Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 471
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Food Sci ; 89(4): 2277-2291, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38488738

RESUMEN

Calcium peptide chelates are developed as efficient supplements for preventing calcium deficiency. Spent hen meat (SHM) contains a high percentage of proteins but is generally wasted due to the disadvantages such as hard texture. We chose the underutilized SHM to produce peptides to bind calcium by proteolysis and aimed to investigate chelation between calcium and peptides in hydrolysate for a sustainable purpose. The optimized proteolysis conditions calculated from the result of response surface methodology for two-step hydrolysis were 0.30% (wenzyme/wmeat) for papain with a hydrolysis time of 3.5 h and 0.18% (wenzyme/wmeat) for flavourzyme with a hydrolysis time of 2.8 h. The enzymatic hydrolysate (EH) showed a binding capacity of 63.8 ± 1.8 mg calcium/g protein. Ethanol separation for EH improved the capacity up to a higher value of 68.6 ± 0.6 mg calcium/g protein with a high association constant of 420 M-1 (25°C) indicating high stability. The separated fraction with a higher amount of Glu, Asp, Lys, and Arg had higher calcium-binding capacity, which was related to the number of ─COOH and ─NH2 groups in peptide side chains according to the result from amino acid analysis and Fourier transform infrared spectroscopy. Two-step enzymatic hydrolysis and ethanol separation were an efficient combination to produce peptide mixtures derived from SHM with high calcium-binding capacity. The high percentage of hydrophilic amino acids in the separated fraction was concluded to increase calcium-binding capacity. This work provides foundations for increasing spent hen utilization and developing calcium peptide chelates based on underutilized meat.


Asunto(s)
Calcio , Pollos , Animales , Femenino , Calcio/metabolismo , Pollos/metabolismo , Hidrolisados de Proteína/química , Péptidos/química , Hidrólisis , Papaína/química , Aminoácidos , Calcio de la Dieta/metabolismo , Proteínas de Unión al GTP/metabolismo , Carne , Etanol
2.
Food Res Int ; 180: 114065, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38395582

RESUMEN

Tartary buckwheat is rich in nutrients and its protein supports numerous biological functions. However, the digestibility of Tartary buckwheat protein (TBP) poses a significant limitation owing to its inherent structure. This study aimed to assess the impact of high moisture extrusion (HME, 60 % moisture content) on the structural and physicochemical attributes, as well as the in vitro digestibility of TBP. Our results indicated that TBP exhibited unfolded and amorphous microstructures after HME. The protein molecular weight of TBP decreased after HME, and a greater degradation was observed at 70 °C than 100 °C. In particular, HME at 70 °C caused an almost complete disappearance of bands near 35 kDa compared with HME at 100 °C. In addition, compared with native TBP (NTBP, 44.53 µmol/g protein), TBP subjected to HME at 70 °C showed a lower disulfide bond (SS) content (42.67 µmol/g protein), whereas TBP subjected to HME at 100 °C demonstrated a higher SS content (45.70 µmol/g protein). These changes endowed TBP with good solubility (from 55.96 % to 83.31 % at pH 7), foaming ability (20.00 %-28.57 %), and surface hydrophobicity (8.34-23.07). Furthermore, the emulsifying activity (EA) and in vitro digestibility are closely related to SS content. Notably, extruded TBP (ETBP) obtained at 70 °C exhibited higher EA and digestibility than NTBP, whereas ETBP obtained at 100 °C showed the opposite trend. Consequently, HME (especially at 70 °C) demonstrated significant potential as a processing technique for improving the functional and digestive properties of TBP.


Asunto(s)
Fagopyrum , Fagopyrum/química , Solubilidad , Digestión , Proteínas de Unión al GTP/metabolismo
3.
Cells ; 12(20)2023 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-37887313

RESUMEN

De novo mutations in GNAO1, the gene encoding the major neuronal G protein Gαo, cause a spectrum of pediatric encephalopathies with seizures, motor dysfunction, and developmental delay. Of the >80 distinct missense pathogenic variants, many appear to uniformly destabilize the guanine nucleotide handling of the mutant protein, speeding up GTP uptake and deactivating GTP hydrolysis. Zinc supplementation emerges as a promising treatment option for this disease, as Zn2+ ions reactivate the GTP hydrolysis on the mutant Gαo and restore cellular interactions for some of the mutants studied earlier. The molecular etiology of GNAO1 encephalopathies needs further elucidation as a prerequisite for the development of efficient therapeutic approaches. In this work, we combine clinical and medical genetics analysis of a novel GNAO1 mutation with an in-depth molecular dissection of the resultant protein variant. We identify two unrelated patients from Norway and France with a previously unknown mutation in GNAO1, c.509C>G that results in the production of the Pro170Arg mutant Gαo, leading to severe developmental and epileptic encephalopathy. Molecular investigations of Pro170Arg identify this mutant as a unique representative of the pathogenic variants. Its 100-fold-accelerated GTP uptake is not accompanied by a loss in GTP hydrolysis; Zn2+ ions induce a previously unseen effect on the mutant, forcing it to lose the bound GTP. Our work combining clinical and molecular analyses discovers a novel, biochemically distinct pathogenic missense variant of GNAO1 laying the ground for personalized treatment development.


Asunto(s)
Encefalopatías , Humanos , Niño , Mutación/genética , Proteínas de Unión al GTP/metabolismo , Iones/metabolismo , Guanosina Trifosfato , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo
4.
Environ Pollut ; 334: 122211, 2023 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-37454720

RESUMEN

As a metabolic disruptor, bisphenol A (BPA) has been widely reported to disrupt lipid balance. Moreover, BPA has gained significant attention due to its estrogenic activity. While both ferroptosis and the G-protein-coupled estrogen receptor (GPER) have been implicated in lipid metabolism, their link to BPA-induced lipid accumulation remains unclear. In this study, chickens were randomly assigned to three groups and housed them for 4 weeks: a control group (0 µg/L BPA), a low dose group (50 µg/L BPA) and a high dose group (5000 µg/L BPA) to investigate the underlying mechanism of BPA-induced hepatotoxicity. Our results showed that BPA exposure significantly increased the contents of TG, TC, and LDL-C while decreasing HDL-C levels. We also found that BPA treatment altered the levels of genes involved in fatty acid ß-oxidation (ampkα, cpt-1, and ppaα), synthesis (acc, fas, scd-1, and srebp-1) and absorption (lpl and cd36). Moreover, the results showed that the BPA group had higher levels of IL-1ß, IL-18 and TNF-α. These results indicated that BPA exposure disrupted lipid metabolism and induced inflammation in the liver. We also demonstrated that BPA caused hepatic ferroptosis by raising iron content and the expression of genes related to lipid peroxidation (lpcat3, acsl4 and alox15), while reducing the expression of antioxidant system-associated genes (gpx4, slc7a11 and slc3a2). Importantly, BPA remarkably activated GPER expression in the liver. Interestingly, inhibition of GPER remarkably ameliorated BPA-induced lipid metabolism disorder, inflammatory response, and ferroptosis, indicating the crucial role of GPER in BPA-induced liver abnormalities. These findings highlight the link between GPER and ferroptosis in BPA-induced hepatotoxicity, providing new insights into the potential hazard of BPA.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas , Ferroptosis , Animales , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Metabolismo de los Lípidos , Pollos/metabolismo , Hígado/metabolismo , Estrógenos/metabolismo , Compuestos de Bencidrilo/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Proteínas de Unión al GTP/metabolismo , Lípidos
5.
Altern Ther Health Med ; 29(5): 238-241, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37052970

RESUMEN

Objective: The study aims to explore the correlation between the expression levels of peptidylarginine deiminase 4 (PADI4), guanylate binding protein 1 (GBP1), miR-215, and tumor mutational burden (TMB) and clinical features and prognosis of lung cancer. Methods: A total of 156 patients with lung cancer admitted to our hospital from July 2021 to March 2022 were selected. Clinical characteristics of patients were collected and PADI4, GBP1, miR-215, and TMB levels were detected. The correlation between the expression levels of PADI4, GBP1, miR-215, and TMB and the clinical characteristics of lung cancer was analyzed. The predictive value of the expression levels of PADI4, GBP1, miR-215, and TMB for lung cancer prognosis was analyzed by the receiver operator characteristic (ROC) curve. Results: The expression levels of PADI4 and GBP1 were significantly different with respect to smoking history and histopathological type of lung cancer (P < .05). The expression levels of miR-215 and TMB were significantly different in terms of age, smoking history, lymph node metastasis, and tumor node metastasis (TNM) stage of lung cancer (P < .05). ROC curve results showed that the area under the curve (AUC) of PADI4, GBP1, miR-215, and TMB combined to predict the prognosis of lung cancer was 0.814 (0.789-0.912), which was higher than the diagnostic efficacy of single biomarker (P < .05). Its sensitivity and specificity were 85.75% and 89.34%, respectively. Conclusions: The expression levels of PADI4, GBP1, miR-215, and TMB are correlated with the clinical characteristics and prognosis of lung cancer, and can be used as prognostic markers.


Asunto(s)
Neoplasias Pulmonares , MicroARNs , Humanos , Biomarcadores de Tumor/genética , Proteínas de Unión al GTP/metabolismo , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/genética , MicroARNs/genética , MicroARNs/metabolismo , Pronóstico , Sensibilidad y Especificidad
6.
Med ; 4(5): 311-325.e7, 2023 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-37001522

RESUMEN

BACKGROUND: The GNAO1 gene, encoding the major neuronal G protein Gαo, is mutated in a subset of pediatric encephalopathies. Most such mutations consist of missense variants. METHODS: In this study, we present a precision medicine workflow combining next-generation sequencing (NGS) diagnostics, molecular etiology analysis, and personalized drug discovery. FINDINGS: We describe a patient carrying a de novo intronic mutation (NM_020988.3:c.724-8G>A), leading to epilepsy-negative encephalopathy with motor dysfunction from the second decade. Our data show that this mutation creates a novel splice acceptor site that in turn causes an in-frame insertion of two amino acid residues, Pro-Gln, within the regulatory switch III region of Gαo. This insertion misconfigures the switch III loop and creates novel interactions with the catalytic switch II region, resulting in increased GTP uptake, defective GTP hydrolysis, and aberrant interactions with effector proteins. In contrast, intracellular localization, Gßγ interactions, and G protein-coupled receptor (GPCR) coupling of the Gαo[insPQ] mutant protein remain unchanged. CONCLUSIONS: This in-depth analysis characterizes the heterozygous c.724-8G>A mutation as partially dominant negative, providing clues to the molecular etiology of this specific pathology. Further, this analysis allows us to establish and validate a high-throughput screening platform aiming at identifying molecules that could correct the aberrant biochemical functions of the mutant Gαo. FUNDING: This work was supported by the Joint Seed Money Funding scheme between the University of Geneva and the Hebrew University of Jerusalem.


Asunto(s)
Proteínas de Unión al GTP , Ensayos Analíticos de Alto Rendimiento , Humanos , Niño , Evaluación Preclínica de Medicamentos , Mutación/genética , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Guanosina Trifosfato , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/química , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo
7.
Phytother Res ; 37(7): 2771-2786, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36809683

RESUMEN

Natural dual farnesyl X receptor (FXR)/G protein-coupled bile acid receptor 1 (TGR5) activators have received little attention in the management of metabolic diseases. Deoxyschizandrin (DS), a natural lignan, occurs in S. chinensis fruit and has potent hepatoprotective effects, whereas its protective roles and mechanisms against obesity and non-alcoholic fatty liver disease (NAFLD) are largely elusive. Here, we identified DS as a dual FXR/TGR5 agonist using luciferase reporter and cyclic adenosine monophosphate (cAMP) assays. DS was orally or intracerebroventricularly administrated to high-fat diet-induced obesity (DIO) mice, and methionine and choline-deficient L-amino acid diet (MCD diet)-induced non-alcoholic steatohepatitis to evaluate its protective effects. Exogenous leptin treatment was employed to investigate the sensitization effect of DS on leptin. The molecular mechanism of DS was explored by Western blot, quantitative real-time PCR analysis, and ELISA. The results showed that DS activated FXR/TGR5 signaling and effectively reduced NAFLD in DIO and MCD diet-fed mice. DS countered obesity in DIO mice by promoting anorexia and energy expenditure and reversing leptin resistance, involving both peripheral and central TGR5 activation and leptin sensitization. Our findings indicate that DS may be a novel therapeutic approach for alleviating obesity and NAFLD through regulating FXR and TGR5 activities and leptin signaling.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Animales , Ratones , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Leptina/uso terapéutico , Receptores Acoplados a Proteínas G/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Ácidos y Sales Biliares/metabolismo , Proteínas de Unión al GTP/metabolismo , Proteínas de Unión al GTP/farmacología , Proteínas de Unión al GTP/uso terapéutico , Ratones Endogámicos C57BL , Hígado
8.
Protein Expr Purif ; 203: 106215, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36535546

RESUMEN

Apyrase from potato (Solanum tuberosum) is a divalent metal ion-dependent enzyme that catalyzes the hydrolysis of nucleoside di- and tri-phosphates with broad substrate specificity. The enzyme is widely used to manipulate nucleotide levels such as in the G protein-coupled receptor (GPCR) field where it is used to deplete guanine nucleotides to stabilize nucleotide-free ternary agonist-GPCR-G protein complexes. Potato apyrase is available commercially as the native enzyme purified from potatoes or as a recombinant protein, but these are prohibitively expensive for some research applications. Here, we report a relatively simple method for the bacterial production of soluble, active potato apyrase. Apyrase has several disulfide bonds, so we co-expressed the enzyme bearing a C-terminal (His)6 tag with the E. coli disulfide isomerase DsbC at low temperature (18 °C) in the oxidizing cytoplasm of E. coli Origami B (DE3). This allowed low level production of soluble apyrase. A two-step purification procedure involving Ni-affinity followed by Cibacron Blue-affinity chromatography yielded highly purified apyrase at a level of ∼0.5 mg per L of bacterial culture. The purified enzyme was functional for ATP hydrolysis in an ATPase assay and for GTP/GDP hydrolysis in a GPCR-G protein coupling assay. This methodology enables the time- and cost-efficient production of recombinant apyrase for various research applications.


Asunto(s)
Apirasa , Solanum tuberosum , Apirasa/genética , Apirasa/química , Escherichia coli/metabolismo , Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/metabolismo , Proteínas Recombinantes/química , Solanum tuberosum/genética , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo
9.
New Phytol ; 237(2): 615-630, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36266966

RESUMEN

Calcium (Ca) deficiency causes necrotic symptoms of foliar edges known as tipburn; however, the underlying cellular mechanisms have been poorly understood due to the lack of an ideal plant model and research platform. Using a phenotyping system that quantitates growth and tipburn traits in the model bryophyte Marchantia polymorpha, we evaluated metabolic compounds and the Gß-null mutant (gpb1) that modulate the occurrence and expansion of the tipburn. Transcriptomic comparisons between wild-type and gpb1 plants revealed the phenylalanine/phenylpropanoid biosynthesis pathway and reactive oxygen species (ROS) important for Ca deficiency responses. gpb1 plants reduced ROS production possibly through transcriptomic regulations of class III peroxidases and induced the expression of phenylpropanoid pathway enzymes without changing downstream lignin contents. Supplementation of intermediate metabolites and chemical inhibitors further confirmed the cell-protective mechanisms of the phenylpropanoid and ROS pathways. Marchantia polymorpha, Arabidopsis thaliana, and Lactuca sativa showed comparable transcriptomic changes where genes related to phenylpropanoid and ROS pathways were enriched in response to Ca deficiency. In conclusion, our study demonstrated unresolved signaling and metabolic pathways of Ca deficiency response. The phenotyping platform can speed up the discovery of chemical and genetic pathways, which could be widely conserved between M. polymorpha and angiosperms.


Asunto(s)
Arabidopsis , Marchantia , Calcio/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Arabidopsis/genética , Redes y Vías Metabólicas , Proteínas de Unión al GTP/metabolismo , Marchantia/genética
10.
Br J Pharmacol ; 180(8): 1132-1147, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36479683

RESUMEN

BACKGROUND AND PURPOSE: Intravenous infusion of chemotherapy drugs can cause severe chemotherapy-induced phlebitis (CIP) in patients. However, the underlying mechanism of CIP development remains unclear. EXPERIMENTAL APPROACH: RNA-sequencing analysis was used to identify potential disease targets in CIP. Guanylate binding protein-5 (GBP5) genetic deletion approaches also were used to investigate the role of GBP5 in NLR family pyrin domain containing 3 (NLRP3) inflammasome activation in lipopolysaccharide (LPS) primed murine bone-marrow-derived macrophages (BMDMs) induced by vinorelbine (VIN) in vitro and in mouse models of VIN-induced CIP in vivo. The anti-CIP effect of aescin was evaluated, both in vivo and in vivo. KEY RESULTS: Here, we show that the expression of GBP5 was upregulated in human peripheral blood mononuclear cells from CIP patients. Genetic ablation of GBP5 in murine macrophages significantly alleviated VIN-induced CIP in the experimental mouse model. Mechanistically, GBP5 contributed to the inflammatory responses through activating NLRP3 inflammasome and driving the production of the inflammatory cytokine IL-1ß. Moreover, aescin, a mixture of triterpene saponins extracted from horse chestnut seed, can alleviate CIP by inhibiting the GBP5/NLRP3 axis. CONCLUSION AND IMPLICATIONS: These findings suggest that GBP5 is an important regulator of NLRP3 inflammasome in CIP mouse model. Our work further reveals that aescin may serve as a promising candidate in the clinical treatment of CIP.


Asunto(s)
Antineoplásicos , Flebitis , Humanos , Animales , Ratones , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Escina , Leucocitos Mononucleares/metabolismo , Lipopolisacáridos/farmacología , Interleucina-1beta/metabolismo , Proteínas de Unión al GTP/metabolismo
11.
Front Endocrinol (Lausanne) ; 13: 992666, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36246903

RESUMEN

The actions of several bone-mineral ion regulators, namely PTH, FGF23, Klotho and 1,25(OH)2 vitamin D (1,25(OH)2D), control calcium and phosphate metabolism, and each of these molecules has additional biological effects related to cell signaling, metabolism and ultimately survival. Therefore, these factors are tightly regulated at various levels - genetic, epigenetic, protein secretion and cleavage. We review the main determinants of mineral homeostasis including well-established genetic and post-translational regulators and bring attention to the epigenetic mechanisms that affect the function of PTH, FGF23/Klotho and 1,25(OH)2D. Clinically relevant epigenetic mechanisms include methylation of cytosine at CpG-rich islands, histone deacetylation and micro-RNA interference. For example, sporadic pseudohypoparathyroidism type 1B (PHP1B), a disease characterized by resistance to PTH actions due to blunted intracellular cAMP signaling at the PTH/PTHrP receptor, is associated with abnormal methylation at the GNAS locus, thereby leading to reduced expression of the stimulatory G protein α-subunit (Gsα). Post-translational regulation is critical for the function of FGF-23 and such modifications include glycosylation and phosphorylation, which regulate the cleavage of FGF-23 and hence the proportion of available FGF-23 that is biologically active. While there is extensive data on how 1,25(OH)2D and the vitamin D receptor (VDR) regulate other genes, much more needs to be learned about their regulation. Reduced VDR expression or VDR mutations are the cause of rickets and are thought to contribute to different disorders. Epigenetic changes, such as increased methylation of the VDR resulting in decreased expression are associated with several cancers and infections. Genetic and epigenetic determinants play crucial roles in the function of mineral factors and their disorders lead to different diseases related to bone and beyond.


Asunto(s)
Receptores de Calcitriol , Vitamina D , Calcio/metabolismo , Citosina , Epigénesis Genética , Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas de Unión al GTP/metabolismo , Glucuronidasa/metabolismo , Histonas/metabolismo , Minerales/metabolismo , Hormona Paratiroidea/metabolismo , Fosfatos/metabolismo , Fósforo/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Receptores de Calcitriol/metabolismo , Vitamina D/metabolismo , Vitaminas
12.
Life Sci ; 308: 120926, 2022 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-36058264

RESUMEN

Fatty liver hemorrhagic syndrome (FLHS) seriously threatens the layer industry due to it can cause a sudden decline in egg production and acute death, and dietary supplement with bioactive substance is considered an effective way to prevent the FLHS occurrence. Dehydroepiandrosterone (DHEA) is a popular dietary supplement and it possesses anti-oxidative and anti-inflammatory functions; however, the effect and underlying mechanism about DHEA in protecting against the occurrence and development of FLHS remain elucidated. The current results showed that DHEA relieved HELP-induced decrease of egg productivity and liver injury in laying hens. Meanwhile, DHEA markedly enhanced the antioxidant capacity and then alleviated oxidative stress via activation of nuclear factor (erythroid-derived 2)-like 2 (NRF-2) signal in laying hens fed with HELP diets. In addition, DHEA significantly alleviated HELP-stimulated systemic inflammatory response by suppressing the overproduction of hepatic pro-inflammatory factors in laying hens, and further found this beneficial effect was achieved by blocking the activation of NF-κB pathway. Furthermore, we found that DHEA promoted the AMP-activated protein kinase α (AMPKα) activation and increased the G-protein-coupled estrogen receptor (GPER) expression level in laying hens fed with HELP diets. In summary, our data demonstrated that DHEA attenuates oxidative stress and inflammation through the activation of GPER-AMPK signal axis in laying hens fed with HELP diets. These results might facilitate an understanding of the benefits and mechanism of DHEA on the development of FLHS, and provide sufficient data to support it as a dietary supplement to control the FLHS-related metabolic diseases in chickens.


Asunto(s)
Hígado Graso , Enfermedades de las Aves de Corral , Proteínas Quinasas Activadas por AMP/metabolismo , Anomalías Múltiples , Alimentación Animal/análisis , Animales , Antioxidantes/metabolismo , Pollos/metabolismo , Anomalías Craneofaciales , Deshidroepiandrosterona/farmacología , Dieta , Dieta con Restricción de Proteínas , Estrógenos , Hígado Graso/metabolismo , Femenino , Proteínas de Unión al GTP/metabolismo , Trastornos del Crecimiento , Defectos del Tabique Interventricular , Hemorragia/etiología , FN-kappa B/metabolismo , Estrés Oxidativo , Enfermedades de las Aves de Corral/etiología , Enfermedades de las Aves de Corral/metabolismo , Receptores de Estrógenos/metabolismo , Transducción de Señal
13.
Biomed Pharmacother ; 155: 113660, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36095960

RESUMEN

Nonalcoholic fatty liver disease (NAFLD), often associated with obesity, is becoming one of the most common liver diseases worldwide. It is estimated to affect one billion individuals and may be present in approximately 25% of the population globally. NAFLD is viewed as a hepatic manifestation of metabolic syndrome, with humans and animal models presenting dyslipidemia, hypertension, and diabetes. The gut-liver axis has been considered the main pathogenesis branch for NAFLD development. Considering that foods or beverages could modulate the gastrointestinal tract, immune system, energy homeostasis regulation, and even the gut-liver axis, we conducted an exploratory study to analyze the effects of kombucha probiotic on hepatic steatosis, glucose tolerance, and hepatic enzymes involved in carbohydrate and fat metabolism using a pre-clinical model. The diet-induced obese mice presented glucose intolerance, hyperinsulinemia, hepatic steatosis, increased collagen fiber deposition in liver vascular spaces, and upregulated TNF-alpha and SREBP-1 gene expression. Mice receiving the kombucha supplement displayed improved glucose tolerance, reduced hyperinsulinemia, decreased citrate synthase and phosphofructokinase-1 enzyme activities, downregulated G-protein-coupled bile acid receptor, also known as TGR5, and farnesol X receptor gene expression, and attenuated steatosis and hepatic collagen fiber deposition. The improvement in glucose tolerance was accompanied by the recovery of acute insulin-induced liver AKT serine phosphorylation. Thus, it is possible to conclude that this probiotic drink has a beneficial effect in reducing the metabolic alterations associated with diet-induced obesity. This probiotic beverage deserves an extension of studies to confirm or refute its potentially beneficial effects.


Asunto(s)
Resistencia a la Insulina , Té de Kombucha , Enfermedad del Hígado Graso no Alcohólico , Humanos , Ratones , Animales , Ratones Obesos , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Citrato (si)-Sintasa/metabolismo , Farnesol/metabolismo , Farnesol/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Hígado , Obesidad/complicaciones , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Insulina/metabolismo , Glucosa/metabolismo , Ácidos y Sales Biliares/metabolismo , Carbohidratos/farmacología , Serina/metabolismo , Serina/farmacología , Fosfofructoquinasa-1/metabolismo , Proteínas de Unión al GTP/metabolismo , Colágeno/metabolismo , Ratones Endogámicos C57BL , Dieta Alta en Grasa
14.
Sci Rep ; 12(1): 13578, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35945258

RESUMEN

cDNA display is an in vitro display technology based on a covalent linkage between a protein and its corresponding mRNA/cDNA, widely used for the selection of proteins and peptides from large libraries (1012) in a high throughput manner, based on their binding affinity. Here, we developed a platform using cDNA display and next-generation sequencing (NGS) for rapid and comprehensive substrate profiling of transglutaminase 2 (TG2), an enzyme crosslinking glutamine and lysine residues in proteins. After screening and selection of the control peptide library randomized at the reactive glutamine, a combinatorial library of displayed peptides randomized at positions - 1, + 1, + 2, and + 3 from the reactive glutamine was screened followed by NGS and bioinformatic analysis, which indicated a strong preference of TG2 towards peptides with glutamine at position - 1 (Gln-Gln motif), and isoleucine or valine at position + 3. The highly enriched peptides indeed contained the indicated sequence and showed a higher reactivity as TG2 substrates than the peptide previously selected by phage display, thus representing the novel candidate peptide probes for TG2 research. Furthermore, the obtained information on substrate profiling can be used to identify potential TG2 protein targets. This platform will be further used for the substrate profiling of other TG isozymes, as well as for the selection and evolution of larger biomolecules.


Asunto(s)
Proteínas de Unión al GTP , Transglutaminasas , Biología Computacional , ADN Complementario , Proteínas de Unión al GTP/metabolismo , Glutamina/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Biblioteca de Péptidos , Péptidos/química , Proteína Glutamina Gamma Glutamiltransferasa 2 , Especificidad por Sustrato , Transglutaminasas/metabolismo
15.
Phytother Res ; 36(9): 3601-3618, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35871535

RESUMEN

Puerarin, the major bioactive ingredient isolated from the root of Pueraria lobata (Willd.), attenuates body weight gain and reduces lipid levels in high-fat diet-induced obese mice; however, the underlying mechanism responsible for regulating lipid metabolism remains unclear. This study investigated the molecular mechanism(s) underlying the role of puerarin in regulating lipogenesis and lipolysis in human HepG2 cells. In this study, puerarin strongly inhibited the expression of fatty acid synthase (FASN) and sterol regulatory element binding protein 1c (SREBP-1c). Moreover, puerarin significantly induced the expression of adipose triglyceride lipase (ATGL), which is responsible for triacylglycerol hydrolase activity in cells. Puerarin enhanced 5' AMP-activated protein kinase (AMPK) activity, which is a central regulator of hepatic lipid metabolism. Furthermore, this AMPK activation could be mediated by sirtuin 1 (SIRT1) and calcium signaling pathways involved in G protein-coupled estrogen receptor (GPER) signaling. GPER blockage significantly reversed the effect of puerarin on lipid accumulation and the related signaling pathways. Docking studies showed that puerarin could bind in the GPER in a similar manner as GPER agonist G1. Our results suggest that puerarin can improve hepatic steatosis by activating GPER; it's signaling cascade sequentially induced calcium and SIRT1 signaling pathways. Thus, puerarin may be a potential therapeutic agent for the treatment of non-alcoholic fatty liver disease.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Sirtuina 1 , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Calcio/metabolismo , Proteínas de Unión al GTP/metabolismo , Proteínas de Unión al GTP/farmacología , Células Hep G2 , Humanos , Isoflavonas , Metabolismo de los Lípidos , Lípidos , Hígado , Ratones , Ratones Obesos , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Receptores de Estrógenos/metabolismo , Transducción de Señal , Sirtuina 1/metabolismo
16.
J Biol Chem ; 298(7): 102094, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35654137

RESUMEN

The cytosolic iron-sulfur (Fe-S) cluster assembly (CIA) pathway delivers Fe-S clusters to nuclear and cytosolic Fe-S proteins involved in essential cellular functions. Although the delivery process is regulated by the availability of iron and oxygen, it remains unclear how CIA components orchestrate the cluster transfer under varying cellular environments. Here, we utilized a targeted proteomics assay for monitoring CIA factors and substrates to characterize the CIA machinery. We find that nucleotide-binding protein 1 (NUBP1/NBP35), cytosolic iron-sulfur assembly component 3 (CIAO3/NARFL), and CIA substrates associate with nucleotide-binding protein 2 (NUBP2/CFD1), a component of the CIA scaffold complex. NUBP2 also weakly associates with the CIA targeting complex (MMS19, CIAO1, and CIAO2B) indicating the possible existence of a higher order complex. Interactions between CIAO3 and the CIA scaffold complex are strengthened upon iron supplementation or low oxygen tension, while iron chelation and reactive oxygen species weaken CIAO3 interactions with CIA components. We further demonstrate that CIAO3 mutants defective in Fe-S cluster binding fail to integrate into the higher order complexes. However, these mutants exhibit stronger associations with CIA substrates under conditions in which the association with the CIA targeting complex is reduced suggesting that CIAO3 and CIA substrates may associate in complexes independently of the CIA targeting complex. Together, our data suggest that CIA components potentially form a metabolon whose assembly is regulated by environmental cues and requires Fe-S cluster incorporation in CIAO3. These findings provide additional evidence that the CIA pathway adapts to changes in cellular environment through complex reorganization.


Asunto(s)
Proteínas Hierro-Azufre , Hierro , Citosol/metabolismo , Proteínas de Unión al GTP/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hierro/metabolismo , Proteínas Hierro-Azufre/biosíntesis , Proteínas Hierro-Azufre/metabolismo , Oxígeno/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Azufre/metabolismo
17.
Cell Death Dis ; 13(6): 533, 2022 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-35672281

RESUMEN

Benign prostatic hyperplasia (BPH) is the most common and progressive urological disease in elderly men worldwide. Epidemiological studies have suggested that the speed of disease progression varies among individuals, while the pathophysiological mechanisms of accelerated clinical progression in some BPH patients remain to be elucidated. In this study, we defined patients with BPH as belonging to the accelerated progressive group (transurethral resection of the prostate [TURP] surgery at ≤50 years old), normal-speed progressive group (TURP surgery at ≥70 years old), or non-progressive group (age ≤50 years old without BPH-related surgery). We enrolled prostate specimens from the three groups of patients and compared these tissues to determine the histopathological characteristics and molecular mechanisms underlying BPH patients with accelerated progression. We found that the main histopathological characteristics of accelerated progressive BPH tissues were increased stromal components and prostatic fibrosis, which were accompanied by higher myofibroblast accumulation and collagen deposition. Mechanism dissection demonstrated that these accelerated progressive BPH tissues have higher expression of the CYP19 and G protein-coupled estrogen receptor (GPER) with higher estrogen biosynthesis. Estrogen functions via GPER/Gαi signaling to modulate the EGFR/ERK and HIF-1α/TGF-ß1 signaling to increase prostatic stromal cell proliferation and prostatic stromal fibrosis. The increased stromal components and prostatic fibrosis may accelerate the clinical progression of BPH. Targeting this newly identified CYP19/estrogen/GPER/Gαi signaling axis may facilitate the development of novel personalized therapeutics to better suppress the progression of BPH.


Asunto(s)
Hiperplasia Prostática , Resección Transuretral de la Próstata , Anciano , Aromatasa/metabolismo , Estrógenos/metabolismo , Fibrosis , Proteínas de Unión al GTP/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Próstata/metabolismo , Hiperplasia Prostática/metabolismo , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
18.
Am J Physiol Endocrinol Metab ; 322(5): E436-E445, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35344393

RESUMEN

The melanocortin 4 receptor (MC4R) plays an important role in the regulation of appetite and energy expenditure in humans and rodents. Impairment of MC4R signaling causes severe obesity. MC4R mainly couples to the G-protein Gs. Ligand binding to MC4R activates adenylyl cyclase resulting in increased intracellular cAMP levels. cAMP acts as a secondary messenger, regulating various cellular processes. MC4R can also couple with Gq and other signaling pathways. Therefore, the contribution of MC4R/Gs signaling to energy metabolism and appetite remains unclear. To study the effect of Gs signaling activation in MC4R cells on whole body energy metabolism and appetite, we generated a novel mouse strain that expresses a Gs-coupled designer receptors exclusively activated by designer drugs [Gs-DREADD (GsD)] selectively in MC4R-expressing cells (GsD-MC4R mice). Chemogenetic activation of the GsD by a designer drug [deschloroclozapine (DCZ); 0.01∼0.1 mg/kg body wt] in MC4R-expressing cells significantly increased oxygen consumption and locomotor activity. In addition, GsD activation significantly reduced the respiratory exchange ratio, promoting fatty acid oxidation, but did not affect core (rectal) temperature. A low dose of DCZ (0.01 mg/kg body wt) did not suppress food intake, but a high dose of DCZ (0.1 mg/kg body wt) suppressed food intake in MC4R-GsD mice, although either DCZ dose (0.01 or 0.1 mg/kg body wt) did not affect food intake in the control mice. In conclusion, the current study demonstrated that the stimulation of Gs signaling in MC4R-expressing cells increases energy expenditure and locomotor activity and suppresses appetite.NEW & NOTEWORTHY We report that Gs signaling in melanocortin 4 receptor (MC4R)-expressing cells regulates energy expenditure, appetite, and locomotor activity. These findings shed light on the mechanism underlying the regulation of energy metabolism and locomotor activity by MC4R/cAMP signaling.


Asunto(s)
Proteínas de Unión al GTP , Obesidad , Receptor de Melanocortina Tipo 4 , Animales , Ingestión de Alimentos , Metabolismo Energético , Proteínas de Unión al GTP/metabolismo , Locomoción , Ratones , Obesidad/metabolismo , Receptor de Melanocortina Tipo 4/genética
19.
Nutrients ; 13(4)2021 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-33917155

RESUMEN

Exposure to gluten, a protein present in wheat rye and barley, is the major inducer for human Celiac Disease (CD), a chronic autoimmune enteropathy. CD occurs in about 1% worldwide population, in genetically predisposed individuals bearing human leukocyte antigen (HLA) DQ2/DQ8. Gut epithelial cell stress and the innate immune activation are responsible for the breaking oral tolerance to gliadin, a gluten component. To date, the only treatment available for CD is a long-term gluten-free diet. Several studies have shown that an altered composition of the intestinal microbiota (dysbiosis) could play a key role in the pathogenesis of CD through the modulation of intestinal permeability and the regulation of the immune system. Here, we show that gliadin induces a chronic endoplasmic reticulum (ER) stress condition in the small intestine of a gluten-sensitive mouse model and that the coadministration of probiotics efficiently attenuates both the unfolded protein response (UPR) and gut inflammation. Moreover, the composition of probiotics formulations might differ in their activity at molecular level, especially toward the three axes of the UPR. Therefore, probiotics administration might potentially represent a new valuable strategy to treat gluten-sensitive patients, such as those affected by CD.


Asunto(s)
Suplementos Dietéticos , Estrés del Retículo Endoplásmico , Intolerancia Alimentaria/terapia , Tracto Gastrointestinal/patología , Gliadina/efectos adversos , Glútenes/efectos adversos , Inflamación/patología , Probióticos/uso terapéutico , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Células CACO-2 , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Proteínas de Unión al GTP/metabolismo , Tracto Gastrointestinal/efectos de los fármacos , Humanos , Ratones Endogámicos BALB C , Permeabilidad , Probióticos/administración & dosificación , Proteína Glutamina Gamma Glutamiltransferasa 2 , Transglutaminasas/metabolismo , Regulación hacia Arriba
20.
Biol Pharm Bull ; 44(3): 379-388, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33390389

RESUMEN

Lipopolysaccharide (LPS)-induced inflammation is the leading cause of multiple organ failure in sepsis. Pyruvate kinase 2 (PKM2) is a protein kinase and transcriptional coactivator that plays an important role in glycolysis. Recent studies have confirmed that glycolysis maintains the M1 differentiation and induces immune activation in macrophages. Lycium barbarum polysaccharide (LBP), the main bioactive component of Chinese wolfberry, suppresses glycolysis and inflammation. Here, RAW264.7 macrophages were treated with LBP for evaluating its effects against LPS-induced inflammation. The differentiation of M1/M2 macrophages was assessed by flow cytometry for assessing the cell surface markers, CD86 and CD206. The enrichment of hypoxia inducible factor (HIF)-1α and ubiquitin in the PKM2 protein complex was determined by co-immunoprecipitation. LBP suppressed LPS-induced glycolysis, differentiation of M1 macrophages, and the production of interleukin (IL)-1ß, tumor necrosis factor (TNF)-α, and high mobility group (HMG) 1 proteins. The suppressive effects of LBP were similar to those of PKM2 knockdown, but were abolished by the overexpression of PKM2. LPS elevated the mRNA and protein levels of PKM2. LBP reduced the LPS-induced expression of PKM2 protein, but had no effects on the expression of PKM2 mRNA. LPS inhibited the ubiquitination of PKM2, probably by downregulating the expression of ubiquitin ligases, including Nedd4L, Nedd4, and Gnb2. LBP interfered with the inhibition of PKM2 ubiquitination by upregulating the expression of Nedd4L, Nedd4, and Gnb2. In conclusion, LBP suppressed the LPS-induced inflammation by altering glycolysis and the M1 differentiation of macrophages. The effects of LBP were mediated by the downregulation of PKM2 via enhanced ubiquitination.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Glucólisis/efectos de los fármacos , Piruvato Quinasa/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Glucosa/metabolismo , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/metabolismo , Ácido Láctico/metabolismo , Lipopolisacáridos , Ratones , Ubiquitina-Proteína Ligasas Nedd4/genética , Ubiquitina-Proteína Ligasas Nedd4/metabolismo , Proteolisis/efectos de los fármacos , Piruvato Quinasa/genética , Células RAW 264.7 , Ubiquitinación/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA