Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 159
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 626(7998): 411-418, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38297130

RESUMEN

Ferroptosis, a form of regulated cell death that is driven by iron-dependent phospholipid peroxidation, has been implicated in multiple diseases, including cancer1-3, degenerative disorders4 and organ ischaemia-reperfusion injury (IRI)5,6. Here, using genome-wide CRISPR-Cas9 screening, we identified that the enzymes involved in distal cholesterol biosynthesis have pivotal yet opposing roles in regulating ferroptosis through dictating the level of 7-dehydrocholesterol (7-DHC)-an intermediate metabolite of distal cholesterol biosynthesis that is synthesized by sterol C5-desaturase (SC5D) and metabolized by 7-DHC reductase (DHCR7) for cholesterol synthesis. We found that the pathway components, including MSMO1, CYP51A1, EBP and SC5D, function as potential suppressors of ferroptosis, whereas DHCR7 functions as a pro-ferroptotic gene. Mechanistically, 7-DHC dictates ferroptosis surveillance by using the conjugated diene to exert its anti-phospholipid autoxidation function and shields plasma and mitochondria membranes from phospholipid autoxidation. Importantly, blocking the biosynthesis of endogenous 7-DHC by pharmacological targeting of EBP induces ferroptosis and inhibits tumour growth, whereas increasing the 7-DHC level by inhibiting DHCR7 effectively promotes cancer metastasis and attenuates the progression of kidney IRI, supporting a critical function of this axis in vivo. In conclusion, our data reveal a role of 7-DHC as a natural anti-ferroptotic metabolite and suggest that pharmacological manipulation of 7-DHC levels is a promising therapeutic strategy for cancer and IRI.


Asunto(s)
Deshidrocolesteroles , Ferroptosis , Humanos , Membrana Celular/metabolismo , Colesterol/biosíntesis , Colesterol/metabolismo , Sistemas CRISPR-Cas/genética , Deshidrocolesteroles/metabolismo , Genoma Humano , Enfermedades Renales/metabolismo , Membranas Mitocondriales/metabolismo , Metástasis de la Neoplasia , Neoplasias/metabolismo , Neoplasias/patología , Fosfolípidos/metabolismo , Daño por Reperfusión/metabolismo
2.
Mol Cell ; 81(13): 2736-2751.e8, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33932349

RESUMEN

Cholesterol metabolism is tightly associated with colorectal cancer (CRC). Nevertheless, the clinical benefit of statins, the inhibitor of cholesterol biogenesis mevalonate (MVA) pathway, is inconclusive, possibly because of a lack of patient stratification criteria. Here, we describe that YAP-mediated zinc finger MYND-type containing 8 (ZMYND8) expression sensitizes intestinal tumors to the inhibition of the MVA pathway. We show that the oncogenic activity of YAP relies largely on ZMYND8 to enhance intracellular de novo cholesterol biogenesis. Disruption of the ZMYND8-dependent MVA pathway greatly restricts the self-renewal capacity of Lgr5+ intestinal stem cells (ISCs) and intestinal tumorigenesis. Mechanistically, ZMYND8 and SREBP2 drive the enhancer-promoter interaction to facilitate the recruitment of Mediator complex, thus upregulating MVA pathway genes. Together, our results establish that the epigenetic reader ZMYND8 endows YAP-high intestinal cancer with metabolic vulnerability.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias Colorrectales/metabolismo , Ácido Mevalónico/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Ratones , Ratones Transgénicos , Proteínas Supresoras de Tumor/genética , Proteínas Señalizadoras YAP
3.
EMBO Rep ; 24(4): e56932, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-36862324

RESUMEN

Obesity is associated with metabolic disorders and chronic inflammation. However, the obesity-associated metabolic contribution to inflammatory induction remains elusive. Here, we show that, compared with lean mice, CD4+ T cells from obese mice exhibit elevated basal levels of fatty acid ß-oxidation (FAO), which promote T cell glycolysis and thus hyperactivation, leading to enhanced induction of inflammation. Mechanistically, the FAO rate-limiting enzyme carnitine palmitoyltransferase 1a (Cpt1a) stabilizes the mitochondrial E3 ubiquitin ligase Goliath, which mediates deubiquitination of calcineurin and thus enhances activation of NF-AT signaling, thereby promoting glycolysis and hyperactivation of CD4+ T cells in obesity. We also report the specific GOLIATH inhibitor DC-Gonib32, which blocks this FAO-glycolysis metabolic axis in CD4+ T cells of obese mice and reduces the induction of inflammation. Overall, these findings establish a role of a Goliath-bridged FAO-glycolysis axis in mediating CD4+ T cell hyperactivation and thus inflammation in obese mice.


Asunto(s)
Ácidos Grasos , Inflamación , Animales , Ratones , Ratones Obesos , Ácidos Grasos/metabolismo , Inflamación/metabolismo , Obesidad/metabolismo , Glucólisis , Ubiquitina-Proteína Ligasas/metabolismo , Oxidación-Reducción
4.
Nature ; 569(7757): 581-585, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31043749

RESUMEN

Methylation of cytosine to 5-methylcytosine (5mC) is a prevalent DNA modification found in many organisms. Sequential oxidation of 5mC by ten-eleven translocation (TET) dioxygenases results in a cascade of additional epigenetic marks and promotes demethylation of DNA in mammals1,2. However, the enzymatic activity and function of TET homologues in other eukaryotes remains largely unexplored. Here we show that the green alga Chlamydomonas reinhardtii contains a 5mC-modifying enzyme (CMD1) that is a TET homologue and catalyses the conjugation of a glyceryl moiety to the methyl group of 5mC through a carbon-carbon bond, resulting in two stereoisomeric nucleobase products. The catalytic activity of CMD1 requires Fe(II) and the integrity of its binding motif His-X-Asp, which is conserved in Fe-dependent dioxygenases3. However, unlike previously described TET enzymes, which use 2-oxoglutarate as a co-substrate4, CMD1 uses L-ascorbic acid (vitamin C) as an essential co-substrate. Vitamin C donates the glyceryl moiety to 5mC with concurrent formation of glyoxylic acid and CO2. The vitamin-C-derived DNA modification is present in the genome of wild-type C. reinhardtii but at a substantially lower level in a CMD1 mutant strain. The fitness of CMD1 mutant cells during exposure to high light levels is reduced. LHCSR3, a gene that is critical for the protection of C. reinhardtii from photo-oxidative damage under high light conditions, is hypermethylated and downregulated in CMD1 mutant cells compared to wild-type cells, causing a reduced capacity for photoprotective non-photochemical quenching. Our study thus identifies a eukaryotic DNA base modification that is catalysed by a divergent TET homologue and unexpectedly derived from vitamin C, and describes its role as a potential epigenetic mark that may counteract DNA methylation in the regulation of photosynthesis.


Asunto(s)
5-Metilcitosina/metabolismo , Proteínas Algáceas/metabolismo , Ácido Ascórbico/metabolismo , Biocatálisis , Chlamydomonas reinhardtii/enzimología , ADN/química , ADN/metabolismo , 5-Metilcitosina/química , Dióxido de Carbono/metabolismo , Metilación de ADN , Glioxilatos/metabolismo , Nucleósidos/química , Nucleósidos/metabolismo , Fotosíntesis
5.
Hepatology ; 2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-38051951

RESUMEN

BACKGROUND AND AIMS: Cross talk between tumor cells and immune cells enables tumor cells to escape immune surveillance and dictate responses to immunotherapy. Previous studies have identified that downregulation of the glycolytic enzyme fructose-1,6-bisphosphate aldolase B (ALDOB) in tumor cells orchestrated metabolic programming to favor HCC. However, it remains elusive whether and how ALDOB expression in tumor cells affects the tumor microenvironment in HCC. APPROACH AND RESULTS: We found that ALDOB downregulation was negatively correlated with CD8 + T cell infiltration in human HCC tumor tissues but in a state of exhaustion. Similar observations were made in mice with liver-specific ALDOB knockout or in subcutaneous tumor models with ALDOB knockdown. Moreover, ALDOB deficiency in tumor cells upregulates TGF-ß expression, thereby increasing the number of Treg cells and impairing the activity of CD8 + T cells. Consistently, a combination of low ALDOB and high TGF-ß expression exhibited the worst overall survival for patients with HCC. More importantly, the simultaneous blocking of TGF-ß and programmed cell death (PD) 1 with antibodies additively inhibited tumorigenesis induced by ALDOB deficiency in mice. Further mechanistic experiments demonstrated that ALDOB enters the nucleus and interacts with lysine acetyltransferase 2A, leading to inhibition of H3K9 acetylation and thereby suppressing TGFB1 transcription. Consistently, inhibition of lysine acetyltransferase 2A activity by small molecule inhibitors suppressed TGF-ß and HCC. CONCLUSIONS: Our study has revealed a novel mechanism by which a metabolic enzyme in tumor cells epigenetically modulates TGF-ß signaling, thereby enabling cancer cells to evade immune surveillance and affect their response to immunotherapy.

6.
PLoS Biol ; 18(12): e3000803, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33275593

RESUMEN

Loss of hepatic fructose-1, 6-bisphosphate aldolase B (Aldob) leads to a paradoxical up-regulation of glucose metabolism to favor hepatocellular carcinogenesis (HCC), but the upstream signaling events remain poorly defined. Akt is highly activated in HCC, and targeting Akt is being explored as a potential therapy for HCC. Herein, we demonstrate that Aldob suppresses Akt activity and tumor growth through a protein complex containing Aldob, Akt, and protein phosphatase 2A (PP2A), leading to inhibition of cell viability, cell cycle progression, glucose uptake, and metabolism. Interestingly, Aldob directly interacts with phosphorylated Akt (p-Akt) and promotes the recruitment of PP2A to dephosphorylate p-Akt, and this scaffolding effect of Aldob is independent of its enzymatic activity. Loss of Aldob or disruption of Aldob/Akt interaction in Aldob R304A mutant restores Akt activity and tumor-promoting effects. Consistently, Aldob and p-Akt expression are inversely correlated in human HCC tissues, and Aldob down-regulation coupled with p-Akt up-regulation predicts a poor prognosis for HCC. We have further discovered that Akt inhibition or a specific small-molecule activator of PP2A (SMAP) efficiently attenuates HCC tumorigenesis in xenograft mouse models. Our work reveals a novel nonenzymatic role of Aldob in negative regulation of Akt activation, suggesting that directly inhibiting Akt activity or through reactivating PP2A may be a potential therapeutic approach for HCC treatment.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Fructosa-Bifosfato Aldolasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/fisiopatología , Línea Celular Tumoral , Supervivencia Celular/genética , China , Fructosa-Bifosfato Aldolasa/biosíntesis , Fructosa-Bifosfato Aldolasa/genética , Glucosa/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Ratones Desnudos , Fosforilación , Proteína Fosfatasa 2/metabolismo , Proteína Fosfatasa 2/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Nature ; 548(7665): 112-116, 2017 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-28723898

RESUMEN

The major energy source for most cells is glucose, from which ATP is generated via glycolysis and/or oxidative metabolism. Glucose deprivation activates AMP-activated protein kinase (AMPK), but it is unclear whether this activation occurs solely via changes in AMP or ADP, the classical activators of AMPK. Here, we describe an AMP/ADP-independent mechanism that triggers AMPK activation by sensing the absence of fructose-1,6-bisphosphate (FBP), with AMPK being progressively activated as extracellular glucose and intracellular FBP decrease. When unoccupied by FBP, aldolases promote the formation of a lysosomal complex containing at least v-ATPase, ragulator, axin, liver kinase B1 (LKB1) and AMPK, which has previously been shown to be required for AMPK activation. Knockdown of aldolases activates AMPK even in cells with abundant glucose, whereas the catalysis-defective D34S aldolase mutant, which still binds FBP, blocks AMPK activation. Cell-free reconstitution assays show that addition of FBP disrupts the association of axin and LKB1 with v-ATPase and ragulator. Importantly, in some cell types AMP/ATP and ADP/ATP ratios remain unchanged during acute glucose starvation, and intact AMP-binding sites on AMPK are not required for AMPK activation. These results establish that aldolase, as well as being a glycolytic enzyme, is a sensor of glucose availability that regulates AMPK.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Fructosa-Bifosfato Aldolasa/metabolismo , Fructosadifosfatos/metabolismo , Glucosa/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Adenosina Difosfato/metabolismo , Adenosina Monofosfato/metabolismo , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Proteína Axina/metabolismo , Sitios de Unión , Activación Enzimática , Fibroblastos , Fructosa-Bifosfato Aldolasa/genética , Glucosa/deficiencia , Humanos , Masculino , Ratones , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo
8.
Ann Rheum Dis ; 81(8): 1189-1193, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35277390

RESUMEN

OBJECTIVES: COVID-19 vaccination often triggers a constellation of transitory inflammatory symptoms. Gout is associated with several comorbidities linked to poor outcomes in COVID-19, and gout flares can be triggered by some vaccinations. We analysed the risk of gout flares in the first 3 months after COVID-19 vaccination with inactivated virus, and whether colchicine can prevent gout flares following post-COVID-19 vaccination. METHODS: A clinical delivery population-based cross-sectional study was conducted in the Gout Clinic at the Affiliated Hospital of Qingdao University between February and October 2021. Study participants were selected using a systematic random sampling technique among follow-up patients with gout. We collected data, including vaccinations and potential risk factors, using a combination of interviews, health QR codes and medical records. Logistic regression was used to adjust for covariates. RESULTS: We enrolled 549 gout participants (median age 39 years, 84.2% vaccinated). For the 462 patients who received COVID-19 vaccine, 203 (43.9%) developed at least one gout flare in the 3 months after vaccination. Most of these flares were experienced within 1 month after the first (99/119 (83.2%)) or second (70/115 (60.9%)) dose of vaccine. Compared with unvaccinated participants, COVID-19 vaccination was associated with higher odds of gout flare within 3 months (adjusted OR 6.02; 95% CI 3.00 to 12.08). Colchicine use was associated with 47% less likelihood of postvaccine gout flare. CONCLUSION: COVID-19 vaccination was associated with increased odds of gout flare, which developed mainly in month 1 after each vaccine dose, and was negatively associated with colchicine prophylaxis.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Colchicina , Supresores de la Gota , Gota , Brote de los Síntomas , Adulto , COVID-19/prevención & control , Vacunas contra la COVID-19/efectos adversos , Colchicina/uso terapéutico , Estudios Transversales , Gota/tratamiento farmacológico , Supresores de la Gota/uso terapéutico , Humanos , Vacunación , Vacunas/uso terapéutico
9.
Hepatology ; 74(6): 3037-3055, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34292642

RESUMEN

BACKGROUND AND AIMS: Insulin receptor (IR) transduces cell surface signal through phosphoinositide 3-kinase (PI3K)-AKT pathways or translocates to the nucleus and binds to the promoters to regulate genes associated with insulin actions, including de novo lipogenesis (DNL). Chronic activation of IR signaling drives malignant transformation, but the underlying mechanisms remain poorly defined. Down-regulation of fructose-1,6-bisphosphate aldolase (ALDO) B in hepatocellular carcinoma (HCC) is correlated with poor prognosis. We aim to study whether and how ALDOB is involved in IR signaling in HCC. APPROACH AND RESULTS: Global or liver-specific ALDOB knockout (L-ALDOB-/- ) mice were used in N-diethylnitrosamine (DEN)-induced HCC models, whereas restoration of ALDOB expression was achieved in L-ALDOB-/- mice by adeno-associated virus (AAV). 13 C6 -glucose was employed in metabolic flux analysis to track the de novo fatty acid synthesis from glucose, and nontargeted lipidomics and targeted fatty acid analysis using mass spectrometry were performed. We found that ALDOB physically interacts with IR and attenuates IR signaling through down-regulating PI3K-AKT pathways and suppressing IR nuclear translocation. ALDOB depletion or disruption of IR/ALDOB interaction in ALDOB mutants promotes DNL and tumorigenesis, which is significantly attenuated with ALDOB restoration in L-ALDOB-/- mice. Notably, attenuated IR/ALDOB interaction in ALDOB-R46A mutant exhibits more significant tumorigenesis than releasing ALDOB/AKT interaction in ALDOB-R43A, whereas knockdown IR sufficiently diminishes tumor-promoting effects in both mutants. Furthermore, inhibiting phosphorylated AKT or fatty acid synthase significantly attenuates HCC in L-ALDOB-/- mice. Consistently, ALDOB down-regulation is correlated with up-regulation of IR signaling and DNL in human HCC tumor tissues. CONCLUSIONS: Our study reports a mechanism by which loss of ALDOB activates IR signaling primarily through releasing IR/ALDOB interaction to promote DNL and HCC, highlighting a potential therapeutic strategy in HCC.


Asunto(s)
Carcinogénesis/genética , Fructosa-Bifosfato Aldolasa/metabolismo , Lipogénesis/genética , Neoplasias Hepáticas Experimentales/genética , Receptor de Insulina/metabolismo , Animales , Carcinogénesis/inducido químicamente , Carcinogénesis/patología , Línea Celular Tumoral , Dietilnitrosamina/administración & dosificación , Regulación hacia Abajo , Ácidos Grasos/biosíntesis , Fructosa-Bifosfato Aldolasa/genética , Regulación Neoplásica de la Expresión Génica , Lipidómica , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Masculino , Ratones Noqueados , Fosforilación
10.
Reproduction ; 164(1): 1-8, 2022 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-35521903

RESUMEN

In Brief: Polycystic ovary syndrome (PCOS) is a common cause of anovulatory infertility in women. This study identified changes in free fatty acids profiles in the follicular fluid that may lead to better diagnosis and management of infertility in PCOS women. Abstract: Polycystic ovary syndrome (PCOS) is a heterogeneous disease characterized by various endocrine/metabolic disorders and impaired reproductive potential. Alterations in oocyte competence are considered potentially causative factors for infertility in PCOS women and analyzing the composition of follicular fluid in these patients may help to identify which changes have the potential to alter oocyte quality. In this study, free fatty acid metabolic signatures in follicular fluid were performed to identify changes that may impact oocyte competence in non-obese PCOS women. Sixty-four non-obese women (32 with PCOS and 32 age- and BMI-matched controls) undergoing in vitro fertilization were recruited. Embryo quality was morphologically assessed. Free fatty acid metabolic profiling in follicular fluid was performed using gas/liquid chromatography-mass spectrometry. Principal component analysis and orthogonal partial least squares-discriminant analysis models were further constructed. Nine free fatty acids and 24 eicosanoids were identified and several eicosanoids synthesized by the cyclooxygenase pathway were significantly elevated in PCOS patients compared to controls. The combination of PGE2, PGF2α, PGJ2, and TXB2 had an area under the curve of 0.867 (0.775-0.960) for PCOS discrimination. Furthermore, follicular fluid levels of PGE2 and PGJ2 were negatively correlated with high-quality embryo rate in PCOS patients (P < 0.05). Metabolomic analysis revealed that follicular fluid lipidomic profiles undergo changes in non-obese PCOS women, which suggests that identifying changes in important metabolic signatures may give us a better understanding of the pathogenesis of PCOS. Furthermore, elevated PGE2 and PGJ2 concentrations may contribute to impaired oocyte competence in non-obese PCOS patients.


Asunto(s)
Infertilidad Femenina , Síndrome del Ovario Poliquístico , Dinoprostona/metabolismo , Ácidos Grasos no Esterificados , Femenino , Líquido Folicular/metabolismo , Humanos , Infertilidad Femenina/metabolismo , Oocitos/metabolismo , Síndrome del Ovario Poliquístico/complicaciones , Síndrome del Ovario Poliquístico/metabolismo
11.
J Nutr ; 152(4): 1118-1129, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35020917

RESUMEN

BACKGROUND: Few studies have assessed the integrative effects of diet, BMI, and exercise on postprandial changes in energy and circulating metabolic profiles. OBJECTIVES: We aimed to assess the collective effects of 3 isocaloric meals high in carbohydrate (74.2% energy), fat (64.6% energy), or protein (39.5% energy) on energy expenditure and clinical and metabolomic biomarkers under resting and exercise conditions in normal-weight and overweight/obese men. METHODS: This crossover controlled acute trial included 20 normal-weight (BMI, 18.5 to <24 kg/m2) and 20 overweight/obese (BMI ≥24 kg/m2) men aged 18-45 years. Each of 3 test meals was provided for 2 continuous days: a resting day without exercise, followed by an exercise day with a bicycling exercise of 50% maximal oxygen consumption (postprandial 90-120 minutes). Energy expenditure (exploratory outcome of primary interest) was measured using indirect calorimetry. Fasting and postprandial 2-hour serum clinical and metabolomic biomarkers (secondary interest) were measured. Mixed models were used to examine the effects of meal, time, and/or BMI category. RESULTS: On the resting day, no significant between-meal differences were detected for energy expenditure. However, high-carbohydrate and high-fat meals induced the highest postprandial 2-hour increase in glucose (0.34 ± 0.15 mmol/L) and triglyceride (0.95 ± 0.09 mmol/L), respectively, while the high-protein meal reduced glucose (-0.48 ± 0.08 mmol/L) and total cholesterol (-0.01 ± 0.03 mmol/L; all Pmeal values < 0.001). On the exercise day, a high-carbohydrate meal significantly promoted the carbohydrate oxidation rate but suppressed the fat oxidation rate (Pmeal < 0.05), while its postprandial glucose response was attenuated by bicycling (-0.31 ± 0.03 mmol/L; Pexercise < 0.001). We identified 69 metabolites as key features in discriminating between the 3 meals, and overweight/obese men had more varieties of metabolites than normal-weight men. CONCLUSIONS: Three isocaloric meals induced unique postprandial changes in clinical and metabolomic biomarkers, while exercise prevented the hyperglycemia induced by a high-carbohydrate meal. Overweight/obese men were more responsive to the meal challenges than normal-weight men. This trial was registered at clinicaltrials.gov as NCT03231618.


Asunto(s)
Adiposidad , Sobrepeso , Adolescente , Adulto , Biomarcadores , Glucemia/metabolismo , Estudios Cruzados , Metabolismo Energético , Humanos , Insulina , Masculino , Comidas , Persona de Mediana Edad , Obesidad/metabolismo , Sobrepeso/metabolismo , Periodo Posprandial/fisiología , Adulto Joven
12.
J Nutr ; 152(4): 1118-1129, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-36967169

RESUMEN

BACKGROUND: Few studies have assessed the integrative effects of diet, BMI, and exercise on postprandial changes in energy and circulating metabolic profiles. OBJECTIVES: We aimed to assess the collective effects of 3 isocaloric meals high in carbohydrate (74.2% energy), fat (64.6% energy), or protein (39.5% energy) on energy expenditure and clinical and metabolomic biomarkers under resting and exercise conditions in normal-weight and overweight/obese men. METHODS: This crossover controlled acute trial included 20 normal-weight (BMI, 18.5 to <24 kg/m2) and 20 overweight/obese (BMI ≥24 kg/m2) men aged 18-45 years. Each of 3 test meals was provided for 2 continuous days: a resting day without exercise, followed by an exercise day with a bicycling exercise of 50% maximal oxygen consumption (postprandial 90-120 minutes). Energy expenditure (exploratory outcome of primary interest) was measured using indirect calorimetry. Fasting and postprandial 2-hour serum clinical and metabolomic biomarkers (secondary interest) were measured. Mixed models were used to examine the effects of meal, time, and/or BMI category. RESULTS: On the resting day, no significant between-meal differences were detected for energy expenditure. However, high-carbohydrate and high-fat meals induced the highest postprandial 2-hour increase in glucose (0.34 ± 0.15 mmol/L) and triglyceride (0.95 ± 0.09 mmol/L), respectively, while the high-protein meal reduced glucose (-0.48 ± 0.08 mmol/L) and total cholesterol (-0.01 ± 0.03 mmol/L; all Pmeal values < 0.001). On the exercise day, a high-carbohydrate meal significantly promoted the carbohydrate oxidation rate but suppressed the fat oxidation rate (Pmeal < 0.05), while its postprandial glucose response was attenuated by bicycling (-0.31 ± 0.03 mmol/L; Pexercise < 0.001). We identified 69 metabolites as key features in discriminating between the 3 meals, and overweight/obese men had more varieties of metabolites than normal-weight men. CONCLUSIONS: Three isocaloric meals induced unique postprandial changes in clinical and metabolomic biomarkers, while exercise prevented the hyperglycemia induced by a high-carbohydrate meal. Overweight/obese men were more responsive to the meal challenges than normal-weight men. This trial was registered at clinicaltrials.gov as NCT03231618.


Asunto(s)
Adiposidad , Sobrepeso , Masculino , Humanos , Sobrepeso/metabolismo , Obesidad/metabolismo , Metabolismo Energético , Glucosa , Proteínas/metabolismo , Biomarcadores , Comidas , Periodo Posprandial/fisiología , Estudios Cruzados , Glucemia/metabolismo
13.
PLoS Biol ; 17(8): e3000420, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31433805

RESUMEN

Dendritic cells (DCs) play pivotal roles in T-cell homeostasis and activation, and metabolic programing has been recently linked to DC development and function. However, the metabolic underpinnings corresponding to distinct DC functions remain largely unresolved. Here, we demonstrate a special metabolic-epigenetic coupling mechanism orchestrated by tuberous sclerosis complex subunit 1 (TSC1)-mechanistic target of rapamycin (mTOR) for homeostatic DC function. Specific ablation of Tsc1 in the DC compartment (Tsc1DC-KO) largely preserved DC development but led to pronounced reduction in naïve and memory-phenotype cluster of differentiation (CD)8+ T cells, a defect fully rescued by concomitant ablation of mTor or regulatory associated protein of MTOR, complex 1 (Rptor) in DCs. Moreover, Tsc1DC-KO mice were unable to launch efficient antigen-specific CD8+ T effector responses required for containing Listeria monocytogenes and B16 melanomas. Mechanistically, our data suggest that the steady-state DCs tend to tune down de novo fatty acid synthesis and divert acetyl-coenzyme A (acetyl-CoA) for histone acetylation, a process critically controlled by TSC1-mTOR. Correspondingly, TSC1 deficiency elevated acetyl-CoA carboxylase 1 (ACC1) expression and fatty acid synthesis, leading to impaired epigenetic imprinting on selective genes such as major histocompatibility complex (MHC)-I and interleukin (IL)-7. Remarkably, tempering ACC1 activity was able to divert cytosolic acetyl-CoA for histone acetylation and restore the gene expression program compromised by TSC1 deficiency. Taken together, our results uncover a crucial role for TSC1-mTOR in metabolic programing of the homeostatic DCs for T-cell homeostasis and implicate metabolic-coupled epigenetic imprinting as a paradigm for DC specification.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteína 1 del Complejo de la Esclerosis Tuberosa/metabolismo , Animales , Antígenos/metabolismo , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/fisiología , Células Dendríticas/inmunología , Epigénesis Genética , Homeostasis , Listeria monocytogenes , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/inmunología , Proteína 1 del Complejo de la Esclerosis Tuberosa/genética , Proteína 1 del Complejo de la Esclerosis Tuberosa/inmunología , Proteínas Supresoras de Tumor/genética
14.
Eur J Nutr ; 61(6): 3235-3246, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35445833

RESUMEN

PURPOSE: There is limited and inconsistent evidence about the relationships of erythrocyte polyunsaturated fatty acids (PUFAs) with stroke and stroke types, particularly in China where the stroke rates are high. We aimed to investigate the associations of different erythrocyte PUFAs with incidence of total stroke, ischemic stroke (IS), and intracerebral hemorrhage (ICH) in Chinese adults. METHODS: In the prospective China Kadoorie Biobank, erythrocyte PUFAs were measured using gas chromatography in 10,563 participants who attended 2013-14 resurvey. After a mean follow-up of 3.8 years, 412 incident stroke cases (342 IS, 53 ICH) were recorded among 8,159 participants without prior vascular diseases or diabetes. Cox regression yielded adjusted hazard ratios (HRs) for stroke associated with 13 PUFAs. RESULTS: Overall, the mean body mass index was 24.0 (3.4) kg/m2 and the mean age was 58.1 (9.9) years. In multivariable analyses, 18:2n-6 was positively associated with ICH (HR = 2.33 [95% CIs 1.41, 3.82] for top versus bottom quintile, Ptrend = 0.007), but inversely associated with IS (0.69 [0.53,0.90], Ptrend = 0.027), while 20:3n-6 was positively associated with risk of IS (1.64 [1.32,2.04], Ptrend < 0.001), but not with ICH. Inverted-U shape curve associations were observed of 20:5n-3 with IS (Pnonlinear = 0.002) and total stroke (Pnonlinear = 0.008), with a threshold at 0.70%. After further adjustment for conventional CVD risk factors and dietary factors, these associations remained similar. CONCLUSION: Among relatively lean Chinese adults, erythrocyte PUFAs 18:2n-6, 20:3n-6 and 20:5n-3 showed different associations with risks of IS and ICH. These results would improve the understanding of stroke etiology.


Asunto(s)
Ácidos Grasos Insaturados , Accidente Cerebrovascular , Adulto , China/epidemiología , Eritrocitos , Humanos , Incidencia , Persona de Mediana Edad , Estudios Prospectivos , Factores de Riesgo , Accidente Cerebrovascular/epidemiología
15.
J Neurosci ; 39(10): 1930-1943, 2019 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-30626699

RESUMEN

Mitochondrial energy production is essential for normal brain function. Traumatic brain injury (TBI) increases brain energy demands, results in the activation of mitochondrial respiration, associated with enhanced generation of reactive oxygen species. This chain of events triggers neuronal apoptosis via oxidation of a mitochondria-specific phospholipid, cardiolipin (CL). One pathway through which cells can avoid apoptosis is via elimination of damaged mitochondria by mitophagy. Previously, we showed that externalization of CL to the mitochondrial surface acts as an elimination signal in cells. Whether CL-mediated mitophagy occurs in vivo or its significance in the disease processes are not known. In this study, we showed that TBI leads to increased mitophagy in the human brain, which was also detected using TBI models in male rats. Knockdown of CL synthase, responsible for de novo synthesis of CL, or phospholipid scramblase-3, responsible for CL translocation to the outer mitochondrial membrane, significantly decreased TBI-induced mitophagy. Inhibition of mitochondrial clearance by 3-methyladenine, mdivi-1, or phospholipid scramblase-3 knockdown after TBI led to a worse outcome, suggesting that mitophagy is beneficial. Together, our findings indicate that TBI-induced mitophagy is an endogenous neuroprotective process that is directed by CL, which marks damaged mitochondria for elimination, thereby limiting neuronal death and behavioral deficits.SIGNIFICANCE STATEMENT Traumatic brain injury (TBI) increases energy demands leading to activation of mitochondrial respiration associated with enhanced generation of reactive oxygen species and resultant damage to mitochondria. We demonstrate that the complete elimination of irreparably damaged organelles via mitophagy is activated as an early response to TBI. This response includes translocation of mitochondria phospholipid cardiolipin from the inner membrane to the outer membrane where externalized cardiolipin mediates targeted protein light chain 3-mediated autophagy of damaged mitochondria. Our data on targeting phospholipid scramblase and cardiolipin synthase in genetically manipulated cells and animals strongly support the essential role of cardiolipin externalization mechanisms in the endogenous reparative plasticity of injured brain cells. Furthermore, successful execution and completion of mitophagy is beneficial in the context of preservation of cognitive functions after TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Encéfalo/metabolismo , Cardiolipinas/metabolismo , Mitofagia/fisiología , Neuronas/metabolismo , Animales , Apoptosis/fisiología , Encéfalo/ultraestructura , Lesiones Traumáticas del Encéfalo/patología , Humanos , Masculino , Membranas Mitocondriales/metabolismo , Neuronas/ultraestructura , Ratas Sprague-Dawley , Transducción de Señal
16.
J Lipid Res ; 61(4): 560-569, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32029512

RESUMEN

This article focuses on the establishment of an accurate and sensitive quantitation method for the analysis of furan fatty acids. In particular, the sensitivity of GC/MS and UPLC/ESI/MS/MS was compared for the identification and quantification of furan fatty acids. Different methylation methods were tested with respect to GC/MS analysis. Special attention needs to be paid to the methylation of furan fatty acids, as acidic catalysts might lead to the degradation of the furan ring. GC/MS analysis in full-scan mode demonstrated that the limit of quantitation was 10 µM. UPLC/ESI/MS/MS in multiple reaction monitoring mode displayed a higher detection sensitivity than GC/MS. Moreover, the identification of furan fatty acids with charge-reversal derivatization was tested in the positive mode with two widely used pyridinium salts. Significant oxidation was unexpectedly observed using N-(4-aminomethylphenyl) pyridinium as a derivatization agent. The formed 3-acyl-oxymethyl-1-methylpyridinium iodide derivatized by 2-bromo-1-methylpyridinium iodide and 3-carbinol-1-methylpyridinium iodide improved the sensitivity more than 2,000-fold compared with nonderivatization in the negative mode by UPLC/ESI/MS/MS. This charge-reversal derivatization enabled the targeted quantitation of furan fatty acids in human plasma. Thus, it is anticipated that this protocol could greatly contribute to the clarification of pathological mechanisms related to furan fatty acids and their metabolites.


Asunto(s)
Análisis Químico de la Sangre/métodos , Ácidos Grasos/sangre , Ácidos Grasos/química , Furanos/química , Límite de Detección , Ayuno/sangre , Voluntarios Sanos , Humanos
17.
Reproduction ; 159(2): 159-169, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31770100

RESUMEN

Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in reproductive-age women usually accompanied by lipid metabolic disorders. However, it remains unknown whether arachidonic acid (AA) and its metabolites in follicular fluid (FF) were altered in PCOS patients. This study was intended to measure the levels of AA and its metabolites in the FF of non-obese PCOS patients that underwent in vitro fertilization (IVF) and to explore the possible causes of the alterations. Thirty-nine non-obese women with PCOS and 30 non-obese women without PCOS were enrolled. AA and its metabolites were measured by liquid chromatography-mass spectrometry. The levels of AA metabolites generated via cyclooxygenase-2 (COX-2) pathway and cytochrome P450 epoxygenase pathway but not lipoxygenase (LOX) pathway were significantly higher in the FF of PCOS patients. The metabolites generated via COX-2 pathway were significantly correlated with levels of testosterone and fasting insulin in serum. The in vitro study further demonstrated that insulin but not testosterone could promote the IL-1ß and hCG-induced COX-2 expression and prostaglandin E2 (PGE2) secretion in primary human granulosa cells. In conclusion, there was an elevation in AA metabolites in FF of PCOS patients. Insulin played a pivotal role in the increased AA metabolites generated via COX-2, which could be interpreted as another novel molecular pathophysiological mechanism of PCOS.

18.
Gynecol Endocrinol ; 36(6): 508-512, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31793360

RESUMEN

This study aimed to identify potential lipid biomarkers in women with polycystic ovary syndrome (PCOS) and determine their predictive value for PCOS. Eighteen women with PCOS and 17 healthy controls were enrolled. A multi-dimensional mass spectrometry-based shotgun lipidomics approach was employed to analyze serum lipid profiles. Shotgun lipidomics revealed that the concentrations of ceramide (Cer) and phosphatidylcholine (PC) were higher (PC: 831.6 ± 217.4 vs. 605.2 ± 164.2 µmol/l; Cer: 3,387.6 ± 829.9 vs. 2,552.2 ± 679.4 nmol/l, respectively), whereas that of lysophosphatidylcholine was lower, in PCOS women than in healthy controls (82.02 ± 39.49 vs. 133.62 ± 65.36 µmol/l, respectively). Receiver operating characteristic analysis showed that the combination of Cer (OH_N16:0/N18:0) and Cer (N22:0) had the greatest discriminatory power to differentiate between women with and without PCOS (area under the curve: 0.889, 95% confidence interval: 0.784-0.994). These results indicate that the combination of Cer (OH_N16:0/N18:0) and Cer (N22:0) may represent a novel lipid predictor of PCOS.


Asunto(s)
Biomarcadores/sangre , Ceramidas/sangre , Lipidómica/métodos , Síndrome del Ovario Poliquístico/sangre , Adolescente , Adulto , Estudios de Casos y Controles , Ceramidas/clasificación , China , Estudios Transversales , Femenino , Humanos , Lípidos/sangre , Proyectos Piloto , Síndrome del Ovario Poliquístico/diagnóstico , Valor Predictivo de las Pruebas , Regulación hacia Arriba , Adulto Joven
20.
FASEB J ; 32(10): 5413-5425, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29723062

RESUMEN

Mechanical insults, such as stent implantation, can induce endothelial injury, vascular inflammation, and ultimately lead to vascular neointimal hyperplasia. Resolvin E1 (RvE1), derived from the ω3 fatty acid eicosapentaenoic acid, can facilitate the resolution of inflammation in many settings. We therefore aimed to determine if there was a role for RvE1 in preventing neointimal formation after arterial injury and to understand the underlying mechanisms. Vascular inflammation and neointimal hyperplasia were induced by wire injury in the femoral arteries of mice. Administration of exogenous RvE1 and endogenously generated RvE1 via dietary supplementation with eicosapentaenoic acid and aspirin markedly reduced vascular neointima formation in this model. Mechanistically, RvE1 was found to inhibit vascular neutrophil infiltration, promote macrophage polarization toward an M2-like phenotype, suppress T-cell trafficking by reducing RANTES secretion from vascular smooth muscle cells, and inhibit vascular smooth muscle cell migration. In summary, RvE1 demonstrated a protective role against vascular inflammation and remodeling in response to mechanical injury, suggesting that it may serve as an adjuvant therapeutic agent for percutaneous coronary interventions, such as stent implantation.-Liu, G., Gong, Y., Zhang, R., Piao, L., Li, X., Liu, Q., Yan, S., Shen, Y., Guo, S., Zhu, M., Yin, H., Funk, C. D., Zhang, J., Yu, Y. Resolvin E1 attenuates injury-induced vascular neointimal formation by inhibition of inflammatory responses and vascular smooth muscle cell migration.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Ácido Eicosapentaenoico/análogos & derivados , Arteria Femoral , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Neointima/prevención & control , Animales , Modelos Animales de Enfermedad , Ácido Eicosapentaenoico/farmacología , Arteria Femoral/lesiones , Arteria Femoral/metabolismo , Arteria Femoral/patología , Inflamación/metabolismo , Inflamación/patología , Inflamación/prevención & control , Masculino , Ratones , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Neointima/metabolismo , Neointima/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA