Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Gut Microbes ; 16(1): 2333413, 2024.
Article in English | MEDLINE | ID: mdl-38561312

ABSTRACT

Urinary tract infections (UTIs) are among the most common late-onset infections in preterm infants, characterized by nonspecific symptoms and a pathogenic spectrum that diverges from that of term infants and older children, which present unique diagnostic and therapeutic challenges. Existing data on the role of gut microbiota in UTI pathogenesis in this demographic are limited. This study aims to investigate alterations in gut microbiota and fecal calprotectin levels and their association with the development of UTIs in hospitalized preterm infants. A longitudinal case-control study was conducted involving preterm infants admitted between January 2018 and October 2020. Fecal samples were collected weekly and analyzed for microbial profiles and calprotectin levels. Propensity score matching, accounting for key perinatal factors including age and antibiotic use, was utilized to match samples from UTI-diagnosed infants to those from non-UTI counterparts. Among the 151 preterm infants studied, 53 were diagnosed with a UTI, predominantly caused by Enterobacteriaceae (79.3%) and Enterococcaceae (19.0%). Infants with UTIs showed a significantly higher abundance of these families compared to non-UTI infants, for both Gram-negative and positive pathogens, respectively. Notably, there was a significant pre-UTI increase in the abundance of pathogen-specific taxa in infants later diagnosed with UTIs, offering high predictive value for early detection. Shotgun metagenomic sequencing further confirmed the dominance of specific pathogenic species pre-UTI and revealed altered virulence factor profiles associated with Klebsiella aerogenes and Escherichia coli infections. Additionally, a decline in fecal calprotectin levels was observed preceding UTI onset, particularly in cases involving Enterobacteriaceae. The observed pathogen-specific alterations in the gut microbiota preceding UTI onset offer novel insight into the UTI pathogenesis and promising early biomarkers for UTIs in preterm infants, potentially enhancing the timely management of this common infection. However, further validation in larger cohorts is essential to confirm these findings.


Subject(s)
Gastrointestinal Microbiome , Urinary Tract Infections , Infant , Child , Humans , Infant, Newborn , Adolescent , Case-Control Studies , Escherichia coli , Infant, Premature , Anti-Bacterial Agents/therapeutic use , Enterobacteriaceae , Leukocyte L1 Antigen Complex
2.
Nutrients ; 15(22)2023 Nov 20.
Article in English | MEDLINE | ID: mdl-38004243

ABSTRACT

BACKGROUND: Feeding intolerance (FI) is a significant concern in the care of preterm infants, impacting their growth and development. We previously reported that FI is linked to lower fecal calprotectin (FC) levels. This study aims to explore the postnatal dynamics and interplay between microbiota, metabolic profiles, and host immunity in preterm infants with and without FI. METHODS: Infants with gestational age <32 weeks or birth weight <1500 g were enrolled at the Children's Hospital of Fudan University between January 2018 and October 2020. Weekly fecal samples were analyzed for bacterial profiling, metabolome, and calprotectin levels, exploring their longitudinal development and interrelationships. RESULTS: Of the 118 very preterm infants studied, 48 showed FI. These infants experienced an interrupted microbial-immune trajectory, particularly at 3-4 weeks of age, marked by a reduced bacterial abundance, alpha diversity, and FC levels. Metabolic changes in FI were pronounced between 3 and 6 weeks. Pantothenic acid and two polyamine metabolites were closely associated with bacterial abundance and FC levels and negatively correlated with the duration to attain full enteral feeding. CONCLUSIONS: FI infants demonstrated compromised microbiome-immune interactions, potentially influenced by specific metabolites. This research underscored the importance of early microbial and metabolic development in the pathogenesis of FI in very preterm infants.


Subject(s)
Gastrointestinal Microbiome , Infant, Premature, Diseases , Infant , Child , Infant, Newborn , Humans , Infant, Premature , Leukocyte L1 Antigen Complex , Infant, Very Low Birth Weight , Bacteria , Metabolome
3.
Phenomics ; 3(3): 268-284, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37325710

ABSTRACT

The gut microbiota refers to the gross collection of microorganisms, estimated trillions of them, which reside within the gut and play crucial roles in the absorption and digestion of dietary nutrients. In the past decades, the new generation 'omics' (metagenomics, transcriptomics, proteomics, and metabolomics) technologies made it possible to precisely identify microbiota and metabolites and describe their variability between individuals, populations and even different time points within the same subjects. With massive efforts made, it is now generally accepted that the gut microbiota is a dynamically changing population, whose composition is influenced by the hosts' health conditions and lifestyles. Diet is one of the major contributors to shaping the gut microbiota. The components in the diets vary in different countries, religions, and populations. Some special diets have been adopted by people for hundreds of years aiming for better health, while the underlying mechanisms remain largely unknown. Recent studies based on volunteers or diet-treated animals demonstrated that diets can greatly and rapidly change the gut microbiota. The unique pattern of the nutrients from the diets and their metabolites produced by the gut microbiota has been linked with the occurrence of diseases, including obesity, diabetes, nonalcoholic fatty liver disease, cardiovascular disease, neural diseases, and more. This review will summarize the recent progress and current understanding of the effects of different dietary patterns on the composition of gut microbiota, bacterial metabolites, and their effects on the host's metabolism.

4.
EMBO Mol Med ; 15(4): e17450, 2023 04 11.
Article in English | MEDLINE | ID: mdl-36847712

ABSTRACT

Premature ovarian insufficiency (POI) is a disease featured by early menopause before 40 years of age, accompanied by an elevation of follicle-stimulating hormone. Though POI affects many aspects of women's health, its major causes remain unknown. Many clinical studies have shown that POI patients are generally underweight, indicating a potential correlation between POI and metabolic disorders. To understand the pathogenesis of POI, we performed metabolomics analysis on serum and identified branch-chain amino acid (BCAA) insufficiency-related metabolic disorders in two independent cohorts from two clinics. A low BCAA diet phenotypically reproduced the metabolic, endocrine, ovarian, and reproductive changes of POI in young C57BL/6J mice. A mechanism study revealed that the BCAA insufficiency-induced POI is associated with abnormal activation of the ceramide-reactive oxygen species (ROS) axis and consequent impairment of ovarian granulosa cell function. Significantly, the dietary supplement of BCAA prevented the development of ROS-induced POI in female mice. The results of this pathogenic study will lead to the development of specific therapies for POI.


Subject(s)
Menopause, Premature , Primary Ovarian Insufficiency , Humans , Female , Mice , Animals , Reactive Oxygen Species , Amino Acids , Mice, Inbred C57BL , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/pathology , Primary Ovarian Insufficiency/therapy
6.
Nutrients ; 14(11)2022 May 26.
Article in English | MEDLINE | ID: mdl-35684023

ABSTRACT

The diet-induced obesity (DIO) mouse model has been widely used for obesity studies. The effects of storage conditions on the composition of nutrients in high-fat diets (HFDs) and their impact on metabolic homeostasis have not been systemically investigated. In the current study, we tested the effects of HFDs stored under different conditions and found that mice fed a HFD stored in the fridge (HFDfri) gained less weight than those fed HFDs stored in the freezer (HFDfre). Further analysis revealed that changes in the relative abundance of medium-chain triglyceride (MCT) in the HFDfri, which have much lower intestinal absorption rates, contributed to the body weight differences. In contrast, exacerbated liver damage and elevated levels of unfolded protein response (UPR) was observed in the mice fed by HFDfri. Depletion of the UPR-regulated gene Nnmt alleviated liver damage via the inhibition of the integrated stress response (ISR). Our study, for the first time, provides evidence that HFD storage conditions can have a significant impact on both body weight changes and liver damage in the DIO model.


Subject(s)
Diet, High-Fat , Liver , Animals , Diet, High-Fat/adverse effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Obesity/metabolism , Triglycerides/metabolism
8.
Mol Nutr Food Res ; 66(1): e2100342, 2022 01.
Article in English | MEDLINE | ID: mdl-34788490

ABSTRACT

SCOPE: Necrotizing enterocolitis (NEC) is a leading cause of morbidity and mortality in preterm infants, occurring more often in formula-fed infants than in breastfed infants. Recent animal studies have shown that cells in fresh breast milk survive in the newborns' digestive tract. However, no clinical studies have been conducted on the effects of human milk cells, and their biological roles in the infants' intestines remain unexplored. METHODS AND RESULTS: Twenty premature infants are enrolled. Cells from fresh milk of their own mothers are enriched and fed to infants with Bell's Stage I NEC once a day for 7 days since the onset of NEC. Fecal samples are collected at enrollment and 2 weeks later. Fecal sphingolipids are observed to be enriched in NEC patients and positively correlated with calprotectin levels. After intervention with enriched human milk cells, inflammation-associated sphingolipids and microbiome profiles are altered and resembled those of the controls. CONCLUSION: These preliminary findings reveal the potential impacts of enriched human milk cells on premature infants with Bell's Stage I NEC and provide insight into the roles of fecal sphingolipid metabolism in the neonates' intestinal inflammation. However, the limited sample size of the study indicates the need for further investigation.


Subject(s)
Enterocolitis, Necrotizing , Gastrointestinal Microbiome , Animals , Enterocolitis, Necrotizing/metabolism , Humans , Infant, Newborn , Infant, Premature , Metabolome , Milk, Human/metabolism , Pilot Projects
9.
Mol Metab ; 54: 101329, 2021 12.
Article in English | MEDLINE | ID: mdl-34454092

ABSTRACT

OBJECTIVE: The loss of forkhead box protein O1 (FoxO1) signaling in response to metabolic stress contributes to the etiology of type II diabetes, causing the dedifferentiation of pancreatic beta cells to a cell type reminiscent of endocrine progenitors. Lack of methods to easily model this process in vitro, however, have hindered progress into the identification of key downstream targets and potential inhibitors. We therefore aimed to establish such an in vitro cellular dedifferentiation model and apply it to identify novel agents involved in the maintenance of beta-cell identity. METHODS: The murine beta-cell line, Min6, was used for primary experiments and high-content screening. Screens encompassed a library of small-molecule drugs representing the chemical and target space of all FDA-approved small molecules with an automated immunofluorescence readout. Validation experiments were performed in a murine alpha-cell line as well as in primary murine and human diabetic islets. Developmental effects were studied in zebrafish and C. elegans models, while diabetic db/db mouse models were used to elucidate global glucose metabolism outcomes. RESULTS: We show that short-term pharmacological FoxO1 inhibition can model beta-cell dedifferentiation by downregulating beta-cell-specific transcription factors, resulting in the aberrant expression of progenitor genes and the alpha-cell marker glucagon. From a high-content screen, we identified loperamide as a small molecule that can prevent FoxO inhibitor-induced glucagon expression and further stimulate insulin protein processing and secretion by altering calcium levels, intracellular pH, and FoxO1 localization. CONCLUSIONS: Our study provides novel models, molecular targets, and drug candidates for studying and preventing beta-cell dedifferentiation.


Subject(s)
Forkhead Box Protein O1/metabolism , Glucagon/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Adult , Animals , Cell Dedifferentiation , Cells, Cultured , Female , Humans , Male , Mice , Middle Aged
10.
Adv Sci (Weinh) ; 7(19): 2000963, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33042738

ABSTRACT

It has been reported that Cavin1 deficiency causes lipodystrophy in both humans and mice by affecting lipid metabolism. The ablation of Cavin1 in rodents also causes a significant deviation from Mendelian ratio at weaning in a background-dependent manner, suggesting the presence of undiscovered functions of Cavin1. In the current study, the results show that Cavin1 deficiency causes neonatal death in C57BL/6J mice by dampening the storage and mobilization of glycogen in the liver, which leads to lethal neonatal hypoglycemia. Further investigation by electron microscopy reveals that Cavin1 deficiency impairs the fenestration in liver sinusoidal endothelial cells (LSECs) and impacts the permeability of endothelial barrier in the liver. Mechanistically, Cavin1 deficiency inhibits the RhoA-Rho-associated protein kinase 2-LIM domain kinase-Cofilin signaling pathway and suppresses the dynamics of the cytoskeleton, and eventually causes the reduction of fenestrae in LSECs. In addition, the defect of fenestration in LSECs caused by Cavin1 deficiency can be rescued by treatment with the F-actin depolymerization reagent latrunculin A. In summary, the current study reveals a novel function of Cavin1 on fenestrae formation in LSECs and liver glycogen metabolism, which provide an explanation for the neonatal death of Cavin1 null mice and a potential mechanism for metabolic disorders in patients with Cavin1 mutation.

11.
Cell Metab ; 32(4): 665-675.e6, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32941798

ABSTRACT

The thiazolidinediones (TZDs) are ligands of PPARγ that improve insulin sensitivity, but their use is limited by significant side effects. Recently, we demonstrated a mechanism wherein TZDs improve insulin sensitivity distinct from receptor agonism and adipogenesis: reversal of obesity-linked phosphorylation of PPARγ at serine 273. However, the role of this modification hasn't been tested genetically. Here we demonstrate that mice encoding an allele of PPARγ that cannot be phosphorylated at S273 are protected from insulin resistance, without exhibiting differences in body weight or TZD-associated side effects. Indeed, hyperinsulinemic-euglycemic clamp experiments confirm insulin sensitivity. RNA-seq in these mice reveals reduced expression of Gdf3, a BMP family member. Ectopic expression of Gdf3 is sufficient to induce insulin resistance in lean, healthy mice. We find Gdf3 inhibits BMP signaling and insulin signaling in vitro. Together, these results highlight the diabetogenic role of PPARγ S273 phosphorylation and focus attention on a putative target, Gdf3.


Subject(s)
Growth Differentiation Factor 3/metabolism , Obesity/drug therapy , PPAR gamma/metabolism , Thiazolidinediones/pharmacology , Alleles , Animals , Cells, Cultured , Growth Differentiation Factor 3/genetics , Humans , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , PPAR gamma/genetics , Phosphorylation/drug effects
12.
J Nutr Biochem ; 80: 108377, 2020 06.
Article in English | MEDLINE | ID: mdl-32278117

ABSTRACT

Deteriorated nicotinamide adenine dinucleotide (NAD+)/sirtuins (SIRTs) metabolism in adipose tissue is implicated in diet-induced obesity, while calorie restriction (CR)-induced beneficial effects require sufficient NAD+ biosynthesis. Mechanistic links have not been defined. This study aims to identify changes of specific components of NAD+/SIRTs system in white adipose tissue (WAT) and brown adipose tissue (BAT) of mice upon energy imbalance, focusing on key enzymes in NAD+ salvage (Nampt, Nmnat1, Nrk1), clearance (Nnmt, Aox1, Cyp2e1) and consumption pathways (Sirt1, Sirt2, Sirt3, Sirt6, Parp1). Male C57BL/6J mice were fed ad libitum with the standard laboratory chow diet, high-fat diet (HFD) or 40% CR diet, respectively. The epididymal and inguinal WAT (eWAT and iWAT) and interscapular BAT (iBAT) were harvested for histological, NAD+ assay, gene and protein expression analysis after 16 weeks of dietary regimen. HFD decreased, while CR increased, the NAD+ and NADH levels in eWAT, iWAT and iBAT. NAD+ content negatively correlated with plasma cholesterol, TNF-α levels and calorie intake, while it positively correlated with plasma adiponectin level. The change trend of SIRT1 is quite the same as that of NAD+/NADH ratio. Nmnat1 gene is sensitive to energy imbalance in WAT but not in BAT. Nrk1 gene expression was decreased in eWAT and iWAT but increased in iBAT of HFD mice. Nnmt mRNA and protein abundance was increased in iWAT of HFD mice. Nampt, Cyp2e1 and Sirt3 were the most robust genes responding to energy imbalance. In summary, adipose tissue responds to long-term energy excess or shortage with depot-specific transcriptional activation or repression of NAD+/SIRTs metabolic components.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Caloric Restriction/methods , Diet, High-Fat/methods , NAD/metabolism , Sirtuins/metabolism , Adiponectin/blood , Animals , Cholesterol/blood , Energy Intake , Energy Metabolism/drug effects , Gene Expression , Male , Mice , Mice, Inbred C57BL , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Obesity/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Tumor Necrosis Factor-alpha/blood
13.
J Sport Health Sci ; 9(1): 53-73, 2020 01.
Article in English | MEDLINE | ID: mdl-31921481

ABSTRACT

Background: In recent years, much evidence has emerged to indicate that exercise can benefit people when performed properly. This review summarizes the exercise interventions used in studies involving mice as they are related to special diseases or physiological status. To further understand the effects of exercise interventions in treating or preventing diseases, it is important to establish a template for exercise interventions that can be used in future exercise-related studies. Methods: PubMed was used as the data resource for articles. To identify studies related to the effectiveness of exercise interventions for treating various diseases and organ functions in mice, we used the following search language: (exercise [Title] OR training [Title] OR physical activity [Title]) AND (mice [title/abstract] OR mouse [title/abstract] OR mus [title/abstract]). To limit the range of search results, we included 2 filters: one that limited publication dates to "in 10 years" and one that sorted the results as "best match". Then we grouped the commonly used exercise methods according to their similarities and differences. We then evaluated the effectiveness of the exercise interventions for their impact on diseases and organ functions in 8 different systems. Results: A total of 331 articles were included in the analysis procedure. The articles were then segmented into 8 systems for which the exercise interventions were used in targeting and treating disorders: motor system (60 studies), metabolic system (45 studies), cardio-cerebral vascular system (58 studies), nervous system (74 studies), immune system (32 studies), respiratory system (7 studies), digestive system (1 study), and the system related to the development of cancer (54 studies). The methods of exercise interventions mainly involved the use of treadmills, voluntary wheel-running, forced wheel-running, swimming, and resistance training. It was found that regardless of the specific exercise method used, most of them demonstrated positive effects on various systemic diseases and organ functions. Most diseases were remitted with exercise regardless of the exercise method used, although some diseases showed the best remission effects when a specific method was used. Conclusion: Our review strongly suggests that exercise intervention is a cornerstone in disease prevention and treatment in mice. Because exercise interventions in humans typically focus on chronic diseases, national fitness, and body weight loss, and typically have low intervention compliance rates, it is important to use mice models to investigate the molecular mechanisms underlying the health benefits from exercise interventions in humans.


Subject(s)
Animal Diseases/prevention & control , Models, Animal , Physical Conditioning, Animal , Animal Diseases/physiopathology , Animal Diseases/therapy , Animals , Bone Density , Mice , Mitochondria, Muscle/physiology , Muscle, Skeletal/blood supply , Muscle, Skeletal/physiology , Muscular Atrophy/prevention & control , Neovascularization, Physiologic , Osteoporosis/prevention & control , Physical Conditioning, Animal/methods , Sarcopenia/prevention & control
14.
J Ginseng Res ; 43(4): 580-588, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31695564

ABSTRACT

BACKGROUND: Ginsenoside Rg1 has been shown to clear senescence-associated beta-galactosidase (SA-ß-gal) in cultured cells. It remains unknown whether Rg1 can influence SA-ß-gal in exercising human skeletal muscle. METHODS: To examine SA-ß-gal change, 12 young men (age 21 ± 0.2 years) were enrolled in a randomized double-blind placebo controlled crossover study, under two occasions: placebo (PLA) and Rg1 (5 mg) supplementations 1 h prior to a high-intensity cycling (70% VO2max). Muscle samples were collected by multiple biopsies before and after cycling exercise (0 h and 3 h). To avoid potential effect of muscle biopsy on performance assessment, cycling time to exhaustion test (80% VO2max) was conducted on another 12 participants (age 23 ± 0.5 years) with the same experimental design. RESULTS: No changes of SA-ß-gal were observed after cycling in the PLA trial. On the contrary, nine of the 12 participants showed complete elimination of SA-ß-gal in exercised muscle after cycling in the Rg1 trial (p < 0.05). Increases in apoptotic DNA fragmentation (PLA: +87% vs. Rg1: +133%, p < 0.05) and CD68+ (PLA: +78% vs. Rg1: +121%, p = 0.17) occurred immediately after cycling in both trials. During the 3-h recovery, reverses in apoptotic nuclei content (PLA: +5% vs. Rg1: -32%, p < 0.01) and increases in inducible nitrate oxide synthase and interleukin 6 mRNA levels of exercised muscle were observed only in the Rg1 trial (p < 0.01). CONCLUSION: Rg1 supplementation effectively eliminates senescent cells in exercising human skeletal muscle and improves high-intensity endurance performance.

15.
Article in English | MEDLINE | ID: mdl-31440207

ABSTRACT

Hypoxic training has been reported to lower obesity morbidity without clear underlying mechanisms. This study investigates the effect of hypoxic training on metabolic changes, particularly, on liver metabolism of high fat diet (HFD)-induced obese mice. We compared the hypoxic training group with normoxic sedentary, normoxic training, and hypoxic sedentary groups. Body weight, fat mass, glucose tolerance and liver physiology were determined after 4 weeks intervention. In both normoxic training and hypoxic training groups, body weight was lower than the normoxic sedentary group, with less fat mass. Insulin sensitivity was improved after hypoxic training. Moreover, liver metabolomics revealed insights into the protective effect of hypoxic training on HFD-induced fatty liver. Taken together, these findings provide a molecular metabolic mechanism for hypoxic training.

16.
Dev Cell ; 48(2): 277-286.e6, 2019 01 28.
Article in English | MEDLINE | ID: mdl-30639055

ABSTRACT

Interactions between tumors and host tissues play essential roles in tumor-induced systemic wasting and cancer cachexia, including muscle wasting and lipid loss. However, the pathogenic molecular mechanisms of wasting are still poorly understood. Using a fly model of tumor-induced organ wasting, we observed aberrant MEK activation in both tumors and host tissues of flies bearing gut-yki3SA tumors. We found that host MEK activation results in muscle wasting and lipid loss, while tumor MEK activation is required for tumor growth. Strikingly, host MEK suppression alone is sufficient to abolish the wasting phenotypes without affecting tumor growth. We further uncovered that yki3SA tumors produce the vein (vn) ligand to trigger autonomous Egfr/MEK-induced tumor growth and produce the PDGF- and VEGF-related factor 1 (Pvf1) ligand to non-autonomously activate host Pvr/MEK signaling and wasting. Altogether, our results demonstrate the essential roles and molecular mechanisms of differential MEK activation in tumor-induced host wasting.


Subject(s)
Cachexia/metabolism , Ligands , MAP Kinase Signaling System/physiology , Signal Transduction/physiology , Animals , Cell Line, Tumor , ErbB Receptors/metabolism , Mice , Muscle, Skeletal/metabolism , Phosphorylation
17.
Biochemistry ; 57(40): 5775-5779, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30226369

ABSTRACT

Methyl donor balance is critical for epigenetic regulation in cells and is maintained by the so-called methionine cycle proteins that regenerate S-adenosylmethionine (SAM), the universal methyl donor, from homocysteine formed by the activity of methyltransferases. Nnmt is a liver enzyme that methylates nicotinamide, but its role in regulating methyl donor balance in the liver is unclear. In this study, we assessed the effect of altered Nnmt expression on various aspects of methyl donor metabolism in the liver. We found that Nnmt overexpression decreased SAM levels and the SAM/ S-adenosylhomocysteine (SAH) ratio both in vivo and in vitro. Nnmt knockdown did not change methyl donor balance in mouse primary hepatocytes but increased SAM levels and the SAM/SAH ratio when Gnmt, the dominantly expressed methyltransferase in liver, was simultaneously knocked down. Paradoxically, expression of enzymatically deficient Nnmt increased the SAM/SAH ratio, suggesting that Nnmt can regulate methyl donor balance independent of its methyltransferase activity. Proteomics analysis of Nnmt-interacting proteins in the liver identified Bhmt, Mat1a, and Ahcy, all components of the methionine cycle, and functional experiments showed that mutant Nnmt increased the level of remethylation of homocysteine to SAM. In summary, we show that the function of Nnmt in hepatic methyl donor balance is multifactorial. On one hand, Nnmt decreases methyl donor balance, consistent with its activity as a methyltransferase consuming methyl donors. On the other hand, by co-opting the enzymes of the methionine cycle, Nnmt aids the recycling of homocysteine to SAM for another round of methylation.


Subject(s)
Glycine N-Methyltransferase/metabolism , Hepatocytes/enzymology , Liver/enzymology , Nicotinamide N-Methyltransferase/metabolism , S-Adenosylmethionine/metabolism , Animals , Gene Knockdown Techniques , Glycine N-Methyltransferase/genetics , Hepatocytes/cytology , Mice , Nicotinamide N-Methyltransferase/genetics , S-Adenosylhomocysteine/metabolism
18.
Mol Metab ; 12: 25-38, 2018 06.
Article in English | MEDLINE | ID: mdl-29661693

ABSTRACT

OBJECTIVE: The inappropriate release of free fatty acids from obese adipose tissue stores has detrimental effects on metabolism, but key molecular mechanisms controlling FFA release from adipocytes remain undefined. Although obesity promotes systemic inflammation, we find activation of the inflammation-associated Mitogen Activated Protein kinase ERK occurs specifically in adipose tissues of obese mice, and provide evidence that adipocyte ERK activation may explain exaggerated adipose tissue lipolysis observed in obesity. METHODS AND RESULTS: We provide genetic and pharmacological evidence that inhibition of the MEK/ERK pathway in human adipose tissue, mice, and flies all effectively limit adipocyte lipolysis. In complementary findings, we show that genetic and obesity-mediated activation of ERK enhances lipolysis, whereas adipose tissue specific knock-out of ERK2, the exclusive ERK1/2 protein in adipocytes, dramatically impairs lipolysis in explanted mouse adipose tissue. In addition, acute inhibition of MEK/ERK signaling also decreases lipolysis in adipose tissue and improves insulin sensitivity in obese mice. Mice with decreased rates of adipose tissue lipolysis in vivo caused by either MEK or ATGL pharmacological inhibition were unable to liberate sufficient White Adipose Tissue (WAT) energy stores to fuel thermogenesis from brown fat during a cold temperature challenge. To identify a molecular mechanism controlling these actions, we performed unbiased phosphoproteomic analysis of obese adipose tissue at different time points following acute pharmacological MEK/ERK inhibition. MEK/ERK inhibition decreased levels of adrenergic signaling and caused de-phosphorylation of the ß3-adrenergic receptor (ß3AR) on serine 247. To define the functional implications of this phosphorylation, we showed that CRISPR/Cas9 engineered cells expressing wild type ß3AR exhibited ß3AR phosphorylation by ERK2 and enhanced lipolysis, but this was not seen when serine 247 of ß3AR was mutated to alanine. CONCLUSION: Taken together, these data suggest that ERK activation in adipocytes and subsequent phosphorylation of the ß3AR on S247 are critical regulatory steps in the enhanced adipocyte lipolysis of obesity.


Subject(s)
Adipocytes, White/metabolism , Lipolysis , MAP Kinase Signaling System , Obesity/metabolism , Receptors, Adrenergic, beta-3/metabolism , 3T3 Cells , Animals , Drosophila melanogaster , Humans , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Receptors, Adrenergic, beta-3/chemistry , Serine/metabolism
19.
Mol Metab ; 6(10): 1212-1225, 2017 10.
Article in English | MEDLINE | ID: mdl-29031721

ABSTRACT

OBJECTIVES: Understanding how loci identified by genome wide association studies (GWAS) contribute to pathogenesis requires new mechanistic insights. Variants within CDKAL1 are strongly linked to an increased risk of developing type 2 diabetes and obesity. Investigations in mouse models have focused on the function of Cdkal1 as a tRNALys modifier and downstream effects of Cdkal1 loss on pro-insulin translational fidelity in pancreatic ß-cells. However, Cdkal1 is broadly expressed in other metabolically relevant tissues, including adipose tissue. In addition, the Cdkal1 homolog Cdk5rap1 regulates mitochondrial protein translation and mitochondrial function in skeletal muscle. We tested whether adipocyte-specific Cdkal1 deletion alters systemic glucose homeostasis or adipose mitochondrial function independently of its effects on pro-insulin translation and insulin secretion. METHODS: We measured mRNA levels of type 2 diabetes GWAS genes, including Cdkal1, in adipose tissue from lean and obese mice. We then established a mouse model with adipocyte-specific Cdkal1 deletion. We examined the effects of adipose Cdkal1 deletion using indirect calorimetry on mice during a cold temperature challenge, as well as by measuring cellular and mitochondrial respiration in vitro. We also examined brown adipose tissue (BAT) mitochondrial morphology by electron microscopy. Utilizing co-immunoprecipitation followed by mass spectrometry, we performed interaction mapping to identify new CDKAL1 binding partners. Furthermore, we tested whether Cdkal1 loss in adipose tissue affects total protein levels or accurate Lys incorporation by tRNALys using quantitative mass spectrometry. RESULTS: We found that Cdkal1 mRNA levels are reduced in adipose tissue of obese mice. Using adipose-specific Cdkal1 KO mice (A-KO), we demonstrated that mitochondrial function is impaired in primary differentiated brown adipocytes and in isolated mitochondria from A-KO brown adipose tissue. A-KO mice displayed decreased energy expenditure during 4 °C cold challenge. Furthermore, mitochondrial morphology was highly abnormal in A-KO BAT. Surprisingly, we found that lysine codon representation was unchanged in Cdkal1 A-KO adipose tissue. We identified novel protein interactors of CDKAL1, including SLC25A4/ANT1, an inner mitochondrial membrane ADP/ATP translocator. ANT proteins can account for the UCP1-independent basal proton leak in BAT mitochondria. Cdkal1 A-KO mice had increased ANT1 protein levels in their white adipose tissue. CONCLUSIONS: Cdkal1 is necessary for normal mitochondrial morphology and function in adipose tissue. These results suggest that the type 2 diabetes susceptibility gene CDKAL1 has novel functions in regulating mitochondrial activity.


Subject(s)
Mitochondria/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Adiposity , Animals , Diabetes Mellitus, Type 2/genetics , Genetic Predisposition to Disease , Glucose/metabolism , Insulin/genetics , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Obesity/genetics , Obesity/metabolism , tRNA Methyltransferases
20.
FASEB J ; 31(10): 4482-4491, 2017 10.
Article in English | MEDLINE | ID: mdl-28646016

ABSTRACT

Subclinical hypothyroidism is known to be associated with increased serum cholesterol. Since thyroid-stimulating hormone (TSH) exerts an inductor effect on cholesterol biosynthesis, we aimed to investigate the relationship between TSH mRNA and cholesterol metabolism in human adipose tissue (AT). Cross-sectionally, AT TSH-ß (TSHB) mRNA was evaluated in 4 independent cohorts in association with serum total and LDL cholesterol, and AT lipidomics. Longitudinally, the effects of statins and of diet and exercise on AT TSHB mRNA were also examined. The bidirectional relationship between cholesterol and TSHB were studied in isolated human adipocytes. TSHB mRNA was consistently detected in AT from euthyroid subjects, and positively associated with serum total- and LDL-cholesterol, and with AT-specific cholesterol metabolism-associated lipids [arachidonoyl cholesteryl ester, C8-dihydroceramide, N-stearoyl-d-sphingosine, and GlcCer(18:0, 24:1)]. Reduction of cholesterol with statins and with diet and exercise interventions led to decreased TSHB mRNA in human AT, whereas excess cholesterol up-regulated TSHB mRNA in human adipocytes. In addition, recombinant human TSH α/ß administration resulted in increased HMGCR mRNA levels in human adipocytes. In mice, subcutaneous AT Tshb expression levels correlated directly with circulating cholesterol levels. In summary, current results provide novel evidence of TSHB as a paracrine factor that is modulated in parallel with cholesterol metabolism in human AT.-Moreno-Navarrete, J. M., Moreno, M., Ortega, F., Xifra, G., Hong, S., Asara, J. M., Serrano, J. C. E., Jové, M., Pissios, P., Blüher, M., Ricart, W., Portero-Otin, M., Fernández-Real, J. M. TSHB mRNA is linked to cholesterol metabolism in adipose tissue.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Lipid Metabolism/physiology , Lipids/blood , Thyrotropin, beta Subunit/genetics , Thyrotropin/metabolism , Animals , Cholesterol/metabolism , Humans , Hypothyroidism/metabolism , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...