Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 260
Filtrar
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(2): 166929, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37918679

RESUMO

The kynurenine pathway (KP) is the principal metabolic route for the essential amino acid tryptophan (TRP). Recent advances have highlighted a pivotal role for several KP metabolites in inflammatory diseases, including ulcerative colitis (UC). However, the alterations of KP enzymes and their functional impact in UC remain poorly defined. Here, we focused on kynurenine 3-monooxygenase (KMO) and kynureninase (KYNU), which serve as critical branching enzymes in the KP. We observed that dextran sodium sulfate (DSS)-induced colitis mice exhibited disturbed TRP metabolism along with KMO and KYNU upregulated. In patients with active UC, both the expression of KMO and KYNU were positively correlated with inflammatory factors TNF-α and IL-1ß. Pharmacological blockade of KMO or genetic silencing of KYNU suppressed IL-1ß-triggered proinflammatory cytokines expression in intestinal epithelial cells. Furthermore, blockage of KMO by selective inhibitor Ro 61-8048 alleviated the symptoms of DSS-induced colitis in mice, accompanied by an expanded NAD+ pool and redox balance restoration. The protective role of Ro 61-8048 may be partly due to its effect on KP regulation, particularly in enhancing kynurenic acid production. In summary, our study provides new evidence for the proinflammatory property of KMO and KYNU in intestinal inflammation, hinting at a promising therapeutic approach in UC through targeting these enzymes.


Assuntos
Colite Ulcerativa , Colite , Humanos , Animais , Camundongos , Cinurenina/metabolismo , Quinurenina 3-Mono-Oxigenase/genética , Quinurenina 3-Mono-Oxigenase/metabolismo , Colite Ulcerativa/induzido quimicamente , Colite Ulcerativa/genética , Regulação para Cima , Colite/induzido quimicamente , Colite/genética , Inflamação/genética
2.
Anticancer Res ; 43(12): 5275-5282, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38030171

RESUMO

Kynurenine 3-monooxygenase (KMO), a key enzyme within the kynurenine (KYN) pathway of tryptophan (TRY) metabolism, enables the excess production of toxic metabolites (such as 3-hydroxykynurenine, xanthurenic acid, 3-hydroxyanthranilic acid and quinolinic acid), and modulates the balance between these toxic molecules and the protective metabolite, kynurenic acid (KYNA). Despite its importance, KMO suppression as a treatment for cancer has not been fully explored. Instead, researchers have focused on prevention of KYN pathway activity by inhibition of enzymes indoleamine 2,3-dioxygenase (IDO1 and IDO2) or tryptophan 2,3-dioxygenase (TDO, also known as TDO2). However, studies using IDO/TDO inhibitors against cancer have not yet shown that this type of treatment can be successful. We argue that KMO suppression can be an effective strategy for treatment of cancer by 1) decreasing toxic metabolites within the KYN pathway and 2) increasing levels of KYNA, which has important protective and anticancer properties. This strategy may be beneficial in the treatment of aggressive breast cancer, particularly in patients with triple-negative breast cancer. A major challenge to this strategy, when searching for an effective treatment for tumors, especially tumors like breast carcinoma that often metastasize to the brain, is finding KMO inhibitors that adequately cross the blood-brain barrier.


Assuntos
Quinurenina 3-Mono-Oxigenase , Neoplasias de Mama Triplo Negativas , Humanos , Quinurenina 3-Mono-Oxigenase/metabolismo , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/metabolismo , Triptofano , Cinurenina/metabolismo , Encéfalo/metabolismo , Resultado do Tratamento , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo
3.
J Cell Mol Med ; 27(16): 2290-2307, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37482908

RESUMO

Protocatechuic acid (3,4-dihydroxybenzoic acid) prevents oxidative stress, inflammation and cardiac hypertrophy. This study aimed to investigate the therapeutic effects of protocatechuic acid in an isoproterenol-induced heart failure mouse model and to identify the underlying mechanisms. To establish the heart failure model, C57BL/6NTac mice were given high-dose isoproterenol (80 mg/kg body weight) for 14 days. Echocardiography revealed that protocatechuic acid reversed the isoproterenol-induced downregulation of fractional shortening and ejection fraction. Protocatechuic acid attenuated cardiac hypertrophy as evidenced by the decreased heart-weight-to-body-weight ratio and the expression of Nppb. RNA sequencing analysis identified kynurenine-3-monooxygenase (Kmo) as a potential target of protocatechuic acid. Protocatechuic acid treatment or transfection with short-interfering RNA against Kmo ameliorated transforming growth factor ß1-induced upregulation of Kmo, Col1a1, Col1a2 and Fn1 in vivo or in neonatal rat cardiac fibroblasts. Kmo knockdown attenuated the isoproterenol-induced increase in cardiomyocyte size, as well as Nppb and Col1a1 expression in H9c2 cells or primary neonatal rat cardiomyocytes. Moreover, protocatechuic acid attenuated Kmo overexpression-induced increases in Nppb mRNA levels. Protocatechuic acid or Kmo knockdown decreased isoproterenol-induced ROS generation in vivo and in vitro. Thus, protocatechuic acid prevents heart failure by downregulating Kmo. Therefore, protocatechuic acid and Kmo constitute a potential novel therapeutic agent and target, respectively, against heart failure.


Assuntos
Insuficiência Cardíaca , Quinurenina 3-Mono-Oxigenase , Camundongos , Ratos , Animais , Isoproterenol/toxicidade , Quinurenina 3-Mono-Oxigenase/genética , Quinurenina 3-Mono-Oxigenase/metabolismo , Quinurenina 3-Mono-Oxigenase/farmacologia , Cinurenina/metabolismo , Cinurenina/farmacologia , Cinurenina/uso terapêutico , Camundongos Endogâmicos C57BL , Insuficiência Cardíaca/induzido quimicamente , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/prevenção & controle , Cardiomegalia/induzido quimicamente , Cardiomegalia/tratamento farmacológico , Cardiomegalia/prevenção & controle , Miócitos Cardíacos/metabolismo
4.
Pancreatology ; 23(6): 589-600, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37438173

RESUMO

INTRODUCTION: Infected pancreatic necrosis (IPN) is a major cause of mortality in acute pancreatitis (AP). Currently, no specific strategies are available to predict the development of IPN. Earlier we reported that persistent down-regulation of HLA-DR increases risk of developing IPN. Altered kynurenine pathway (KP) metabolites showed poor prognosis in sepsis. Here we evaluated the role of HLA-DR and KP in IPN. METHODS: Patients with ANP and healthy controls were enrolled. Demographic and clinical parameters were recorded. Circulating interleukin (IL)-8, 6, 1ß, 10, Tumor necrosis factor-α were quantified using flowcytometry. Plasma procalcitonin, endotoxin, and KP (tryptophan, kynurenine) concentrations were estimated using ELISA. qRT-PCR was conducted to evaluate mRNA expression of HLA-DR, IL-10, Toll like receptor-4 (TLR-4), and kynurenine-3-monooxygenase (KMO) genes on peripheral blood mononuclear cells. Plasma metabolites were quantified using gas chromatography mass spectrometry (GC-MS/MS). Standard statistical methods were used to compare study groups. Metaboanalyst was used to analyse/visualize the metabolomics data. RESULTS: We recruited 56 patients in Cohort-1 (IPN:26,Non-IPN:30), 78 in Cohort-2 (IPN:57,Non-IPN:21), 26 healthy controls. Increased cytokines, endotoxin, and procalcitonin were observed in patients with IPN compared to Non-IPN. HLA-DR and KMO gene expressions were significantly down-regulated in IPN groups, showed positive correlation with one another but negatively correlated with IL-6 and endotoxin concentrations. Increased IDO and decreased plasma tryptophan were observed in IPN patients. Metabolome analysis showed significant reduction in several essential amino acids including tryptophan in IPN patients. Tryptophan, at a concentration of 9 mg/ml showed an AUC of 91.9 (95%CI 86.5-97.4) in discriminating IPN. CONCLUSION: HLA-DR downregulation and KP alteration are related to IPN. The KP metabolite plasma tryptophan can act as a potential biomarker for IPN.


Assuntos
Cinurenina , Pancreatite Necrosante Aguda , Humanos , Cinurenina/metabolismo , Triptofano/metabolismo , Pró-Calcitonina , Espectrometria de Massas em Tandem , Doença Aguda , Leucócitos Mononucleares , Biomarcadores , Antígenos HLA-DR/genética , Quinurenina 3-Mono-Oxigenase/genética , Quinurenina 3-Mono-Oxigenase/metabolismo , Necrose , Endotoxinas
5.
Int J Biol Sci ; 19(10): 3077-3098, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37416768

RESUMO

Looking for early diagnostic markers and therapeutic targets is the key to ensuring prompt treatment of myocardial ischemia (MI). Here, a novel biomarker xanthurenic acid (XA) was identified based on metabolomics and exhibited high sensitivity and specificity in the diagnosis of MI patients. Additionally, the elevation of XA was proved to induce myocardial injury in vivo by promoting myocardial apoptosis and ferroptosis. Combining metabolomics and transcriptional data further revealed that kynurenine 3-monooxygenase (KMO) profoundly increased in MI mice, and was closely associated with the elevation of XA. More importantly, pharmacological or heart-specific inhibition of KMO obviously suppressed the elevation of XA and profoundly ameliorated the OGD-induced cardiomyocytes injury and the ligation-induced MI injury. Mechanistically, KMO inhibition effectively restrained myocardial apoptosis and ferroptosis by modulating mitochondrial fission and fusion. In addition, virtual screening and experimental validation were adopted to identify ginsenoside Rb3 as a novel inhibitor of KMO and exhibited great cardioprotective effects by regulating mitochondrial dynamical balance. Taken together, targeting KMO may provide a new approach for the clinical treatment of MI through maintaining mitochondrial fusion and fission balance, and ginsenoside Rb3 showed great potential to be developed as a novel therapeutic drug targeting KMO.


Assuntos
Doença da Artéria Coronariana , Isquemia Miocárdica , Camundongos , Animais , Dinâmica Mitocondrial , Quinurenina 3-Mono-Oxigenase/farmacologia , Isquemia Miocárdica/tratamento farmacológico , Miócitos Cardíacos
6.
Cell Rep ; 42(8): 112763, 2023 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-37478012

RESUMO

Kynurenine monooxygenase (KMO) blockade protects against multiple organ failure caused by acute pancreatitis (AP), but the link between KMO and systemic inflammation has eluded discovery until now. Here, we show that the KMO product 3-hydroxykynurenine primes innate immune signaling to exacerbate systemic inflammation during experimental AP. We find a tissue-specific role for KMO, where mice lacking Kmo solely in hepatocytes have elevated plasma 3-hydroxykynurenine levels that prime inflammatory gene transcription. 3-Hydroxykynurenine synergizes with interleukin-1ß to cause cellular apoptosis. Critically, mice with elevated 3-hydroxykynurenine succumb fatally earlier and more readily to experimental AP. Therapeutically, blockade with the highly selective KMO inhibitor GSK898 rescues the phenotype, reducing 3-hydroxykynurenine and protecting against critical illness and death. Together, our findings establish KMO and 3-hydroxykynurenine as regulators of inflammation and the innate immune response to sterile inflammation. During critical illness, excess morbidity and death from multiple organ failure can be rescued by systemic KMO blockade.


Assuntos
Cinurenina , Pancreatite , Camundongos , Animais , Estado Terminal , Insuficiência de Múltiplos Órgãos , Doença Aguda , Camundongos Knockout , Inflamação , Quinurenina 3-Mono-Oxigenase/genética
7.
Phytopathology ; 113(3): 484-496, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36173285

RESUMO

Sugarcane smut is a serious disease caused by Sporisorium scitamineum, which causes significant losses to the sugar industry. It is critical to reveal the molecular pathogenic mechanism of S. scitamineum to explore a new control strategy for sugarcane smut. On the basis of transcriptome sequencing data of two S. scitamineum strains with different pathogenicity, we identified the gene, SsCI51640, which was predicted to encode kynurenine 3-monooxygenase. In this study, we obtained knockout mutants and complementary mutants of this gene and identified gene function. The results showed that the sporidial growth rate and acid production ability of knockout mutants were significantly higher and stronger than those of the wild-type and complementary mutants. The growth of knockout mutants under abiotic stress (osmotic stress and cell wall stress) was significantly inhibited. In addition, the sexual mating ability and pathogenicity of knockout mutants were significantly reduced, while this phenomenon could be restored by adding exogenous cyclic adenosine monophosphate (cAMP). It is thus speculated that the SsCI51640 gene may regulate sexual mating and pathogenicity of S. scitamineum by the cAMP signaling pathway. Moreover, the SsCI51640 gene enhanced the sporidial environmental adaptability, which promoted sexual mating and development of pathogenicity. This study provides a theoretical basis for the molecular pathogenesis of S. scitamineum.


Assuntos
Basidiomycota , Saccharum , Ustilaginales , Quinurenina 3-Mono-Oxigenase/metabolismo , Doenças das Plantas , Ustilaginales/genética , Saccharum/genética
8.
Biochem Biophys Res Commun ; 629: 142-151, 2022 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-36116377

RESUMO

Phencyclidine (PCP) causes mental symptoms that closely resemble schizophrenia through the inhibition of the glutamatergic system. The kynurenine (KYN) pathway (KP) generates metabolites that modulate glutamatergic systems such as kynurenic acid (KA), quinolinic acid (QA), and xanthurenic acid (XA). Kynurenine 3-monooxygenase (KMO) metabolizes KYN to 3-hydroxykynurenine (3-HK), an upstream metabolite of QA and XA. Clinical studies have reported lower KMO mRNA and higher KA levels in the postmortem brains of patients with schizophrenia and exacerbation of symptoms in schizophrenia by PCP. However, the association between KMO deficiency and PCP remains elusive. Here, we demonstrated that a non-effective dose of PCP induced impairment of prepulse inhibition (PPI) in KMO KO mice. KA levels were increased in the prefrontal cortex (PFC) and hippocampus (HIP) of KMO KO mice, but 3-HK levels were decreased. In wild-type C57BL/6 N mice, the PPI impairment induced by PCP is exacerbated by KA, while attenuated by 3-HK, QA and XA. Taken together, KMO KO mice were vulnerable to the PPI impairment induced by PCP through an increase in KA and a decrease in 3-HK, suggesting that an increase in the ratio of KA to 3-HK (QA and XA) may play an important role in the pathophysiology of schizophrenia.


Assuntos
Quinurenina 3-Mono-Oxigenase , Cinurenina , Animais , Ácido Cinurênico/metabolismo , Cinurenina/metabolismo , Quinurenina 3-Mono-Oxigenase/genética , Quinurenina 3-Mono-Oxigenase/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fenciclidina , Inibição Pré-Pulso , Ácido Quinolínico/metabolismo , RNA Mensageiro
9.
J Cell Physiol ; 237(12): 4339-4355, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36088660

RESUMO

Kynurenine-3-monooxygenase (KMO) is a mitochondrial enzyme involved in the eukaryotic kynurenine pathway (KP), which is the major catabolic route of tryptophan. KMO can convert the substrate kynurenine into the neurotoxin 3-hydroxykynurenine and quinolinic acid, which promote the production of toxic metabolites and formation of free radical in the blood, while decrease the neuroprotective metabolite kynurenic acid. As a result of branch point, KMO is predicted as an attractive drug target for several diseases, especially neurodegenerative diseases, psychosis, and cancer. This review mainly pays attention to KMO structure and the research of mechanisms and functions, with a particular emphasis on the roles of KMO in the pathogenesis of various conditions. Furthermore, we also summarized important KMO inhibitors to supporting their effects on these diseases, indicating the prospect to find novel KMO inhibitors for diseases therapy.


Assuntos
Quinurenina 3-Mono-Oxigenase , Doenças Neurodegenerativas , Humanos , Progressão da Doença , Ácido Cinurênico/metabolismo , Cinurenina/metabolismo , Quinurenina 3-Mono-Oxigenase/química , Quinurenina 3-Mono-Oxigenase/metabolismo , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/metabolismo , Triptofano/metabolismo
10.
Biochim Biophys Acta Mol Basis Dis ; 1868(11): 166509, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-35914653

RESUMO

Type 2 diabetes is associated with an inflammatory phenotype in the pancreatic islets. We previously demonstrated that proinflammatory cytokines potently activate the tryptophan/kynurenine pathway (TKP) in INS-1 cells and in normal rat islets. Here we examined: (1) the TKP enzymes expression in the diabetic GK islets; (2) the TKP enzymes expression profiles in the GK islets before and after the onset of diabetes; (3) The glucose-stimulated insulin secretion (GSIS) in vitro in GK islets after KMO knockdown using specific morpholino-oligonucleotides against KMO or KMO blockade using the specific inhibitor Ro618048; (4) The glucose tolerance and GSIS after acute in vivo exposure to Ro618048 in GK rats. We report a remarkable induction of the kmo gene in GK islets and in human islets exposed to proinflammatory conditions. It occurred prominently in beta cells. The increased expression and activity of KMO reflected an acquired adaptation. Both KMO knockdown and specific inhibitor Ro618048 enhanced GSIS in vitro in GK islets. Moreover, acute administration of Ro618048 in vivo improved glucose tolerance, GSIS and basal blood glucose levels in GK rats. These results demonstrate that targeting islet TKP is able to correct defective GSIS. KMO inhibition could represent a potential therapeutic strategy for type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Animais , Glicemia/metabolismo , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Glucose/metabolismo , Glucose/farmacologia , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Cinurenina/metabolismo , Quinurenina 3-Mono-Oxigenase/metabolismo , Morfolinos , Ratos , Ratos Wistar , Triptofano/metabolismo
11.
Mar Biotechnol (NY) ; 24(5): 942-955, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36030481

RESUMO

The "Wanlihong" Meretrix meretrix (WLH-M) clam is a new variety of this species that has a red shell and stronger Vibrio tolerance than ordinary M. meretrix (ORI-M). To investigate the molecular mechanisms responsible for the WLH-M strain's tolerance to Vibrio, we challenged clams with Vibrio parahaemolyticus and then assessed physiological indexes and conducted transcriptome analysis and RNA interference experiments. The mortality, tissue bacterial load, and hemocyte reactive oxygen species level of ORI-M were significantly higher than those of WLH-M, whereas the content and activity of lysozyme were significantly lower. Gene Ontology functional annotation analysis and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis revealed that immune and metabolic pathways were enriched in Vibrio-challenged clams. The expressions of the heat shock protein 70 (Hsp70) and serine protease (SP) genes, which are involved in antibacterial immunity, were significantly upregulated in WLH-M but not in ORI-M, while the expression of the kynurenine 3-monooxygenase gene, a proinflammatory factor, was significantly downregulated in WLH-M. RNA interference experiments confirmed that Hsp70 and SP downregulation could result in increased mortality of WLH-M. Therefore, we speculate that Hsp70 and SP may be involved in the antibacterial immunity of WLH-M in vivo. Our data provided a valuable resource for further studies of the antibacterial mechanism of WLH-M and provided a foundation for the breeding of pathogen-resistant strains.


Assuntos
Bivalves , Vibrio parahaemolyticus , Animais , Antibacterianos , Bivalves/genética , Proteínas de Choque Térmico HSP70/genética , Imunidade Inata/genética , Quinurenina 3-Mono-Oxigenase/genética , Muramidase/genética , Interferência de RNA , RNA-Seq , Espécies Reativas de Oxigênio , Serina Proteases/genética , Vibrio parahaemolyticus/genética
12.
Neuroreport ; 33(13): 569-576, 2022 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-35894672

RESUMO

OBJECTIVES: The overactivation of the kynurenine pathway, the major metabolic pathway of tryptophan, induced by inflammation and oxidative stress, might bring about excessive neurotoxic metabolites. This study aimed to investigate whether kynurenine pathway is overactivated in blast-related traumatic brain injury (bTBI) and whether inhibitors of kynureninase and kynurenine-3-monooxygenase (KMO), important enzymes in kynurenine pathway, could alleviate bTBI in rats. METHODS: A shock tube was used to establish the bTBI animal models. Pathological changes in the hippocampus were observed using Nissl, propidium iodide and TdT-mediated dUTP Nick-End Labeling (TUNEL) staining. Immunohistochemistry was used to evaluate the expression levels of kynureninase and KMO. After the establishment of bTBI rat models, they were treated with KMO inhibitor (Ro 61-8048) and kynureninase inhibitor (benserazide hydrochloride), and the animals' behavioral performance was assessed using an elevated plus maze (EPM). RESULTS: After blast exposure, the number of neurons decreased, whereas the expression of kynureninase and KMO increased in the CA1 area of the rat hippocampus. In vitro, KMO inhibitor (Ro) and kynureninase inhibitor (benserazide hydrochloride) intervention could reduce the proportion of TUNEL-positive neurons in the hippocampus. In vivo, after Ro treatment, the behavior of the bTBI rats was significantly improved, and more neurons survived in the hippocampus CA1 region; however, following benserazide hydrochloride treatment, the behavior of bTBI rats was not significantly improved, and neuron survival could not be improved in the hippocampal CA1 region. CONCLUSION: The expression levels of KMO and kynureninase were increased in the hippocampus of the bTBI rats, suggesting that these factors might mediate the bTBI damage. Furthermore, the KMO inhibitor showed a significant protective effect on bTBI.


Assuntos
Lesões Encefálicas Traumáticas , Cinurenina , Animais , Benserazida , Quinurenina 3-Mono-Oxigenase/metabolismo , Triptofano
13.
Neuropharmacology ; 215: 109169, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35753430

RESUMO

Kynurenine pathway, a neuroimmunological pathway plays a substantial role in depression. Consistently, increased levels of neurotoxic metabolite of kynurenine pathway; quinolinic acid (QA) found in the suicidal patients and remitted major depressive patients. QA, an endogenous modulator of N-methyl-d-aspartate receptor is produced by microglial cells, may serve as a potential candidate for a link between antioxidant defence system and immune changes in depression. Further, nuclear factor (erythroid-derived 2) like 2 (Nrf2), an endogenous antioxidant transcription factor plays a significant role in maintaining antioxidant homeostasis during basal and stress conditions. The present study was designed to explore the effects of KMO-inhibition (Kynurenine monooxygenase) and association of reduced QA on Keap1/Nrf2/ARE pathway activity in olfactory bulbectomized mice (OBX-mice). KMO catalysis the neurotoxic branch of kynurenine pathway directing the synthesis of QA. KMO inhibitionshowed significant reversal of depressive-like behaviour, restored Keap-1 and Nrf2 mRNA expression, and associated antioxidant levels in cortex and hippocampus of OBX-mice. KMO inhibition also increased PI3K/AKT mRNA expression in OBX-mice. KMO inhibition and associated reduced QA significantly decreased inflammatory markers, kynurenine and increased the 5-HT, 5-HIAA and tryptophan levels in OBX-mice. Furthermore, molecular docking studies has shown good binding affinity of QA towards ubiquitin proteasome complex and PI3K protein involved in Keap-1 dependent and independent proteasome degradation of Nrf2 respectively supporting our in-vivo findings. Hence, QA might act as pro-oxidant through downregulating Nrf2/ARE pathway along with modulating other pathways and KMO inhibition could be a potential therapeutic target for depression treatment.


Assuntos
Transtorno Depressivo Maior , Ácido Quinolínico , Animais , Antioxidantes , Depressão/tratamento farmacológico , Modelos Animais de Doenças , Humanos , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Cinurenina/metabolismo , Quinurenina 3-Mono-Oxigenase/metabolismo , Camundongos , Simulação de Acoplamento Molecular , Fator 2 Relacionado a NF-E2/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ácido Quinolínico/metabolismo , RNA Mensageiro
14.
PLoS Pathog ; 18(3): e1010366, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35235615

RESUMO

Tryptophan (Trp) metabolism through the kynurenine pathway (KP) is well known to play a critical function in cancer, autoimmune and neurodegenerative diseases. However, its role in host-pathogen interactions has not been characterized yet. Herein, we identified that kynurenine-3-monooxygenase (KMO), a key rate-limiting enzyme in the KP, and quinolinic acid (QUIN), a key enzymatic product of KMO enzyme, exerted a novel antiviral function against a broad range of viruses. Mechanistically, QUIN induced the production of type I interferon (IFN-I) via activating the N-methyl-d-aspartate receptor (NMDAR) and Ca2+ influx to activate Calcium/calmodulin-dependent protein kinase II (CaMKII)/interferon regulatory factor 3 (IRF3). Importantly, QUIN treatment effectively inhibited viral infections and alleviated disease progression in mice. Furthermore, kmo-/- mice were vulnerable to pathogenic viral challenge with severe clinical symptoms. Collectively, our results demonstrated that KMO and its enzymatic product QUIN were potential therapeutics against emerging pathogenic viruses.


Assuntos
Quinurenina 3-Mono-Oxigenase , Viroses , Animais , Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Cinurenina/metabolismo , Quinurenina 3-Mono-Oxigenase/metabolismo , Camundongos , Ácido Quinolínico/metabolismo , Ácido Quinolínico/farmacologia , Viroses/tratamento farmacológico
16.
Br J Pharmacol ; 179(14): 3711-3726, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35189673

RESUMO

BACKGROUND AND PURPOSE: The kynurenine pathway has been proposed as a target for modulating drug abuse. We previously demonstrated that inhibition of kynurenine 3-monooxygenase (KMO), using Ro 61-8048, reduces ethanol consumption in a binge drinking model. Here, we investigate the effect of the kynurenine pathway modulation in ethanol-dependent mice. EXPERIMENTAL APPROACH: Adult male and female mice were subjected to a Chronic Intermittent Ethanol (CIE) paradigm. On the last day of CIE, mice were treated with Ro 61-8048, Ro 61-8048 + PNU-120596, a positive allosteric modulator of α7nAChR, and Ro 61-8048 + L-leucine or probenecid, which blocks the influx or efflux of kynurenine from the brain, respectively. Ethanol, water consumption and preference were measured and kynurenine levels in plasma and limbic forebrain were determined. KEY RESULTS: Ro 61-8048 decreases consumption and preference for ethanol in both sexes exposed to the CIE model, an effect that was prevented by PNU-120596. The Ro 61-8048-induced decrease in ethanol consumption depends on the influx of kynurenine into the brain. CONCLUSION AND IMPLICATIONS: Inhibition of KMO reduces ethanol consumption and preference in both male and female mice subjected to CIE model by a mechanism involving α7nAChR. Moreover, this centrally-mediated effect depends on the influx of peripheral kynurenine to the brain and can be prolonged by blocking the efflux of kynurenine from the brain. Here, for the first time, we demonstrate that the modulation of the kynurenine pathway is an effective strategy for the treatment of ethanol dependence in both sexes.


Assuntos
Etanol , Cinurenina , Animais , Encéfalo/metabolismo , Feminino , Cinurenina/metabolismo , Quinurenina 3-Mono-Oxigenase/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sulfonamidas , Tiazóis , Receptor Nicotínico de Acetilcolina alfa7/metabolismo
17.
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
18.
Cell Rep Med ; 2(10): 100409, 2021 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-34755129

RESUMO

Fibrosing chronic graft-versus-host disease (cGVHD) is a debilitating complication of allogeneic stem cell transplantation (alloSCT). A driver of fibrosis is the kynurenine (Kyn) pathway, and Kyn metabolism patterns and cytokines may influence cGVHD severity and manifestation (fibrosing versus gastrointestinal [GI] cGVHD). Using a liquid chromatography-tandem mass spectrometry approach on sera obtained from 425 patients with allografts, we identified high CXCL9, high indoleamine-2,3-dioxygenase (IDO) activity, and an activated Kyn pathway as common characteristics in all cGVHD subtypes. Specific Kyn metabolism patterns could be identified for non-severe cGVHD, severe GI cGVHD, and fibrosing cGVHD, respectively. Specifically, fibrosing cGVHD was associated with a distinct pathway shift toward anthranilic and kynurenic acid, correlating with reduced activity of the vitamin-B2-dependent kynurenine monooxygenase, low vitamin B6, and increased interleukin-18. The Kyn metabolite signature is a candidate biomarker for severe fibrosing cGVHD and provides a rationale for translational trials on prophylactic vitamin B2/B6 supplementation for cGVHD prevention.


Assuntos
Doença Enxerto-Hospedeiro/sangue , Ácido Cinurênico/sangue , Cinurenina/sangue , Riboflavina/sangue , Transplante de Células-Tronco , Vitamina B 6/sangue , Adolescente , Adulto , Idoso , Quimiocina CXCL9/sangue , Quimiocina CXCL9/genética , Feminino , Fibrose , Regulação da Expressão Gênica , Doença Enxerto-Hospedeiro/genética , Doença Enxerto-Hospedeiro/patologia , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/sangue , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Interleucina-18/sangue , Interleucina-18/genética , Quinurenina 3-Mono-Oxigenase/sangue , Quinurenina 3-Mono-Oxigenase/genética , Leucemia/genética , Leucemia/metabolismo , Leucemia/patologia , Leucemia/terapia , Linfoma/genética , Linfoma/metabolismo , Linfoma/patologia , Linfoma/terapia , Masculino , Redes e Vias Metabólicas/genética , Pessoa de Meia-Idade , Estudos Retrospectivos , Índice de Gravidade de Doença , Transdução de Sinais , Transplante Homólogo , Triptofano/sangue , ortoaminobenzoatos/sangue
19.
J Phys Chem A ; 125(43): 9459-9477, 2021 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-34676771

RESUMO

Recently, two new mechanistic proposals for the kynurenine 3-monooxygenase (KMO) catalyzed hydroxylation reaction of l-Kynurenine (l-Kyn) have been proposed. According to the first proposal, instead of the distal oxygen, the proximal oxygen of the hydroperoxide intermediate of flavin adenine dinucleotide (FAD) is transferred to the substrate ring. The second study proposes that l-Kyn participates in its base form in the reaction. To address these proposals, the reaction was reconsidered with a 386 atom quantum cluster model that is based on a recent X-ray structure (PDB id: 6FOX). The computations were carried out at the UB3LYP/6-311+G(2d,2p)//UB3LYP/6-31G(d,p) level with solvation (polarizable continuum model) and dispersion (DFT-D3(BJ)) corrections. To supplement the results of the density functional theory (DFT) calculations, molecular dynamics (MD) simulations of the protein-substrate complex were employed. The comparison of a proximal oxygen transfer mechanism to the distal oxygen transfer mechanism revealed that the former requires too high of a barrier energy while the latter validated our previous results. According to the MD simulations, the hydroperoxy moiety does not favor an alignment that might promote the proximal oxygen transfer mechanism. In the second part of the study, hydroxylation reaction with the base form of l-Kyn was sought. Although DFT calculations confirmed a much more facile reaction with the base form of l-Kyn, a mechanism which would allow the deprotonation of the l-Kyn before the oxygen transfer could not be determined with the X-ray-based positions. A concerted mechanism with both the oxygen transfer and the deprotonation required a high barrier energy. A stepwise mechanism involving the deprotonation of l-Kyn was found, starting from an MD frame. The overall barrier of the oxygen transfer step of this model was found to be in the range of that of with neutral l-Kyn. MD simulations supported the idea of ineffectiveness of the nearby shell surrounding the utilized active site core on the deprotonation of l-Kyn.


Assuntos
Quinurenina 3-Mono-Oxigenase/química , Quinurenina 3-Mono-Oxigenase/metabolismo , Simulação de Dinâmica Molecular , Domínio Catalítico , Hidroxilação , Cinurenina/química , Cinurenina/metabolismo , Modelos Moleculares , Estrutura Molecular , Oxirredução , Conformação Proteica
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...