Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(14)2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-39062992

RESUMEN

[123I]ß-methyl-p-iodophenyl-pentadecanoic acid ([123I]BMIPP), which is used for nuclear medicine imaging of myocardial fatty acid metabolism, accumulates in cancer cells. However, the mechanism of accumulation remains unknown. Therefore, this study aimed to elucidate the accumulation and accumulation mechanism of [123I]BMIPP in cancer cells. We compared the accumulation of [123I]BMIPP in cancer cells with that of [18F]FDG and found that [123I]BMIPP was a much higher accumulation than [18F]FDG. The accumulation of [123I]BMIPP was evaluated in the presence of sulfosuccinimidyl oleate (SSO), a CD36 inhibitor, and lipofermata, a fatty acid transport protein (FATP) inhibitor, under low-temperature conditions and in the presence of etomoxir, a carnitine palmitoyl transferase I (CPT1) inhibitor. The results showed that [123I]BMIPP accumulation was decreased in the presence of SSO and lipofermata in H441, LS180, and DLD-1 cells, suggesting that FATPs and CD36 are involved in [123I]BMIPP uptake in cancer cells. [123I]BMIPP accumulation in all cancer cell lines was significantly decreased at 4 °C compared to that at 37 °C and increased in the presence of etomoxir in all cancer cell lines, suggesting that the accumulation of [123I]BMIPP in cancer cells is metabolically dependent. In a biological distribution study conducted using tumor-bearing mice transplanted with LS180 cells, [123I]BMIPP highly accumulated in not only LS180 cells but also normal tissues and organs (including blood and muscle). The tumor-to-intestine or large intestine ratios of [123I]BMIPP were similar to those of [18F]FDG, and the tumor-to-large-intestine ratios exceeded 1.0 during 30 min after [123I]BMIPP administration in the in vivo study. [123I]BMIPP is taken up by cancer cells via CD36 and FATP and incorporated into mitochondria via CPT1. Therefore, [123I]BMIPP may be useful for imaging cancers with activated fatty acid metabolism, such as colon cancer. However, the development of novel imaging radiotracers based on the chemical structure analog of [123I]BMIPP is needed.


Asunto(s)
Neoplasias del Colon , Yodobencenos , Animales , Humanos , Neoplasias del Colon/diagnóstico por imagen , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Ratones , Línea Celular Tumoral , Yodobencenos/química , Antígenos CD36/metabolismo , Radiofármacos/química , Radiofármacos/metabolismo , Radioisótopos de Yodo , Ácidos Oléicos/química , Miocardio/metabolismo , Distribución Tisular , Proteínas de Transporte de Ácidos Grasos/metabolismo , Fluorodesoxiglucosa F18/química , Fluorodesoxiglucosa F18/metabolismo , Ácidos Grasos
2.
Placenta ; 139: 181-189, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37421872

RESUMEN

INTRODUCTION: Expression of nutrient transporters in the placenta affects fetal growth. This study reports the protein expression of nutrient transporters in the syncytial membranes [microvillous membrane (MVM) and basal membrane (BM)] of normotensive control and preeclampsia placentae. METHODS: Placentae were collected from fourteen normotensive control women and fourteen women with preeclampsia. The syncytiotrophoblast MVM and BM membranes were isolated. The protein expression of glucose transporter (GLUT1), vitamin B12 transporter (CD320) and fatty acid transporters (FATP2, FATP4) was assessed in both the membranes. RESULTS: Comparison between membranes demonstrates similar CD320 protein expression in normotensive group whereas, in preeclampsia placentae it was higher in the BM as compared to MVM (p < 0.05). FATP2&4 protein expression was higher in the BM as compared to their respective MVM fraction in both the groups (p < 0.01 for both). Comparison between groups demonstrates higher GLUT1 expression in the MVM (p < 0.05) and BM (p < 0.05) whereas lower CD320 expression in the MVM (p < 0.05) of preeclampsia placentae as compared to their respective membranes in normotensive control. Furthermore, GLUT1 protein expression was positively associated and CD320 protein expression was negatively associated with maternal body mass index (BMI) (p < 0.05 for both). No difference was observed in the FATP2&4 protein expression. However, FATP4 protein expression was negatively associated with maternal blood pressure (p < 0.05 for MVM; p = 0.060 for BM) and birth weight (p < 0.05 for both membranes). DISCUSSION: The current study for the first time demonstrates differential expression of various transporters in the syncytiotrophoblast membranes of the preeclampsia placentae which may influence fetal growth.


Asunto(s)
Preeclampsia , Trofoblastos , Embarazo , Femenino , Humanos , Trofoblastos/metabolismo , Preeclampsia/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Placenta/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Nutrientes
3.
J Anim Sci ; 1012023 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-37086180

RESUMEN

Metabolic syndrome is a worldwide health issue. Previous research has revealed that low-birth weight (LBW) swine fed a high-fat (HF) diet were susceptible to insulin resistance (IR) and developed a preferential intestinal lipid absorption, hypertriglyceridemia, and muscle steatosis. We hypothesized that fatty acid transporters such as CD36, FATP4, and FABP2 could potentially explain the development of these conditions. In addition, dairy-derived fatty acids have been shown to be valid biomarkers to assess dairy intake, which can be utilized to investigate muscle lipid deposition in LBW swine. The overall aim of this study was to delineate molecular transport candidates responsible for intestinal lipid absorption and muscle lipid deposition in LBW swine; and secondly to determine what dietary fatty acids might accumulate preferentially in pork muscle when consuming dairy products. At 5 weeks of age, normal birth weight (NBW) and LBW piglets were randomly assigned to three experimental diets: 1-chow diet, 2-HF diet, or 3-isocaloric HF diet supplemented with full fat dairy products. At 12 weeks of age, piglets were euthanized, and carcass, fasting plasma, biceps femoris and jejunum mucosal scrapings were collected. Results showed that HF-fed LBW swine exhibited early signs of IR (fasting glucose, P < 0.05; fasting insulin, P = 0.091; HOMA-IR, P = 0.086) compared with NBW-Chow, which were attenuated with increased dairy intake. Muscle samples from HF-fed LBW swine contained significantly more triglyceride compared to Chow-fed NBW swine (P < 0.05). Increased dairy intake significantly increased myristic acid (C14:0) and DPA (C22:5n3) relative to HF feeding alone (P < 0.05). All HF-fed LBW swine (regardless of dairy intake) exhibited an upregulation of CD36 expression (but not FABP2) compared with NBW littermates in both the small intestine and muscle (P < 0.05). Interestingly, increased dairy intake significantly increased the Canadian Lean Yield percentage in LBW swine fed an HF diet (P < 0.05). Findings from this study provide evidence on the mechanistic pathway of intestinal and muscle lipid metabolism in an innovative LBW swine model. We have also revealed that increasing dairy intake can enhance the incorporation of dietary long-chain polyunsaturated fatty acids into pork, as well as increasing the predicted lean yield of the carcass.


Metabolic syndrome affects millions of people worldwide, and large animal models represent a unique opportunity for research advancement. Intensive swine production can induce low-birth weight (LBW) litters. We have developed an innovative LBW swine model to investigate insulin resistance and dyslipidemia. We present evidence to explain how LBW swine can upregulate lipid intestinal absorption as well as preferentially increase pork marbling. We have also identified a potential added value approach to increase healthy fatty acids in pork and/or increase the carcass lean yield in LBW swine.


Asunto(s)
Resistencia a la Insulina , Enfermedades de los Porcinos , Porcinos , Animales , Peso al Nacer/fisiología , Ácidos Grasos/metabolismo , Regulación hacia Arriba , Canadá , Músculos/metabolismo , Dieta Alta en Grasa , Resistencia a la Insulina/fisiología , Enfermedades de los Porcinos/metabolismo
4.
Phytomedicine ; 111: 154661, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36682299

RESUMEN

BACKGROUND: More than 70% of patients with type 2 diabetes (T2DM) concomitantly suffer from Non-alcoholic fatty liver disease (NAFLD), and the coexistence and interaction of them increases the intractability of NAFLD. With the protective effect against hepatic steatosis and liver fibrosis, SIRT6 is becoming a notable target of NAFLD. Diosgenin, an active monomer from Chinese herbs, has been reported to protect against NAFLD. PURPOSE: This study aims to figure out the mechanism how diosgenin alleviate NAFLD in T2DM and the relationship with SIRT6. METHODS: In vivo studies used spontaneous diabetic db/db mice and divided them into two parts. The first part included four groups consisting of control (Con) group, model (Mod) group, low dose of diosgenin (DL) group and high dose of diosgenin (DH) group. The second part included four groups consisting of Con group, Mod group, DH+OSS (OSS_128167, inhibitor of SIRT6) group, MDL (MDL800, agonist of SIRT6) group. HepG2 cell line was selected in study in vitro, which was mainly composed of six groups including Con group, palmitic acid (PA) group, PA+DL group, PA+DH group, PA+DH+OSS group, PA+MDL group. OGTT, Biochemical biomarker (including TG, TC, AST, ALT), inflammatory biomarker (including IL-6 and TNF-α) were measured. HE, Oil Red O, and DHE staining were conducted. Immunohistochemistry, immunofluorescence, mRNA-seq, and qPCR were used to explore the mechanism. RESULTS: Results in the first part of study in vivo indicated that diosgenin protected against lipid accumulation, oxidative stress, cell injury, and light inflammatory of liver in db/db mice and regulated the expression of SIRT6 and fatty acid transporter including CD36, FATP2, FABP1. The effect of diosgenin could be reversed in DH+OSS group and the same effect was observed in MDL group in the second part of study in vivo. The same results were also noted in followed study in vitro. Diosgenin inhibited the fatty acids uptake and regulated the expression of SIRT6 and fatty acid transporter including CD36, FATP2, and FABP1 in PA-induced hepG2 cells, and which was reversed in DH+OSS group and resembled in MDL group. CONCLUSIONS: Diosgenin could attenuate non-alcoholic fatty liver disease in type 2 diabetes through regulating SIRT6-related fatty acid uptake.


Asunto(s)
Diabetes Mellitus Tipo 2 , Diosgenina , Enfermedad del Hígado Graso no Alcohólico , Sirtuinas , Ratones , Animales , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Ácidos Grasos/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Diosgenina/farmacología , Diosgenina/metabolismo , Metabolismo de los Lípidos , Hígado , Sirtuinas/metabolismo
5.
Appl Environ Microbiol ; 88(20): e0129422, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36169310

RESUMEN

Due to the barrier effect of lipopolysaccharide (LPS) in the outer membrane of Gram-negative bacteria, transporters are required for hydrophobic alkane uptake. However, there are few reports on long-chain alkane transporters. In this study, a potential long-chain alkane transporter (AltL) was screened in Acinetobacter venetianus RAG-1 by comparative transcriptome analysis. Growth and degradation experiments showed that altL deletion led to the loss of n-octacosane utilization capacity of RAG-1. To identify the function of AltL, we measured the existence and accumulation of alkanes in cells through the constructed alkane detection system and isotope transport experiment, which proved its long-chain alkane transport function. Growth experiments using different chain-length n-alkanes and fatty acids as substrates showed that AltL was responsible for the transport of (very) long-chain n-alkanes (C20 to C38) and fatty acids (C18A to C28A) and was also involved in the uptake of medium-chain n-alkanes (C16 to C18). Subsequently, we analyzed the distribution of AltL in bacteria, and found that AltL homologs are widespread in Gamma-, Beta-, and Deltaproteobacteria. An AltL homolog in Pseudomonas aeruginosa was also identified to participate in long-chain alkane transport by a gene deletion and growth assay. We also found that overexpression of altL in Pseudomonas aeruginosa enhanced the degradation of C16 to C32 n-alkanes. In addition, structure analysis showed that AltL has longer extracellular loops than other FadL family members, which may be involved in the binding of alkanes. These results showed that AltL is a novel transporter and that it is mainly responsible for the transport of long-chain n-alkanes and (very) long-chain fatty acids and has broad application potential. IMPORTANCE Petroleum pollution has caused great harm to the natural environment, and alkanes are the main components of petroleum. Many Gram-negative bacteria can use alkanes as carbon and energy sources, which is an important strategy for oil pollution remediation. Alkane uptake is the first step for its utilization. Hence, the characterization of transport proteins is of great significance for the recovery of oil pollution and other potential applications in industrial engineering bacteria. At present, some short- and medium-chain alkane transporters have been identified, but stronger hydrophobic long-chain alkane transporters have received little attention. In this study, the broad-spectrum transporter AltL, identified in RAG-1, makes up for the lack of research on the transport of long-chain alkanes and (very) long-chain fatty acids. Meanwhile, the structural features of longer extracellular loops might be related to its unique transport function on more hydrophobic and larger substrates, indicating it is a novel type alkane transporter.


Asunto(s)
Lipopolisacáridos , Petróleo , Lipopolisacáridos/metabolismo , Ácidos Grasos/metabolismo , Biodegradación Ambiental , Alcanos/metabolismo , Petróleo/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Pseudomonas aeruginosa/genética , Bacterias/metabolismo , Carbono/metabolismo
6.
Mol Biol Rep ; 49(12): 11643-11652, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36169895

RESUMEN

BACKGROUND: Polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) promote tumor immune tolerance and cause tumor immunotherapy failure. In this study, we found that high PMN-MDSCs infiltration, overexpressed fatty acid transporter protein 2 (FATP2) and underexpressed receptor-interacting protein kinase 3 (RIPK3) existed in the mouse and human bladder cancer tissues. However, the related mechanisms remain largely unknown. METHODS AND RESULTS: Both FATP2 and RIPK3 expressions were associated with clinical stage. FATP2 knockout or up-regulating RIPK3 reduced the synthesis of prostaglandin E2 (PGE2) in PMN-MDSCs, attenuated the suppressive activity of PMN-MDSCs on CD8+ T cells functions and inhibited the tumor growth. There was a PGE2-mediated feedback loop between FATP2 and RIPK3 pathways, which markedly promoted the immunosuppressive activity of PMN-MDSCs. Combination therapy with inhibition of FATP2 and activation of RIPK3 can effectively inhibit tumor growth. CONCLUSIONS: This study demonstrated that a feedback loop between FATP2 and RIPK3 pathways in PMN-MDSCs significantly promoted the synthesis of PGE2, which severely impaired the CD8+ T cell functions. This study may provide new ideas for immunotherapy of human bladder cancer.


Asunto(s)
Proteínas de Transporte de Ácidos Grasos , Células Supresoras de Origen Mieloide , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Neoplasias de la Vejiga Urinaria , Animales , Humanos , Ratones , Linfocitos T CD8-positivos , Dinoprostona/metabolismo , Proteínas de Transporte de Ácidos Grasos/metabolismo , Ácidos Grasos/metabolismo , Células Supresoras de Origen Mieloide/metabolismo , Neutrófilos/metabolismo , Neoplasias de la Vejiga Urinaria/metabolismo , Retroalimentación Fisiológica , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
7.
Int J Mol Sci ; 23(15)2022 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-35955521

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is found in up to 30% of the world's population and can lead to hepatocellular carcinoma (HCC), which has a poor 5-year relative survival rate of less than 40%. Clinical therapeutic strategies are not very successful. The co-occurrence of metabolic disorders and inflammatory environments during the development of steatohepatitis thus needs to be more specifically diagnosed and treated to prevent fatal HCC development. To improve diagnostic and therapeutic strategies, the identification of molecules and/or pathways responsible for the initiation and progression of chronic liver disease has been explored in many studies, but further study is still required. Transmembrane 4 L six family member 5 (TM4SF5) has been observed to play roles in the regulation of metabolic functions and activities in hepatocytes using in vitro cell and in vivo animal models without or with TM4SF5 expression in addition to clinical liver tissue samples. TM4SF5 is present on the membranes of different organelles or vesicles and cooperates with transporters for fatty acids, amino acids, and monocarbohydrates, thus regulating nutrient uptake into hepatocytes and metabolism and leading to phenotypes of chronic liver diseases. In addition, TM4SF5 can remodel the immune environment by interacting with immune cells during TM4SF5-mediated chronic liver diseases. Because TM4SF5 may act as an NAFLD biomarker, this review summarizes crosstalk between TM4SF5 and nutrient transporters in hepatocytes, which is related to chronic liver diseases.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Enfermedades Metabólicas , Enfermedad del Hígado Graso no Alcohólico , Animales , Carcinoma Hepatocelular/metabolismo , Hepatocitos/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Enfermedades Metabólicas/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo
8.
Vet World ; 15(4): 911-918, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35698512

RESUMEN

Background and Aim: As a new feed additive, bile acid (BA) can promote the absorption and transport of lipids and fat-soluble vitamins. In recent years, BAs have been widely used in animal feed to promote fat absorption. Therefore, this study aimed to investigate the effect of bile salt supplementation in the diet of yellow-feathered broilers on messenger RNA (mRNA) expression of sterol regulatory element-binding protein 1 (SREBF1), fatty acid synthase (FAS), acetyl-coenzyme A carboxylase (ACC), and fatty acid transport protein 4 (FATP4). Materials and Methods: Four hundred and twenty commercial male chicks were randomly divided into seven groups (with four replicates per group and 15 chickens per replicate). They were fed diets supplemented with bile salts at 0, 1.5, 2.5, 3.5, 4.5, 5.5 mg/kg, and 2 mg/kg tylosin for 30 days. Changes in SREBF1, fatty acid transporter 4, FAS, and acetyl-CoA carboxylase genes in intestinal mucosa and liver of yellow-feathered broilers were determined using a quantitative fluorescence polymerase chain reaction. Results: mRNA expression of SREBF1, FAS, ACC, and FATP4 in the small intestine decreased in chicks fed diets supplemented with 3.5 and 4.5 mg/kg bile salts (p<0.05) compared with the control group on 7 days and 14 d. The mRNA expressions of SREBF1, FAS, ACC, and FATP4 in liver tissue decreased in chicks fed diets supplemented with 4.5 and 5.5 mg/kg bile salts (p<0.05) compared to the control group on 7 days. The mRNA expression of SREBF1, FAS, ACC, and FATP4 in the liver at 14 days and the small intestine on 21 days also decreased in chicks fed diets supplemented with 4.5 mg/kg bile salts (p<0.05) compared to the control group. When contrasted with the control group on day 21, the mRNA expression of SRWBF1, FAS, ACC, and FATP4 detected in the liver was lower in chicks fed diets supplemented with bile salts (p<0.05). Conclusion: The dietary supplementation of bile salts at 4.5 mg/kg effectively regulates the expression of fat metabolism genes, such as SREBF1, FAS, ACC, and FATP4 mRNA. At this concentration, bile salts promote fat catabolism, inhibit fat synthesis, and play an essential role in improving the fat deposition of broilers.

9.
Toxicol Lett ; 363: 1-10, 2022 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-35589016

RESUMEN

Glucocorticoids such as dexamethasone (DEX) are widely prescribed to treat numerous conditions and diseases. However, glucocorticoid-induced liver lipid metabolism disorder, even nonalcoholic fatty liver disease, has caused extensive attention. Since fatty acid transporters such as CD36 and FATP play crucial roles in hepatic fatty acid uptake, this work examined their potential involvement in DEX-induced liver lipid accumulation. Chronic DEX administration (1-5 mg/kg/day over 28 days) induced hepatic lipid accumulation in mice. Fatty acid uptake in HepG2 cells and mouse primary hepatocytes was also stimulated after incubation with 0.5-2 µM DEX. Meanwhile, qPCR and western blotting demonstrated dose-dependent upregulation of CD36 expression by DEX in the mouse liver and in cultured hepatocytes. Glucocorticoid receptor (GR) inhibition with mifepristone (RU486) and siRNA-mediated GR knockdown attenuated lipid accumulation in hepatocytes by inhibiting DEX-induced CD36 upregulation, and direct binding of GR to the CD36 promoter was demonstrated by luciferase reporter and chromatin immunoprecipitation assays. These results indicate that DEX promotes free fatty acid uptake leading to hepatic steatosis by upregulating CD36 expression via activation of GR. Thus, strategies aimed at inhibiting GR/CD36 expression or activity might help prevent or reduce the onset and progression of hepatic lipid metabolism disorders induced by glucocorticoid drugs.


Asunto(s)
Trastornos del Metabolismo de los Lípidos , Enfermedad del Hígado Graso no Alcohólico , Animales , Antígenos CD36/genética , Dexametasona/toxicidad , Ácidos Grasos/metabolismo , Glucocorticoides/metabolismo , Metabolismo de los Lípidos , Trastornos del Metabolismo de los Lípidos/metabolismo , Hígado/metabolismo , Ratones , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Receptores de Glucocorticoides/metabolismo , Regulación hacia Arriba
10.
Cell Biosci ; 12(1): 69, 2022 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-35619150

RESUMEN

INTRODUCTION: Blood-brain barrier (BBB) dysfunction may occur at the onset of Alzheimer's disease (AD). Pericytes are a vital part of the neurovascular unit and the BBB, acting as gatekeepers of the BBB. Amyloid ß (Aß) deposition and neurofibrillary tangles in the brain are the central pathological features of AD. CD36 promotes vascular amyloid deposition and leads to vascular brain damage, neurovascular dysfunction, and cognitive deficits. However, the molecular mechanism by which pericytes of the BBB are disrupted remains unclear. OBJECTIVES: To investigate the effect of low-dose Aß1-40 administration on pericyte outcome and the molecular mechanism of BBB injury. METHODS: We selected 6-month-old and 9-month-old APP/PS1 mice and wild-type (WT) mice of the same strain, age, and sex as controls. We assessed the BBB using PET/CT. Brain pericytes were extracted and cocultured with endothelial cells (bEnd.3) to generate an in vitro BBB model to observe the effect of Aß1-40 on the BBB. Furthermore, we explored the intracellular degradation and related molecular mechanisms of Aß1-40 in cells. RESULTS: BBB permeability and the number of pericytes decreased in APP/PS1 mice. Aß1-40 increased BBB permeability in an in vivo model and downregulated the expression of CD36, which reversed the Aß-induced changes in BBB permeability. Aß1-40 was uptaked in pericytes with high CD36 expression. We observed that this molecule inhibited pericyte proliferation, caused mitochondrial damage, and increased mitophagy. Finally, we confirmed that Aß1-40 induced pericyte mitophagy-dependent ferroptosis through the CD36/PINK1/Parkin pathway. CONCLUSION: PDGFRß (a marker of pericytes), CD36, and Aß colocalized in vitro and in vivo, and Aß1-40 caused BBB disruption by upregulating CD36 expression in pericytes. The mechanism by which Aß1-40 destroys the BBB involves the induction of pericyte mitophagy-dependent ferroptosis through the CD36/PINK1/Parkin pathway.

11.
Nutrients ; 13(8)2021 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-34444927

RESUMEN

The rise in prevalence of obesity in women of reproductive age in developed and developing countries might propagate intergenerational cycles of detrimental effects on metabolic health. Placental lipid metabolism is disrupted by maternal obesity, which possibly affects the life-long health of the offspring. Here, we investigated placental lipid metabolism in women with pre-gestational obesity as a sole pregnancy complication and compared it to placental responses of lean women. Open profile and targeted lipidomics were used to assess placental lipids and oxidised products of docosahexaenoic (DHA) and arachidonic acid (AA), respectively, neuroprostanes and isoprostanes. Despite no overall signs of lipid accumulation, DHA and AA levels in placentas from obese women were, respectively, 2.2 and 2.5 times higher than those from lean women. Additionally, a 2-fold increase in DHA-derived neuroprostanes and a 1.7-fold increase in AA-derived isoprostanes were seen in the obese group. These changes correlated with a 70% decrease in placental FABP1 protein. Multivariate analyses suggested that neuroprostanes and isoprostanes are associated with maternal and placental inflammation and with birth weight. These results might shed light on the molecular mechanisms associated with altered placental fatty acid metabolism in maternal pre-gestational obesity, placing these oxidised fatty acids as novel mediators of placental function.


Asunto(s)
Proteínas de Unión a Ácidos Grasos/metabolismo , Isoprostanos/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos/genética , Neuroprostanos/metabolismo , Obesidad Materna/metabolismo , Adulto , Peso al Nacer , Femenino , Humanos , Inflamación , Metabolismo de los Lípidos , Placenta/metabolismo , Embarazo
12.
Reprod Med Biol ; 20(3): 289-298, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34262396

RESUMEN

PURPOSE: The present study was undertaken to clarify whether bovine sperm could take up fatty acids (FAs) and produce ATP to maintain linear motility. METHODS: Frozen bovine semen was thawed in media containing either lipid mixture (LM) or FAs, and sperm motility was analyzed. The kinetic changes in FA levels in sperm were detected using gas chromatography-mass spectrometry. The mitochondrial activity of sperm thawed in media containing LM or FAs was analyzed based on the fluorescence intensity of JC-1 staining and the oxygen consumption rate. FA transporters were observed using whole-mounted immunofluorescence. RESULTS: Sperm linear motility was significantly (P < .05) increased after thawing in media with LM and FA. Moreover, saturated fatty acids were predominant in sperm thawed in media with LM. Notably, our study revealed that frozen bovine sperm possessed FA transporters in the midpiece where the fluorescence signals were detected after treatment with fluorescence-tagged FA. Treatment with FA activated electron transport in mitochondria through ß-oxidation. CONCLUSIONS: Sperm linear motility is facilitated by FAs in the thawing media used for frozen bovine sperm. This might provide a new approach for upgrading the artificial insemination technique used in both livestock animals and human infertility care.

13.
Sci Total Environ ; 756: 144130, 2021 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-33288249

RESUMEN

Polycyclic aromatic hydrocarbon exposure accelerates the initiation and progression of lung cancer through aryl hydrocarbon receptor (AHR) signaling. Metabolic reprogramming is a hallmark of cancer. However, how AHR reprograms metabolism related to the malignant transformation in of benzo[a]pyrene (BaP)-exposed lung cells remains unclear. After confirming that BaP exposure activated AHR signaling and relevant downstream factors and then promoted epithelial-mesenchymal transition, an untargeted metabolomics approach was employed to discover AHR-mediated metabolic reprogramming and potential therapeutic targets in BaP-exposed BEAS-2B cells. We found that 52 metabolites were significantly altered in BaP-exposed BEAS-2B cells and responsive to resveratrol (RSV) intervention. Pathway analysis revealed that 28 and 30 metabolic pathways were significantly altered in response to BaP exposure and RSV intervention, respectively. Notably, levels of most amino acids were significantly decreased, while those of most fatty acids were significantly increased in BaP-exposed BEAS-2B cells, and above changes were abolished by RSV intervention. Besides, levels of amino acids and fatty acids were highly correlated with those of many metabolites and AHR signaling upon BaP exposure and RSV intervention (the absolute values of Pearson correlation coefficients above 0.8). We further discovered a decrease in peroxisome proliferator-activated receptor (PPAR) A/G signaling and an increase in fatty acid import by the transporter FATP1 in BaP-exposed BEAS-2B cells. Furthermore, inhibition of AHR signaling by CH-223191 abolished BaP-induced repression of PPARA/G signaling and activation of FATP1 in BEAS-2B cells, demonstrating the regulatory role of AHR signaling in fatty acid accumulation via mediating PPARA/G-FATP1 signaling. These data suggested amino acid and fatty acid metabolism, AHR and PPAR-FATP1 signaling as potential therapeutic targets for intervening BaP-induced toxicity and related diseases. As far as we known, fatty acid accumulation and high correlations of AHR signaling with amino acid and fatty acid metabolism are novel phenomena discovered in BaP-exposed lung epithelial cells.


Asunto(s)
Benzo(a)pireno , Receptores de Hidrocarburo de Aril , Benzo(a)pireno/toxicidad , Células Epiteliales , Humanos , Metabolismo de los Lípidos , Pulmón/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo
14.
Animal ; : 1-8, 2020 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-32148213

RESUMEN

Glucocorticoids (GCs) are counterregulatory hormones with broad effects on the digestion and absorption of dietary carbohydrates, lipids and proteins, but the underlying molecular mechanisms of these effects remain unclear. The present experiment was conducted to investigate the main expression sites of nutrient transporters and the effects of GCs on the gene expression of these transporters in the rabbit small intestine. The results showed that peptide transporter 1 (PepT1), facultative amino acid transporter (rBAT), neutral amino acid transporter (B0AT), excitatory amino acid transporter 3 (EAAT3), sodium-glucose transporter 1 (SGLT1) and glucose transporter 5 (GLUT5) were mainly expressed in the distal segment, glucose transporter 2 (GLUT2) and fatty-acid-binding protein 4 (FATP4) were mainly expressed in the proximal segment and cationic amino acid transporter 1 (CAT1) was mainly expressed in the middle segment of the rabbit small intestine. In addition, we analysed the effects of 3 h (short-term) or 7 days (long-term) dexamethasone (DEX) treatment on the gene expression of most nutrient transporters. The results showed that short-term DEX treatment significantly decreased PepT1, B0AT, EAAT3, rBAT and SGLT1 expressions in all small intestinal segments, while it significantly decreased GLUT2 in the duodenum and FATP4 in the duodenum and ileum (P < 0.05). Long-term DEX treatment also significantly decreased PepT1, CAT1, B0AT, EAAT3, rBAT and SGLT1 in all small intestinal segments and significantly decreased GLUT2 in the jejunum and FATP4 in the ileum (P < 0.05). In conclusion, DEX could decrease the gene expression of most nutrient transporters (except GLUT5) and affect the transport of intestinal amino acids, monosaccharides and fatty acids.

15.
J Med Food ; 23(3): 281-288, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32119806

RESUMEN

The aim of this study was to investigate the effect of a high-fat diet (HFD) on energy substrate utilization during long-term endurance exercise in mice. Male ICR mice (n = 32; 6 weeks old) were divided into two groups: low-fat diet (LFD, n = 16) and HFD (n = 16) and acclimatized to LFD or HFD feeding over 12 weeks. After 12 weeks, the two dietary groups were each divided into two groups with or without exercise (EX): LF-CON, LF-EX, HF-CON, and HF-EX groups. The exercise groups were trained to run on a treadmill for 12 weeks. At the end of the experimental protocol, energy metabolism in the whole body was measured at rest for 24 h and during exercise for 1 h using respiratory gas analysis. Furthermore, molecules involved in skeletal muscle fat metabolism were analyzed. Substrate utilization for energy metabolism in the whole body indicated that fat utilization was high in HFD intake. Notably, when HFD intake and exercise were combined, fat utilization was markedly increased during endurance exercise. In contrast, exercise showed no effect when combined with LFD intake. The gene expressions of Fat/Cd36, Fatp1, Fabp-pm, and Cpt1 were upregulated by HFD intake, with Fat/Cd36 and Cpt1 considerably elevated during long-term endurance exercise. In contrast, exercise showed no effect when combined with LFD intake. These results suggest that HFD intake effectively increased fat utilization as an energy substrate during long-term endurance exercise.


Asunto(s)
Grasas de la Dieta/metabolismo , Ácidos Grasos/metabolismo , Músculo Esquelético/metabolismo , Animales , Dieta Alta en Grasa , Grasas de la Dieta/análisis , Metabolismo Energético , Proteínas de Transporte de Ácidos Grasos/genética , Proteínas de Transporte de Ácidos Grasos/metabolismo , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Oxidación-Reducción , Condicionamiento Físico Animal
16.
Reprod Sci ; 27(9): 1758-1769, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32072607

RESUMEN

Fetal growth and development are dependent on placental nutrient transport. The syncytiotrophoblast (ST) and its two polarized plasma membranes, the maternal-facing microvillous membrane (MVM) and fetal-facing basal membrane (BM), represent the primary barrier in the human placenta, controlling transplacental transfer of small solutes. MVM and BM nutrient transporter expression and activity are increased in obese mothers delivering large babies. However, placental nutrient transporter expression and activity in early gestation in normal and obese women are largely unknown. Placentas from normal BMI and obese women at 6-24 weeks of gestation, and term placentas from normal BMI women, were collected and ST plasma membranes isolated. The activity and protein expression of amino acid, glucose, and fatty acid transporters was assessed. No significant differences were observed in placental nutrient transporter protein expression between normal BMI and obese women in early pregnancy. In the MVM, system A amino acid activity (p = 0.02), SNAT2 (p < 0.0001), SNAT4 (p < 0.001), and GLUT1 (p = 0.01) protein expression were higher at term compared with early gestation. In contrast, MVM system L activity (p = 0.001), FATP4 (p = 0.03), and FATP6 (p = 0.009) protein expression were lower at term compared with early pregnancy. In the BM, there was no change in system L activity across gestation; however, BM FATP6 (p = 0.002) protein expression was lower at term compared with early pregnancy. These results suggest that placental transport of amino acids, glucose, and fatty acids are subjected to coordinated regulation across gestation to meet a fetal nutrient demand that changes with advancing pregnancy.


Asunto(s)
Sistema de Transporte de Aminoácidos A/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Obesidad/metabolismo , Placenta/metabolismo , Primer Trimestre del Embarazo/metabolismo , Segundo Trimestre del Embarazo/metabolismo , Adulto , Índice de Masa Corporal , Femenino , Humanos , Embarazo , Trofoblastos/metabolismo
17.
Oral Dis ; 26(4): 745-755, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-31603606

RESUMEN

OBJECTIVE: Recent studies have demonstrated the pro-tumour role of CD36 in multiple cancer types. However, its role has not been well elucidated in oral squamous cell carcinoma (OSCC). Here, we aimed to evaluate the role of CD36 in proliferation and migration of OSCC cells. METHODS: Human OSCC cell lines HSC-2, HSC-3, HSC-4 and Ca9-22 were assessed for proliferation by staining with the cell proliferation marker Ki-67. We also assessed migration activity, and the expression of cell adhesion molecules such as E-cadherin and ß-catenin and platelet-derived growth factor receptors (PDGFRs) of CD36-positive cells. RESULTS: CD36-positive cells showed increased expression of Ki-67 and migration activity compared with CD36-negative cells. Moreover, CD36-positive cells showed reduced expression of E-cadherin and ß-catenin, whereas the expression of PDGFRs increased compared with that in CD36-negative cells. CONCLUSIONS: Our results strongly suggest that CD36 has an important role in facilitating the proliferation and migration activity of OSCC cells, indicating its usefulness in the diagnosis of high-grade tumour and targeted therapy of oral cancer.


Asunto(s)
Antígenos CD36/metabolismo , Carcinoma de Células Escamosas/metabolismo , Movimiento Celular , Proliferación Celular , Neoplasias de la Boca/metabolismo , Antígenos CD/metabolismo , Cadherinas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Humanos , Antígeno Ki-67/metabolismo , Neoplasias de la Boca/patología , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , beta Catenina/metabolismo
18.
Mol Cell Biochem ; 461(1-2): 195-204, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31414336

RESUMEN

Recent studies on mice with null mutation of the angiotensin type 2 receptor (AT2R) gene have implicated the involvement of AT2R in regulating adipocyte size and obesity, a major risk factor for metabolic syndrome. However, the outcome from these studies remains inconclusive. Therefore, current study was designed to test whether pharmacological activation of AT2R regulates adiposity and lipid metabolism. Male mice (5-weeks old) were pre-treated with vehicle or AT2R agonist (C21, 0.3 mg/kg, i.p., daily, for 4 days) and fed normal diet (ND). Then these animals were subdivided into ND and high-fat diet (HFD) regimen and concomitantly treated with vehicle or C21 through day 14. Vehicle-treated HFD-fed mice demonstrated an increase in epididymal white adipose tissue (eWAT) weight and adipocyte size, which were associated with increased eWAT expression of the lipogenic regulators, fatty acid binding protein and fatty acid synthase, decreased expression of adipose triglyceride lipase and increased expression of hormone-sensitive lipase. Interestingly, C21 pre-treatment altered HFD-induced changes in lipogenic and lipolytic regulators. C21 pre-treatment prevented decrease in expression of uncoupler protein-1 in brown adipose in HFD-fed mice, which was associated with increased core temperature. In addition, C21 pre-treatment ameliorated plasma-free fatty acids, triglycerides, insulin and tumor necrosis factor-α in HFD-fed mice. Ex-vivo study in isolated primary epididymal adipocytes revealed that C21 inhibits long chain fatty acid transporter, via a nitric oxide synthase/guanylate cyclase/protein kinase G-dependent pathway. Collectively, we propose pharmacological activation of AT2R regulates fatty acid metabolism and thermogenesis and prevents HFD-induced adiposity in mice.


Asunto(s)
Adiposidad , Metabolismo de los Lípidos , Receptor de Angiotensina Tipo 2/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Adiponectina/sangre , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Temperatura Corporal , Peso Corporal , Tamaño de la Célula , Ingestión de Energía , Epidídimo/anatomía & histología , Ácidos Grasos/sangre , Insulina/sangre , Masculino , Ratones Endogámicos C57BL , Triglicéridos/sangre , Factor de Necrosis Tumoral alfa/sangre , Proteína Desacopladora 1/metabolismo
19.
Toxicology ; 423: 112-122, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31152847

RESUMEN

Cadmium (Cd) is one of the environmental pollutants, which has multiple toxic effects on fetuses and placentas. Placental fatty acid (FA) uptake and transport are critical for the fetal and placental development. We aimed to analyze the triglyceride (TG) level, the expression patterns of several key genes involved in FA uptake and transport, and the molecular mechanisms for the altered gene expressions in placentas in response to Cd treatment. Our results showed that the placental TG level was significantly decreased in the Cd-exposed placentas. Fatty acid transporting protein 1 (FATP1), FATP6 and fatty acid binding protein 3 (FABP3) were significantly down-regulated in the placentas from Cd-exposed mice. The expression level of phospho-p38 MAPK was increased by Cd treatment, while the protein level of total p38 MAPK remained unchanged. The expression levels of peroxisome proliferator-activated receptor-γ (PPAR-γ) and the hypoxia-inducible factor-1α (HIF-1α) were significantly decreased in the Cd-exposed placentas. The methylation levels of the promoter regions of FATP1, FATP6 and FABP3 showed no significant differences between the treatment and control groups. In addition, the circulating non-esterified fatty acid (NEFA), total cholesterol (TC), and TG levels were not decreased in the maternal serum from the Cd-exposed mice. Therefore, our results suggest Cd exposure dose not reduce the maternal FA supply, but reduces the placental TG level. Cd treatment also downregulates the placental expressions of FATP1, FATP6 and FABP3, respectively associated with p38-MAPK, p38 MAPK/PPAR-γ and HIF-1α pathways.


Asunto(s)
Cadmio/toxicidad , Proteínas de Transporte de Ácidos Grasos/metabolismo , Retardo del Crecimiento Fetal/metabolismo , Intercambio Materno-Fetal , Placenta/efectos de los fármacos , Animales , Línea Celular , Regulación hacia Abajo , Proteínas de Transporte de Ácidos Grasos/genética , Femenino , Retardo del Crecimiento Fetal/inducido químicamente , Retardo del Crecimiento Fetal/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones Endogámicos C57BL , PPAR gamma/genética , PPAR gamma/metabolismo , Ácido Palmítico/farmacología , Placenta/metabolismo , Embarazo , ARN Mensajero/metabolismo , Triglicéridos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
J Anim Sci ; 97(6): 2342-2356, 2019 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-30958881

RESUMEN

Bacterial infection causes nutrient malabsorption in small intestine. KR-32, a kind of synthetic antimicrobial peptide, has the bacteriostatic effect. In the present study, 2 experiments were designed to analyze the effects of KR-32 on fat absorption of piglets with or without Escherichia coli infection. In Exp. 1, 12 weaning piglets (21 d old) were allocated to 2 groups: piglets with an intraperitoneal (i.p.) injection of antimicrobial peptide KR-32 (APK) and piglets with an i.p. injection of an equivalent volume (1 mL) of phosphate-buffered saline (PBS) (CON-1). Results showed that after 7 d of growth, KR-32 did not significantly change growth performance and apparent total tract digestibility (ATTD) of feed nutrients of normal pigs. To confirm whether KR-32 affects those of enterotoxigenic Escherichia coli (ETEC) K88-challenged pigs, we performed Exp. 2, in which 18 piglets (28 d old) were divided into the following 3 groups: 1) piglets orally challenged with 1 × 1010 cfu ETEC K88 on day 1 followed by an i.p. injection of 0.6 mg/kg KR-32 (K88 + APK); 2) piglets orally challenged with 1 × 1010 cfu ETEC K88 on day 1 followed by an i.p. injection of an equivalent volume (1 mL) of PBS (K88); and 3) piglets with an oral administration of fresh Luria-Bertani broth (50 mL) followed by an i.p. injection of an equivalent volume of PBS (CON-2). Results showed that ETEC K88 challenge led to poor ADFI, ADG, and G:F in piglets; decreased ATTD of feed nutrients, especially CP and ether extract (EE); and intestinal morphology disorder. After i.p. injection of KR-32, ADG and ATTD of CP and EE were greatly increased, G:F was significantly reduced (P < 0.05), and, especially, ATTD of EE returned to a normal level compared with group CON-2. Fatty acid absorption also highly increased after KR-32 injection. Then we focused on fat digestion and fatty acid uptake. The pH in the intestine and pancreas lipase showed no difference among the 3 treatment groups, whereas fatty acid transporter protein 4 (FATP4) expression was remarkably improved (P < 0.05) and the epithelial barrier was recovered after i.p. injection of KR-32. In conclusion, KR-32, given to ETEC K88-challenged piglets, improved growth performance, ATTD of EE, fatty acid absorption, and intestinal morphology, which indicated that KR-32 was likely to improve the expression of FATP4 and by repairing the epithelial barrier, thereby alleviating fatty acid malabsorption.


Asunto(s)
Antiinfecciosos/farmacología , Escherichia coli Enterotoxigénica/efectos de los fármacos , Infecciones por Escherichia coli/veterinaria , Proteínas de Transporte de Ácidos Grasos/genética , Ácidos Grasos/metabolismo , Enfermedades de los Porcinos/tratamiento farmacológico , Animales , Digestión/efectos de los fármacos , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/patología , Proteínas de Transporte de Ácidos Grasos/efectos de los fármacos , Proteínas de Transporte de Ácidos Grasos/metabolismo , Ácidos Grasos/sangre , Heces/microbiología , Femenino , Absorción Intestinal/efectos de los fármacos , Intestinos/efectos de los fármacos , Intestinos/ultraestructura , Péptidos/farmacología , Distribución Aleatoria , Porcinos , Enfermedades de los Porcinos/microbiología , Enfermedades de los Porcinos/patología , Destete
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA