Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27.617
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Proteome Res ; 23(7): 2452-2473, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38965921

RESUMEN

Cancer cachexia is an involuntary loss of body weight, mostly of skeletal muscle. Previous research favors the existence of a microbiota-muscle crosstalk, so the aim of the study was to evaluate the impact of microbiota alterations induced by antibiotics on skeletal muscle proteins expression. Skeletal muscle proteome changes were investigated in control (CT) or C26 cachectic mice (C26) with or without antibiotic treatment (CT-ATB or C26-ATB, n = 8 per group). Muscle protein extracts were divided into a sarcoplasmic and myofibrillar fraction and then underwent label-free liquid chromatography separation, mass spectrometry analysis, Mascot protein identification, and METASCAPE platform data analysis. In C26 mice, the atrogen mafbx expression was 353% higher than CT mice and 42.3% higher than C26-ATB mice. No effect on the muscle protein synthesis was observed. Proteomic analyses revealed a strong effect of antibiotics on skeletal muscle proteome outside of cachexia, with adaptative processes involved in protein folding, growth, energy metabolism, and muscle contraction. In C26-ATB mice, proteome adaptations observed in CT-ATB mice were blunted. Differentially expressed proteins were involved in other processes like glucose metabolism, oxidative stress response, and proteolysis. This study confirms the existence of a microbiota-muscle axis, with a muscle response after antibiotics that varies depending on whether cachexia is present.


Asunto(s)
Antibacterianos , Caquexia , Músculo Esquelético , Proteoma , Caquexia/metabolismo , Caquexia/microbiología , Animales , Músculo Esquelético/metabolismo , Músculo Esquelético/efectos de los fármacos , Antibacterianos/farmacología , Antibacterianos/efectos adversos , Proteoma/metabolismo , Proteoma/análisis , Ratones , Neoplasias/metabolismo , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico , Proteínas Musculares/metabolismo , Masculino , Proteómica/métodos , Microbiota/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos
2.
Cell Death Dis ; 15(7): 484, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38969639

RESUMEN

An increasing evidence supports that cell competition, a vital selection and quality control mechanism in multicellular organisms, is involved in tumorigenesis and development; however, the mechanistic contributions to the association between cell competition and tumor drug resistance remain ill-defined. In our study, based on a contructed lenvitinib-resistant hepatocellular carcinoma (HCC) cells display obvious competitive growth dominance over sensitive cells through reprogramming energy metabolism. Mechanistically, the hyperactivation of BCL2 interacting protein3 (BNIP3) -mediated mitophagy in lenvatinib-resistant HCC cells promotes glycolytic flux via shifting energy production from mitochondrial oxidative phosphorylation to glycolysis, by regulating AMP-activated protein kinase (AMPK) -enolase 2 (ENO2) signaling, which perpetually maintaining lenvatinib-resistant HCC cells' competitive advantage over sensitive HCC cells. Of note, BNIP3 inhibition significantly sensitized the anti-tumor efficacy of lenvatinib in HCC. Our findings emphasize a vital role for BNIP3-AMPK-ENO2 signaling in maintaining the competitive outcome of lenvitinib-resistant HCC cells via regulating energy metabolism reprogramming; meanwhile, this work recognizes BNIP3 as a promising target to overcome HCC drug resistance.


Asunto(s)
Carcinoma Hepatocelular , Resistencia a Antineoplásicos , Metabolismo Energético , Neoplasias Hepáticas , Proteínas de la Membrana , Mitofagia , Compuestos de Fenilurea , Quinolinas , Humanos , Quinolinas/farmacología , Mitofagia/efectos de los fármacos , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Proteínas de la Membrana/metabolismo , Metabolismo Energético/efectos de los fármacos , Compuestos de Fenilurea/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Animales , Línea Celular Tumoral , Proteínas Proto-Oncogénicas/metabolismo , Ratones , Ratones Desnudos , Proliferación Celular/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/metabolismo , Ratones Endogámicos BALB C , Reprogramación Metabólica
3.
Biomolecules ; 14(6)2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38927022

RESUMEN

Recent studies increasingly suggest that targeting brown/beige adipose tissues to enhance energy expenditure offers a novel therapeutic approach for treating metabolic diseases. Brown/beige adipocytes exhibit elevated expression of uncoupling protein 1 (UCP1), which is a thermogenic protein that efficiently converts energy into heat, particularly in response to cold stimulation. Polyphenols possess potential anti-obesity properties, but their pharmacological effects are limited by their bioavailability and distribution within tissue. This study discovered 18a, a polyphenol compound with a favorable distribution within adipose tissues, which transcriptionally activates UCP1, thereby promoting thermogenesis and enhancing mitochondrial respiration in brown adipocytes. Furthermore, in vivo studies demonstrated that 18a prevents high-fat-diet-induced weight gain and improves insulin sensitivity. Our research provides strong mechanistic evidence that UCP1 is a complex mediator of 18a-induced thermogenesis, which is a critical process in obesity mitigation. Brown adipose thermogenesis is triggered by 18a via the AMPK-PGC-1α pathway. As a result, our research highlights a thermogenic controlled polyphenol compound 18a and clarifies its underlying mechanisms, thus offering a potential strategy for the thermogenic targeting of adipose tissue to reduce the incidence of obesity and its related metabolic problems.


Asunto(s)
Dieta Alta en Grasa , Obesidad , Polifenoles , Termogénesis , Proteína Desacopladora 1 , Proteína Desacopladora 1/metabolismo , Proteína Desacopladora 1/genética , Termogénesis/efectos de los fármacos , Animales , Obesidad/metabolismo , Obesidad/tratamiento farmacológico , Polifenoles/farmacología , Ratones , Dieta Alta en Grasa/efectos adversos , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Pardo/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Humanos , Metabolismo Energético/efectos de los fármacos
4.
Int J Mol Sci ; 25(12)2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38928027

RESUMEN

A hypothesis is presented to explain how the ageing process might be influenced by optimizing mitochondrial efficiency to reduce intracellular entropy. Research-based quantifications of entropy are scarce. Non-equilibrium metabolic reactions and compartmentalization were found to contribute most to lowering entropy in the cells. Like the cells, mitochondria are thermodynamically open systems exchanging matter and energy with their surroundings-the rest of the cell. Based on the calculations from cancer cells, glycolysis was reported to produce less entropy than mitochondrial oxidative phosphorylation. However, these estimations depended on the CO2 concentration so that at slightly increased CO2, it was oxidative phosphorylation that produced less entropy. Also, the thermodynamic efficiency of mitochondrial respiratory complexes varies depending on the respiratory state and oxidant/antioxidant balance. Therefore, in spite of long-standing theoretical and practical efforts, more measurements, also in isolated mitochondria, with intact and suboptimal respiration, are needed to resolve the issue. Entropy increases in ageing while mitochondrial efficiency of energy conversion, quality control, and turnover mechanisms deteriorate. Optimally functioning mitochondria are necessary to meet energy demands for cellular defence and repair processes to attenuate ageing. The intuitive approach of simply supplying more metabolic fuels (more nutrients) often has the opposite effect, namely a decrease in energy production in the case of nutrient overload. Excessive nutrient intake and obesity accelerate ageing, while calorie restriction without malnutrition can prolong life. Balanced nutrient intake adapted to needs/activity-based high ATP requirement increases mitochondrial respiratory efficiency and leads to multiple alterations in gene expression and metabolic adaptations. Therefore, rather than overfeeding, it is necessary to fine-tune energy production by optimizing mitochondrial function and reducing oxidative stress; the evidence is discussed in this paper.


Asunto(s)
Envejecimiento , Entropía , Mitocondrias , Especies Reactivas de Oxígeno , Mitocondrias/metabolismo , Humanos , Envejecimiento/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Metabolismo Energético , Estrés Oxidativo , Fosforilación Oxidativa
5.
Plant Cell Rep ; 43(7): 180, 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38914787

RESUMEN

KEY MESSAGE: Hydrogen sulfide improved cold resistance of tomato fruits by regulating energy metabolism and delaying cell wall degradation, thereby alleviating the damage of cold storage on fruits. Postharvest cold storage in tomato fruits extended shelf life but caused the appearance of chilling injury (CI), appeared by softness and spots on the surface of the fruits. These changes were linked closely with energy and cell wall metabolisms. Hydrogen sulfide (H2S), as the gaseous fresh-keeping regulator, was used in the present study to investigate the effects of H2S on energy and cell wall metabolisms in tomato fruits during cold storage. Fruits after harvest were fumigated with different concentrations (0, 0.5, 1, 1.5 mM) of sodium hydrosulfide (NaHS) solution as H2S honor for 24 h and stored at 4 °C for 25 days. The results showed that 1 and 1.5 mM NaHS solution fumigation promoted the accumulation of endogenous H2S, followed by the increase in L-cysteine desulfurase (LCD) and D-cysteine desulfurase (DCD) activities in fruits during cold storage. It was also found that 1 and 1.5 mM NaHS treatments improved H+-ATPase, Ca2+-ATPase, cytochrome C oxidase (CCO), and succinic dehydrogenase (SDH) activities. Moreover, the contents of cellulose and hemicellulose were increased by 1 and 1.5 mM NaHS, following down-regulated activities of cellulase (CL), pectin lyase (PL), α-mannosidase (α-man) and ß-Galactosidase (ß-Gal) and down-regulated expression of PL1, PL8, MAN4 and MAN7 genes. Thus, H2S alleviates CI led by cold storage in tomato fruits via regulating energy and cell wall metabolisms.


Asunto(s)
Pared Celular , Frío , Metabolismo Energético , Frutas , Sulfuro de Hidrógeno , Solanum lycopersicum , Pared Celular/metabolismo , Sulfuro de Hidrógeno/farmacología , Sulfuro de Hidrógeno/metabolismo , Solanum lycopersicum/genética , Solanum lycopersicum/metabolismo , Solanum lycopersicum/fisiología , Frutas/metabolismo , Frutas/genética , Frutas/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Regulación de la Expresión Génica de las Plantas/efectos de los fármacos , Proteínas de Plantas/metabolismo , Proteínas de Plantas/genética , Almacenamiento de Alimentos/métodos , Sulfuros/farmacología , Sulfuros/metabolismo
6.
Sci Adv ; 10(26): eadn5229, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38924414

RESUMEN

There is a regional preference around lymph nodes (LNs) for adipose beiging. Here, we show that local LN removal within inguinal white adipose tissue (iWAT) greatly impairs cold-induced beiging, and this impairment can be restored by injecting M2 macrophages or macrophage-derived C-C motif chemokine (CCL22) into iWAT. CCL22 injection into iWAT effectively promotes iWAT beiging, while blocking CCL22 with antibodies can prevent it. Mechanistically, the CCL22 receptor, C-C motif chemokine receptor 4 (CCR4), within eosinophils and its downstream focal adhesion kinase/p65/interleukin-4 signaling are essential for CCL22-mediated beige adipocyte formation. Moreover, CCL22 levels are inversely correlated with body weight and fat mass in mice and humans. Acute elevation of CCL22 levels effectively prevents diet-induced body weight and fat gain by enhancing adipose beiging. Together, our data identify the CCL22-CCR4 axis as an essential mediator for LN-controlled adaptive thermogenesis and highlight its potential to combat obesity and its associated complications.


Asunto(s)
Tejido Adiposo Blanco , Quimiocina CCL22 , Metabolismo Energético , Ganglios Linfáticos , Macrófagos , Termogénesis , Animales , Femenino , Humanos , Masculino , Ratones , Adipocitos Beige/metabolismo , Tejido Adiposo Blanco/metabolismo , Quimiocina CCL22/metabolismo , Eosinófilos/metabolismo , Ganglios Linfáticos/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Obesidad/metabolismo , Receptores CCR4/metabolismo , Transducción de Señal
7.
Proc Natl Acad Sci U S A ; 121(27): e2315944121, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38917002

RESUMEN

Chronic inflammation is epidemiologically linked to the pathogenesis of gastrointestinal diseases, including inflammatory bowel disease (IBD) and colorectal cancer (CRC). However, our understanding of the molecular mechanisms controlling gut inflammation remains insufficient, hindering the development of targeted therapies for IBD and CRC. In this study, we uncovered C15ORF48/miR-147 as a negative regulator of gut inflammation, operating through the modulation of epithelial cell metabolism. C15ORF48/miR-147 encodes two molecular products, C15ORF48 protein and miR-147-3p microRNA, which are predominantly expressed in the intestinal epithelium. C15ORF48/miR-147 ablation leads to gut dysbiosis and exacerbates chemically induced colitis in mice. C15ORF48 and miR-147-3p work together to suppress colonocyte metabolism and inflammation by silencing NDUFA4, a subunit of mitochondrial complex IV (CIV). Interestingly, the C15ORF48 protein, a structural paralog of NDUFA4, contains a unique C-terminal α-helical domain crucial for displacing NDUFA4 from CIV and its subsequent degradation. NDUFA4 silencing hinders NF-κB signaling activation and consequently attenuates inflammatory responses. Collectively, our findings have established the C15ORF48/miR-147-NDUFA4 molecular axis as an indispensable regulator of gut homeostasis, bridging mitochondrial metabolism and inflammation.


Asunto(s)
Metabolismo Energético , Microbioma Gastrointestinal , Inflamación , MicroARNs , Animales , MicroARNs/genética , MicroARNs/metabolismo , Ratones , Metabolismo Energético/genética , Humanos , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Colitis/metabolismo , Colitis/microbiología , Colitis/genética , Colitis/inducido químicamente , FN-kappa B/metabolismo , Disbiosis/metabolismo , Disbiosis/microbiología , Transducción de Señal , Ratones Endogámicos C57BL , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/genética
8.
Nat Aging ; 4(6): 839-853, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38858606

RESUMEN

Thermogenic beige adipocytes are recognized as potential therapeutic targets for combating metabolic diseases. However, the metabolic advantages that they offer are compromised with aging. Here we show that treating mice with estrogen (E2), a hormone that decreases with age, can counteract the age-related decline in beige adipogenesis when exposed to cold temperature while concurrently enhancing energy expenditure and improving glucose tolerance in mice. Mechanistically, we found that nicotinamide phosphoribosyl transferase (NAMPT) plays a pivotal role in facilitating the formation of E2-induced beige adipocytes, which subsequently suppresses the onset of age-related endoplasmic reticulum (ER) stress. Furthermore, we found that targeting NAMPT signaling, either genetically or pharmacologically, can restore the formation of beige adipocytes by increasing the number of perivascular adipocyte progenitor cells. Conversely, the absence of NAMPT signaling prevents this process. Together, our findings shed light on the mechanisms regulating the age-dependent impairment of beige adipocyte formation and underscore the E2-NAMPT-controlled ER stress pathway as a key regulator of this process.


Asunto(s)
Adipocitos Beige , Adipogénesis , Envejecimiento , Estrés del Retículo Endoplásmico , Estrógenos , Nicotinamida Fosforribosiltransferasa , Nicotinamida Fosforribosiltransferasa/metabolismo , Animales , Adipogénesis/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Ratones , Envejecimiento/efectos de los fármacos , Envejecimiento/fisiología , Estrógenos/metabolismo , Estrógenos/farmacología , Adipocitos Beige/efectos de los fármacos , Adipocitos Beige/metabolismo , Citocinas/metabolismo , Transducción de Señal/efectos de los fármacos , Femenino , Ratones Endogámicos C57BL , Metabolismo Energético/efectos de los fármacos
9.
J Anim Sci ; 1022024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38864567

RESUMEN

Dietary net energy for maintenance (NEm) and gain (NEg) can be estimated using calculations based on live performance or adjusted-final body weight, which is calculated based on carcass characteristics. These values are commonly referred to as performance-adjusted (pa) NEm (paNEm) and NEg (paNEg). The NEm and NEg of a diet can also be estimated by adding recovered energy (RE) with heat production (HP) derived from an automated head chamber system (AHCS), which we will term gas-adjusted (ga) NEm (gaNEm) and NEg (gaNEg). Furthermore, HP from the Brouwer equation requires an estimate of urinary nitrogen (UN) excretion, which can be calculated based on N intake, blood urea N, UN concentration, and urine creatinine, or it could be zeroed. Alternatively, HP can be calculated using an alternative equation based on the respiratory quotient. Demonstrating agreement between pa and ga derived dietary energy values provides an opportunity to validate using the AHCS for energetic experiments and this comparison has not been conducted previously. Accordingly, the objective of this experiment was to assess the agreement between live and carcass paNEm and paNEg with gaNEm and gaNEg, where HP was calculated using 4 different approaches. Estimates of HP were not different (P = 0.99) between the 4 approaches employed, indicating that all options investigated are appropriate. Live paNEm and paNEg had a higher agreement (Lin's concordance correlation coefficient [CCC] = 0.91) with gaNEm and gaNEg than carcass values (CCC ≤ 0.84). These results suggest that researchers can implement the AHCS to provide good estimates of dietary energy values in finishing beef cattle that are unrestrained.


Automated head chamber systems (AHCS) implemented into beef cattle research allow estimation of gas flux, heat production (HP), and calculated gas-adjusted dietary net energy for maintenance (gaNEm) and gain (gaNEg) values when paired with recovered energy. However, a comparison between AHCS-derived values and performance-adjusted NEm (paNEm) and NEg (paNEg) from either live performance (live paNEm and paNEg) or carcass data (carcass paNEm and paNEg) has not been conducted. Accordingly, the objectives of this experiment were to evaluate the agreement between gaNEm and gaNEg, estimated using different approaches for calculating HP, with live paNEm and paNEg or carcass paNEm and paNEg. Accounting for urinary nitrogen or methane when calculating HP does not appreciably influence HP estimates or subsequent calculations to estimate dietary NEm and NEg. There was excellent agreement between live paNEm and gaNEm, and between paNEg and gaNEg. Measures of precision, accuracy, and agreement were lower for carcass than for live-derived values when compared to gaNEm and gaNEg but were still acceptable. These results suggest that researchers can implement the AHCS to provide estimates of HP, gas flux, and estimates of dietary energy values in unrestrained finishing beef cattle-fed diets ranging in crude protein content (10.8% to 12.5%). Additional research is warranted on the use of the AHCS to conduct energetic studies across varying diets and production systems, particularly grazing systems.


Asunto(s)
Alimentación Animal , Dieta , Metabolismo Energético , Animales , Bovinos/fisiología , Dieta/veterinaria , Alimentación Animal/análisis , Fenómenos Fisiológicos Nutricionales de los Animales , Ingestión de Energía , Masculino
10.
Obesity (Silver Spring) ; 32(7): 1373-1388, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38932722

RESUMEN

OBJECTIVE: Obesity is characterized by dysregulated homeostatic mechanisms resulting in positive energy balance; however, when this dysregulation occurs is unknown. We assessed the time course of alterations to behaviors promoting weight gain in male and female mice switched to an obesogenic high-fat diet (HFD). METHODS: Male and female C57BL/6J mice were housed in metabolic chambers and were switched from chow to a 60% or 45% HFD for 4 and 3 weeks, respectively. Food intake, meal patterns, energy expenditure (EE), and body weight were continuously measured. A separate cohort of male mice was switched from chow to a 60% HFD and was given access to locked or unlocked running wheels. RESULTS: Switching mice to obesogenic diets promotes transient bouts of hyperphagia during the first 2 weeks followed by persistent caloric hyperphagia. EE increases but not sufficiently enough to offset increased caloric intake, resulting in a sustained net positive energy balance. Hyperphagia is associated with consumption of calorically larger meals (impaired satiation) more frequently (impaired satiety), particularly during the light cycle. Running wheel exercise delays weight gain in male mice fed a 60% HFD by enhancing satiation and increasing EE. However, exercise effects on satiation are no longer apparent after 2 weeks, coinciding with weight gain. CONCLUSIONS: Exposure to obesogenic diets engages homeostatic regulatory mechanisms for ~2 weeks that ultimately fail, and consequent weight gain is characterized by impaired satiation and satiety. Insights into the etiology of obesity can be obtained by investigating changes to satiation and satiety mechanisms during the initial ~2 weeks of HFD exposure.


Asunto(s)
Dieta Alta en Grasa , Ingestión de Energía , Metabolismo Energético , Conducta Alimentaria , Hiperfagia , Ratones Endogámicos C57BL , Obesidad , Aumento de Peso , Animales , Masculino , Ratones , Dieta Alta en Grasa/efectos adversos , Femenino , Obesidad/etiología , Obesidad/metabolismo , Hiperfagia/etiología , Conducta Alimentaria/fisiología , Factores de Tiempo , Condicionamiento Físico Animal , Saciedad , Ingestión de Alimentos/fisiología
11.
Front Immunol ; 15: 1411132, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38840928

RESUMEN

Background: Cervical cancer remains a significant gynecologic malignancy in both China and the United States, posing a substantial threat to women's lives and health due to its high morbidity and mortality rates. Altered energy metabolism and dysregulated mitochondrial function play crucial roles in the development, growth, metastasis, and recurrence of malignant tumors. In this study, we aimed to predict prognosis and assess efficacy of anti-tumor therapy in cervical cancer patients based on differential genes associated with mitochondrial metabolism. Methods: Transcriptomic data and clinical profiles of cervical cancer patients were retrieved from the TCGA and GEO databases. Differential gene-related cellular pathways were identified through GO, KEGG, and GSEA analyses. Prognostic indices were constructed using LASSO regression analysis. Immune cell infiltration was assessed using CIBERSORT and ssGSEA, and the correlation between immune checkpoint inhibitor genes and differential genes was examined. Tumor mutation load (TMB) and its association with prognostic indices were analyzed using nucleotide variant data from the TCGA database. Patient response to immunotherapy and sensitivity to antitumor drugs were determined using the TIDE algorithm and the oncoPredic algorithm, respectively. Results: A prognostic index based on metabolism-related differential genes was developed to predict the clinical outcome of cervical cancer patients, enabling their classification into two distinct subtypes. The prognostic index emerged as an independent risk factor for unfavorable prognosis. The high-index group exhibited a significantly worse overall prognosis, along with elevated tumor mutation burden (TMB), increased immune cell infiltration, and lower TIDE scores, indicating a potential benefit from immunotherapy. Conversely, the low-index group demonstrated increased sensitivity to metabolism-related antitumor agents, specifically multikinase inhibitors. Conclusion: The aim of this study was to develop a prognostic index based on differential genes associated with mitochondrial metabolism, which could be used to predict cervical cancer patients' prognoses. When combined with TIDE and TMB analyses, this prognostic index offers insights into the immune cell infiltration landscape, as well as the potential efficacy of immunotherapy and targeted therapy. Our analysis suggests that the Iron-Sulfur Cluster Assembly Enzyme (ISCU) gene holds promise as a biomarker for cervical cancer immunotherapy.


Asunto(s)
Biomarcadores de Tumor , Neoplasias del Cuello Uterino , Humanos , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/inmunología , Neoplasias del Cuello Uterino/mortalidad , Femenino , Pronóstico , Biomarcadores de Tumor/genética , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Regulación Neoplásica de la Expresión Génica , Transcriptoma , Microambiente Tumoral/inmunología , Microambiente Tumoral/genética , Mitocondrias/metabolismo , Mitocondrias/genética , Metabolismo Energético/genética , Bases de Datos Genéticas , Persona de Mediana Edad , Mutación , Perfilación de la Expresión Génica
12.
Adv Exp Med Biol ; 1441: 417-433, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884723

RESUMEN

This chapter will describe basic structural and functional features of the contractile apparatus of muscle cells of the heart, namely, cardiomyocytes and smooth muscle cells. Cardiomyocytes form the contractile myocardium of the heart, while smooth muscle cells form the contractile coronary vessels. Both muscle types have distinct properties and will be considered with respect to their cellular appearance (brick-like cross-striated versus spindle-like smooth), arrangement of contractile proteins (sarcomeric versus non-sarcomeric organization), calcium activation mechanisms (thin-filament versus thick-filament regulation), contractile features (fast and phasic versus slow and tonic), energy metabolism (high oxygen versus low oxygen demand), molecular motors (type II myosin isoenzymes with high adenosine diphosphate [ADP]-release rate versus myosin isoenzymes with low ADP-release rates), chemomechanical energy conversion (high adenosine triphosphate [ATP] consumption and short duty ratio versus low ATP consumption and high duty ratio of myosin II cross-bridges [XBs]), and excitation-contraction coupling (calcium-induced calcium release versus pharmacomechanical coupling). Part of the work has been published (Neuroscience - From Molecules to Behavior", Chap. 22, Galizia and Lledo eds 2013, Springer-Verlag; with kind permission from Springer Science + Business Media).


Asunto(s)
Contracción Miocárdica , Miocitos Cardíacos , Humanos , Contracción Miocárdica/fisiología , Animales , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Calcio/metabolismo , Metabolismo Energético , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/fisiología , Acoplamiento Excitación-Contracción/fisiología
13.
Sci Rep ; 14(1): 13862, 2024 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-38879688

RESUMEN

Acute kidney injury (AKI) is a systemic disease that affects energy metabolism in various remote organs in murine models of ischemic AKI. However, AKI-mediated effects in the liver have not been comprehensively assessed. After inducing ischemic AKI in 8-10-week-old, male C57BL/6 mice, mass spectrometry metabolomics revealed that the liver had the most distinct phenotype 24 h after AKI versus 4 h and 7 days. Follow up studies with in vivo [13C6]-glucose tracing on liver and kidney 24 h after AKI revealed 4 major findings: (1) increased flux through glycolysis and the tricarboxylic (TCA) cycle in both kidney and liver; (2) depleted hepatic glutathione levels and its intermediates despite unchanged level of reactive oxygen species, suggesting glutathione consumption exceeds production due to systemic oxidative stress after AKI; (3) hepatic ATP depletion despite unchanged rate of mitochondrial respiration, suggesting increased ATP consumption relative to production; (4) increased hepatic and renal urea cycle intermediates suggesting hypercatabolism and upregulation of the urea cycle independent of impaired renal clearance of nitrogenous waste. Taken together, this is the first study to describe the hepatic metabolome after ischemic AKI in a murine model and demonstrates that there is significant liver-kidney crosstalk after AKI.


Asunto(s)
Lesión Renal Aguda , Metabolismo Energético , Glutatión , Riñón , Hígado , Ratones Endogámicos C57BL , Animales , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/etiología , Hígado/metabolismo , Glutatión/metabolismo , Riñón/metabolismo , Masculino , Ratones , Isquemia/metabolismo , Metabolómica/métodos , Modelos Animales de Enfermedad , Estrés Oxidativo , Glucólisis , Metaboloma
14.
J Exp Clin Cancer Res ; 43(1): 165, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38877560

RESUMEN

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is the most lethal cancer with an aggressive metastatic phenotype and very poor clinical prognosis. Interestingly, a lower occurrence of PDAC has been described in individuals with severe and long-standing asthma. Here we explored the potential link between PDAC and the glucocorticoid (GC) budesonide, a first-line therapy to treat asthma. METHODS: We tested the effect of budesonide and the classical GCs on the morphology, proliferation, migration and invasiveness of patient-derived PDAC cells and pancreatic cancer cell lines, using 2D and 3D cultures in vitro. Furthermore, a xenograft model was used to investigate the effect of budesonide on PDAC tumor growth in vivo. Finally, we combined genome-wide transcriptome analysis with genetic and pharmacological approaches to explore the mechanisms underlying budesonide activities in the different environmental conditions. RESULTS: We found that in 2D culture settings, high micromolar concentrations of budesonide reduced the mesenchymal invasive/migrating features of PDAC cells, without affecting proliferation or survival. This activity was specific and independent of the Glucocorticoid Receptor (GR). Conversely, in a more physiological 3D environment, low nanomolar concentrations of budesonide strongly reduced PDAC cell proliferation in a GR-dependent manner. Accordingly, we found that budesonide reduced PDAC tumor growth in vivo. Mechanistically, we demonstrated that the 3D environment drives the cells towards a general metabolic reprogramming involving protein, lipid, and energy metabolism (e.g., increased glycolysis dependency). This metabolic change sensitizes PDAC cells to the anti-proliferative effect of budesonide, which instead induces opposite changes (e.g., increased mitochondrial oxidative phosphorylation). Finally, we provide evidence that budesonide inhibits PDAC growth, at least in part, through the tumor suppressor CDKN1C/p57Kip2. CONCLUSIONS: Collectively, our study reveals that the microenvironment influences the susceptibility of PDAC cells to GCs and provides unprecedented evidence for the anti-proliferative activity of budesonide on PDAC cells in 3D conditions, in vitro and in vivo. Our findings may explain, at least in part, the reason for the lower occurrence of pancreatic cancer in asthmatic patients and suggest a potential suitability of budesonide for clinical trials as a therapeutic approach to fight pancreatic cancer.


Asunto(s)
Budesonida , Proliferación Celular , Metabolismo Energético , Neoplasias Pancreáticas , Humanos , Budesonida/farmacología , Budesonida/uso terapéutico , Ratones , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Metabolismo Energético/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Animales , Línea Celular Tumoral , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Ensayos Antitumor por Modelo de Xenoinjerto , Movimiento Celular/efectos de los fármacos
15.
J Ovarian Res ; 17(1): 118, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38822408

RESUMEN

In women who are getting older, the quantity and quality of their follicles or oocytes and decline. This is characterized by decreased ovarian reserve function (DOR), fewer remaining oocytes, and lower quality oocytes. As more women choose to delay childbirth, the decline in fertility associated with age has become a significant concern for modern women. The decline in oocyte quality is a key indicator of ovarian aging. Many studies suggest that age-related changes in oocyte energy metabolism may impact oocyte quality. Changes in oocyte energy metabolism affect adenosine 5'-triphosphate (ATP) production, but how related products and proteins influence oocyte quality remains largely unknown. This review focuses on oocyte metabolism in age-related ovarian aging and its potential impact on oocyte quality, as well as therapeutic strategies that may partially influence oocyte metabolism. This research aims to enhance our understanding of age-related changes in oocyte energy metabolism, and the identification of biomarkers and treatment methods.


Asunto(s)
Envejecimiento , Metabolismo Energético , Oocitos , Ovario , Oocitos/metabolismo , Humanos , Femenino , Envejecimiento/metabolismo , Ovario/metabolismo , Animales , Adenosina Trifosfato/metabolismo
16.
Circ Res ; 134(12): 1824-1840, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38843291

RESUMEN

Immunometabolism is an emerging field at the intersection of immunology and metabolism. Immune cell activation plays a critical role in the pathogenesis of cardiovascular diseases and is integral for regeneration during cardiac injury. We currently possess a limited understanding of the processes governing metabolic interactions between immune cells and cardiomyocytes. The impact of this intercellular crosstalk can manifest as alterations to the steady state flux of metabolites and impact cardiac contractile function. Although much of our knowledge is derived from acute inflammatory response, recent work emphasizes heterogeneity and flexibility in metabolism between cardiomyocytes and immune cells during pathological states, including ischemic, cardiometabolic, and cancer-associated disease. Metabolic adaptation is crucial because it influences immune cell activation, cytokine release, and potential therapeutic vulnerabilities. This review describes current concepts about immunometabolic regulation in the heart, focusing on intercellular crosstalk and intrinsic factors driving cellular regulation. We discuss experimental approaches to measure the cardio-immunologic crosstalk, which are necessary to uncover unknown mechanisms underlying the immune and cardiac interface. Deeper insight into these axes holds promise for therapeutic strategies that optimize cardioimmunology crosstalk for cardiac health.


Asunto(s)
Miocitos Cardíacos , Humanos , Animales , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/inmunología , Metabolismo Energético , Cardiomiopatías/metabolismo , Cardiomiopatías/inmunología , Miocardio/metabolismo , Miocardio/inmunología , Miocardio/patología
17.
J Physiol Sci ; 74(1): 32, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38849720

RESUMEN

We investigated whether calorie restriction (CR) enhances metabolic adaptations to endurance training (ET). Ten-week-old male Institute of Cancer Research (ICR) mice were fed ad libitum or subjected to 30% CR. The mice were subdivided into sedentary and ET groups. The ET group performed treadmill running (20-25 m/min, 30 min, 5 days/week) for 5 weeks. We found that CR decreased glycolytic enzyme activity and monocarboxylate transporter (MCT) 4 protein content, while enhancing glucose transporter 4 protein content in the plantaris and soleus muscles. Although ET and CR individually increased citrate synthase activity in the plantaris muscle, the ET-induced increase in respiratory chain complex I protein content was counteracted by CR. In the soleus muscle, mitochondrial enzyme activity and protein levels were increased by ET, but decreased by CR. It has been suggested that CR partially interferes with skeletal muscle adaptation to ET.


Asunto(s)
Restricción Calórica , Metabolismo Energético , Hígado , Transportadores de Ácidos Monocarboxílicos , Músculo Esquelético , Condicionamiento Físico Animal , Animales , Músculo Esquelético/metabolismo , Masculino , Ratones , Restricción Calórica/métodos , Hígado/metabolismo , Condicionamiento Físico Animal/fisiología , Metabolismo Energético/fisiología , Transportadores de Ácidos Monocarboxílicos/metabolismo , Ratones Endogámicos ICR , Entrenamiento Aeróbico/métodos , Transportador de Glucosa de Tipo 4/metabolismo , Adaptación Fisiológica/fisiología , Citrato (si)-Sintasa/metabolismo , Proteínas Musculares
18.
Mol Biol Rep ; 51(1): 729, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38862809

RESUMEN

BACKGROUND: Preclinical and clinical studies over the past several decades have indicated the potential value of metformin, a widely utilized treatment for Type 2 diabetes, in prostate cancer therapy. Notably, these studies demonstrated metformin's pleiotropic effects on several molecular and metabolic pathways, such as androgen signaling, cell cycle, and cellular bioenergetics. In this study we investigated the role of metformin in regulating intracellular redox status and cell survival in LNCaP prostate cancer cells. METHODS AND RESULTS: The cytotoxic effects of metformin with or without the presence of SBI0206965 (AMPK inhibitor) on LNCaP cells were determined using MTT and trypan blue exclusion assays. Seahorse XP extracellular analysis, Liquid Chromatography/ Mass Spectrophotometry (LC/MS), and 2,7- and Dichlorofluoresin diacetate (DCFDA) assay were used to assess the effects of metformin on cellular bioenergetics, redox status, and redox-related metabolites. mRNA expression and protein concentration of redox-related enzymes were measured using Real Time-qPCR and ELISA assay, respectively. Independently of AMP-activated protein kinase, metformin exhibited a dose- and time-dependent inhibition of LNCaP cell survival, a response mitigated by glutathione or N-acetylcysteine (ROS scavengers) treatment. Notably, these findings were concomitant with a decline in ATP levels and the inhibition of oxidative phosphorylation. The results further indicated metformin's induction of reactive oxygen species, which significantly decreased glutathione levels and the ratio of reduced to oxidized glutathione, as well as the transsulfuration metabolite, cystathionine. Consistent with an induction of oxidative stress condition, metformin increased mRNA levels of the master redox transcription factor Nrf-2 (nuclear factor erythroid-derived 2-like), as well as transsulfuration enzymes cystathionine beta-synthase and cystathionase and GSH synthesis enzymes γ-glutamylcysteine synthetase and glutathione synthetase. CONCLUSION: Our findings highlight multiple mechanisms by which metformin-induced formation of reactive oxygen species may contribute to its efficacy in prostate cancer treatment, including promotion of oxidative stress, Nrf2 activation, and modulation of redox-related pathways, leading to its anti-survival action.


Asunto(s)
Supervivencia Celular , Metformina , Estrés Oxidativo , Neoplasias de la Próstata , Especies Reactivas de Oxígeno , Metformina/farmacología , Humanos , Masculino , Estrés Oxidativo/efectos de los fármacos , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Supervivencia Celular/efectos de los fármacos , Línea Celular Tumoral , Especies Reactivas de Oxígeno/metabolismo , Oxidación-Reducción/efectos de los fármacos , Glutatión/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Metabolismo Energético/efectos de los fármacos
19.
Mol Biol Rep ; 51(1): 727, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38861014

RESUMEN

BACKGROUND: The presence of granulocyte-macrophage colony-stimulating factor (GM-CSF) and its receptor in various testicular cells and spermatozoa suggests a potential role in enhancing spermatogonial and postmeiotic cell development. Moreover, GM-CSF activates the pivotal pathways implicated in sperm motility regulation and glucose metabolism. However, the impact of GM-CSF on testicular biopsies from patients with obstructive azoospermia (OA) remains unexplored. Therefore, this study aimed to investigate the in vitro effects of GM-CSF on the expression of genes related to glucose transporters and signaling pathways, sperm motility, and viability in testicular biopsies. METHODS AND RESULTS: Following testicular sperm extraction from 20 patients diagnosed with OA, each sample was divided into two parts: the experimental samples were incubated with medium containing 2 ng/ml GM-CSF at 37 °C for 60 min, and the control samples were incubated with medium without GM-CSF. Subsequently, the oocytes retrieved from the partner were injected with sperm from the treatment and control groups. The sperm parameters (motility and viability), the expression levels of sperm motility-related genes (PIK3R1, PIK3CA, and AKT1), and the expression levels of sperm energy metabolism-related genes (GLUT1, GLUT3, and GLUT14) were assessed. Furthermore, the fertilization and day 3 embryo development rate and embryo quality were evaluated. Compared with those in the nontreated group, the motility parameters and the mRNA expression levels of PIK3R1, AKT1, and GLUT3 in testicular sperm supplemented with GM-CSF were significantly greater (p < 0.05). However, no significant differences in the mRNA expression of PIK3CA, GLUT1, or GLUT14 were detected. According to the ICSI results, compared with the control group, the GM-CSF treatment group exhibited significantly greater fertilization rates (p = 0.027), Day 3 embryo development rate (p = 0.001), and proportions of good-quality embryos (p = 0.002). CONCLUSIONS: GM-CSF increased the expression of genes related to motility and the energy metabolism pathway and effectively promoted the motility of testis-extracted spermatozoa, consequently yielding positive clinical outcomes.


Asunto(s)
Azoospermia , Metabolismo Energético , Factor Estimulante de Colonias de Granulocitos y Macrófagos , Inyecciones de Esperma Intracitoplasmáticas , Motilidad Espermática , Espermatozoides , Humanos , Masculino , Motilidad Espermática/efectos de los fármacos , Motilidad Espermática/genética , Azoospermia/genética , Azoospermia/tratamiento farmacológico , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Inyecciones de Esperma Intracitoplasmáticas/métodos , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Espermatozoides/metabolismo , Espermatozoides/efectos de los fármacos , Adulto , Testículo/metabolismo , Testículo/efectos de los fármacos , Transportador de Glucosa de Tipo 3/genética , Transportador de Glucosa de Tipo 3/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Femenino , Regulación de la Expresión Génica/efectos de los fármacos
20.
J Nanobiotechnology ; 22(1): 318, 2024 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-38849914

RESUMEN

Mitochondria occupy a central role in the biology of most eukaryotic cells, functioning as the hub of oxidative metabolism where sugars, fats, and amino acids are ultimately oxidized to release energy. This crucial function fuels a variety of cellular activities. Disruption in mitochondrial metabolism is a common feature in many diseases, including cancer, neurodegenerative conditions and cardiovascular diseases. Targeting tumor cell mitochondrial metabolism with multifunctional nanosystems emerges as a promising strategy for enhancing therapeutic efficacy against cancer. This review comprehensively outlines the pathways of mitochondrial metabolism, emphasizing their critical roles in cellular energy production and metabolic regulation. The associations between aberrant mitochondrial metabolism and the initiation and progression of cancer are highlighted, illustrating how these metabolic disruptions contribute to oncogenesis and tumor sustainability. More importantly, innovative strategies employing nanomedicines to precisely target mitochondrial metabolic pathways in cancer therapy are fully explored. Furthermore, key challenges and future directions in this field are identified and discussed. Collectively, this review provides a comprehensive understanding of the current state and future potential of nanomedicine in targeting mitochondrial metabolism, offering insights for developing more effective cancer therapies.


Asunto(s)
Mitocondrias , Nanomedicina , Neoplasias , Humanos , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Nanomedicina/métodos , Animales , Metabolismo Energético/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Nanopartículas/química , Sistemas de Liberación de Medicamentos/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA