Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 15.411
Filtrar
1.
J Zhejiang Univ Sci B ; 25(9): 756-772, 2024 Sep 12.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-39308066

RESUMEN

Doxorubicin (DOX) is a commonly administered chemotherapy drug for treating hematological malignancies and solid tumors; however, its clinical application is limited by significant cardiotoxicity. Cynaroside (Cyn) is a flavonoid glycoside distributed in honeysuckle, with confirmed potential biological functions in regulating inflammation, pyroptosis, and oxidative stress. Herein, the effects of Cyn were evaluated in a DOX-induced cardiotoxicity (DIC) mouse model, which was established by intraperitoneal injections of DOX (5 mg/kg) once a week for three weeks. The mice in the treatment group received dexrazoxane, MCC950, and Cyn every two days. Blood biochemistry, histopathology, immunohistochemistry, reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and western blotting were conducted to investigate the cardioprotective effects and potential mechanisms of Cyn treatment. The results demonstrated the significant benefits of Cyn treatment in mitigating DIC; it could effectively alleviate oxidative stress to a certain extent, maintain the equilibrium of cell apoptosis, and enhance the cardiac function of mice. These effects were realized via regulating the transcription levels of pyroptosis-related genes, such as nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3), caspase-1, and gasdermin D (GSDMD). Mechanistically, for DOX-induced myocardial injury, Cyn could significantly modulate the expression of pivotal genes, including adenosine monophosphate-activated protein kinase (AMPK), peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), sirtuin 3 (SIRT3), and nuclear factor erythroid 2-related factor 2 (Nrf2). We attribute it to the mediation of AMPK/SIRT3/Nrf2 pathway, which plays a central role in preventing DOX-induced cardiomyocyte injury. In conclusion, the present study confirms the therapeutic potential of Cyn in DIC by regulating the AMPK/SIRT3/Nrf2 pathway.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Cardiotoxicidad , Doxorrubicina , Miocitos Cardíacos , Factor 2 Relacionado con NF-E2 , Piroptosis , Transducción de Señal , Sirtuina 3 , Animales , Doxorrubicina/efectos adversos , Piroptosis/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Sirtuina 3/metabolismo , Cardiotoxicidad/prevención & control , Cardiotoxicidad/tratamiento farmacológico , Masculino , Transducción de Señal/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Ratones Endogámicos C57BL
2.
Funct Integr Genomics ; 24(5): 161, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39285026

RESUMEN

Epigenetic regulation plays a role in Parkinson's disease (PD), and ten-eleven translocation methylcytosine dioxygenase 1 (TET1) catalyzes the first step in DNA demethylation by converting 5-methylcytosine to 5-hydroxymethylcytosine. We investigated whether TET1 binds to the promoter of the transient receptor potential cation channel subfamily V member 1 (TRPV1) and regulates its expression, thereby controlling oxidative stress in PD. TRPV1 was identified as an oxidative stress-associated gene in the GSE20186 dataset including substantia nigra from 14 patients with PD and 14 healthy controls and the Genecards database. Lentiviral vectors were used to manipulate Trpv1 expression in rats, followed by 6-hydroxydopamine hydrochloride (6-OHDA) injection for modeling. Behavioral tests, immunofluorescence, Nissl staining, western blot assays, DHE fluorescent probe, biochemical analysis, and ELISA were conducted to assess oxidative stress and neurotoxicity. Trpv1 expression was significantly reduced in the brain tissues of 6-OHDA-treated Parkinsonian rats. Trpv1 alleviated behavioral dysfunction, oxidative stress, and dopamine neuron loss in rats. TET1 mediated TRPV1 hydroxymethylation to promote its expression, and Trpv1 inhibition reversed the mitigating effect of Tet1 on oxidative stress and behavioral dysfunction in PD. TRPV1 activated the AMPK signaling by promoting AMPK phosphorylation to alleviate neurotoxicity and oxidative stress in SH-SY5Y cells. Tet1-mediated Trpv1 hydroxymethylation modification promotes the Ampk signaling activation, thereby eliciting neuroprotection in 6-OHDA-treated Parkinsonian rats. These findings provide experimental evidence that targeting the TET1/TRPV1 axis may be neuroprotective for PD by acting on the AMPK signaling.


Asunto(s)
Metilación de ADN , Enfermedad de Parkinson , Transducción de Señal , Canales Catiónicos TRPV , Animales , Humanos , Masculino , Ratas , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Dioxigenasas , Modelos Animales de Enfermedad , Epigénesis Genética , Oxigenasas de Función Mixta/metabolismo , Oxigenasas de Función Mixta/genética , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Oxidopamina , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/genética , Ratas Sprague-Dawley , Canales Catiónicos TRPV/metabolismo , Canales Catiónicos TRPV/genética
3.
Biomed Pharmacother ; 179: 117361, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39243432

RESUMEN

Berberine (BBR) is a major active component of traditional Chinese medicine Rhizoma Coptidis and Cortex Phellodendri, which have been frequently used to treat liver diseases. Oxidative stress and inflammation are two pivotal hepatic pathological hallmarks. This study aimed to explore the potential effect and underlying mechanism of BBR on fructose-induced rat liver injury model, and hepatocyte damage in HepG2 and BRL-3A cells. Our results indicated that BBR effectively reversed fructose-induced body weight gain, glucose intolerance, and insulin resistance, observably attenuated abnormal histopathological alterations and ameliorated serum activities of ALT and AST. In vivo and in vitro, BBR significantly alleviated the secretion of pro-inflammatory cytokines IL-6 and TNF-α, and elevated levels of anti-inflammatory cytokine IL-10. BBR also attenuated oxidative stress by markedly decreasing intracellular contents of ROS and MDA, and increasing SOD enzymatic activity and GSH level. Furthermore, BBR substantially upregulated the protein expression of Nrf2, HO-1 and p-AMPK, and the fluorescence level of p-AMPK. In addition, BBR significantly increased the level of AMP, the ratio of AMP/ATP, and promoted the expression of ADK. Nevertheless, siADK abolished the benefits exerted by BBR on HepG2 and BRL-3A cells. Conclusively, the hepatoprotective effect of BBR was believed to be intimately associated with anti-inflammatory and antioxidant action mediated, at least partially, via ADK/AMPK/Nrf2 signaling. This work provided further support for the traditional application of Rhizoma Coptidis and Cortex Phellodendri in liver protection and might shed novel dimension to the clinical application of BBR, providing a promising lead compound for drug design.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Berberina , Enfermedad Hepática Inducida por Sustancias y Drogas , Fructosa , Factor 2 Relacionado con NF-E2 , Ratas Sprague-Dawley , Transducción de Señal , Berberina/farmacología , Animales , Humanos , Factor 2 Relacionado con NF-E2/metabolismo , Masculino , Células Hep G2 , Proteínas Quinasas Activadas por AMP/metabolismo , Transducción de Señal/efectos de los fármacos , Ratas , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Estrés Oxidativo/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/patología , Hígado/metabolismo , Antioxidantes/farmacología
4.
J Agric Food Chem ; 72(38): 21052-21064, 2024 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-39268842

RESUMEN

Succinate is an important metabolite and a critical chemical with diverse applications in the food, pharmaceutical, and agriculture industries. Recent studies have demonstrated several protective or detrimental functions of succinate in diseases; however, the effect of succinate on lipid metabolism is still unclear. Here, we identified a role of succinate in nonobese nonalcoholic fatty liver disease (NAFLD). Specifically, the level of succinate is increased in the livers and serum of mice with hepatic steatosis. The administration of succinate promotes triglyceride (TG) deposition and hepatic steatosis by suppressing fatty acid oxidation (FAO) in nonobese NAFLD mouse models. RNA-Seq revealed that succinate suppressed fibroblast growth factor 21 (FGF21) expression. Then, the restoration of FGF21 was sufficient to alleviate hepatic steatosis and FAO inhibition induced by succinate treatment in vitro and in vivo. Furthermore, the inhibition of FGF21 expression and FAO mediated by succinate was dependent on the AMPK/PPARα axis. This study provides evidence linking succinate exposure to abnormal hepatic lipid metabolism and the progression of nonobese NAFLD.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Ácidos Grasos , Factores de Crecimiento de Fibroblastos , Hígado , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico , Oxidación-Reducción , PPAR alfa , Ácido Succínico , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Animales , PPAR alfa/metabolismo , PPAR alfa/genética , Ratones , Masculino , Ácidos Grasos/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Humanos , Ácido Succínico/metabolismo , Hígado/metabolismo , Hígado/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Hígado Graso/metabolismo , Hígado Graso/genética
5.
Phytomedicine ; 134: 156009, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39260136

RESUMEN

BACKGROUND: Targeting white adipose tissue (WAT) browning to increase systemic energy expenditure is a promising therapeutic strategy to combat obesity. Actein from Actaea cimicifuga L. has recently been reported to ameliorate high fat-induced hepatic steatosis. However, the effect of actein on diet-induced obesity merits more and further investigation. PURPOSE: We aimed to examine the anti-obesity potential of actein and unravel its actions on WAT browning. METHODS: The effect of actein on diet-induced obesity was evaluated using a high-fat diet model in C57BL/6 mice. Systemic energy expenditure of mice was measured with a combined indirect calorimetry system. Quantitative real-time PCR analyses were performed to investigate the mRNA levels of genes involved in thermogenesis, browning, and lipolysis. The protein levels were assessed by Western blot. Moreover, WAT explants and a transwell co-culture system consisting of SVFs and adipocytes were constructed to study the mechanisms of actein on promoting WAT browning and lipolysis. RESULTS: At a dosage of 5 mg/kg/d, actein not only protected mice against diet-induced obesity and insulin resistance, but also reversed pre-established obesity and glucose intolerance in mice. Meanwhile, actein facilitated systemic energy expenditure by activating WAT lipolysis and browning. Further, mechanistic studies revealed that actein indirectly induced epididymal adipocyte lipolysis and directly promoted a white-to-beige conversion of subcutaneous adipocytes by activating the AMPK signaling. CONCLUSION: Actein ameliorated diet-induced obesity and was discovered as a natural lead compound directly targeting white-to-beige conversion of subcutaneous adipocytes, suggesting the potential of developing new therapies for obesity and associated metabolic disorders.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Tejido Adiposo Pardo , Tejido Adiposo Blanco , Dieta Alta en Grasa , Metabolismo Energético , Lipólisis , Ratones Endogámicos C57BL , Obesidad , Termogénesis , Animales , Obesidad/tratamiento farmacológico , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Masculino , Lipólisis/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/metabolismo , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Termogénesis/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Ratones , Resistencia a la Insulina , Fármacos Antiobesidad/farmacología , Adipocitos/efectos de los fármacos
6.
J Transl Med ; 22(1): 844, 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39285426

RESUMEN

BACKGROUND: Ocular toxicity is a severe adverse effect that limits the chronic clinical use of the antiarrhythmic drug amiodarone. Here, we aimed to evaluate the cytoprotective effect of artemisinin and explore the potential signalling pathways in human retinal pigment epithelial (RPE) cell cultures. METHODS: D407 cell cultures were exposed to amiodarone and the impact of artemisinin was evaluated. The key parameters included lactate dehydrogenase (LDH) release, intracellular reactive oxygen species (ROS) generation, and the mitochondrial membrane potential (MMP). We also assessed the protein levels of cleaved caspase-3, cleaved poly (ADP-ribose) polymerase (PARP), phosphorylated adenosine monophosphate-activated protein kinase (AMPK)ɑ (p-AMPK), calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2), and nuclear factor erythroid 2-related factor 2 (Nrf2). RESULTS: Artemisinin reduced the cytotoxicity induced by amiodarone, as reflected by decreased LDH release, ROS generation, and MMP disruption. Additionally, artemisinin increased p-AMPK, CaMKK2, and Nrf2 protein levels. Inhibition of AMPK, CaMKK2, or Nrf2 abolished the cytoprotective effect of artemisinin. AMPK activation and Nrf2 knockdown further supported its protective role. CONCLUSIONS: Artemisinin protected RPE cells from amiodarone-induced damage via the CaMKK2/AMPK/Nrf2 pathway. The in vivo experiments in mice confirmed its efficacy in preventing retinal injury caused by amiodarone. These results suggest that an artemisinin-based eye formulation could be repurposed for treating amiodarone-induced ocular toxicity.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Amiodarona , Artemisininas , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina , Citoprotección , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Especies Reactivas de Oxígeno , Epitelio Pigmentado de la Retina , Transducción de Señal , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Humanos , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/patología , Epitelio Pigmentado de la Retina/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/efectos de los fármacos , Citoprotección/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Amiodarona/efectos adversos , Amiodarona/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Artemisininas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Línea Celular , Ratones , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología
7.
Mol Biol Rep ; 51(1): 1003, 2024 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-39305364

RESUMEN

BACKGROUND: Poricoic acid A (PAA), a major triterpenoid component of Poria cocos with anti-tumor, anti-fibrotic, anti-inflammatory, and immune-regulating activities, has been shown to induce podocyte autophagy in diabetic kidney disease (DKD) by downregulating FUN14 domain containing 1 (FUNDC1). This study aimed to identify the role of adenosine monophosphate-activated protein kinase alpha (AMPKα) in PAA-mediated phosphorylation of FUNDC1 in podocyte injury occurring in the pathogenesis of DKD. METHODS AND RESULTS: A cellular model of renal podocyte injury was established by culturing MPC5 cells under high-glucose (HG) conditions. MPC5 cells were subjected to transfection with small interfering RNA (siRNA) targeting AMPKα or siRNA targeting FUNDC1, an AMPKα activator, or PAA. PAA treatment induced the phosphorylation of AMPKα in HG-cultured podocytes. AMPKα activation was implicated in the inhibitory effect of PAA on FUNDC phosphorylation in HG-cultured podocytes. Treatment targeting the AMPKα activator also significantly augmented proliferation, migration, mitochondrial membrane potential, and autophagy levels, while reducing apoptosis levels, inhibiting oxidative stress, and suppressing the release of proinflammatory factors in HG-cultured MPC5 cells. In contrast, insufficient expression of AMPKα reversed the effects of PAA on the proliferation, migration, and apoptosis of podocytes and further exacerbated the reduction of phosphorylated FUNDC1 expression in podocytes under HG conditions. CONCLUSIONS: AMPKα is involved in the regulation of FUNDC1 phosphorylation by PAA in HG-induced podocyte injury. Furthermore, the AMPKα/FUNDC1 pathway plays a crucial regulatory role in HG-induced podocyte injury. These findings support AMPKα, FUNDC1, and the AMPKα/FUNDC1 pathway as targets for PAA intervention.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Autofagia , Nefropatías Diabéticas , Glucosa , Proteínas de la Membrana , Podocitos , Transducción de Señal , Triterpenos , Podocitos/efectos de los fármacos , Podocitos/metabolismo , Podocitos/patología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Transducción de Señal/efectos de los fármacos , Ratones , Autofagia/efectos de los fármacos , Fosforilación/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Triterpenos/farmacología , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/patología , Apoptosis/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos
8.
Int J Biol Sci ; 20(11): 4476-4495, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39247815

RESUMEN

Chronic stress is closely associated with gastrointestinal disorders. However, the impact of stress-related neurotransmitters such as serotonin (5-hydroxytryptamine, 5-HT) on the intestines under chronic stress conditions remains poorly understood. This study aims to elucidate the mechanisms by which 5-HT affects mitochondrial biogenesis and intestinal barrier integrity during chronic stress. Employing a chronic restraint stress (CRS) mouse model, we observed elevated intestinal 5-HT levels, altered colonic mucosal structure, and disrupted tight junctions. The increase in 5-HT was associated with up-regulated serotonin synthesis enzymes and downregulated serotonin reuptake transporters, indicating an imbalance in serotonin homeostasis imbalance caused by chronic stress. Furthermore, serotonin exacerbated oxidative stress and impaired tight junction protein expression, highlighting its role in promoting intestinal barrier dysfunction. Experiments with cells in vitro demonstrated that 5-HT impairs mitochondrial biogenesis by inhibiting the AMPK-PGC-1α axis via 5-HT7 receptors and the cAMP-PKA pathway. Pharmacological inhibition of serotonin synthesis or 5-HT7 receptors alleviated the intestinal barrier damage caused by 5-HT and chronic stress, restoring mitochondrial biogenesis. These findings provide compelling evidence that serotonin exacerbates chronic stress-induced intestinal barrier disruption by inhibiting the AMPK-PGC-1α axis, paving the way for novel therapeutic interventions targeting the detrimental effects of serotonin on the intestine, particularly under chronic stress conditions.


Asunto(s)
Mitocondrias , Biogénesis de Organelos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Serotonina , Serotonina/metabolismo , Animales , Ratones , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Mitocondrias/metabolismo , Células Epiteliales/metabolismo , Mucosa Intestinal/metabolismo , Masculino , Proteínas Quinasas Activadas por AMP/metabolismo , Ratones Endogámicos C57BL
9.
Cell Mol Biol (Noisy-le-grand) ; 70(8): 148-152, 2024 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-39262249

RESUMEN

Cerebrovascular disease, one of the high-risk diseases worldwide, is high in morbidity, disability, mortality, and recurrence rates, which brings many harms to human beings such as physical and mental harm, economic losses, and impairment of social relations. Cerebral ischemia-reperfusion injury (CIRI) is one of the most common pathological manifestations, with mild hypothermia therapy being the most commonly used treatment in clinical practice. In this study, the research team established a CIRI animal model and found that the neuronal apoptosis rate was significantly increased, accompanied by significant ferroptosis, increased inflammation and oxidative stress damage in brain tissue, and obviously inhibited SIRT1/AMPK pathway. However, after mild hypothermia treatment, the pathological changes of CIRI rats were significantly reversed, and the SIRT1/AMPK pathway was reactivated. Therefore, mild hypothermia may achieve the purpose of CIRI repair by activating the SIRT1/AMPK signaling pathway, and targeted regulation of the SIRT1/AMPK signaling pathway may be a research direction for optimizing mild hypothermia therapy or developing new treatment plans for CIRI.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Apoptosis , Hipotermia Inducida , Neuronas , Estrés Oxidativo , Daño por Reperfusión , Transducción de Señal , Sirtuina 1 , Sirtuina 1/metabolismo , Daño por Reperfusión/terapia , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Animales , Hipotermia Inducida/métodos , Neuronas/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Masculino , Isquemia Encefálica/terapia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Ratas Sprague-Dawley , Ratas , Modelos Animales de Enfermedad
10.
Hum Exp Toxicol ; 43: 9603271241266106, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39291655

RESUMEN

OBJECTIVE: This study aimed to explore the expression and biological functions of SIRT3 in colorectal cancer cells (HCT-116), the impacts of sulforaphane on the ferroptosis of HCT-116 cells and the involvement of the SIRT3/AMPK/mTOR axis in those effects. METHODS: SIRT3-overexpressing (OE) and SIRT3-knockout (KO) cell lines were treated with different concentrations of sulforaphane, RSL-3, and IKE. Cell viability, intracellular ROS, MDA, iron levels, as well as mRNA and protein expressions of target genes were measured. RESULTS: SIRT3 expression in HCT-116 cells was increased by ferroptosis inducers and decreased by ferroptosis inhibitors. SIRT3 overexpression reduced cell viability and increased intracellular levels of ROS, MDA, and iron, whereas SIRT3 knockdown achieved the opposite effects. SIRT3 overexpression suppressed SLC7A11 expression and promoted the activation of AMPK/mTOR pathway. Restoration of SLC7A11 expression blocked the effects of SIRT3 on ferroptosis induction and cell viability inhibition. SIRT3 effects on cell viability and ferroptosis were antagonized by inhibitors of AMPK or mTOR. Moreover, sulforaphane triggered the ferroptosis of HCT-116 cells by activating the SIRT3/AMPK/mTOR axis. CONCLUSIONS: SIRT3 triggered SLC7A11-mediated ferroptosis in HCT-116 cells, reducing cell viability by activating the AMPK/mTOR pathway, and sulforaphane targets it to inhibit colorectal cancer.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Neoplasias Colorrectales , Ferroptosis , Isotiocianatos , Transducción de Señal , Sirtuina 3 , Sulfóxidos , Serina-Treonina Quinasas TOR , Humanos , Isotiocianatos/farmacología , Sirtuina 3/metabolismo , Sirtuina 3/genética , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Ferroptosis/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Transducción de Señal/efectos de los fármacos , Células HCT116 , Anticarcinógenos/farmacología , Supervivencia Celular/efectos de los fármacos
11.
Trends Cell Biol ; 34(9): 695-697, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39241754

RESUMEN

In neurodegeneration, neurons release lipids that accumulate in glial lipid droplets (LDs). But what controls lipid transport and how does this affect glia? A recent study by Li et al. discovered that the loss of neuronal AMP-activated protein kinase (AMPK) activity promotes lipid efflux, which drives a proinflammatory state in microglia.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Microglía , Neuronas , Animales , Humanos , Proteínas Quinasas Activadas por AMP/metabolismo , Transporte Biológico , Gotas Lipídicas/metabolismo , Metabolismo de los Lípidos , Microglía/metabolismo , Neuronas/metabolismo , Ratones
12.
Drug Des Devel Ther ; 18: 3925-3938, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39247793

RESUMEN

Background: GLP-1 receptor agonists (GLP-1 RA) have been proven to treat several metabolic diseases; however, the effects of GLP-1 RA on polycystic ovary syndrome (PCOS) remain unclear. Here, we aimed to investigate whether semaglutide, a novel GLP-1 RA, could alleviate ovarian inflammation in PCOS mice. Methods: Female C57BL/6J mice were subcutaneously injected with dehydroepiandrosterone for 21 days to establish the PCOS model. Then the mice were randomly divided into three groups: PCOS group (n = 6), S-0.42 group (semaglutide 0.42 mg/kg/w, n = 6), and S-0.84 group (semaglutide 0.84 mg/kg/w, n = 6). The remaining six mice were used as controls (NC). After 28 days of intervention, serum sex hormones and inflammatory cytokine levels were measured. Hematoxylin and eosin staining was used to observe the ovarian morphology. Immunohistochemical staining was used to detect the relative expression of CYP19A1, TNF-α, IL-6, IL-1ß, and NF-κB in ovaries. CYP17A1 and StAR were detected using immunofluorescence staining. Finally, the relative expressions of AMPK, pAMPK, SIRT1, NF-κB, IκBα, pIκBα, TNF-α, IL-6, and IL-1ß were measured using Western blotting. Results: First, after intervention with semaglutide, the weight of the mice decreased, insulin resistance improved, and the estrous cycle returned to normal. Serum testosterone and IL-1ß levels decreased significantly, whereas estradiol and progestin levels increased significantly. Follicular cystic dilation significantly improved. The expression of TNF-α, IL-6, IL-1ß, NF-κB, CYP17A1, and StAR in the ovary was significantly downregulated, whereas CYP19A1 expression was upregulated after the intervention. Finally, we confirmed that semaglutide alleviates ovarian tissue inflammation and improves PCOS through the AMPK/SIRT1/NF-κB signaling pathway. Conclusion: Semaglutide alleviates ovarian inflammation via the AMPK/SIRT1/NF­κB signaling pathway in PCOS mice.


Asunto(s)
Agonistas Receptor de Péptidos Similares al Glucagón , Péptidos Similares al Glucagón , Inflamación , Síndrome del Ovario Poliquístico , Transducción de Señal , Animales , Femenino , Ratones , Proteínas Quinasas Activadas por AMP/metabolismo , Modelos Animales de Enfermedad , Péptidos Similares al Glucagón/farmacología , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Ovario/efectos de los fármacos , Ovario/patología , Ovario/metabolismo , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/patología , Transducción de Señal/efectos de los fármacos , Sirtuina 1/metabolismo , Agonistas Receptor de Péptidos Similares al Glucagón/farmacología
13.
Clin Exp Hypertens ; 46(1): 2402258, 2024 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-39277848

RESUMEN

BACKGROUND: Irisin, as a myokine, plays a protective role against cardiovascular disease, including myocardial infarction, atherosclerosis and hypertension. However, whether irisin attenuates salt-sensitive hypertension and the related underlying mechanisms is unknown. METHODS: Male Dahl salt-resistant (DSR) and Dahl salt-sensitive (DSS) (12 weeks) rats were fed a high salt diet (8% NaCl) with or without irisin treatment by intraperitoneal injection for 8 weeks. RESULTS: Compared with DSR rats, DSS rats showed higher systolic blood pressure (SBP), impaired natriuresis and diuresis and renal dysfunction. In addition, it was accompanied by downregulation of renal p-AMPKα and upregulation of renal RAC1 and nuclear mineralocorticoid receptor (MR). Irisin intervention could significantly up-regulated renal p-AMPKα level and down-regulated renal RAC1-MR signal, thereby improving renal sodium excretion and renal function, and ultimately reducing blood pressure in DSS rats. Ex vivo treatment with irisin reduced the expression of RAC1 and nuclear MR in primary renal distal convoluted tubule cells from DSS rats and the effects of irisin were abolished by cotreatment of compound C (AMPK inhibitor), indicating that the regulation of RAC1-MR signals by irisin depended on the activation of AMPK. CONCLUSIONS: Irisin administration lowered salt-sensitive hypertension through regulating RAC1-MR signaling via activation of AMPK, which may be a promising therapeutic approach for salt-sensitive hypertension.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Presión Sanguínea , Fibronectinas , Hipertensión , Riñón , Ratas Endogámicas Dahl , Transducción de Señal , Proteína de Unión al GTP rac1 , Animales , Masculino , Ratas , Proteínas Quinasas Activadas por AMP/metabolismo , Presión Sanguínea/efectos de los fármacos , Fibronectinas/metabolismo , Hipertensión/metabolismo , Hipertensión/fisiopatología , Hipertensión/tratamiento farmacológico , Riñón/efectos de los fármacos , Riñón/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Receptores de Mineralocorticoides/metabolismo , Transducción de Señal/efectos de los fármacos , Cloruro de Sodio Dietético
14.
Sci Rep ; 14(1): 21275, 2024 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-39261583

RESUMEN

Hepatocellular carcinoma (HCC) associated with viral or metabolic liver diseases is a growing cancer without effective therapy. AMPK is downregulated in HCC and its activation diminishes tumor growth. Alpha lipoic acid (ALA), an indirect AMPK activator that inhibits hepatic steatosis, shows antitumor effects in different cancers. We aimed to study its putative action in liver-cancer derived cell lines through AMPK signaling. We performed cytometric studies for apoptosis and cell cycle, and 2D and 3D migration analysis in HepG2/C3A and Hep3B cells. ALA led to significant inhibition of cell migration/invasion only in HepG2/C3A cells. We showed that these effects depended on AMPK, and ALA also increased the levels and nuclear compartmentalization of the AMPK target p53. The anti-invasive effect of ALA was abrogated in stable-silenced (shTP53) versus isogenic-TP53 HepG2/C3A cells. Furthermore, ALA inhibited epithelial-mesenchymal transition (EMT) in control HepG2/C3A but not in shTP53 nor in Hep3B cells. Besides, we spotted that in patients from the HCC-TCGA dataset some EMT genes showed different expression patterns or survival depending on TP53. ALA emerges as a potent activator of AMPK-p53 axis in HCC cells, and it decreases migration/invasion by reducing EMT which could mitigate the disease in wild-type TP53 patients.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Carcinoma Hepatocelular , Movimiento Celular , Transición Epitelial-Mesenquimal , Neoplasias Hepáticas , Ácido Tióctico , Proteína p53 Supresora de Tumor , Humanos , Ácido Tióctico/farmacología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/tratamiento farmacológico , Movimiento Celular/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/tratamiento farmacológico , Células Hep G2 , Proteínas Quinasas Activadas por AMP/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Invasividad Neoplásica , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos
15.
BMC Cancer ; 24(1): 1079, 2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-39223494

RESUMEN

BACKGROUND: Due to the complex pathophysiological mechanisms involved in cancer progression and metastasis, current therapeutic approaches lack efficacy and have significant adverse effects. Therefore, it is essential to establish novel strategies for combating cancer. Phytochemicals, which possess multiple biological activities, such as antioxidant, anti-inflammatory, antimutagenic, immunomodulatory, antiproliferative, anti-angiogenesis, and antimetastatic properties, can regulate cancer progression and interfere in various stages of cancer development by suppressing various signaling pathways. METHODS: The current systematic and comprehensive review was conducted based on Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) criteria, using electronic databases, including PubMed, Scopus, and Science Direct, until the end of December 2023. After excluding unrelated articles, 111 related articles were included in this systematic review. RESULTS: In this current review, the major signaling pathways of cancer metabolism are highlighted with the promising anticancer role of phytochemicals. This was through their ability to regulate the AMP-activated protein kinase (AMPK)/peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α) signaling pathway. The AMPK/PGC-1α signaling pathway plays a crucial role in cancer cell metabolism via targeting energy homeostasis and mitochondria biogenesis, glucose oxidation, and fatty acid oxidation, thereby generating ATP for cell growth. As a result, targeting this signaling pathway may represent a novel approach to cancer treatment. Accordingly, alkaloids, phenolic compounds, terpene/terpenoids, and miscellaneous phytochemicals have been introduced as promising anticancer agents by regulating the AMPK/PGC-1α signaling pathway. Novel delivery systems of phytochemicals targeting the AMPK/PGC-1α pathway in combating cancer are also highlighted in this review.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Neoplasias , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Fitoquímicos , Transducción de Señal , Humanos , Fitoquímicos/uso terapéutico , Fitoquímicos/farmacología , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Transducción de Señal/efectos de los fármacos
16.
Int J Mol Sci ; 25(17)2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39273120

RESUMEN

PRKAG2 cardiomyopathy is a rare genetic disorder that manifests early in life with an autosomal dominant inheritance pattern. It harbors left ventricular hypertrophy (LVH), ventricular pre-excitation and progressively worsening conduction system defects. Its estimated prevalence among patients with LVH ranges from 0.23 to about 1%, but it is likely an underdiagnosed condition. We report the association of the PRKAG2 missense variant c.1006G>A p. (Val336Ile) with LVH, conduction abnormalities (short PR interval and incomplete right bundle branch bock) and early-onset arterial hypertension (AH) in a 44-year-old Caucasian patient. While cardiac magnetic resonance (CMR) showed a mild hypertrophic phenotype with maximal wall thickness of 17 mm in absence of tissue alterations, the electric phenotype was relevant including brady-tachy syndrome and recurrent syncope. The same variant has been detected in the patient's sister and daughter, with LVH + early-onset AH and electrocardiographic (ECG) alterations + lipothymic episodes, respectively. Paying close attention to the coexistence of LVH and ECG alterations in the proband has been helpful in directing genetic tests to exclude primary cardiomyopathy. Hence, identifying the genetic basis in the patient allowed for familial screening as well as a proper follow-up and therapeutic management of the affected members. A review of the PRKAG2 cardiomyopathy literature is provided alongside the case report.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Hipertrofia Ventricular Izquierda , Mutación Missense , Humanos , Adulto , Hipertrofia Ventricular Izquierda/genética , Femenino , Proteínas Quinasas Activadas por AMP/genética , Electrocardiografía , Masculino , Linaje
17.
Int J Mol Sci ; 25(17)2024 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-39273671

RESUMEN

Glucose-dependent insulinotropic polypeptide (GIP) of the incretin group has been shown to exert pleiotropic actions. There is growing evidence that advanced glycation end products (AGEs), senescent macromolecules formed at an accelerated rate under chronic hyperglycemic conditions, play a role in the pathogenesis of atherosclerotic cardiovascular disease in diabetes. However, whether and how GIP could inhibit the AGE-induced foam cell formation of macrophages, an initial step of atherosclerosis remains to be elucidated. In this study, we address these issues. We found that AGEs increased oxidized low-density-lipoprotein uptake into reactive oxygen species (ROS) generation and Cdk5 and CD36 gene expressions in human U937 macrophages, all of which were significantly blocked by [D-Ala2]GIP(1-42) or an inhibitor of NADPH oxidase activity. An inhibitor of AMP-activated protein kinase (AMPK) attenuated all of the beneficial effects of [D-Ala2]GIP(1-42) on AGE-exposed U937 macrophages, whereas an activator of AMPK mimicked the effects of [D-Ala2]GIP(1-42) on foam cell formation, ROS generation, and Cdk5 and CD36 gene expressions in macrophages. The present study suggests that [D-Ala2]GIP(1-42) could inhibit the AGE-RAGE-induced, NADPH oxidase-derived oxidative stress generation in U937 macrophages via AMPK activation and subsequently suppress macrophage foam cell formation by reducing the Cdk5-CD36 pathway.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Quinasa 5 Dependiente de la Ciclina , Células Espumosas , Polipéptido Inhibidor Gástrico , Productos Finales de Glicación Avanzada , NADPH Oxidasas , Estrés Oxidativo , Especies Reactivas de Oxígeno , Humanos , Células Espumosas/metabolismo , Células Espumosas/efectos de los fármacos , Productos Finales de Glicación Avanzada/metabolismo , Estrés Oxidativo/efectos de los fármacos , NADPH Oxidasas/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Quinasa 5 Dependiente de la Ciclina/metabolismo , Quinasa 5 Dependiente de la Ciclina/genética , Polipéptido Inhibidor Gástrico/metabolismo , Polipéptido Inhibidor Gástrico/farmacología , Antígenos CD36/metabolismo , Antígenos CD36/genética , Células U937 , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Lipoproteínas LDL
18.
Int J Mol Sci ; 25(17)2024 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-39273677

RESUMEN

8-Prenylgenistein (8PG), a genistein derivative, is present in fermented soybeans (Glycine max), including cheonggukjang (CGJ), and exhibits osteoprotective, osteogenic, and antiadipogenic properties. However, the hepatoprotective effects of 8PG and its underlying molecular mechanisms remain largely unexplored. Here, we identified the high binding affinity of 8PG with AMP-activated protein kinase (AMPK) and sirtuin 1 (SIRT1), which acts as a potent AMPK activator that counteracts hepatic steatosis. Notably, 8PG exhibited better pharmacokinetics with greater absorption and higher plasma binding than the positive controls for the target proteins. Moreover, 8PG exerted non-carcinogenic activity in rats and significantly increased AMPK phosphorylation. Compound C, an AMPK inhibitor, did not antagonize 8PG-activated AMPK in HepG2 cells. 8PG significantly attenuated palmitate-induced lipid accumulation and enhanced phosphorylated AMPK and its downstream target, acetyl-CoA carboxylase. Further, 8PG activated nuclear SIRT1 at the protein level, which promoted fatty acid oxidation in palmitate-treated HepG2 cells. Overall, 8PG acts as a potent AMPK activator, further attenuating hepatic steatosis via the SIRT1-mediated pathway and providing new avenues for dietary interventions to treat metabolic dysfunction-associated steatotic liver disease (MASLD).


Asunto(s)
Proteínas Quinasas Activadas por AMP , Isoflavonas , Sirtuina 1 , Sirtuina 1/metabolismo , Animales , Humanos , Proteínas Quinasas Activadas por AMP/metabolismo , Células Hep G2 , Ratas , Masculino , Isoflavonas/farmacología , Isoflavonas/uso terapéutico , Hígado Graso/tratamiento farmacológico , Hígado Graso/metabolismo , Transducción de Señal/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Ratas Sprague-Dawley , Glycine max/química , Genisteína/farmacología
19.
Int J Mol Sci ; 25(17)2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39273687

RESUMEN

The global obesity epidemic, exacerbated by the sedentary lifestyle fostered by the COVID-19 pandemic, presents a growing socioeconomic burden due to decreased physical activity and increased morbidity. Current obesity treatments show promise, but they often come with expensive medications, frequent injections, and potential side effects, with limited success in improving obesity through increased energy expenditure. This study explores the potential of a refined sulfated polysaccharide (SPSL), derived from the brown seaweed Scytosiphon lomentaria (SL), as a safe and effective anti-obesity treatment by promoting energy expenditure. Chemical characterization revealed that SPSL, rich in sulfate and L-fucose content, comprises nine distinct sulfated glycan structures. In vitro analysis demonstrated potent anti-lipogenic properties in adipocytes, mediated by the downregulation of key adipogenic modulators, including 5' adenosine monophosphate-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor γ (PPARγ) pathways. Inhibiting AMPK attenuated the anti-adipogenic effects of SPSL, confirming its involvement in the mechanism of action. Furthermore, in vivo studies using zebrafish models showed that SPSL increased energy expenditure and reduced lipid accumulation. These findings collectively highlight the therapeutic potential of SPSL as a functional food ingredient for mitigating obesity-related metabolic dysregulation by promoting energy expenditure. Further mechanistic and preclinical investigations are warranted to fully elucidate its mode of action and evaluate its efficacy in obesity management, potentially offering a novel, natural therapeutic avenue for this global health concern.


Asunto(s)
Adipogénesis , Metabolismo Energético , Fucosa , Alimentos Funcionales , Obesidad , Polisacáridos , Algas Marinas , Pez Cebra , Animales , Metabolismo Energético/efectos de los fármacos , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Polisacáridos/química , Polisacáridos/farmacología , Algas Marinas/química , Fucosa/metabolismo , Adipogénesis/efectos de los fármacos , Ratones , Adipocitos/metabolismo , Adipocitos/efectos de los fármacos , Humanos , Sulfatos/química , Sulfatos/metabolismo , PPAR gamma/metabolismo , Fármacos Antiobesidad/farmacología , Fármacos Antiobesidad/química , Fármacos Antiobesidad/uso terapéutico , Células 3T3-L1 , Proteínas Quinasas Activadas por AMP/metabolismo
20.
Int J Cardiol ; 416: 132505, 2024 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-39222886

RESUMEN

BACKGROUND: Myocardial ischemia-reperfusion(I/R)injury constitute the fundamental pathophysiology of acute myocardial infarction (AMI). Ischemic heart releases macrophage migration inhibitory factor (MIF), which activates MIF- AMPK signaling pathway. Depression is a significant risk factor for AMI. In a state of depression, peripheral expression of cannabinoid receptor 2 (CNR2) genes was downregulated. AIMS: We investigated the mechanism by which depression exacerbates myocardial I/R injury through the CNR2 and MIF-AMPK signaling pathways. METHODS: We established mouse models of depression and myocardial I/R. Left ventricular function was assessed using cardiac ultrasound and TTC staining. The protein levels of myocardial CNR2, MIF, AMPK, and ACC were determined by Western blot, while the expression level of CNR2 was measured using RT-qPCR. Additionally, MIF content in peripheral blood was quantified using ELISA. RESULTS: After I/R, the expression level of CNR2 was found to be lower in the depression group, leading to a deterioration in left heart function. Depressed mice exhibited lower secretion of MIF, accompanied by a decrease in the activation of the MIF-AMPK signaling pathway. However, injection of CNR2 agonist JWH133 prior to ischemia increased the activation of the MIF-AMPK signaling pathway, while CNR2 inhibitor AM630 decreased the activation. LIMITATIONS: Further research is needed to investigate the specific neuroendocrine mechanism affecting myocardial CNR2 expression in depression. And these experimental conclusions require further verification at the cellular level. CONCLUSIONS: The activation of CNR2 in myocardium following I/R is impeded by depression, thereby exacerbating myocardial I/R injury through attenuation of the MIF-AMPK signaling pathway activation.


Asunto(s)
Oxidorreductasas Intramoleculares , Factores Inhibidores de la Migración de Macrófagos , Ratones Endogámicos C57BL , Daño por Reperfusión Miocárdica , Receptor Cannabinoide CB2 , Transducción de Señal , Animales , Factores Inhibidores de la Migración de Macrófagos/genética , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Receptor Cannabinoide CB2/genética , Receptor Cannabinoide CB2/metabolismo , Receptor Cannabinoide CB2/biosíntesis , Ratones , Transducción de Señal/fisiología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/genética , Masculino , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Depresión/metabolismo , Depresión/etiología , Depresión/genética , Modelos Animales de Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA