Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Proteome Res ; 2024 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-39024330

RESUMEN

Ferroptosis adversely affects the viability, differentiation, and metabolic integrity of C2C12 myoblasts, contributing to the decline in skeletal muscle health. The intricate mechanisms behind this process are not fully understood. In this study, we induced ferroptosis in myoblasts using targeted inducers and found a marked decrease in specific redox metabolites, particularly taurine. Taurine supplementation effectively reversed the deleterious effects of ferroptosis, significantly increased cellular glutathione levels, reduced MDA and ROS levels, and rejuvenated impaired myogenic differentiation. Furthermore, taurine downregulated HO-1 expression and decreased intracellular Fe2+ levels, thereby stabilizing the labile iron pool. Using NMR metabolomic analysis, we observed that taurine profoundly promoted glycerophospholipid metabolism, which is critical for cell membrane repair, and enhanced mitochondrial bioenergetics, thereby increasing the energy reserves essential for muscle satellite cell regeneration. These results suggest that taurine is a potent ferroptosis inhibitor that attenuates key drivers of this process, strengthens oxidative defenses, and improves redox homeostasis. This combined effect protects cells from ferroptosis-induced damage. This study highlights the potential of taurine as a valuable ferroptosis inhibitor that protects skeletal muscle from ferroptosis-induced damage and provides a basis for therapeutic strategies to rejuvenate and facilitate the regeneration of aging skeletal muscle.

2.
J Cell Physiol ; 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38686599

RESUMEN

Lactate can serve as both an energy substrate and a signaling molecule, exerting diverse effects on skeletal muscle physiology. Due to the apparently positive effects, it would be interesting to consider it as a sports supplement. However, the mechanism behind these effects are yet to be comprehensively understood. In this study, we observed that lactate administration could improve the ability of antifatigue, and we further found that lactate upregulated the expression of myosin heavy chain (MYHC I) and MYHC IIa, while downregulating the expression of MYHC IIb. Besides, transcriptomics and metabolomics revealed significant changes in the metabolic profile of gastrocnemius muscle following lactate administration. Furthermore, lactate enhanced the activities of metabolic enzymes, including HK, LDHB, IDH, SDM, and MDH, and promoted the expression of lactate transport-related proteins MCT1 and CD147, thereby improving the transport and utilization of lactate in both vivo and vitro. More importantly, lactate administration increased cellular Ca2+ concentration and facilitated nuclear translocation of nuclear factor of activated T cells (NFATC1) in myotubes, whereas inhibition of NFATC1 significantly attenuated the effects of lactate treatment on NFATC1 nuclear translocation and MyHC expression. Our results elucidate the ability of lactate to induce metabolic remodeling in skeletal muscle and promote myofiber-type transitions by activating the Ca2+-NFATC1 signaling pathway. This study is useful in exploring the potential of lactate as a nutritional supplement for skeletal muscle adaptation and contributing to a mechanistic understanding of the central role of lactate in exercise physiology.

3.
Appl Microbiol Biotechnol ; 108(1): 170, 2024 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-38265689

RESUMEN

The deep-sea environment is an extremely difficult habitat for microorganisms to survive in due to its intense hydrostatic pressure. However, the mechanisms by which these organisms adapt to such extreme conditions remain poorly understood. In this study, we investigated the metabolic adaptations of Microbacterium sediminis YLB-01, a cold and stress-tolerant microorganism isolated from deep-sea sediments, in response to high-pressure conditions. YLB-01 cells were cultured at normal atmospheric pressure and 28 ℃ until they reached the stationary growth phase. Subsequently, the cells were exposed to either normal pressure or high pressure (30 MPa) at 4 ℃ for 7 days. Using NMR-based metabolomic and proteomic analyses of YLB-01 cells exposed to high-pressure conditions, we observed significant metabolic changes in several metabolic pathways, including amino acid, carbohydrate, and lipid metabolism. In particular, the high-pressure treatment stimulates cell division and triggers the accumulation of UDP-glucose, a critical factor in cell wall formation. This finding highlights the adaptive strategies used by YLB-01 cells to survive in the challenging high-pressure environments of the deep sea. Specifically, we discovered that YLB-01 cells regulate amino acid metabolism, promote carbohydrate metabolism, enhance cell wall synthesis, and improve cell membrane fluidity in response to high pressure. These adaptive mechanisms play essential roles in supporting the survival and growth of YLB-01 in high-pressure conditions. Our study offers valuable insights into the molecular mechanisms underlying the metabolic adaptation of deep-sea microorganisms to high-pressure environments. KEY POINTS: • NMR-based metabolomic and proteomic analyses were conducted on Microbacterium sediminis YLB-01 to investigate the significant alterations in several metabolic pathways in response to high-pressure treatment. • YLB-01 cells used adaptive strategies (such as regulated amino acid metabolism, promoted carbohydrate metabolism, enhanced cell wall synthesis, and improved cell membrane fluidity) to survive in the challenging high-pressure environment of the deep sea. • High-pressure treatment stimulated cell division and triggered the accumulation of UDP-glucose, a critical factor in cell wall formation, in Microbacterium sediminis YLB-01 cells.


Asunto(s)
Actinomycetales , Proteómica , Aminoácidos , Glucosa , Uridina Difosfato , Microbacterium
4.
Molecules ; 29(10)2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38792078

RESUMEN

Disuse muscle atrophy (DMA) is a significant healthcare challenge characterized by progressive loss of muscle mass and function resulting from prolonged inactivity. The development of effective strategies for muscle recovery is essential. In this study, we established a DMA mouse model through hindlimb suspension to evaluate the therapeutic potential of lactate in alleviating the detrimental effects on the gastrocnemius muscle. Using NMR-based metabolomic analysis, we investigated the metabolic changes in DMA-injured gastrocnemius muscles compared to controls and evaluated the beneficial effects of lactate treatment. Our results show that lactate significantly reduced muscle mass loss and improved muscle function by downregulating Murf1 expression, decreasing protein ubiquitination and hydrolysis, and increasing myosin heavy chain levels. Crucially, lactate corrected perturbations in four key metabolic pathways in the DMA gastrocnemius: the biosynthesis of phenylalanine, tyrosine, and tryptophan; phenylalanine metabolism; histidine metabolism; and arginine and proline metabolism. In addition to phenylalanine-related pathways, lactate also plays a role in regulating branched-chain amino acid metabolism and energy metabolism. Notably, lactate treatment normalized the levels of eight essential metabolites in DMA mice, underscoring its potential as a therapeutic agent against the consequences of prolonged inactivity and muscle wasting. This study not only advances our understanding of the therapeutic benefits of lactate but also provides a foundation for novel treatment approaches aimed at metabolic restoration and muscle recovery in conditions of muscle wasting.


Asunto(s)
Ácido Láctico , Metabolómica , Músculo Esquelético , Animales , Ratones , Metabolómica/métodos , Ácido Láctico/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/efectos de los fármacos , Atrofia Muscular/metabolismo , Atrofia Muscular/etiología , Atrofia Muscular/tratamiento farmacológico , Atrofia Muscular/patología , Modelos Animales de Enfermedad , Espectroscopía de Resonancia Magnética , Masculino , Proteínas Musculares/metabolismo , Trastornos Musculares Atróficos/metabolismo , Trastornos Musculares Atróficos/tratamiento farmacológico , Trastornos Musculares Atróficos/patología , Ubiquitina-Proteína Ligasas/metabolismo , Metaboloma/efectos de los fármacos , Suspensión Trasera , Proteínas de Motivos Tripartitos/metabolismo , Ratones Endogámicos C57BL , Cadenas Pesadas de Miosina/metabolismo
5.
Molecules ; 29(4)2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38398511

RESUMEN

Trimethylamine N-oxide (TMAO) has attracted interest because of its association with cardiovascular disease and diabetes, and evidence for the beneficial effects of TMAO is accumulating. This study investigates the role of TMAO in improving exercise performance and elucidates the underlying molecular mechanisms. Using C2C12 cells, we established an oxidative stress model and administered TMAO treatment. Our results indicate that TMAO significantly protects myoblasts from oxidative stress-induced damage by increasing the expression of Nrf2, heme oxygenase-1 (HO-1), NAD(P)H dehydrogenase (NQO1), and catalase (CAT). In particular, suppression of Nrf2 resulted in a loss of the protective effects of TMAO and a significant decrease in the expression levels of Nrf2, HO-1, and NQO1. In addition, we evaluated the effects of TMAO in an exhaustive swimming test in mice. TMAO treatment significantly prolonged swimming endurance, increased glutathione and taurine levels, enhanced glutathione peroxidase activity, and increased the expression of Nrf2 and its downstream antioxidant genes, including HO-1, NQO1, and CAT, in skeletal muscle. These findings underscore the potential of TMAO to counteract exercise-induced oxidative stress. This research provides new insights into the ability of TMAO to alleviate exercise-induced oxidative stress via the Nrf2 signaling pathway, providing a valuable framework for the development of sports nutrition supplements aimed at mitigating oxidative stress.


Asunto(s)
Metilaminas , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Ratones , Animales , Factor 2 Relacionado con NF-E2/metabolismo , Antioxidantes/farmacología , Antioxidantes/metabolismo , Transducción de Señal , Hemo-Oxigenasa 1/metabolismo
6.
Amino Acids ; 55(12): 1867-1878, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37814030

RESUMEN

Hepatic stellate cell (HSC) activation is the key process in hepatic fibrosis (HF) development. Targeted death of HSCs could be effective in the prevention and treatment of HF. Phosphatidylethanolamine-binding protein (PEBP)1 can trigger ferroptosis by mediating peroxide production, but how it modulates HSC ferroptosis is not known. We screened natural small molecules that could bind with PEBP1, and investigated the mechanism by which it promotes HSC ferroptosis. The maximum binding energy of berberine with PEBP1 was - 8.51 kcal/mol, indicating that berberine could bind strongly with PEBP1. Berberine binding to PEBP1 could promote HSC ferroptosis via synergy of its actions with those of sorafenib, but it could not induce ferroptosis alone. Combined administration of berberine enhanced the ferroptotic effects of low-dose sorafenib upon HSCs. Herein, we revealed that PEBP1 might be a target that could enhance the effects of sorafenib, which could provide a new therapeutic approach for HF treatment.


Asunto(s)
Berberina , Ferroptosis , Humanos , Sorafenib/farmacología , Sorafenib/metabolismo , Sorafenib/uso terapéutico , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Berberina/farmacología , Berberina/metabolismo , Berberina/uso terapéutico , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/metabolismo , Proteínas de Unión a Fosfatidiletanolamina/genética , Proteínas de Unión a Fosfatidiletanolamina/metabolismo
7.
Acta Biochim Biophys Sin (Shanghai) ; 55(12): 1913-1924, 2023 12 25.
Artículo en Inglés | MEDLINE | ID: mdl-37705348

RESUMEN

Cancer cachexia (CAC) is a debilitating condition that often arises from noncachexia cancer (NCAC), with distinct metabolic characteristics and medical treatments. However, the metabolic changes and underlying molecular mechanisms during cachexia progression remain poorly understood. Understanding the progression of CAC is crucial for developing diagnostic approaches to distinguish between CAC and NCAC stages, facilitating appropriate treatment for cancer patients. In this study, we establish a mouse model of colon CAC and categorize the mice into three groups: CAC, NCAC and normal control (NOR). By performing nuclear magnetic resonance (NMR)-based metabolomic profiling on mouse sera, we elucidate the metabolic properties of these groups. Our findings unveil significant differences in the metabolic profiles among the CAC, NCAC and NOR groups, highlighting significant impairments in energy metabolism and amino acid metabolism during cachexia progression. Additionally, we observe the elevated serum levels of lysine and acetate during the transition from the NCAC to CAC stages. Using multivariate ROC analysis, we identify lysine and acetate as potential biomarkers for distinguishing between CAC and NCAC stages. These biomarkers hold promise for the diagnosis of CAC from noncachexia cancer. Our study provides novel insights into the metabolic mechanisms underlying cachexia progression and offers valuable avenues for the diagnosis and treatment of CAC in clinical settings.


Asunto(s)
Caquexia , Neoplasias del Colon , Humanos , Animales , Ratones , Caquexia/diagnóstico , Caquexia/etiología , Caquexia/metabolismo , Lisina , Metabolómica , Neoplasias del Colon/complicaciones , Neoplasias del Colon/diagnóstico , Biomarcadores , Acetatos
8.
Int J Mol Sci ; 24(17)2023 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-37686153

RESUMEN

Trehalose, a naturally occurring non-toxic disaccharide, has attracted considerable attention for its potential in alleviating oxidative stress in skeletal muscle. In this study, our aim was to elucidate the metabolic mechanisms underlying the protective effects of trehalose against hydrogen peroxide (H2O2)-induced oxidative stress in C2C12 myoblasts. Our results show that both trehalose treatment and pretreatment effectively alleviate the H2O2-induced decrease in cell viability, reduce intracellular reactive oxygen species (ROS), and attenuate lipid peroxidation. Furthermore, using NMR-based metabolomics analysis, we observed that trehalose treatment and pretreatment modulate the metabolic profile of myoblasts, specifically regulating oxidant metabolism and amino acid metabolism, contributing to their protective effects against oxidative stress. Importantly, our results reveal that trehalose treatment and pretreatment upregulate the expression levels of P62 and Nrf2 proteins, thereby activating the Nrf2-NQO1 axis and effectively reducing oxidative stress. These significant findings highlight the potential of trehalose supplementation as a promising and effective strategy for alleviating oxidative stress in skeletal muscle and provide valuable insights into its potential therapeutic applications.


Asunto(s)
Peróxido de Hidrógeno , Trehalosa , Trehalosa/farmacología , Factor 2 Relacionado con NF-E2 , Metabolómica , Estrés Oxidativo , Mioblastos
9.
Molecules ; 28(9)2023 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-37175250

RESUMEN

Skeletal muscle is closely linked to energy metabolism, but it is inevitably deprived of energy. Cellular differentiation is an essential and energy-demanding process in skeletal muscle development. Much attention has been paid to identifying beneficial factors that promote skeletal muscle satellite cell differentiation and further understanding the underlying regulatory mechanisms. As a critical metabolic substrate or regulator, α-ketoglutarate (AKG) has been recognized as a potential nutritional supplement or therapeutic target for skeletal muscle. We have previously found beneficial effects of AKG supplementation on the proliferation of C2C12 myoblasts cultured under both normal and energy-deficient conditions and have further elucidated the underlying metabolic mechanisms. However, it remains unclear what role AKG plays in myotube formation in different energy states. In the present study, we investigated the effects of AKG supplementation on the differentiation of C2C12 myoblasts cultured in normal medium (Nor myotubes) and low glucose medium (Low myotubes) and performed NMR-based metabonomic profiling to address AKG-induced metabolic changes in both Nor and Low myotubes. Significantly, AKG supplementation promoted myotube formation and induced metabolic remodeling in myotubes under normal medium and low glucose medium, including improved energy metabolism and enhanced antioxidant capacity. Specifically, AKG mainly altered amino acid metabolism and antioxidant metabolism and upregulated glycine levels and antioxidase expression. Our results are typical for the mechanistic understanding of the effects of AKG supplementation on myotube formation in the two energy states. This study may be beneficial for further exploring the applications of AKG supplementation in sports, exercise, and therapy.


Asunto(s)
Antioxidantes , Ácidos Cetoglutáricos , Antioxidantes/metabolismo , Ácidos Cetoglutáricos/farmacología , Ácidos Cetoglutáricos/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Suplementos Dietéticos , Glucosa
10.
Int J Mol Sci ; 23(22)2022 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-36430479

RESUMEN

Lactate is a general compound fuel serving as the fulcrum of metabolism, which is produced from glycolysis and shuttles between different cells, tissues and organs. Lactate is usually accumulated abundantly in muscles during exercise. It remains unclear whether lactate plays an important role in the metabolism of muscle cells. In this research, we assessed the effects of lactate on myoblasts and clarified the underlying metabolic mechanisms through NMR-based metabonomic profiling. Lactate treatment promoted the proliferation and differentiation of myoblasts, as indicated by significantly enhanced expression levels of the proteins related to cellular proliferation and differentiation, including p-AKT, p-ERK, MyoD and myogenin. Moreover, lactate treatment profoundly regulated metabolisms in myoblasts by promoting the intake and intracellular utilization of lactate, activating the TCA cycle, and thereby increasing energy production. For the first time, we found that lactate treatment evidently promotes AMPK signaling as reflected by the elevated expression levels of p-AMPK and p-ACC. Our results showed that lactate as a metabolic regulator activates AMPK, remodeling the cellular metabolic profile, and thereby promoting the proliferation and differentiation of myoblasts. This study elucidates molecular mechanisms underlying the effects of lactate on skeletal muscle in vitro and may be of benefit to the exploration of lactate acting as a metabolic regulator.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Ácido Láctico , Mioblastos , Proliferación Celular , Músculo Esquelético , Metaboloma
11.
Molecules ; 27(18)2022 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-36144851

RESUMEN

Viral myocarditis (VMC), which is defined as inflammation of the myocardium with consequent myocardial injury, may develop chronic disease eventually leading to dilated cardiomyopathy (DCM). Molecular mechanisms underlying the progression from acute VMC (aVMC), to chronic VMC (cVMC) and finally to DCM, are still unclear. Here, we established mouse models of VMC and DCM with Coxsackievirus B3 infection and conducted NMR-based metabolomic analysis of aqueous metabolites extracted from cardiac tissues of three histologically classified groups including aVMC, cVMC and DCM. We showed that these three pathological groups were metabolically distinct from their normal counterparts and identified three impaired metabolic pathways shared by these pathological groups relative to normal controls, including nicotinate and nicotinamide metabolism; alanine, aspartate and glutamate metabolism; and D-glutamine and D-glutamate metabolism. We also identified two extra impaired metabolic pathways in the aVMC group, including glycine, serine and threonine metabolism; and taurine and hypotaurine metabolism Furthermore, we identified potential cardiac biomarkers for metabolically distinguishing these three pathological stages from normal controls. Our results indicate that the metabolomic analysis of cardiac tissues can provide valuable insights into the molecular mechanisms underlying the progression from acute VMC to DCM.


Asunto(s)
Cardiomiopatía Dilatada , Infecciones por Coxsackievirus , Miocarditis , Niacina , Alanina , Animales , Ácido Aspártico , Biomarcadores , Cardiomiopatía Dilatada/metabolismo , Infecciones por Coxsackievirus/metabolismo , Infecciones por Coxsackievirus/patología , Enterovirus Humano B , Ácido Glutámico , Glutamina , Glicina , Ratones , Ratones Endogámicos BALB C , Miocarditis/metabolismo , Miocarditis/patología , Niacinamida , Serina , Taurina , Treonina
12.
Int J Mol Sci ; 22(13)2021 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-34201416

RESUMEN

Hypoxia is associated with clinical diseases. Extreme hypoxia leads to multiple organs failure. However, the different effects of hypoxia on brain and visceral organs still need to be clarified, and moreover, characteristics in vulnerable organs suffering from hypoxia remain elusive. In the present study, we first aimed to figure out the hypoxic sensitivity of organs. Adult male mice were exposed to 6% O2 or 8% O2 for 6 h. Control mice were raised under normoxic conditions. In vivo and in vitro imaging of anti-HIF-1α-NMs-cy5.5 nanocomposites showed that the expression level of hypoxia-inducible factor (HIF-1α) was the highest in the liver, followed by kidney and brain. HIF-1α was detected in the hepatocytes of liver, distal convoluted tubules of kidney and neurons of cerebral cortex. The liver, kidney and brain showed distinct metabolic profiles but an identical change in glutamate. Compared with kidney and brain, the liver had more characteristic metabolites and more disturbed metabolic pathways related to glutaminolysis and glycolysis. The level of O-phosphocholine, GTP, NAD and aspartate were upregulated in hypoxic mice brain, which displayed significant positive correlations with the locomotor activity in control mice, but not in hypoxic mice with impaired locomotor activities. Taken together, the liver, kidney and brain are the three main organs of the body that are strongly respond to acute hypoxia, and the liver exhibited the highest hypoxic sensitivity. The metabolic disorders appear to underlie the physiological function changes.


Asunto(s)
Encéfalo/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Riñón/metabolismo , Hígado/metabolismo , Animales , Conducta Animal , Western Blotting , Carbocianinas/química , Carbocianinas/farmacocinética , Hipoxia/fisiopatología , Espectroscopía de Resonancia Magnética , Masculino , Ratones Endogámicos BALB C , Imagen Molecular , Nanocompuestos/química
13.
Molecules ; 26(7)2021 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-33805924

RESUMEN

α-Ketoglutarate (AKG) is attracting much attention from researchers owing to its beneficial effects on anti-aging and cancer suppression, and, more recently, in nutritional supplements. Given that glucose is the main source of energy to maintain normal physiological functions of skeletal muscle, the effects of AKG supplementation for improving muscle performance are closely related to the glucose level in skeletal muscle. The differences of AKG-induced effects in skeletal muscle between two states of normal energy and energy deficiency are unclear. Furthermore, AKG-induced metabolic changes in skeletal muscles in different energy states also remain elusive. Here, we assessed the effects of AKG supplementation on mouse C2C12 myoblast cells cultured both in normal medium (Nor cells) and in low-glucose medium (Low cells), which were used to mimic two states of normal energy and energy deficiency, respectively. We further performed NMR-based metabolomic analysis to address AKG-induced metabolic changes in Nor and Low cells. AKG supplementation significantly promoted the proliferation and differentiation of cells in the two energy states through glutamine metabolism, oxidative stress, and energy metabolism. Under normal culture conditions, AKG up-regulated the intracellular glutamine level, changed the cellular energy status, and maintained the antioxidant capacity of cells. Under low-glucose culture condition, AKG served as a metabolic substrate to reduce the glutamine-dependence of cells, remarkably enhanced the antioxidant capacity of cells and significantly elevated the intracellular ATP level, thereby ensuring the normal growth and metabolism of cells in the state of energy deficiency. Our results provide a mechanistic understanding of the effects of AKG supplements on myoblasts in both normal energy and energy deficiency states. This work may be beneficial to the exploitation of AKG applications in clinical treatments and nutritional supplementations.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Ácidos Cetoglutáricos/farmacología , Espectroscopía de Resonancia Magnética , Metabolómica , Mioblastos Esqueléticos/metabolismo , Animales , Línea Celular , Ratones
14.
Molecules ; 26(17)2021 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-34500559

RESUMEN

Endothelial dysfunction plays key roles in the pathological process of contrast media (CM)-induced acute kidney injury (CI-AKI) in patients undergoing vascular angiography or intervention treatment. Previously, we have demonstrated that an apolipoprotein A-I (apoA-I) mimetic peptide, D-4F, inhibits oxidative stress and improves endothelial dysfunction caused by CM through the AMPK/PKC pathway. However, it is unclear whether CM induce metabolic impairments in endothelial cells and whether D-4F ameliorates these metabolic impairments. In this work, we evaluated vitalities of human umbilical vein endothelial cells (HUVECs) treated with iodixanol and D-4F and performed nuclear magnetic resonance (NMR)-based metabolomic analysis to assess iodixanol-induced metabolic impairments in HUVECs, and to address the metabolic mechanisms underlying the protective effects of D-4F for ameliorating these metabolic impairments. Our results showed that iodixanol treatment distinctly impaired the vitality of HUVECs, and greatly disordered the metabolic pathways related to energy production and oxidative stress. Iodixanol activated glucose metabolism and the TCA cycle but inhibited choline metabolism and glutathione metabolism. Significantly, D-4F pretreatment could improve the iodixanol-impaired vitality of HUVECs and ameliorate the iodixanol-induced impairments in several metabolic pathways including glycolysis, TCA cycle and choline metabolism in HUVECs. Moreover, D-4F upregulated the glutathione level and hence enhanced antioxidative capacity and increased the levels of tyrosine and nicotinamide adenine dinucleotide in HUVECs. These results provided the mechanistic understanding of CM-induced endothelial impairments and the protective effects of D-4F for improving endothelial cell dysfunction. This work is beneficial to further exploring D-4F as a potential pharmacological agent for preventing CM-induced endothelial impairment and acute kidney injury.


Asunto(s)
Apolipoproteína A-I/metabolismo , Medios de Contraste/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Espectroscopía de Resonancia Magnética/métodos , Metabolómica/métodos , Péptidos/metabolismo , Enfermedades Vasculares/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Células Cultivadas , Humanos , Redes y Vías Metabólicas/fisiología , Estrés Oxidativo/fisiología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología
15.
Cancer Sci ; 111(9): 3195-3209, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32369664

RESUMEN

Gastric cancer (GC) is one of the deadliest cancers worldwide, and the progression of gastric carcinogenesis (GCG) covers multiple complicated pathological stages. Molecular mechanisms of GCG are still unclear. Here, we undertook NMR-based metabolomic analysis of aqueous metabolites extracted from gastric tissues in an established rat model of GCG. We showed that the metabolic profiles were clearly distinguished among 5 histologically classified groups: control, gastritis, low-grade gastric dysplasia, high-grade gastric dysplasia (HGD), and GC. Furthermore, we carried out metabolic pathway analysis based on identified significant metabolites and revealed significantly disturbed metabolic pathways closely associated with the 4 pathological stages, including oxidation stress, choline phosphorylation, amino acid metabolism, Krebs cycle, and glycolysis. Three metabolic pathways were continually disturbed during the progression of GCG, including taurine and hypotaurine metabolism, glutamine and glutamate metabolism, alanine, aspartate, and glutamate metabolism. Both the Krebs cycle and glycine, serine, and threonine metabolism were profoundly impaired in both the HGD and GC stages, potentially due to abnormal energy supply for tumor cell proliferation and growth. Furthermore, valine, leucine, and isoleucine biosynthesis and glycolysis were significantly disturbed in the GC stage for higher energy requirement of the rapid growth of tumor cells. Additionally, we identified potential gastric tissue biomarkers for metabolically discriminating the 4 pathological stages, which also showed good discriminant capabilities for their serum counterparts. This work sheds light on the molecular mechanisms of GCG and is of benefit to the exploration of potential biomarkers for clinically diagnosing and monitoring the progression of GCG.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Espectroscopía de Resonancia Magnética , Metabolómica , Neoplasias Gástricas/etiología , Neoplasias Gástricas/metabolismo , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Metabolismo Energético , Humanos , Espectroscopía de Resonancia Magnética/métodos , Redes y Vías Metabólicas , Metaboloma , Metabolómica/métodos , Ratas , Neoplasias Gástricas/diagnóstico , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Appl Microbiol Biotechnol ; 104(1): 277-289, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31728583

RESUMEN

The most wide-spread "hostile" environmental factor for marine microorganisms is low temperature, which is usually accompanied by high hydrostatic pressure (HHP). Metabolic mechanisms of marine microorganisms adapting to prolonged low temperature under HHP remain to be clarified. To reveal the underlying metabolic mechanisms, we performed NMR-based metabolomic analysis of aqueous extracts derived from a psychrotolerant Microbacterium sediminis YLB-01, which was isolated from deep-sea sediment and possess great biotechnology potentials. The YLB-01 cells were firstly cultivated at the optimal condition (28 °C, 0.1 MPa) for either 18 h (logarithmic phase) or 24 h (stationary phase), then continually cultivated at either 28 °C or 4 °C under HHP (30 MPa) for 7 days. The cells cultivated at low temperature, which experienced cold stress, were distinctly distinguished from those at normal temperature. Cold stress primarily induced metabolic changes in amino acid metabolism and carbohydrate metabolism. Furthermore, the logarithmic and stationary phase cells cultivated at low temperature exhibited distinct metabolic discrimination, which was mostly reflected in the significantly disturbed carbohydrate metabolism. The logarithmic phase cells displayed suppressed TCA cycle, while the stationary phase cells showed decreased pyruvate and increased lactate. In addition, we performed transcriptome analysis for the stationary phase cells to support the metabolomic analysis. Our results suggest that the cold adaptation of the psychrotroph YLB-01 is closely associated with profoundly altered amino acid metabolism and carbohydrate metabolism. Our work provides a mechanistic understanding of the metabolic adaptation of marine psychrotrophs to prolonged low temperature under HHP.


Asunto(s)
Actinobacteria/metabolismo , Adaptación Fisiológica , Frío , Presión Hidrostática , Metabolómica , Actinobacteria/genética , Actinobacteria/crecimiento & desarrollo , Aminoácidos/metabolismo , Organismos Acuáticos/genética , Organismos Acuáticos/metabolismo , Metabolismo de los Hidratos de Carbono , Ciclo del Ácido Cítrico , Respuesta al Choque por Frío , Perfilación de la Expresión Génica , Sedimentos Geológicos/microbiología
17.
J Proteome Res ; 18(4): 1880-1892, 2019 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-30888184

RESUMEN

Cachexia is a complex metabolic derangement syndrome that affects approximately 50-80% of cancer patients. So far, few works have been reported to provide a global overview of gastric cancer cachexia (GCC)-related metabolic changes. We established a GCC murine model by orthotopicly implanting BGC823 cell line and conducted NMR-based metabolomic analysis of gastric tissues, sera, and gastrocnemius. The model with typical cachexia symptoms, confirmed by significant weight loss and muscle atrophy, showed distinctly distinguished metabolic profiles of tumors, sera, and gastrocnemius from sham mice. We identified 20 differential metabolites in tumors, 13 in sera, and 14 in gastrocnemius. Tumor extracts displayed increased pyruvate and lactate, and decreased hypoxanthine, inosine, and inosinate, indicating significantly altered glucose and nucleic acid metabolisms. Cachectic mice exhibited up-regulated serum lactate and glycerol, and down-regulated glucose, which were closely related to hyperlipidemia and hypoglycemia. Furthermore, gastrocnemius transcriptomic and metabolomic data revealed that GCC induced perturbed pathways mainly concentrated on carbohydrate and amino acid metabolism. Specifically, cachectic gastrocnemius exhibited increased α-ketoglutarate and decreased glucose. In vitro study indicated that α-ketoglutarate could prompt myoblasts proliferation and reduce glucose deficiency-induced myotubes atrophy. Overall, this work provides a global metabolic overview to understand the metabolic alterations associated with GCC-induced muscle atrophy.


Asunto(s)
Caquexia/metabolismo , Metaboloma/fisiología , Músculo Esquelético/metabolismo , Neoplasias Gástricas/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Ácidos Cetoglutáricos/metabolismo , Ácidos Cetoglutáricos/farmacología , Masculino , Metabolómica , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Músculo Esquelético/química , Neoplasias Gástricas/química
18.
Cell Physiol Biochem ; 46(4): 1668-1682, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29694977

RESUMEN

BACKGROUND/AIMS: Mitochondrial perturbation is a well-established cause of cognitive decline, but as yet it is unclear how mitochondria-associated neuronal abnormalities in type 1 diabetic (T1DM) brain contribute to cognitive decline. METHODS: The streptozotocin (STZ)-induced mouse model of T1DM was used. The Morris water maze test was applied to assess the effect of T1DM on learning and memory. We detected changes in mitochondrial morphology, function and dynamics. Furthermore, we employed metabolomic analysis to reveal the underlying mechanisms of mitochondrial perturbation which contribute to cognitive decline. RESULTS: Our results show that T1DM impairs mitochondrial dynamics, morphology and function in neurons, associated with a decline in cognitive ability. Metabolomic analyses revealed that T1DM mainly affects metabolic pathways involved in mitochondrial energy failure and impairs the antioxidative system. CONCLUSION: These results lay the basis for understanding the underlying mitochondria-associated causes of T1DM-associated cognitive decline and may provide a potential treatment strategy for this condition in future.


Asunto(s)
Disfunción Cognitiva/etiología , Diabetes Mellitus Experimental/patología , Mitocondrias/metabolismo , Animales , Corteza Cerebelosa/metabolismo , Disfunción Cognitiva/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/complicaciones , Análisis Discriminante , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Análisis de los Mínimos Cuadrados , Espectroscopía de Resonancia Magnética , Masculino , Aprendizaje por Laberinto , Metaboloma , Metabolómica , Análisis de Componente Principal , Ratas , Ratas Sprague-Dawley , Estreptozocina/toxicidad
19.
J Mol Cell Cardiol ; 105: 77-88, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28274624

RESUMEN

Apolipoprotein A-I (apoA-I) mimetic peptide exerts many anti-atherogenic properties. However, the underlying mechanisms related to the endothelial protective effects remain elusive. In this study, the apoA-I mimetic peptide, D-4F, was used. Proliferation assay, wound healing, and transwell migration experiments showed that D-4F improved the impaired endothelial proliferation and migration resulting from ox-LDL. Endothelial adhesion molecules expression and monocyte adhesion assay demonstrated that D-4F inhibited endothelial inflammation. Caspase-3 activation and TUNEL stain indicated that D-4F reduced endothelial cell apoptosis. A pivotal anti-oxidant enzyme, heme oxygenase-1 (HO-1) was upregulated by D-4F. The Akt/AMPK/eNOS pathways were involved in the expression of HO-1 induced by D-4F. Moreover, the anti-oxidation, pro-proliferation, and pro-migration capacities of D-4F were diminished by the inhibitors of both eNOS (L-NAME) and HO-1 (Znpp). Additionally, downregulation of ATP-binding cassette transporter A1 (ABCA1) by siRNA abolished the activation of Akt, AMPK and eNOS, and reduced the upregulation of HO-1 triggered by D-4F. Furthermore, D-4F promoted the reendothelialization of injured intima in carotid artery injury model of C57BL/6J mice in vivo. In summary, these findings suggested that D-4F might be a powerful candidate in the protection of endothelial cells and the prevention of cardiovascular disease (CVD).


Asunto(s)
Apolipoproteína A-I/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Hemo-Oxigenasa 1/metabolismo , Lipoproteínas LDL/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Adhesión Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hemo-Oxigenasa 1/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/metabolismo , Oxidación-Reducción/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Regeneración , Cicatrización de Heridas
20.
J Cell Mol Med ; 21(12): 3810-3820, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28767201

RESUMEN

D-4F, an apolipoprotein A-I (apoA-I) mimetic peptide, possesses distinctly anti-atherogenic effects. However, the biological functions and mechanisms of D-4F on the hyperplasia of vascular smooth muscle cells (VSMCs) remain unclear. This study aimed to determine its roles in the proliferation and migration of VSMCs. In vitro, D-4F inhibited VSMC proliferation and migration induced by ox-LDL in a dose-dependent manner. D-4F up-regulated heme oxygenase-1 (HO-1) expression in VSMCs, and the PI3K/Akt/AMP-activated protein kinase (AMPK) pathway was involved in these processes. HO-1 down-regulation with siRNA or inhibition with zinc protoporphyrin (Znpp) impaired the protective effects of D-4F on the oxidative stress and the proliferation and migration of VSMCs. Moreover, down-regulation of ATP-binding cassette transporter A1 (ABCA1) abolished the activation of Akt and AMPK, the up-regulation of HO-1 and the anti-oxidative effects of D-4F. In vivo, D-4F restrained neointimal formation and oxidative stress of carotid arteries in balloon-injured Sprague Dawley rats. And inhibition of HO-1 with Znpp decreased the inhibitory effects of D-4F on neointimal formation and ROS production in arteries. In conclusion, D-4F inhibited VSMC proliferation and migration in vitro and neointimal formation in vivo through HO-1 up-regulation, which provided a novel prophylactic and therapeutic strategy for anti-restenosis of arteries.


Asunto(s)
Apolipoproteína A-I/farmacología , Aterosclerosis/prevención & control , Hemo-Oxigenasa 1/genética , Músculo Liso Vascular/efectos de los fármacos , Neointima/prevención & control , Sustancias Protectoras/farmacología , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Transportador 1 de Casete de Unión a ATP/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Animales , Aorta Torácica/citología , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación de la Expresión Génica , Hemo-Oxigenasa 1/metabolismo , Lipoproteínas LDL/antagonistas & inhibidores , Lipoproteínas LDL/farmacología , Masculino , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Neointima/genética , Neointima/metabolismo , Neointima/patología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA