Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 170
Filtrar
1.
Int J Oncol ; 64(6)2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38757341

RESUMEN

Ferroptosis, a recently discovered type of programmed cell death triggered by excessive accumulation of iron­dependent lipid peroxidation, is linked to several malignancies, including non­small cell lung cancer. Long non­coding RNAs (lncRNAs) are involved in ferroptosis; however, data on their role and mechanism in cancer therapy remains limited. Therefore, the aim of the present study was to identify ferroptosis­associated mRNAs and lncRNAs in A549 lung cancer cells treated with RAS­selective lethal 3 (RSL3) and ferrostatin­1 (Fer­1) using RNA sequencing. The results demonstrated that lncRNA lung cancer­associated transcript 1 (LUCAT1) was significantly upregulated in lung adenocarcinoma and lung squamous cell carcinoma tissues. Co­expression analysis of differentially expressed mRNAs and lncRNAs suggested that LUCAT1 has a crucial role in ferroptosis. LUCAT1 expression was markedly elevated in A549 cells treated with RSL3, which was prevented by co­incubation with Fer­1. Functionally, overexpression of LUCAT1 facilitated cell proliferation and reduced the occurrence of ferroptosis induced by RSL3 and Erastin, while inhibition of LUCAT1 expression reduced cell proliferation and increased ferroptosis. Mechanistically, downregulation of LUCAT1 resulted in the downregulation of both GTP cyclohydrolase 1 (GCH1) and ferroptosis suppressor protein 1 (FSP1). Furthermore, inhibition of LUCAT1 expression upregulated microRNA (miR)­34a­5p and then downregulated GCH1. These results indicated that inhibition of LUCAT1 expression promoted ferroptosis by modulating the downregulation of GCH1, mediated by miR­34a­5p. Therefore, the combination of knocking down LUCAT1 expression with ferroptosis inducers may be a promising strategy for lung cancer treatment.


Asunto(s)
Regulación hacia Abajo , Ferroptosis , GTP Ciclohidrolasa , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares , MicroARNs , ARN Largo no Codificante , Humanos , Ferroptosis/genética , MicroARNs/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Células A549 , GTP Ciclohidrolasa/genética , GTP Ciclohidrolasa/metabolismo , Proliferación Celular , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Masculino , Línea Celular Tumoral , Femenino , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Adenocarcinoma del Pulmón/metabolismo
2.
Exp Mol Med ; 56(5): 1107-1122, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38689083

RESUMEN

Genotoxic therapy triggers reactive oxygen species (ROS) production and oxidative tissue injury. S-nitrosylation is a selective and reversible posttranslational modification of protein thiols by nitric oxide (NO), and 5,6,7,8-tetrahydrobiopterin (BH4) is an essential cofactor for NO synthesis. However, the mechanism by which BH4 affects protein S-nitrosylation and ROS generation has not been determined. Here, we showed that ionizing radiation disrupted the structural integrity of BH4 and downregulated GTP cyclohydrolase I (GCH1), which is the rate-limiting enzyme in BH4 biosynthesis, resulting in deficiency in overall protein S-nitrosylation. GCH1-mediated BH4 synthesis significantly reduced radiation-induced ROS production and fueled the global protein S-nitrosylation that was disrupted by radiation. Likewise, GCH1 overexpression or the administration of exogenous BH4 protected against radiation-induced oxidative injury in vitro and in vivo. Conditional pulmonary Gch1 knockout in mice (Gch1fl/fl; Sftpa1-Cre+/- mice) aggravated lung injury following irradiation, whereas Gch1 knock-in mice (Gch1lsl/lsl; Sftpa1-Cre+/- mice) exhibited attenuated radiation-induced pulmonary toxicity. Mechanistically, lactate dehydrogenase (LDHA) mediated ROS generation downstream of the BH4/NO axis, as determined by iodoacetyl tandem mass tag (iodoTMT)-based protein quantification. Notably, S-nitrosylation of LDHA at Cys163 and Cys293 was regulated by BH4 availability and could restrict ROS generation. The loss of S-nitrosylation in LDHA after irradiation increased radiosensitivity. Overall, the results of the present study showed that GCH1-mediated BH4 biosynthesis played a key role in the ROS cascade and radiosensitivity through LDHA S-nitrosylation, identifying novel therapeutic strategies for the treatment of radiation-induced lung injury.


Asunto(s)
Biopterinas , GTP Ciclohidrolasa , Lesión Pulmonar , Especies Reactivas de Oxígeno , Animales , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Ratones , Lesión Pulmonar/metabolismo , Lesión Pulmonar/etiología , GTP Ciclohidrolasa/metabolismo , GTP Ciclohidrolasa/genética , Humanos , Tolerancia a Radiación/genética , Lactato Deshidrogenasa 5/metabolismo , Ratones Noqueados , Óxido Nítrico/metabolismo , L-Lactato Deshidrogenasa/metabolismo , L-Lactato Deshidrogenasa/genética , Procesamiento Proteico-Postraduccional , Radiación Ionizante
3.
Int J Mol Sci ; 24(2)2023 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-36674724

RESUMEN

The GTP cyclohydrolase 1 enzyme (GTPCH1) is the rate-limiting enzyme of the tetrahydrobiopterin (BH4) biosynthetic pathway. Physiologically, BH4 plays a crucial role as an essential cofactor for the production of catecholamine neurotransmitters, including epinephrine, norepinephrine and dopamine, as well as the gaseous signaling molecule, nitric oxide. Pathological levels of the cofactor have been reported in a number of disease states, such as inflammatory conditions, neuropathic pain and cancer. Targeting the GTPCH1 enzyme has great potential in the management of a number of disease pathologies associated with dysregulated BH4 physiology. This study is an in silico investigation of the human GTPCH1 enzyme using virtual screening and molecular dynamic simulation to identify molecules that can be repurposed to therapeutically target the enzyme. A three-tier molecular docking protocol was employed in the virtual screening of a comprehensive library of over 7000 approved medications and nutraceuticals in order to identify hit compounds capable of binding to the GTPCH1 binding pocket with the highest affinity. Hit compounds were further verified by molecular dynamic simulation studies to provide a detailed insight regarding the stability and nature of the binding interaction. In this study, we identify a number of drugs and natural compounds with recognized anti-inflammatory, analgesic and cytotoxic effects, including the aminosalicylate olsalazine, the antiepileptic phenytoin catechol, and the phlorotannins phlorofucofuroeckol and eckol. Our results suggest that the therapeutic and clinical effects of hit compounds may be partially attributed to the inhibition of the GTPCH1 enzyme. Notably, this study offers an understanding of the off-target effects of a number of compounds and advocates the potential role of aminosalicylates in the regulation of BH4 production in inflammatory disease states. It highlights an in silico drug repurposing approach to identify a potential means of safely targeting the BH4 biosynthetic pathway using established therapeutic agents.


Asunto(s)
GTP Ciclohidrolasa , Neuralgia , Humanos , GTP Ciclohidrolasa/genética , GTP Ciclohidrolasa/química , Biopterinas/farmacología , Reposicionamiento de Medicamentos , Simulación del Acoplamiento Molecular , Neuralgia/metabolismo , Óxido Nítrico/metabolismo
4.
Sci Transl Med ; 14(660): eabj1531, 2022 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-36044597

RESUMEN

Increased tetrahydrobiopterin (BH4) generated in injured sensory neurons contributes to increased pain sensitivity and its persistence. GTP cyclohydrolase 1 (GCH1) is the rate-limiting enzyme in the de novo BH4 synthetic pathway, and human single-nucleotide polymorphism studies, together with mouse genetic modeling, have demonstrated that decreased GCH1 leads to both reduced BH4 and pain. However, little is known about the regulation of Gch1 expression upon nerve injury and whether this could be modulated as an analgesic therapeutic intervention. We performed a phenotypic screen using about 1000 bioactive compounds, many of which are target-annotated FDA-approved drugs, for their effect on regulating Gch1 expression in rodent injured dorsal root ganglion neurons. From this approach, we uncovered relevant pathways that regulate Gch1 expression in sensory neurons. We report that EGFR/KRAS signaling triggers increased Gch1 expression and contributes to neuropathic pain; conversely, inhibiting EGFR suppressed GCH1 and BH4 and exerted analgesic effects, suggesting a molecular link between EGFR/KRAS and pain perception. We also show that GCH1/BH4 acts downstream of KRAS to drive lung cancer, identifying a potentially druggable pathway. Our screen shows that pharmacologic modulation of GCH1 expression and BH4 could be used to develop pharmacological treatments to alleviate pain and identified a critical role for EGFR-regulated GCH1/BH4 expression in neuropathic pain and cancer in rodents.


Asunto(s)
Neoplasias Pulmonares , Neuralgia , Analgésicos/farmacología , Analgésicos/uso terapéutico , Animales , Biopterinas/análogos & derivados , Receptores ErbB/genética , Receptores ErbB/metabolismo , GTP Ciclohidrolasa/genética , GTP Ciclohidrolasa/metabolismo , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Ratones , Neuralgia/tratamiento farmacológico , Neuralgia/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo
5.
CNS Neurosci Ther ; 28(1): 36-45, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34845843

RESUMEN

AIMS: Neuropathic pain after spinal cord injury is one of the most difficult clinical problems after the loss of mobility, and pharmacological or neuromodulation therapy showed limited efficacy. In this study, we examine the possibility of pain modulation by a recombinant adeno-associated virus (rAAV) encoding small-hairpin RNA against GCH1 (rAAV-shGCH1) in a spinal cord injury model in which neuropathic pain was induced by a spinothalamic tract (STT) lesion. METHODS: Micro-electric lesioning was used to damage the left STT in rats (n = 32), and either rAAV-shGCH1 (n = 19) or rAAV control (n = 6) was injected into the dorsal horn of the rats at the same time. On postoperative days 3, 7, and 14, we evaluated neuropathic pain using a behavioral test and microglial activation by immunohistochemical staining. RESULTS: A pain modulation effect of shGCH1 was observed from postoperative days 3 to 14. The mechanical withdrawal threshold was 13.0 ± 0.95 in the shGCH1 group, 4.3 ± 1.37 in the control group, and 3.49 ± 0.85 in sham on postoperative day 3 (p < 0.0001) and continued to postoperative day 14 (shGCH1 vs. control: 11.4 ± 1.1 vs. 2.05 ± 0.60, p < 0.001 and shGCH1 vs. sham: 11.4 ± 1.1 vs. 1.43 ± 0.54, p < 0.001). Immunohistochemical staining of the spinal cord dorsal horn showed deactivation of microglia in the shGCH1 group without any change of delayed pattern of astrocyte activation as in STT model. CONCLUSIONS: Neuropathic pain after spinal cord injury can be modulated bilaterally by deactivating microglial activation after a unilateral injection of rAAV-shGCH1 into the dorsal horn of a STT lesion spinal cord pain model. This new attempt would be another therapeutic approach for NP after SCI, which once happens; there is no clear curative options still now.


Asunto(s)
Dependovirus/genética , GTP Ciclohidrolasa/genética , Microglía/fisiología , Neuralgia/prevención & control , ARN Interferente Pequeño/metabolismo , Traumatismos de la Médula Espinal/fisiopatología , Tractos Espinotalámicos/lesiones , Animales , GTP Ciclohidrolasa/metabolismo , Hiperalgesia/patología , Masculino , Neuralgia/fisiopatología , Neuralgia/terapia , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Asta Dorsal de la Médula Espinal , Tractos Espinotalámicos/fisiopatología
6.
Antioxid Redox Signal ; 37(1-3): 171-183, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34806400

RESUMEN

Significance: Tetrahydrobiopterin (BH4) is most well known as a required cofactor for enzymes regulating cellular redox homeostasis, aromatic amino acid metabolism, and neurotransmitter synthesis. Less well known are the effects dependent on the cofactor's availability, factors governing its synthesis and recycling, redox implications of the cofactor itself, and protein-protein interactions that underlie cell death. This review provides an understanding of the recent advances implicating BH4 in the mechanisms of cell death and suggestions of possible therapeutic interventions. Recent Advances: The levels of BH4 often reflect the sum of synthetic and recycling enzyme activities. Enhanced expression of GTP cyclohydrolase, the rate-limiting enzyme in biosynthesis, increases BH4, leading to improved cell function and survival. Pharmacologically increasing BH4 levels has similar beneficial effects, leading to enhanced production of neurotransmitters and nitric oxide or reducing oxidant levels. The GTP cyclohydrolase-BH4 pairing has been implicated in a type of cell death, ferroptosis. At the cellular level, BH4 counteracts anticancer therapies directed to enhance ferroptosis via glutathione peroxidase 4 (GPX4) activity inhibition. Critical Issues: Because of the multitude of intertwined mechanisms, a clear relationship between BH4 and cell death is not well understood yet. The possibility that the cofactor directly influences cell viability has not been excluded in previous studies when modulating BH4-producing enzymes. Future Directions: The importance of cellular BH4 variations and BH4 biosynthetic enzymes to cell function and viability makes it essential to better characterize temporal changes, cofactor activity, and the influence on redox status, which in turn would help develop novel therapies. Antioxid. Redox Signal. 37, 171-183.


Asunto(s)
Biopterinas , GTP Ciclohidrolasa , Biopterinas/análogos & derivados , Biopterinas/metabolismo , GTP Ciclohidrolasa/genética , Óxido Nítrico/metabolismo , Oxidación-Reducción
7.
Biosci Rep ; 41(9)2021 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-34402860

RESUMEN

Neuropathic pain (NP) is a chronic pain directly caused by injury or disease of the somatosensory nervous system. Previous studies suggest that GTP cyclohydrolase I (GCH1) may play a pivotal role in microglial activation, which has been shown to be essential for NP. However, its underlying mechanisms in microglial activation remain unclear. A wide range of microRNAs (miRNAs) have been found to be involved in microglial activation-induced NP. To identify the miRNAs regulated by GCH1 and predict their functions in the progression of microglial activation, we analyzed the miRNA expression profiles of GCH1-knockdown (KD) BV2 microglial cells. Small RNA-sequencing analysis revealed 13 differentially expressed (DE) miRNAs in GCH1-KD cells. The target genes of DE miRNAs mainly participate in PI3K-Akt signaling pathway, peroxisome and ferroptosis. The miRNA-mRNA regulatory network analysis showed that GCH1, MAP4K5 and YWHAB acted as hub genes. qRT-PCR results further verified the expression levels of mmu-miR-1a-3p, mmu-miR-133a-3p, mmu-miR-7a-5p and mmu-miR-10a-5p in GCH1-KD cells, which were consistent with the sequencing data. In addition, our data indicated that overexpression of mmu-miR-133a-3p alleviated the pro-inflammatory cytokines IL-1ß and IL-6 production induced by lipopolysaccharide (LPS), indicating that mmu-miR-133a-3p has a negative effect on microglial activation. Taken together, our findings suggest that many miRNAs regulated by GCH1 may be involved in microglial activation, which may provide new potential targets for GCH1 in the pathogenesis of NP.


Asunto(s)
GTP Ciclohidrolasa/metabolismo , MicroARNs/metabolismo , Microglía/enzimología , Neuralgia/enzimología , Transcriptoma , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Animales , Línea Celular , GTP Ciclohidrolasa/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Mediadores de Inflamación/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Ratones , MicroARNs/genética , Microglía/efectos de los fármacos , Neuralgia/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo
8.
J Cell Biol ; 220(9)2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34328510

RESUMEN

Ferroptosis is a form of iron-dependent regulated cell death driven by uncontrolled lipid peroxidation. Mitochondria are double-membrane organelles that have essential roles in energy production, cellular metabolism, and cell death regulation. However, their role in ferroptosis has been unclear and somewhat controversial. In this Perspective, I summarize the diverse metabolic processes in mitochondria that actively drive ferroptosis, discuss recently discovered mitochondria-localized defense systems that detoxify mitochondrial lipid peroxides and protect against ferroptosis, present new evidence for the roles of mitochondria in regulating ferroptosis, and outline outstanding questions on this fascinating topic for future investigations. An in-depth understanding of mitochondria functions in ferroptosis will have important implications for both fundamental cell biology and disease treatment.


Asunto(s)
Ferroptosis/genética , GTP Ciclohidrolasa/antagonistas & inhibidores , Hierro/metabolismo , Mitocondrias/genética , Fosfolípido Hidroperóxido Glutatión Peroxidasa/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Sistema de Transporte de Aminoácidos y+/antagonistas & inhibidores , Sistema de Transporte de Aminoácidos y+/genética , Sistema de Transporte de Aminoácidos y+/metabolismo , Células Cultivadas , Ferroptosis/efectos de los fármacos , GTP Ciclohidrolasa/genética , GTP Ciclohidrolasa/metabolismo , Regulación de la Expresión Génica , Glutatión/antagonistas & inhibidores , Glutatión/biosíntesis , Humanos , Peroxidación de Lípido/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Oxidantes/farmacología , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Especies Reactivas de Oxígeno/agonistas , Especies Reactivas de Oxígeno/metabolismo , Proteína de Unión al Calcio S100A4/antagonistas & inhibidores , Proteína de Unión al Calcio S100A4/genética , Proteína de Unión al Calcio S100A4/metabolismo , Transducción de Señal , Ubiquinona/antagonistas & inhibidores , Ubiquinona/metabolismo
9.
J Neurol Neurosurg Psychiatry ; 91(11): 1210-1218, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32732384

RESUMEN

Loss of nigrostriatal dopaminergic projection neurons is a key pathology in Parkinson's disease, leading to abnormal function of basal ganglia motor circuits and the accompanying characteristic motor features. A number of intraparenchymally delivered gene therapies designed to modify underlying disease and/or improve clinical symptoms have shown promise in preclinical studies and subsequently were evaluated in clinical trials. Here we review the challenges with surgical delivery of gene therapy vectors that limited therapeutic outcomes in these trials, particularly the lack of real-time monitoring of vector administration. These challenges have recently been addressed during the evolution of novel techniques for vector delivery that include the use of intraoperative MRI. The preclinical development of these techniques are described in relation to recent clinical translation in an adeno-associated virus serotype 2-mediated human aromatic L-amino acid decarboxylase gene therapy development programme. This new paradigm allows visualisation of the accuracy and adequacy of viral vector delivery within target structures, enabling intertrial modifications in surgical approaches, cannula design, vector volumes and dosing. The rapid, data-driven evolution of these procedures is unique and has led to improved vector delivery.


Asunto(s)
Cuerpo Estriado , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Imagen por Resonancia Magnética , Procedimientos Neuroquirúrgicos/métodos , Enfermedad de Parkinson/terapia , Sustancia Negra , Animales , Descarboxilasas de Aminoácido-L-Aromático/genética , Ganglios Basales , Dependovirus , Medicina Basada en la Evidencia , GTP Ciclohidrolasa/genética , Glutamato Descarboxilasa/genética , Humanos , Cuidados Intraoperatorios/métodos , Lentivirus , Neurturina/genética , Parvovirinae , Primates , Cirugía Asistida por Computador , Tirosina 3-Monooxigenasa/genética
10.
Biochem Pharmacol ; 180: 114102, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32562786

RESUMEN

Radiation enteritis (RE) is a common side effect after radiotherapy for abdominal cancer. RE pathogenesis is complicated, with no drugs available for prevention or treatments. Intestinal ischemia is a key factor in the occurrence and development of enteritis. The effect of ionizing radiation (IR) on intestinal ischemia is unknown. Deficiency of tetrahydrobiopterin (BH4) produced by GTP-cyclohydrolase 1 (Gch1) is important in ischemic diseases. This study focused on the relationship of Gch1/BH4 between intestinal ischemia in radiation enteritis. BH4 levels were analyzed by high-performance liquid chromatography in humans and rats after radiotherapy. Intestinal blood perfusion was measured by laser doppler flow imaging. Vascular ring tests determined the diastolic functions of rat mesenteric arteries. Gene, protein, and immunohistochemical staining experiments and inhibitor interventions were used to investigate Gch1 and endothelial NOS (eNOS) in rat mesenteric arteries and endothelial cells. The results showed that IR decreased BH4 levels in patients and rats after radiotherapy and decreased intestinal blood perfusion in rats. The degree of change in intestinal ischemia was consistent with intestinal villus injury. Gch1 mRNA and protein levels and nitric oxide (NO) production significantly decreased, while eNOS uncoupling in arterial and vascular endothelial cells strongly increased. BH4 supplementation improved eNOS uncoupling and NO levels in vascular endothelia after IR. The results of this study showed that downregulation of Gch1 in intestinal blood vessels after IR is an important target in RE. BH4 supplementation may prevent intestinal ischemia and improve vascular endothelial function after IR. These findings have clinical significance for the prevention and treatment of RE.


Asunto(s)
Enteritis/prevención & control , GTP Ciclohidrolasa/genética , Intestinos/irrigación sanguínea , Fenilcetonurias/sangre , Traumatismos por Radiación/prevención & control , Radioterapia/efectos adversos , Anciano , Anciano de 80 o más Años , Animales , Biopterinas/análogos & derivados , Biopterinas/farmacología , Regulación hacia Abajo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/efectos de la radiación , Enteritis/sangre , Enteritis/genética , Enteritis/patología , Femenino , GTP Ciclohidrolasa/antagonistas & inhibidores , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/efectos de la radiación , Persona de Mediana Edad , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fenilcetonurias/etiología , Traumatismos por Radiación/sangre , Traumatismos por Radiación/genética , Traumatismos por Radiación/patología , Traumatismos Experimentales por Radiación/sangre , Traumatismos Experimentales por Radiación/genética , Traumatismos Experimentales por Radiación/prevención & control , Ratas , Ratas Sprague-Dawley , Vasodilatación/efectos de los fármacos , Vasodilatación/efectos de la radiación
11.
Diabetes ; 69(1): 99-111, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31676569

RESUMEN

Endothelial dysfunction plays a crucial role in the progress of diabetic vasculopathy. C1q/tumor necrosis factor-related protein 13 (CTRP13) is a secreted adipokine that can ameliorate atherosclerosis and vascular calcification. However, the role of CTRP13 in regulating endothelial function in diabetes has yet to be explored. In this study, CTRP13 treatment improved endothelium-dependent relaxation in the aortae and mesenteric arteries of both db/db mice and streptozotocin-injected mice. CTRP13 supplement also rescued the impaired endothelium-dependent relaxation ex vivo in the db/db mouse aortae and in high glucose (HG)-treated mouse aortae. Additionally, CTRP13 treatment reduced reactive oxygen species overproduction and improved nitric oxide (NO) production and endothelial NO synthase (eNOS) coupling in the aortae of diabetic mice and in HG-treated human umbilical vein endothelial cells. Mechanistically, CTRP13 could increase GTP cyclohydrolase 1 (GCH1) expression and tetrahydrobiopterin (BH4) levels to ameliorate eNOS coupling. More importantly, CTRP13 rescued HG-induced inhibition of protein kinase A (PKA) activity. Increased PKA activity enhanced phosphorylation of the peroxisome proliferator-activated receptor α and its recruitment to the GCH1 promoter, thus activating GCH1 transcription and, ultimately, endothelial relaxation. Together, these results suggest that CTRP13 preserves endothelial function in diabetic mice by regulating GCH1/BH4 axis-dependent eNOS coupling, suggesting the therapeutic potential of CTRP13 against diabetic vasculopathy.


Asunto(s)
Adipoquinas/fisiología , Complemento C1q/fisiología , Diabetes Mellitus/fisiopatología , Endotelio Vascular/fisiología , GTP Ciclohidrolasa/genética , Células Endoteliales de la Vena Umbilical Humana/fisiología , Animales , Células Cultivadas , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Angiopatías Diabéticas/genética , Angiopatías Diabéticas/fisiopatología , Angiopatías Diabéticas/prevención & control , GTP Ciclohidrolasa/metabolismo , Regulación Enzimológica de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
12.
Cell Rep ; 28(1): 218-230.e7, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31269442

RESUMEN

Classical activation of macrophages (M(LPS+IFNγ)) elicits the expression of inducible nitric oxide synthase (iNOS), generating large amounts of NO and inhibiting mitochondrial respiration. Upregulation of glycolysis and a disrupted tricarboxylic acid (TCA) cycle underpin this switch to a pro-inflammatory phenotype. We show that the NOS cofactor tetrahydrobiopterin (BH4) modulates IL-1ß production and key aspects of metabolic remodeling in activated murine macrophages via NO production. Using two complementary genetic models, we reveal that NO modulates levels of the essential TCA cycle metabolites citrate and succinate, as well as the inflammatory mediator itaconate. Furthermore, NO regulates macrophage respiratory function via changes in the abundance of critical N-module subunits in Complex I. However, NO-deficient cells can still upregulate glycolysis despite changes in the abundance of glycolytic intermediates and proteins involved in glucose metabolism. Our findings reveal a fundamental role for iNOS-derived NO in regulating metabolic remodeling and cytokine production in the pro-inflammatory macrophage.


Asunto(s)
Ciclo del Ácido Cítrico , Inflamación/metabolismo , Macrófagos/metabolismo , Óxido Nítrico/metabolismo , Succinatos/metabolismo , Animales , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Ciclo del Ácido Cítrico/efectos de los fármacos , Transporte de Electrón/efectos de los fármacos , Endotoxemia/inducido químicamente , Endotoxemia/metabolismo , GTP Ciclohidrolasa/genética , GTP Ciclohidrolasa/metabolismo , Glucólisis/efectos de los fármacos , Interferón gamma/farmacología , Interleucina-1beta/metabolismo , Isocitrato Deshidrogenasa/metabolismo , Lipopolisacáridos/farmacología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Ratones , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Mitocondrias/metabolismo , Infecciones por Mycobacterium/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fragmentos de Péptidos/metabolismo , Proteoma/genética , Proteoma/metabolismo , Ácido Succínico/metabolismo , Espectrometría de Masas en Tándem
13.
BMC Pediatr ; 19(1): 199, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-31202265

RESUMEN

BACKGROUND: Tetrahydrobiopterin (BH4) deficiencies are disorders affecting phenylalanine homeostasis, and catecholamine and serotonin biosynthesis. GTP-Cyclohydrolase I deficiency (OMIM 600225) is an extremely rare variant of inborn error of BH4 synthesis which exists in recessive and dominant forms. The recessive form presents with complex neurological and autonomic dysfunction whilst the dominant form presents as Dopa-responsive dystonia. CASE PRESENTATION: We describe a South Asian child who initially presented with neurological dysfunction and recurrent vomiting and later developed recurrent hyperthermia for several months. The child did not have screening for hyperphenylalaninemia at birth and was found to have marked hyperphenylalaninemia after clinical presentation at 5 months. Further evaluation revealed BH4 deficiency. GTP-Cyclohydrolase I deficiency (GTPCH) was identified based on normal dihydro pteridine reductase activity and markedly reduced neopterin in dried blood spot test. After institution of treatment and control of high phenylalanine levels, clinical deterioration decelerated yet with noticeable residual neurological dysfunction. CONCLUSION: To authors' knowledge, this is first report of GTPCH deficiency in a South Asian child. The case highlights practical issues regarding diagnosis of GTPCH deficiency, especially in countries without broader universal newborn screening programs for early detection of inherited metabolic disorders. Testing for GTPCH deficiency should be considered for patients with unexplained neurological and autonomic symptoms following initial metabolic screen.


Asunto(s)
Fiebre/etiología , GTP Ciclohidrolasa/deficiencia , Enfermedades del Sistema Nervioso/etiología , Fenilcetonurias/etiología , Vómitos/etiología , Encéfalo/diagnóstico por imagen , Consanguinidad , GTP Ciclohidrolasa/genética , Humanos , Lactante , Masculino , Fenilalanina/sangre , Recurrencia , Sri Lanka
14.
Brain ; 142(8): 2402-2416, 2019 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-31243443

RESUMEN

Ever since its introduction 40 years ago l-3,4-dihydroxyphenylalanine (l-DOPA) therapy has retained its role as the leading standard medication for patients with Parkinson's disease. With time, however, the shortcomings of oral l-DOPA treatment have become apparent, particularly the motor fluctuations and troublesome dyskinetic side effects. These side effects, which are caused by the excessive swings in striatal dopamine caused by intermittent oral delivery, can be avoided by delivering l-DOPA in a more continuous manner. Local gene delivery of the l-DOPA synthesizing enzymes, tyrosine hydroxylase and guanosine-tri-phosphate-cyclohydrolase-1, offers a new approach to a more refined dopaminergic therapy where l-DOPA is delivered continuously at the site where it is needed i.e. the striatum. In this study we have explored the therapeutic efficacy of adeno-associated viral vector-mediated l-DOPA delivery to the putamen in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated rhesus monkeys, the standard non-human primate model of Parkinson's disease. Viral vector delivery of the two enzymes, tyrosine hydroxylase and guanosine-5'-tri-phosphate-cyclohydrolase-1, bilaterally into the dopamine-depleted putamen, induced a significant, dose-dependent improvement of motor behaviour up to a level identical to that obtained with the optimal dose of peripheral l-DOPA. Importantly, this improvement in motor function was obtained without any adverse dyskinetic effects. These results provide proof-of-principle for continuous vector-mediated l-DOPA synthesis as a novel therapeutic strategy for Parkinson's disease. The constant, local supply of l-DOPA obtained with this approach holds promise as an efficient one-time treatment that can provide long-lasting clinical improvement and at the same time prevent the appearance of motor fluctuations and dyskinetic side effects associated with standard oral dopaminergic medication.


Asunto(s)
Antiparkinsonianos/administración & dosificación , GTP Ciclohidrolasa/administración & dosificación , Vectores Genéticos/uso terapéutico , Levodopa/biosíntesis , Trastornos Parkinsonianos/terapia , Putamen/metabolismo , Tirosina 3-Monooxigenasa/administración & dosificación , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/efectos adversos , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/análogos & derivados , Animales , Antiparkinsonianos/uso terapéutico , Dependovirus/genética , Evaluación Preclínica de Medicamentos , Femenino , GTP Ciclohidrolasa/análisis , GTP Ciclohidrolasa/genética , GTP Ciclohidrolasa/metabolismo , Genes Reporteros , Genes Sintéticos , Vectores Genéticos/administración & dosificación , Humanos , Macaca mulatta , Masculino , Actividad Motora/efectos de los fármacos , Trastornos Parkinsonianos/inducido químicamente , Porción Compacta de la Sustancia Negra/química , Porción Compacta de la Sustancia Negra/patología , Prueba de Estudio Conceptual , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/análisis , Proteínas Recombinantes/uso terapéutico , Tirosina 3-Monooxigenasa/análisis , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
15.
Pigment Cell Melanoma Res ; 32(5): 724-727, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30896066

RESUMEN

Although xanthophores are found widely among poikilothermic species, the developmental and biochemical pathways underlying differentiation of these pteridine- and carotenoid-containing cells remain murky. I have identified a recessive zebrafish mutant, camembert (cmm), which displays defective xanthophore pigmentation during embryonic and larval stages with cells appearing grayish rather than yellow, but as an adult appears to have normally pigmented xanthophores and wild-type stripe pattern. Examination of molecular markers reveals that xanthophores are present in typical numbers and position in cmm embryos; however, the localization of transcripts for the gene GTP cyclohydrolase 2 (gch2), encoding a critical protein in the pteridine biosynthetic pathway, is strikingly altered. RT-PCR analysis indicates that gch2 transcripts in mutant embryos skip an exon or retain the intron upstream and that no correctly spliced mRNA is made. Sequencing of genomic DNA reveals that the skipped exon is intact, but the retained intron contains a deletion of approximately 180 base pairs, just upstream of the splice acceptor. Microinjection of a gch2 BAC clone rescues yellow pigmentation in camembert larvae, confirming that the pigmentation defect is due to mutation of gch2.


Asunto(s)
Embrión no Mamífero/metabolismo , GTP Ciclohidrolasa/metabolismo , Regulación del Desarrollo de la Expresión Génica , Larva/crecimiento & desarrollo , Melanóforos/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Embrión no Mamífero/citología , GTP Ciclohidrolasa/genética , Larva/genética , Larva/metabolismo , Pigmentación , Pez Cebra , Proteínas de Pez Cebra/genética
16.
J Clin Psychopharmacol ; 39(1): 28-38, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30566416

RESUMEN

PURPOSE/BACKGROUND: Interventions for attention-deficit/hyperactivity disorder (ADHD) may be inadequate for some patients. There is evidence that supplementation with L-methylfolate augments antidepressant agent effects and thus might also augment ADHD treatment effects by a common catecholaminergic mechanism. METHODS: Forty-four adults with Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition diagnosis of ADHD participated in a randomized, double-blind, placebo-controlled, 12-week trial of 15 mg of L-methylfolate in combination with osmotic-release oral system methylphenidate. Osmotic-release oral system methylphenidate was dose optimized over the first 6 weeks. We evaluated the effects on ADHD symptoms, self-report on the Behavior Rating Inventory of Executive Function of executive function, methylphenidate dosing, neuropsychological test measures, the Adult ADHD Self-report scale, emotional dysregulation, social adjustment, and work productivity, as well as moderating effects of body mass index, autoantibodies to folate receptors, and select genetic polymorphisms. RESULTS: L-Methylfolate was well tolerated, with no significant effect over placebo except improvement from abnormal measures on the mean adaptive dimension of the ASR scale (χ = 4.36, P = 0.04). Methylphenidate dosing was significantly higher in individuals on L-methylfolate over time (χ = 7.35, P = 0.007). Exploratory analyses suggested that variation in a guanosine triphosphate cyclohydrolase gene predicted association with higher doses of methylphenidate (P < 0.001). CONCLUSIONS: L-Methylfolate was associated with no change in efficacy on measures relevant to neuropsychiatric function in adults with ADHD, other than suggestion of reduced efficacy of methylphenidate. Further investigation would be required to confirm this effect and its mechanism and the genotype prediction of effects on dosing.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/tratamiento farmacológico , Metilfenidato/uso terapéutico , Tetrahidrofolatos/uso terapéutico , Administración Oral , Adulto , Trastorno por Déficit de Atención con Hiperactividad/genética , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Estimulantes del Sistema Nervioso Central/uso terapéutico , Preparaciones de Acción Retardada/uso terapéutico , Dietoterapia , Suplementos Dietéticos , Inhibidores de Captación de Dopamina/administración & dosificación , Inhibidores de Captación de Dopamina/uso terapéutico , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Quimioterapia Combinada , Función Ejecutiva/efectos de los fármacos , Femenino , Receptor 1 de Folato/inmunología , GTP Ciclohidrolasa/genética , Humanos , Masculino , Metilfenidato/administración & dosificación , Pruebas Neuropsicológicas , Proyectos Piloto , Tetrahidrofolatos/efectos adversos , Resultado del Tratamiento , Adulto Joven
17.
Nat Commun ; 9(1): 5409, 2018 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-30573728

RESUMEN

Inducible nitric oxide synthase (iNOS) plays a crucial role in controlling growth of Mycobacterium tuberculosis (M.tb), presumably via nitric oxide (NO) mediated killing. Here we show that leukocyte-specific deficiency of NO production, through targeted loss of the iNOS cofactor tetrahydrobiopterin (BH4), results in enhanced control of M.tb infection; by contrast, loss of iNOS renders mice susceptible to M.tb. By comparing two complementary NO-deficient models, Nos2-/- mice and BH4 deficient Gch1fl/flTie2cre mice, we uncover NO-independent mechanisms of anti-mycobacterial immunity. In both murine and human leukocytes, decreased Gch1 expression correlates with enhanced cell-intrinsic control of mycobacterial infection in vitro. Gene expression analysis reveals that Gch1 deficient macrophages have altered inflammatory response, lysosomal function, cell survival and cellular metabolism, thereby enhancing the control of bacterial infection. Our data thus highlight the importance of the NO-independent functions of Nos2 and Gch1 in mycobacterial control.


Asunto(s)
Biopterinas/análogos & derivados , GTP Ciclohidrolasa/fisiología , Óxido Nítrico Sintasa de Tipo II/fisiología , Óxido Nítrico/biosíntesis , Tuberculosis/inmunología , Animales , Biopterinas/genética , Biopterinas/metabolismo , Biopterinas/fisiología , Supervivencia Celular , GTP Ciclohidrolasa/genética , GTP Ciclohidrolasa/metabolismo , Eliminación de Gen , Perfilación de la Expresión Génica , Humanos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo
18.
Nature ; 563(7732): 564-568, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30405245

RESUMEN

Genetic regulators and environmental stimuli modulate T cell activation in autoimmunity and cancer. The enzyme co-factor tetrahydrobiopterin (BH4) is involved in the production of monoamine neurotransmitters, the generation of nitric oxide, and pain1,2. Here we uncover a link between these processes, identifying a fundamental role for BH4 in T cell biology. We find that genetic inactivation of GTP cyclohydrolase 1 (GCH1, the rate-limiting enzyme in the synthesis of BH4) and inhibition of sepiapterin reductase (the terminal enzyme in the synthetic pathway for BH4) severely impair the proliferation of mature mouse and human T cells. BH4 production in activated T cells is linked to alterations in iron metabolism and mitochondrial bioenergetics. In vivo blockade of BH4 synthesis abrogates T-cell-mediated autoimmunity and allergic inflammation, and enhancing BH4 levels through GCH1 overexpression augments responses by CD4- and CD8-expressing T cells, increasing their antitumour activity in vivo. Administration of BH4 to mice markedly reduces tumour growth and expands the population of intratumoral effector T cells. Kynurenine-a tryptophan metabolite that blocks antitumour immunity-inhibits T cell proliferation in a manner that can be rescued by BH4. Finally, we report the development of a potent SPR antagonist for possible clinical use. Our data uncover GCH1, SPR and their downstream metabolite BH4 as critical regulators of T cell biology that can be readily manipulated to either block autoimmunity or enhance anticancer immunity.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Biopterinas/análogos & derivados , Neoplasias/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Administración Oral , Oxidorreductasas de Alcohol/antagonistas & inhibidores , Oxidorreductasas de Alcohol/metabolismo , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/patología , Biopterinas/biosíntesis , Biopterinas/metabolismo , Biopterinas/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Coenzimas/metabolismo , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Femenino , GTP Ciclohidrolasa/genética , GTP Ciclohidrolasa/metabolismo , Humanos , Hipersensibilidad/inmunología , Hierro/metabolismo , Quinurenina/metabolismo , Quinurenina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo
19.
Sci Rep ; 8(1): 14310, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30254268

RESUMEN

Nitric Oxide (NO) is an intracellular signalling mediator, which affects many biological processes via the posttranslational modification of proteins through S-nitrosation. The availability of NO and NOS-derived reactive oxygen species (ROS) from enzymatic uncoupling are determined by the NO synthase cofactor Tetrahydrobiopterin (BH4). Here, using a global proteomics "biotin-switch" approach, we identified components of the ubiquitin-proteasome system to be altered via BH4-dependent NO signalling by protein S-nitrosation. We show S-nitrosation of ubiquitin conjugating E2 enzymes, in particular the catalytic residue C87 of UBC13/UBE2N, leading to impaired polyubiquitylation by interfering with the formation of UBC13~Ub thioester intermediates. In addition, proteasome cleavage activity in cells also seems to be altered by S-nitrosation, correlating with the modification of cysteine residues within the 19S regulatory particle and catalytic subunits of the 20S complex. Our results highlight the widespread impact of BH4 on downstream cellular signalling as evidenced by the effect of a perturbed BH4-dependent NO-Redox balance on critical processes within the ubiquitin-proteasome system (UPS). These studies thereby uncover a novel aspect of NO associated modulation of cellular homeostasis.


Asunto(s)
Biopterinas/análogos & derivados , Complejo de la Endopetidasa Proteasomal/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina/metabolismo , Animales , Biopterinas/metabolismo , GTP Ciclohidrolasa/deficiencia , GTP Ciclohidrolasa/genética , Técnicas de Silenciamiento del Gen , Ratones , Células 3T3 NIH , Óxido Nítrico/metabolismo , Nitrosación , Transducción de Señal
20.
J Cell Mol Med ; 22(11): 5406-5417, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30091833

RESUMEN

Smoking is a major preventable risk factor for atherosclerosis. However, the causative link between cigarette smoke and atherosclerosis remains to be established. The objective of this study is to characterize the role of GTP cyclohydrolase 1 (GTPCH1), the rate-limiting enzyme for de novo tetrahydrobiopterin (BH4) synthesis, in the smoking-accelerated atherosclerosis and the mechanism involved. In vitro, human umbilical vein endothelial cells were treated with nicotine, a major component of cigarette smoke, which reduced the mRNA and protein levels of GTPCH1 and led to endothelial dysfunction. GTPCH1 overexpression or sepiapterin could attenuate nicotine-reduced nitric oxide and -increased reactive oxygen species levels. Mechanistically, human antigen R (HuR) bound with the adenylateuridylate-rich elements of the GTPCH1 3' untranslated region and increased its stability; nicotine inhibited HuR translocation from the nucleus to cytosol, which downregulated GTPCH1. In vivo, nicotine induced endothelial dysfunction and promoted atherosclerosis in ApoE-/- mice, which were attenuated by GTPCH1 overexpression or BH4 supplement. Our findings may provide a novel and promising approach to atherosclerosis treatment.


Asunto(s)
Aterosclerosis/genética , Proteína 1 Similar a ELAV/genética , GTP Ciclohidrolasa/genética , Nicotina/toxicidad , Animales , Apolipoproteínas E/genética , Aterosclerosis/inducido químicamente , Aterosclerosis/patología , Biopterinas/análogos & derivados , Biopterinas/biosíntesis , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Nicotina/administración & dosificación , Óxido Nítrico/genética , Pterinas/farmacología , ARN Mensajero/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Factores de Riesgo , Fumar/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA