KMO inhibition increased naive T cell frequency and lowered PD-1 expression in naive and memory T cell subsets. Importantly, early PD-1 expression during acute SIV infection predicted clinical outcomes of body weight and CD4+ T cell counts. Our data indicate that KMO inhibition in early acute SIV infection provides clinical benefit and suggest a rationale for testing KMO inhibition as an adjunctive treatment in SIV/HIV infection to slow the progression of the disease and improve immune reconstitution.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Receptor de Morte Celular Programada 1/biossíntese , Pirimidinas/farmacologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Animais , Antirretrovirais/farmacologia , Peso Corporal/efeitos dos fármacos , Linfócitos T CD4-Positivos/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Macaca mulatta , Receptor de Morte Celular Programada 1/efeitos dos fármacos , Síndrome de Imunodeficiência Adquirida dos Símios/metabolismo
7.
Int J Mol Sci ; 22(20)2021 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-34681715

RESUMO

Accumulating evidence suggests the key role of the kynurenine pathway (KP) of the tryptophan metabolism in the pathogenesis of several diseases. Despite extensive research aimed at clarifying the mechanisms underlying the development and maintenance of neuropathic pain, the roles of KP metabolites in this process are still not fully known. Although the function of the peripheral KP has been known for several years, it has only recently been acknowledged that its metabolites within the central nervous system have remarkable consequences related to physiology and behavior. Both the products and metabolites of the KP are involved in the pathogenesis of pain conditions. Apart from the neuroactive properties of kynurenines, the KP regulates several neurotransmitter systems in direct or indirect ways. Some neuroactive metabolites are known to have neuroprotective properties (kynurenic acid, nicotinamide adenine dinucleotide cofactor), while others are toxic (3-hydroxykynurenine, quinolinic acid). Numerous animal models show that modulation of the KP may turn out to be a viable target for the treatment of diseases. Importantly, some compounds that affect KP enzymes are currently described to possess analgesic properties. Additionally, kynurenine metabolites may be useful for assessing response to therapy or as biomarkers in therapeutic monitoring. The following review describes the molecular site of action and changes in the levels of metabolites of the kynurenine pathway in the pathogenesis of various conditions, with a particular emphasis on their involvement in neuropathy. Moreover, the potential clinical implications of KP modulation in chronic pain therapy as well as the directions of new research initiatives are discussed.


Assuntos
Cinurenina/metabolismo , Neuralgia/patologia , Analgésicos/uso terapêutico , Animais , Biomarcadores/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenase/antagonistas & inibidores , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Quinurenina 3-Mono-Oxigenase/metabolismo , Redes e Vias Metabólicas/genética , Neuralgia/tratamento farmacológico , Ácido Quinolínico/química , Ácido Quinolínico/metabolismo , Ácido Quinolínico/uso terapêutico , Triptofano/metabolismo
8.
Molecules ; 26(11)2021 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-34073016

RESUMO

The pathogenesis of several neurodegenerative diseases such as Alzheimer's or Huntington's disease has been associated with metabolic dysfunctions caused by imbalances in the brain and cerebral spinal fluid levels of neuroactive metabolites. Kynurenine monooxygenase (KMO) is considered an ideal therapeutic target for the regulation of neuroactive tryptophan metabolites. Despite significant efforts, the known KMO inhibitors lack blood-brain barrier (BBB) permeability and upon the mimicking of the substrate binding mode, are subject to produce reactive oxygen species as a side reaction. The computational drug design is further complicated by the absence of complete crystal structure information for human KMO (hKMO). In the current work, we performed virtual screening of readily available compounds using several protein-ligand complex pharmacophores. Each of the pharmacophores accounts for one of three distinct reported KMO protein-inhibitor binding conformations. As a result, six novel KMO inhibitors were discovered based on an in vitro fluorescence assay. Compounds VS1 and VS6 were predicted to be BBB permeable and avoid the hydrogen peroxide production dilemma, making them valuable, novel hit compounds for further drug property optimization and advancement in the drug design pipeline.


Assuntos
Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Quinurenina 3-Mono-Oxigenase/metabolismo , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Biologia Computacional/métodos , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores Enzimáticos/farmacologia , Humanos , Cinurenina/metabolismo , Quinurenina 3-Mono-Oxigenase/química , Simulação de Acoplamento Molecular/métodos , Doenças Neurodegenerativas/tratamento farmacológico , Conformação Proteica
9.
J Biol Chem ; 294(28): 11047-11053, 2019 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-31177094

RESUMO

A key metabolic adaptation of some species that face hypoxia as part of their life cycle involves an alternative electron transport chain in which rhodoquinone (RQ) is required for fumarate reduction and ATP production. RQ biosynthesis in bacteria and protists requires ubiquinone (Q) as a precursor. In contrast, Q is not a precursor for RQ biosynthesis in animals such as parasitic helminths, and most details of this pathway have remained elusive. Here, we used Caenorhabditis elegans as a model animal to elucidate key steps in RQ biosynthesis. Using RNAi and a series of C. elegans mutants, we found that arylamine metabolites from the kynurenine pathway are essential precursors for RQ biosynthesis de novo Deletion of kynu-1, encoding a kynureninase that converts l-kynurenine (KYN) to anthranilic acid (AA) and 3-hydroxykynurenine (3HKYN) to 3-hydroxyanthranilic acid (3HAA), completely abolished RQ biosynthesis but did not affect Q levels. Deletion of kmo-1, which encodes a kynurenine 3-monooxygenase that converts KYN to 3HKYN, drastically reduced RQ but not Q levels. Knockdown of the Q biosynthetic genes coq-5 and coq-6 affected both Q and RQ levels, indicating that both biosynthetic pathways share common enzymes. Our study reveals that two pathways for RQ biosynthesis have independently evolved. Unlike in bacteria, where amination is the last step in RQ biosynthesis, in worms the pathway begins with the arylamine precursor AA or 3HAA. Because RQ is absent in mammalian hosts of helminths, inhibition of RQ biosynthesis may have potential utility for targeting parasitic infections that cause important neglected tropical diseases.


Assuntos
Caenorhabditis elegans/metabolismo , Cinurenina/metabolismo , Ubiquinona/análogos & derivados , Animais , Proteínas de Caenorhabditis elegans/antagonistas & inibidores , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Cromatografia Líquida de Alta Pressão , Hidrolases/antagonistas & inibidores , Hidrolases/genética , Hidrolases/metabolismo , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Quinurenina 3-Mono-Oxigenase/genética , Quinurenina 3-Mono-Oxigenase/metabolismo , Espectrometria de Massas , Metiltransferases/antagonistas & inibidores , Metiltransferases/genética , Metiltransferases/metabolismo , Mitocôndrias/metabolismo , Interferência de RNA , RNA de Cadeia Dupla/metabolismo , Tela Subcutânea/metabolismo , Ubiquinona/análise , Ubiquinona/biossíntese , Ubiquinona/metabolismo
10.
FASEB J ; 32(4): 2036-2045, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29208702

RESUMO

The human kynurenine 3-monooxygenase (hKMO) is a potential therapeutic target for neurodegenerative and neurologic disorders. Inhibition of KMO by Ro 61-8048, a potent, selective, and the most widely used inhibitor of KMO, was shown effective in various models of neurodegenerative or neurologic disorders. However, the molecular basis of hKMO inhibition by Ro 61-8048 is not clearly understood. Here, we report biochemistry studies on hKMO and crystal structures of an hKMO homolog, pfKMO from Pseudomonas fluorescens, in complex with the substrate l-kynurenine and Ro 61-8048. We found that the C-terminal ∼110 aa are essential for the enzymatic activity of hKMO and the homologous C-terminal region of pfKMO folds into a distinct, all-α-helical domain, which associates with the N-terminal catalytic domain to form a unique tunnel in proximity to the substrate-binding pocket. The tunnel binds the Ro 61-8048 molecule, which fills most of the tunnel, and Ro 61-8048 is hydrogen bonded with several completely conserved residues, including an essential catalytic residue. Modification of Ro 61-8048 and biochemical studies of the modified Ro 61-8048 derivatives suggested that Ro 61-8048 inhibits the enzyme in an allosteric manner by affecting the conformation of the essential catalytic residue and by blocking entry of the substrate or product release. The unique binding sites distinguish Ro 61-8048 as a noncompetitive and highly selective inhibitor from other competitive inhibitors, which should facilitate further optimization of Ro 61-8048 and the development of new inhibitory drugs to hKMO.-Gao, J., Yao, L., Xia, T., Liao, X., Zhu, D., Xiang, Y. Biochemistry and structural studies of kynurenine 3-monooxygenase reveal allosteric inhibition by Ro 61-8048.


Assuntos
Sítio Alostérico , Proteínas de Bactérias/química , Inibidores Enzimáticos/farmacologia , Quinurenina 3-Mono-Oxigenase/química , Sulfonamidas/farmacologia , Tiazóis/farmacologia , Regulação Alostérica , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/metabolismo , Inibidores Enzimáticos/química , Células HEK293 , Humanos , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Quinurenina 3-Mono-Oxigenase/metabolismo , Ligação Proteica , Pseudomonas fluorescens/enzimologia , Sulfonamidas/química , Tiazóis/química
11.
Nature ; 496(7445): 382-5, 2013 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-23575632

RESUMO

Inhibition of kynurenine 3-monooxygenase (KMO), an enzyme in the eukaryotic tryptophan catabolic pathway (that is, kynurenine pathway), leads to amelioration of Huntington's-disease-relevant phenotypes in yeast, fruitfly and mouse models, as well as in a mouse model of Alzheimer's disease. KMO is a flavin adenine dinucleotide (FAD)-dependent monooxygenase and is located in the outer mitochondrial membrane where it converts l-kynurenine to 3-hydroxykynurenine. Perturbations in the levels of kynurenine pathway metabolites have been linked to the pathogenesis of a spectrum of brain disorders, as well as cancer and several peripheral inflammatory conditions. Despite the importance of KMO as a target for neurodegenerative disease, the molecular basis of KMO inhibition by available lead compounds has remained unknown. Here we report the first crystal structure of Saccharomyces cerevisiae KMO, in the free form and in complex with the tight-binding inhibitor UPF 648. UPF 648 binds close to the FAD cofactor and perturbs the local active-site structure, preventing productive binding of the substrate l-kynurenine. Functional assays and targeted mutagenesis reveal that the active-site architecture and UPF 648 binding are essentially identical in human KMO, validating the yeast KMO-UPF 648 structure as a template for structure-based drug design. This will inform the search for new KMO inhibitors that are able to cross the blood-brain barrier in targeted therapies against neurodegenerative diseases such as Huntington's, Alzheimer's and Parkinson's diseases.


Assuntos
Ciclopropanos/química , Ciclopropanos/farmacologia , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Quinurenina 3-Mono-Oxigenase/química , Saccharomyces cerevisiae/enzimologia , Arginina/metabolismo , Barreira Hematoencefálica/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Desenho de Fármacos , Humanos , Doença de Huntington/tratamento farmacológico , Doença de Huntington/enzimologia , Cinurenina/metabolismo , Quinurenina 3-Mono-Oxigenase/metabolismo , Modelos Moleculares , Terapia de Alvo Molecular , Conformação Proteica , Reprodutibilidade dos Testes , Relação Estrutura-Atividade
12.
Cell Mol Life Sci ; 74(16): 2899-2916, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28314892

RESUMO

Kynurenine pathway (KP) is the primary path of tryptophan (Trp) catabolism in most mammalian cells. The KP generates several bioactive catabolites, such as kynurenine (Kyn), kynurenic acid (KA), 3-hydroxykynurenine (3-HK), xanthurenic acid (XA), and 3-hydroxyanthranilic acid (3-HAA). Increased catabolite concentrations in serum are associated with several cardiovascular diseases (CVD), including heart disease, atherosclerosis, and endothelial dysfunction, as well as their risk factors, including hypertension, diabetes, obesity, and aging. The first catabolic step in KP is primarily controlled by indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO). Following this first step, the KP has two major branches, one branch is mediated by kynurenine 3-monooxygenase (KMO) and kynureninase (KYNU) and is responsible for the formation of 3-HK, 3-HAA, and quinolinic acid (QA); and another branch is controlled by kynurenine amino-transferase (KAT), which generates KA. Uncontrolled Trp catabolism has been demonstrated in distinct CVD, thus, understanding the underlying mechanisms by which regulates KP enzyme expression and activity is paramount. This review highlights the recent advances on the effect of KP enzyme expression and activity in different tissues on the pathological mechanisms of specific CVD, KP is an inflammatory sensor and modulator in the cardiovascular system, and KP catabolites act as the potential biomarkers for CVD initiation and progression. Moreover, the biochemical features of critical KP enzymes and principles of enzyme inhibitor development are briefly summarized, as well as the therapeutic potential of KP enzyme inhibitors against CVD is briefly discussed.


Assuntos
Doenças Cardiovasculares/metabolismo , Cinurenina/metabolismo , Transdução de Sinais , Triptofano/metabolismo , Envelhecimento , Animais , Pressão Sanguínea/efeitos dos fármacos , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/fisiopatologia , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Diabetes Mellitus/fisiopatologia , Descoberta de Drogas , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Inibidores Enzimáticos/farmacologia , Humanos , Hidrolases/antagonistas & inibidores , Hidrolases/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Quinurenina 3-Mono-Oxigenase/metabolismo , Obesidade/metabolismo , Obesidade/patologia , Obesidade/fisiopatologia , Transdução de Sinais/efeitos dos fármacos , Transaminases/antagonistas & inibidores , Transaminases/metabolismo
13.
Bioorg Med Chem Lett ; 27(8): 1705-1708, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28302400

RESUMO

Kynurenine monooxygenase (KMO) is a potential drug target for treatment of neurodegenerative disorders such as Huntington's and Alzheimer's diseases. We have evaluated substituted kynurenines as substrates or inhibitors of KMO from Cytophaga hutchinsonii. Kynurenines substituted with a halogen at the 5-position are excellent substrates, with values of kcat and kcat/Km comparable to or higher than kynurenine. However, kynurenines substituted in the 3-position are competitive inhibitors, with KI values lower than the Km for kynurenine. Bromination also enhances inhibition, and 3,5-dibromokynurenine is a potent competitive inhibitor with a KI value of 1.5µM. A pharmacophore model of KMO was developed, and predicted that 3,4-dichlorohippuric acid would be an inhibitor. The KI for this compound was found to be 34µM, thus validating the pharmacophore model. We are using these results and our model to design more potent inhibitors of KMO.


Assuntos
Cytophaga/enzimologia , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Cinurenina/análogos & derivados , Cinurenina/farmacologia , Inibidores Enzimáticos/metabolismo , Halogenação , Humanos , Cinética , Cinurenina/metabolismo , Quinurenina 3-Mono-Oxigenase/metabolismo , Modelos Moleculares , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/enzimologia , Doenças Neurodegenerativas/metabolismo , Especificidade por Substrato
15.
Bioorg Med Chem Lett ; 26(23): 5712-5718, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27838184

RESUMO

Huntington's disease (HD) is caused by mutation of huntingtin protein (mHtt) leading to neuronal cell death. The mHtt induced toxicity can be rescued by inhibiting the kynurenine monooxygenase (KMO) enzyme. Therefore, KMO is a promising drug target to address the neurodegenerative disorders such as Huntington's diseases. Fiftysix arylpyrimidine KMO inhibitors are structurally explored through regression and classification based multi-QSAR modeling, pharmacophore mapping and molecular docking approaches. Moreover, ten new compounds are proposed and validated through the modeling that may be effective in accelerating Huntington's disease drug discovery efforts.


Assuntos
Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Doença de Huntington/tratamento farmacológico , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Pirimidinas/química , Pirimidinas/farmacologia , Teorema de Bayes , Análise Discriminante , Descoberta de Drogas , Humanos , Doença de Huntington/enzimologia , Doença de Huntington/metabolismo , Quinurenina 3-Mono-Oxigenase/metabolismo , Simulação de Acoplamento Molecular , Redes Neurais de Computação , Relação Quantitativa Estrutura-Atividade , Máquina de Vetores de Suporte
16.
Bioorg Med Chem Lett ; 22(10): 3398-401, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22525315

RESUMO

Ianthellamide A (1), a novel octopamine derivative, was isolated from the Australian marine sponge Ianthella quadrangulata. Compound 1 selectively inhibited the activity of kynurenine 3-hydroxylase with an IC(50) value of 1.5 µM. It also significantly increased the level of endogenous kynurenic acid in rat brain and hence has the potential as a neuroprotective agent in the treatment of neurodegenerative disorders.


Assuntos
Benzenossulfonatos/farmacologia , Inibidores Enzimáticos/farmacologia , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Octopamina/análogos & derivados , Poríferos/química , Animais , Concentração Inibidora 50 , Espectroscopia de Ressonância Magnética , Biologia Marinha , Octopamina/farmacologia
17.
Commun Biol ; 4(1): 159, 2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-33542467

RESUMO

The structural mechanisms of single-pass transmembrane enzymes remain elusive. Kynurenine 3-monooxygenase (KMO) is a mitochondrial protein involved in the eukaryotic tryptophan catabolic pathway and is linked to various diseases. Here, we report the mammalian full-length structure of KMO in its membrane-embedded form, complexed with compound 3 (identified internally) and compound 4 (identified via DNA-encoded chemical library screening) at 3.0 Å resolution. Despite predictions suggesting that KMO has two transmembrane domains, we show that KMO is actually a single-pass transmembrane protein, with the other transmembrane domain lying laterally along the membrane, where it forms part of the ligand-binding pocket. Further exploration of compound 3 led to identification of the brain-penetrant compound, 5. We show that KMO is dimeric, and that mutations at the dimeric interface abolish its activity. These results will provide insight for the drug discovery of additional blood-brain-barrier molecules, and help illuminate the complex biology behind single-pass transmembrane enzymes.


Assuntos
Membrana Celular/enzimologia , Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Quinurenina 3-Mono-Oxigenase/metabolismo , Animais , Sítios de Ligação , Inibidores Enzimáticos/química , Células HEK293 , Humanos , Quinurenina 3-Mono-Oxigenase/química , Quinurenina 3-Mono-Oxigenase/genética , Ligantes , Simulação de Acoplamento Molecular , Mutação , Ligação Proteica , Domínios Proteicos , Ratos , Relação Estrutura-Atividade
18.
Drug Discov Today ; 26(6): 1473-1481, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33609782

RESUMO

The novel respiratory virus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes coronavirus disease 2019 (COVID-19), emerged during late 2019 and spread rapidly across the world. It is now recognised that the nervous system can be affected in COVID-19, with several studies reporting long-term cognitive problems in patients. The metabolic pathway of tryptophan degradation, known as the kynurenine pathway (KP), is significantly activated in patients with COVID-19. KP metabolites have roles in regulating both inflammatory/immune responses and neurological functions. In this review, we speculate on the effects of KP activation in patients with COVID-19, and how modulation of this pathway might impact inflammation and reduce neurological symptoms.


Assuntos
COVID-19 , Cognição , Inflamação/metabolismo , Cinurenina/metabolismo , Sulfonamidas/farmacologia , Tiazóis/farmacologia , Triptofano/metabolismo , Animais , COVID-19/imunologia , COVID-19/psicologia , Cognição/efeitos dos fármacos , Cognição/fisiologia , Humanos , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Fármacos Neuroprotetores/farmacologia , Transdução de Sinais
19.
Immunohorizons ; 5(6): 523-534, 2021 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-34183381

RESUMO

Despite advances in our understanding of endotoxic shock, novel therapeutic interventions that can reduce the burden of sepsis remain elusive. Current treatment options are limited, and it is only through refinements in the ways that we deliver supportive care that mortality has fallen over the years. In this study, the role of kynurenine 3-monooxygenase (KMO) in immune regulation was examined in LPS-induced endotoxemia using KMO-/- and KMO+/+ mice treated with the KMO inhibitor Ro61-8048. We showed that LPS-induced or cecal ligation and puncture-induced mortality and hepatic IL-6 production increased in the absence of KMO, possibly involving increased activating transcription factor 4 (ATF4) signaling in hepatic macrophages. Moreover, treatment of septic mice with 3-hydroxykynurenine reduced mortality rates and inflammatory responses regardless of the presence or absence of KMO. According to our results, the administration of 3-hydroxykynurenine as part of the treatment approach for sepsis or as an adjuvant therapy might reduce the overproduction of IL-6, which is responsible for severe endotoxemia, and ultimately improve the survival rates of patients with sepsis.


Assuntos
Interleucina-6/metabolismo , Quinurenina 3-Mono-Oxigenase/metabolismo , Cinurenina/análogos & derivados , Choque Séptico/tratamento farmacológico , Fator 4 Ativador da Transcrição/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Cinurenina/metabolismo , Cinurenina/uso terapêutico , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Quinurenina 3-Mono-Oxigenase/genética , Lipopolissacarídeos/imunologia , Fígado/citologia , Fígado/imunologia , Fígado/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Choque Séptico/imunologia , Choque Séptico/patologia , Transdução de Sinais/genética , Sulfonamidas/farmacologia , Tiazóis/farmacologia
20.
J Neurochem ; 109(2): 316-25, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19226371

RESUMO

In the mammalian brain, kynurenine aminotransferase II (KAT II) and kynurenine 3-monooxygenase (KMO), key enzymes of the kynurenine pathway (KP) of tryptophan degradation, form the neuroactive metabolites kynurenic acid (KYNA) and 3-hydroxykynurenine (3-HK), respectively. Although physically segregated, both enzymes use the pivotal KP metabolite l-kynurenine as a substrate. We studied the functional consequences of this cellular compartmentalization in vivo using two specific tools, the KAT II inhibitor BFF 122 and the KMO inhibitor UPF 648. The acute effects of selective KAT II or KMO inhibition were studied using a radiotracing method in which the de novo synthesis of KYNA, and of 3-HK and its downstream metabolite quinolinic acid (QUIN), is monitored following an intrastriatal injection of (3)H-kynurenine. In naïve rats, intrastriatal BFF 122 decreased newly formed KYNA by 66%, without influencing 3-HK or QUIN production. Conversely, UPF 648 reduced 3-HK synthesis (by 64%) without affecting KYNA formation. Similar, selective effects of KAT II and KMO inhibition were observed when the inhibitors were applied acutely together with the excitotoxin QUIN, which impairs local KP metabolism. Somewhat different effects of KMO (but not KAT II) inhibition were obtained in rats that had received an intrastriatal QUIN injection 7 days earlier. In these neuron-depleted striata, UPF 648 not only decreased both 3-HK and QUIN production (by 77% and 66%, respectively) but also moderately raised KYNA synthesis (by 27%). These results indicate a remarkable functional segregation of the two pathway branches in the brain, boding well for the development of selective KAT II or KMO inhibitors for cognitive enhancement and neuroprotection, respectively.


Assuntos
Encéfalo/metabolismo , Quinurenina 3-Mono-Oxigenase/química , Cinurenina/metabolismo , Transdução de Sinais/fisiologia , Transaminases/química , Animais , Encéfalo/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Cinurenina/química , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Quinurenina 3-Mono-Oxigenase/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transaminases/antagonistas & inibidores , Transaminases/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA