Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.074
Filtrar
1.
Sci Rep ; 14(1): 12663, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38830939

RESUMEN

Patients with metabolic dysfunction-associated fatty liver disease (MAFLD) often present with concomitant metabolic dysregulation and alcohol consumption, potentially leading to distinct clinical outcomes. We analyzed data from 8043 participants with MAFLD in the Thai National Health Examination Survey with linked mortality records. According to the MAFLD criteria, 1432 individuals (17.2%) were categorized as having the diabetes phenotype, 5894 (71.0%) as the overweight/obesity phenotype, and 978 (11.8%) as the lean metabolic phenotype. Over 71,145 person-years, 916 participants died. Using Cox proportional hazard models adjusting for physiological, lifestyle, and comorbid factors, both diabetes (adjusted hazards ratio [aHR] 1.59, 95% CI 1.18-2.13) and lean metabolic phenotypes (aHR 1.28, 95% CI 1.01-1.64) exhibited significantly higher mortality risk compared to the overweight/obesity phenotype. A J-shaped relationship was observed between daily alcohol consumption and the risk of all-cause mortality. Daily alcohol intake exceeding 50 g for women and 60 g for men increased the all-cause mortality risk among MAFLD individuals with the lean metabolic phenotype (aHR 3.39, 95% CI 1.02-11.29). Our study found that metabolic phenotype and alcohol consumption have interactive effects on the risk of all-cause mortality in patients with MAFLD, indicating that evaluating both factors is crucial for determining prognostic outcomes and management strategies.


Asunto(s)
Consumo de Bebidas Alcohólicas , Fenotipo , Humanos , Masculino , Femenino , Consumo de Bebidas Alcohólicas/efectos adversos , Persona de Mediana Edad , Adulto , Factores de Riesgo , Estudios de Cohortes , Modelos de Riesgos Proporcionales , Obesidad/complicaciones , Obesidad/mortalidad , Obesidad/metabolismo , Anciano , Tailandia/epidemiología , Enfermedades Metabólicas/mortalidad , Enfermedades Metabólicas/metabolismo
2.
J Mol Model ; 30(6): 170, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38753123

RESUMEN

CONTEXT: In the pursuit of novel therapeutic possibilities, repurposing existing drugs has gained prominence as an efficient strategy. The findings from our study highlight the potential of repurposed drugs as promising candidates against receptor for advanced glycation endproducts (RAGE) that offer therapeutic implications in cancer, neurodegenerative conditions and metabolic syndromes. Through careful analyses of binding affinities and interaction patterns, we identified a few promising candidates, ultimately focusing on sertindole and temoporfin. These candidates exhibited exceptional binding affinities, efficacy, and specificity within the RAGE binding pocket. Notably, they displayed a pronounced propensity to interact with the active site of RAGE. Our investigation further revealed that sertindole and temoporfin possess desirable pharmacological properties that highlighted them as attractive candidates for targeted drug development. Overall, our integrated computational approach provides a comprehensive understanding of the interactions between repurposed drugs, sertindole and temoporfin and RAGE that pave the way for future experimental validation and drug development endeavors. METHODS: We present an integrated approach utilizing molecular docking and extensive molecular dynamics (MD) simulations to evaluate the potential of FDA-approved drugs, sourced from DrugBank, against RAGE. To gain deeper insights into the binding mechanisms of the elucidated candidate repurposed drugs, sertindole and temoporfin with RAGE, we conducted extensive all-atom MD simulations, spanning 500 nanoseconds (ns). These simulations elucidated the conformational dynamics and stability of the RAGE-sertindole and RAGE-temoporfin complexes.


Asunto(s)
Reposicionamiento de Medicamentos , Imidazoles , Indoles , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Receptor para Productos Finales de Glicación Avanzada , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Receptor para Productos Finales de Glicación Avanzada/química , Humanos , Indoles/química , Indoles/farmacología , Imidazoles/química , Imidazoles/farmacología , Unión Proteica , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Enfermedades Metabólicas/tratamiento farmacológico , Enfermedades Metabólicas/metabolismo , Sitios de Unión
3.
J Nanobiotechnology ; 22(1): 226, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38711066

RESUMEN

Nanozyme, characterized by outstanding and inherent enzyme-mimicking properties, have emerged as highly promising alternatives to natural enzymes owning to their exceptional attributes such as regulation of oxidative stress, convenient storage, adjustable catalytic activities, remarkable stability, and effortless scalability for large-scale production. Given the potent regulatory function of nanozymes on oxidative stress and coupled with the fact that reactive oxygen species (ROS) play a vital role in the occurrence and exacerbation of metabolic diseases, nanozyme offer a unique perspective for therapy through multifunctional activities, achieving essential results in the treatment of metabolic diseases by directly scavenging excess ROS or regulating pathologically related molecules. The rational design strategies, nanozyme-enabled therapeutic mechanisms at the cellular level, and the therapies of nanozyme for several typical metabolic diseases and underlying mechanisms are discussed, mainly including obesity, diabetes, cardiovascular disease, diabetic wound healing, and others. Finally, the pharmacokinetics, safety analysis, challenges, and outlooks for the application of nanozyme are also presented. This review will provide some instructive perspectives on nanozyme and promote the development of enzyme-mimicking strategies in metabolic disease therapy.


Asunto(s)
Enfermedades Metabólicas , Estrés Oxidativo , Especies Reactivas de Oxígeno , Humanos , Enfermedades Metabólicas/tratamiento farmacológico , Enfermedades Metabólicas/metabolismo , Animales , Especies Reactivas de Oxígeno/metabolismo , Estrés Oxidativo/efectos de los fármacos , Nanoestructuras/química , Nanoestructuras/uso terapéutico , Nanopartículas/química , Enzimas/metabolismo , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/metabolismo , Obesidad/metabolismo , Obesidad/tratamiento farmacológico
4.
Rev Invest Clin ; 76(2): 65-79, 2024 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-38718804

RESUMEN

UNASSIGNED: Excess body weight has become a global epidemic and a significant risk factor for developing chronic diseases, which are the leading causes of worldwide morbidities. Adipose tissue (AT), primarily composed of adipocytes, stores substantial amounts of energy and plays a crucial role in maintaining whole-body glucose and lipid metabolism. This helps prevent excessive body fat accumulation and lipotoxicity in peripheral tissues. In addition, AT contains endothelial cells and a substantial population of immune cells (constituting 60-70% of non-adipocyte cells), including macrophages, T and B lymphocytes, and natural killer cells. These resident immune cells engage in crosstalk with adipocytes, contributing to the maintenance of metabolic and immune homeostasis in AT. An exacerbated inflammatory response or inadequate immune resolution can lead to chronic systemic low-grade inflammation, triggering the development of metabolic alterations and the onset of chronic diseases. This review aims to elucidate the regulatory mechanisms through which immune cells influence AT function and energy homeostasis. We also focus on the interactions and functional dynamics of immune cell populations, highlighting their role in maintaining the delicate balance between metabolic health and obesity-related inflammation. Finally, understanding immunometabolism is crucial for unraveling the pathogenesis of metabolic diseases and developing targeted immunotherapeutic strategies. These strategies may offer innovative avenues in the rapidly evolving field of immunometabolism. (Rev Invest Clin. 2024;76(2):65-79).


Asunto(s)
Tejido Adiposo , Inflamación , Enfermedades Metabólicas , Obesidad , Humanos , Tejido Adiposo/metabolismo , Tejido Adiposo/inmunología , Obesidad/inmunología , Obesidad/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Enfermedades Metabólicas/inmunología , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/etiología , Metabolismo Energético/fisiología , Adipocitos/metabolismo , Adipocitos/inmunología , Metabolismo de los Lípidos/fisiología , Animales , Homeostasis
5.
Int J Mol Sci ; 25(9)2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38732118

RESUMEN

Metabolic dysfunction-associated steatotic liver disease (MASLD) is an increasingly prevalent condition characterized by abnormal fat accumulation in the liver, often associated with metabolic disorders. Emerging evidence suggests a potential link between vitamin D deficiency and the development and progression of MASLD. The current review provides a concise overview of recent studies uncovering novel mechanistic insights into the interplay between vitamin D and MASLD. Several epidemiological studies have highlighted a significant association between low vitamin D levels and an increased risk of MASLD. Vitamin D, traditionally known for its role in bone health, has now been recognized as a key player in various physiological processes, including immune regulation and inflammation. Experimental studies using animal models have demonstrated that vitamin D deficiency exacerbates liver steatosis and inflammation, suggesting a potential protective role against MASLD. Mechanistically, vitamin D appears to modulate MASLD through multiple pathways. Firstly, the vitamin D receptor (VDR) is abundantly expressed in liver cells, indicating a direct regulatory role in hepatic function. Activation of the VDR has been shown to suppress hepatic lipid accumulation and inflammation, providing a mechanistic basis for the observed protective effects. Additionally, vitamin D influences insulin sensitivity, a critical factor in MASLD pathogenesis. Improved insulin sensitivity may mitigate the excessive accumulation of fat in the liver, thus attenuating MASLD progression. In parallel, vitamin D exhibits anti-inflammatory properties by inhibiting pro-inflammatory cytokines implicated in MASLD pathophysiology. Experimental evidence suggests that the immunomodulatory effects of vitamin D extend to the liver, reducing inflammation and oxidative stress, key drivers of MASLD, and the likelihood of hepatocyte injury and fibrosis. Understanding the complex interplay between vitamin D and MASLD provides a basis for exploring targeted therapeutic strategies and preventive interventions. As vitamin D deficiency is a modifiable risk factor, addressing this nutritional concern may prove beneficial in mitigating the burden of MASLD and associated metabolic disorders.


Asunto(s)
Hígado Graso , Receptores de Calcitriol , Deficiencia de Vitamina D , Vitamina D , Humanos , Vitamina D/metabolismo , Animales , Deficiencia de Vitamina D/complicaciones , Deficiencia de Vitamina D/metabolismo , Receptores de Calcitriol/metabolismo , Hígado Graso/metabolismo , Hígado Graso/etiología , Resistencia a la Insulina , Hígado/metabolismo , Hígado/patología , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/etiología
6.
Front Endocrinol (Lausanne) ; 15: 1328139, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38742195

RESUMEN

The topic of human circadian rhythms is not only attracting the attention of clinical researchers from various fields but also sparking a growing public interest. The circadian system comprises the central clock, located in the suprachiasmatic nucleus of the hypothalamus, and the peripheral clocks in various tissues that are interconnected; together they coordinate many daily activities, including sleep and wakefulness, physical activity, food intake, glucose sensitivity and cardiovascular functions. Disruption of circadian regulation seems to be associated with metabolic disorders (particularly impaired glucose tolerance) and cardiovascular disease. Previous clinical trials revealed that disturbance of the circadian system, specifically due to shift work, is associated with an increased risk of type 2 diabetes mellitus. This review is intended to provide clinicians who wish to implement knowledge of circadian disruption in diagnosis and strategies to avoid cardio-metabolic disease with a general overview of this topic.


Asunto(s)
Enfermedades Cardiovasculares , Ritmo Circadiano , Enfermedades Metabólicas , Humanos , Ritmo Circadiano/fisiología , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/fisiopatología , Enfermedades Metabólicas/fisiopatología , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/etiología , Diabetes Mellitus Tipo 2/fisiopatología , Diabetes Mellitus Tipo 2/metabolismo , Trastornos Cronobiológicos/fisiopatología , Trastornos Cronobiológicos/complicaciones
7.
Gut Microbes ; 16(1): 2359515, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38808455

RESUMEN

The intestinal microbiota, consisting of an estimated 10^10-10^11 organisms, regulate physiological processes involved in digestion, metabolism, and immunity. Surprisingly, these intestinal microorganisms have been found to influence tissues that are not directly in contact with the gut, such as adipose tissue, the liver, skeletal muscle, and the brain. This interaction takes place even when intestinal barrier function is uncompromised. An increasing body of evidence suggests that bacterial membrane vesicles (bMVs), in addition to bacterial metabolites such as short-chain fatty acids, are able to mediate effects of the microbiota on these host tissues. The ability of bMVs to dissipate from the intestinal lumen into systemic circulation hereby facilitates the transport and presentation of bacterial components and metabolites to host organs. Importantly, there are indications that the interaction between bMVs and tissues or immune cells may play a role in the etiology of (chronic metabolic) disease. For example, the gut-derived bMV-mediated induction of insulin resistance in skeletal muscle cells and pro-inflammatory signaling by adipocytes possibly underlies diseases such as type 2 diabetes and obesity. Here, we review the current knowledge on bMVs in the microbiota's effects on host energy/substrate metabolism with a focus on etiological roles in the onset and progression of metabolic disease. We furthermore illustrate that vesicle production by bacterial microbiota could potentially be modulated through lifestyle intervention to improve host metabolism.


Asunto(s)
Bacterias , Microbioma Gastrointestinal , Animales , Humanos , Bacterias/metabolismo , Bacterias/clasificación , Bacterias/genética , Vesículas Extracelulares/metabolismo , Microbioma Gastrointestinal/fisiología , Enfermedades Metabólicas/microbiología , Enfermedades Metabólicas/metabolismo , Interacciones Microbiota-Huesped
9.
Biomed Pharmacother ; 175: 116683, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38705130

RESUMEN

OBJECTIVE: Blockade of activin 2 receptor (ACVR2) signaling has been shown to improve insulin sensitivity and aid in weight loss. Inhibition of ACVR2 signaling restores cardiac function in multiple heart failure models. However, its potential in the treatment of obesity-related cardiometabolic disease remains unknown. Here, we investigated targeting ACVR2 signaling in cardiometabolic disease manifested with metabolic dysfunction-associated steatotic liver disease (MASLD). METHODS: Mice were fed a high-fat, high-sugar diet combined with the administration of nitric oxide synthase inhibitor L-NAME in drinking water, which causes hypertensive stress. For the last eight weeks, the mice were treated with the soluble ACVR2B decoy receptor (sACVR2B-Fc). RESULTS: sACVR2B-Fc protected against the development of comorbidities associated with cardiometabolic disease. This was most pronounced in the liver where ACVR2 blockade attenuated the development of MASLD including cessation of pro-fibrotic activation. It also significantly reduced total plasma cholesterol levels, impeded brown adipose tissue whitening, and improved cardiac diastolic function. In vitro, ACVR2 ligands activin A, activin B and GDF11 induced profibrotic signaling and the proliferation of human cardiac fibroblasts. CONCLUSIONS: Blockade of ACVR2B exerts broad beneficial effects for therapy of cardiometabolic disease. By reducing obesity, ameliorating cardiovascular deterioration and restraining MASLD, blockade of ACVR2B signaling proves a potential target in MASLD and its comorbidities.


Asunto(s)
Receptores de Activinas Tipo II , Ratones Endogámicos C57BL , NG-Nitroarginina Metil Éster , Transducción de Señal , Animales , Transducción de Señal/efectos de los fármacos , NG-Nitroarginina Metil Éster/farmacología , Masculino , Ratones , Receptores de Activinas Tipo II/metabolismo , Humanos , Dieta Occidental/efectos adversos , Hígado Graso/tratamiento farmacológico , Hígado Graso/metabolismo , Enfermedades Metabólicas/tratamiento farmacológico , Enfermedades Metabólicas/metabolismo , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/prevención & control , Enfermedades Cardiovasculares/metabolismo , Hígado/metabolismo , Hígado/efectos de los fármacos , Hígado/patología
10.
Biomed Pharmacother ; 175: 116694, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38713943

RESUMEN

The incidence of metabolic diseases has progressively increased, which has a negative impact on human health and life safety globally. Due to the good efficacy and limited side effects, there is growing interest in developing effective drugs to treat metabolic diseases from natural compounds. Kaempferol (KMP), an important flavonoid, exists in many vegetables, fruits, and traditional medicinal plants. Recently, KMP has received widespread attention worldwide due to its good potential in the treatment of metabolic diseases. To promote the basic research and clinical application of KMP, this review provides a timely and comprehensive summary of the pharmacological advances of KMP in the treatment of four metabolic diseases and its potential molecular mechanisms of action, including diabetes mellitus, obesity, non-alcoholic fatty liver disease (NAFLD)/nonalcoholic steatohepatitis (NASH), and atherosclerosis. According to the research, KMP shows remarkable therapeutic effects on metabolic diseases by regulating multiple signaling transduction pathways such as NF-κB, Nrf2, AMPK, PI3K/AKT, TLR4, and ER stress. In addition, the most recent literature on KMP's natural source, pharmacokinetics studies, as well as toxicity and safety are also discussed in this review, thus providing a foundation and evidence for further studies to develop novel and effective drugs from natural compounds. Collectively, our manuscript strongly suggested that KMP could be a promising candidate for the treatment of metabolic diseases.


Asunto(s)
Aterosclerosis , Diabetes Mellitus , Quempferoles , Enfermedad del Hígado Graso no Alcohólico , Obesidad , Humanos , Quempferoles/farmacología , Quempferoles/uso terapéutico , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Animales , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/metabolismo , Enfermedades Metabólicas/tratamiento farmacológico , Enfermedades Metabólicas/metabolismo , Transducción de Señal/efectos de los fármacos
11.
Biomed Pharmacother ; 175: 116735, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38744220

RESUMEN

G-protein-coupled receptors are a diverse class of cell surface receptors that orchestrate numerous physiological functions. The G-protein-coupled receptors, GPR41 and GPR43, sense short-chain fatty acids (SCFAs), which are metabolites of dietary fermentation by the host's intestinal bacteria. These receptors have gained attention as potential therapeutic targets against various diseases because of their SCFA-mediated beneficial effects on the host's intestinal health. Mounting evidence has associated the activity of these receptors with chronic metabolic diseases, including obesity, diabetes, inflammation, and cardiovascular disease. However, despite intensive research using various strategies, including gene knockout (KO) mouse models, evidence about the precise roles of GPR41 and GPR43 in disease treatment remains inconsistent. Here, we comprehensively review the latest findings from functional studies of the signaling mechanisms that underlie the activities of GPR41 and GPR43, as well as highlight their multifaceted roles in health and disease. We anticipate that this knowledge will guide future research priorities and the development of effective therapeutic interventions.


Asunto(s)
Receptores Acoplados a Proteínas G , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Animales , Humanos , Transducción de Señal , Enfermedades Metabólicas/metabolismo , Ácidos Grasos Volátiles/metabolismo
12.
Int J Mol Sci ; 25(10)2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38791478

RESUMEN

This review highlights the role of postbiotics, which may provide an underappreciated avenue doe promising therapeutic alternatives. The discovery of natural compounds obtained from microorganisms needs to be investigated in the future in terms of their effects on various metabolic disorders and molecular pathways, as well as modulation of the immune system and intestinal microbiota in children and adults. However, further studies and efforts are needed to evaluate and describe new postbiotics. This review provides available knowledge that may assist future research in identifying new postbiotics and uncovering additional mechanisms to combat metabolic diseases.


Asunto(s)
Microbioma Gastrointestinal , Humanos , Animales , Enfermedades Metabólicas/microbiología , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/tratamiento farmacológico , Enfermedades Metabólicas/terapia , Probióticos/uso terapéutico , Productos Biológicos/farmacología , Productos Biológicos/uso terapéutico
13.
Front Endocrinol (Lausanne) ; 15: 1379228, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38745956

RESUMEN

Aims: Individuals with lipodystrophies typically suffer from metabolic disease linked to adipose tissue dysfunction including lipoatrophic diabetes. In the most severe forms of lipodystrophy, congenital generalised lipodystrophy, adipose tissue may be almost entirely absent. Better therapies for affected individuals are urgently needed. Here we performed the first detailed investigation of the effects of a glucagon like peptide-1 receptor (GLP-1R) agonist in lipoatrophic diabetes, using mice with generalised lipodystrophy. Methods: Lipodystrophic insulin resistant and glucose intolerant seipin knockout mice were treated with the GLP-1R agonist liraglutide either acutely preceding analyses of insulin and glucose tolerance or chronically prior to metabolic phenotyping and ex vivo studies. Results: Acute liraglutide treatment significantly improved insulin, glucose and pyruvate tolerance. Once daily injection of seipin knockout mice with liraglutide for 14 days led to significant improvements in hepatomegaly associated with steatosis and reduced markers of liver fibrosis. Moreover, liraglutide enhanced insulin secretion in response to glucose challenge with concomitantly improved glucose control. Conclusions: GLP-1R agonist liraglutide significantly improved lipoatrophic diabetes and hepatic steatosis in mice with generalised lipodystrophy. This provides important insights regarding the benefits of GLP-1R agonists for treating lipodystrophy, informing more widespread use to improve the health of individuals with this condition.


Asunto(s)
Modelos Animales de Enfermedad , Receptor del Péptido 1 Similar al Glucagón , Resistencia a la Insulina , Lipodistrofia , Liraglutida , Ratones Noqueados , Animales , Liraglutida/farmacología , Liraglutida/uso terapéutico , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Ratones , Lipodistrofia/tratamiento farmacológico , Lipodistrofia/metabolismo , Masculino , Enfermedades Metabólicas/tratamiento farmacológico , Enfermedades Metabólicas/metabolismo , Glucemia/metabolismo , Insulina/metabolismo , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Ratones Endogámicos C57BL
14.
J Agric Food Chem ; 72(15): 8632-8649, 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38577880

RESUMEN

Our previous studies found that Sea Buckthorn polyphenols (SBP) extract inhibits fatty acid synthase (FAS) in vitro. Thus, we continued to explore possible effects and underlying mechanisms of SBP on complicated metabolic disorders in long-term high-fat-diet (HFD)-fed mice. To reveal that, an integrated approach was developed in this study. Targeted quantitative lipidomics with a total of 904 unique lipids mapping contributes to profiling the comprehensive features of disarranged hepatic lipid homeostasis and discovering a set of newfound lipid-based biomarkers to predict the occurrence and indicate the progression of metabolic disorders beyond current indicators. On the other hand, technologies of intermolecular interactions characterization, especially surface plasmon resonance (SPR) assay, contribute to recognizing targeted bioactive constituents present in SBP. Our findings highlight hepatic lipid homeostasis maintenance and constituent-FAS enzyme interactions, to provide new insights that SBP as a functional food alleviates HFD-induced metabolic disorders in mice via reprograming hepatic lipid homeostasis caused by targeting FAS, owing to four polyphenols directly interacting with FAS and cinaroside binding to FAS with good affinity.


Asunto(s)
Hippophae , Enfermedades Metabólicas , Ratones , Animales , Polifenoles/metabolismo , Hígado/metabolismo , Dieta Alta en Grasa/efectos adversos , Ácido Graso Sintasas/genética , Ácido Graso Sintasas/metabolismo , Lípidos/farmacología , Enfermedades Metabólicas/metabolismo , Homeostasis , Ratones Endogámicos C57BL , Metabolismo de los Lípidos
15.
Biomed Pharmacother ; 174: 116585, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38615611

RESUMEN

Emerging research into metabolic dysfunction-associated steatotic liver disease (MASLD) up until January 2024 has highlighted the critical role of cuproptosis, a unique cell death mechanism triggered by copper overload, in the disease's development. This connection offers new insights into MASLD's complex pathogenesis, pointing to copper accumulation as a key factor that disrupts lipid metabolism and insulin sensitivity. The identification of cuproptosis as a significant contributor to MASLD underscores the potential for targeting copper-mediated pathways for novel therapeutic approaches. This promising avenue suggests that managing copper levels could mitigate MASLD progression, offering a fresh perspective on treatment strategies. Further investigations into how cuproptosis influences MASLD are essential for unraveling the detailed mechanisms at play and for identifying effective interventions. The focus on copper's role in liver health opens up the possibility of developing targeted therapies that address the underlying causes of MASLD, moving beyond symptomatic treatment to tackle the root of the problem. The exploration of cuproptosis in the context of MASLD exemplifies the importance of understanding metal homeostasis in metabolic diseases and represents a significant step forward in the quest for more effective treatments. This research direction lights path for innovative MASLD management and reversal.


Asunto(s)
Apoptosis , Cobre , Hígado Graso , Animales , Humanos , Cobre/metabolismo , Hígado Graso/metabolismo , Resistencia a la Insulina , Metabolismo de los Lípidos , Hígado/metabolismo , Hígado/patología , Enfermedades Metabólicas/metabolismo
16.
Metabolism ; 155: 155911, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38609037

RESUMEN

BACKGROUND: The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) is increasing year by year and has become one of the leading causes of end-stage liver disease worldwide. Triggering Receptor Expressed on Myeloid Cells 2 (Trem2) has been confirmed to play an essential role in the progression of MASLD, but its specific mechanism still needs to be clarified. This study aims to explore the role and mechanism of Trem2 in MASLD. METHODS: Human liver tissues were obtained from patients with MASLD and controls. Myeloid-specific knockout mice (Trem2mKO) and myeloid-specific overexpression mice (Trem2TdT) were fed a high-fat diet, either AMLN or CDAHFD, to establish the MASLD model. Relevant signaling molecules were assessed through lipidomics and RNA-seq analyses after that. RESULTS: Trem2 is upregulated in human MASLD/MASH-associated macrophages and is associated with hepatic steatosis and inflammation progression. Hepatic steatosis and inflammatory responses are exacerbated with the knockout of myeloid Trem2 in MASLD mice, while mice overexpressing Trem2 exhibit the opposite phenomenon. Mechanistically, Trem2mKO can aggravate macrophage pyroptosis through the PI3K/AKT signaling pathway and amplify the resulting inflammatory response. At the same time, Trem2 promotes the inflammation resolution phenotype transformation of macrophages through TGFß1, thereby promoting tissue repair. CONCLUSIONS: Myeloid Trem2 ameliorates the progression of Metabolic dysfunction-associated steatotic liver disease by regulating macrophage pyroptosis and inflammation resolution. We believe targeting myeloid Trem2 could represent a potential avenue for treating MASLD.


Asunto(s)
Progresión de la Enfermedad , Hígado Graso , Inflamación , Macrófagos , Glicoproteínas de Membrana , Ratones Noqueados , Piroptosis , Receptores Inmunológicos , Animales , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/genética , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Ratones , Humanos , Macrófagos/metabolismo , Inflamación/metabolismo , Inflamación/patología , Piroptosis/fisiología , Hígado Graso/metabolismo , Hígado Graso/patología , Hígado Graso/genética , Masculino , Ratones Endogámicos C57BL , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/patología , Enfermedades Metabólicas/genética , Hígado/metabolismo , Hígado/patología
17.
Cell Metab ; 36(5): 912-926, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38608696

RESUMEN

Metabolic dysfunction-associated steatohepatitis (MASH) is a leading etiology of chronic liver disease worldwide, with increasing incidence and prevalence in the setting of the obesity epidemic. MASH is also a leading indication for liver transplantation, given its associated risk of progression to end-stage liver disease. A key challenge in managing MASH is the lack of approved pharmacotherapy. In its absence, lifestyle interventions with a focus on healthy nutrition and regular physical activity have been the cornerstone of therapy. Real-world efficacy and sustainability of lifestyle interventions are low, however. Pharmacotherapy development for MASH is emerging with promising data from several agents with different mechanisms of action (MOAs) in phase 3 clinical trials. In this review, we highlight ongoing challenges and potential solutions in drug development for MASH and provide an overview of available data from emerging therapies across multiple MOAs.


Asunto(s)
Hígado Graso , Humanos , Hígado Graso/terapia , Hígado Graso/metabolismo , Animales , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/terapia
18.
Biomolecules ; 14(4)2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38672429

RESUMEN

In 1992, a transcendental report suggested that the receptor of advanced glycation end-products (RAGE) functions as a cell surface receptor for a wide and diverse group of compounds, commonly referred to as advanced glycation end-products (AGEs), resulting from the non-enzymatic glycation of lipids and proteins in response to hyperglycemia. The interaction of these compounds with RAGE represents an essential element in triggering the cellular response to proteins or lipids that become glycated. Although initially demonstrated for diabetes complications, a growing body of evidence clearly supports RAGE's role in human diseases. Moreover, the recognizing capacities of this receptor have been extended to a plethora of structurally diverse ligands. As a result, it has been acknowledged as a pattern recognition receptor (PRR) and functionally categorized as the RAGE axis. The ligation to RAGE leads the initiation of a complex signaling cascade and thus triggering crucial cellular events in the pathophysiology of many human diseases. In the present review, we intend to summarize basic features of the RAGE axis biology as well as its contribution to some relevant human diseases such as metabolic diseases, neurodegenerative, cardiovascular, autoimmune, and chronic airways diseases, and cancer as a result of exposure to AGEs, as well as many other ligands.


Asunto(s)
Productos Finales de Glicación Avanzada , Inflamación , Receptor para Productos Finales de Glicación Avanzada , Humanos , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Inflamación/metabolismo , Transducción de Señal , Neoplasias/metabolismo , Animales , Enfermedades Cardiovasculares/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Metabólicas/metabolismo , Enfermedades Autoinmunes/metabolismo
19.
Nat Metab ; 6(4): 639-650, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38671149

RESUMEN

Thyroid hormones (THs) are key hormones that regulate development and metabolism in mammals. In man, the major target tissues for TH action are the brain, liver, muscle, heart, and adipose tissue. Defects in TH synthesis, transport, metabolism, and nuclear action have been associated with genetic and endocrine diseases in man. Over the past few years, there has been renewed interest in TH action and the therapeutic potential of THs and thyromimetics to treat several metabolic disorders such as hypercholesterolemia, dyslipidaemia, non-alcoholic fatty liver disease (NAFLD), and TH transporter defects. Recent advances in the development of tissue and TH receptor isoform-targeted thyromimetics have kindled new hope for translating our fundamental understanding of TH action into an effective therapy. This review provides a concise overview of the historical development of our understanding of TH action, its physiological and pathophysiological effects on metabolism, and future therapeutic applications to treat metabolic dysfunction.


Asunto(s)
Hormonas Tiroideas , Humanos , Hormonas Tiroideas/metabolismo , Animales , Enfermedades Metabólicas/metabolismo , Receptores de Hormona Tiroidea/metabolismo
20.
Cell Metab ; 36(5): 893-911, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38614092

RESUMEN

On average, aging is associated with unfavorable changes in cellular metabolism, which are the processes involved in the storage and expenditure of energy. However, metabolic dysregulation may not occur to the same extent in all older individuals as people age at different rates. Those who are aging rapidly are at increased risk of adverse health outcomes and are said to be "frail." Here, we explore the links between frailty and metabolism, including metabolic contributors and consequences of frailty. We examine how metabolic diseases may modify the degree of frailty in old age and suggest that frailty may predispose toward metabolic disease. Metabolic interventions that can mitigate the degree of frailty in people are reviewed. New treatment strategies developed in animal models that are poised for translation to humans are also considered. We suggest that maintaining a youthful metabolism into older age may be protective against frailty.


Asunto(s)
Envejecimiento , Fragilidad , Humanos , Fragilidad/metabolismo , Animales , Envejecimiento/metabolismo , Enfermedades Metabólicas/metabolismo , Anciano , Metabolismo Energético , Anciano Frágil
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA