Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Obesity (Silver Spring) ; 32(3): 593-602, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38410080

ABSTRACT

OBJECTIVE: The objective of this study was to examine the hypothesis that abdominal and gluteal adipocyte turnover, lipid dynamics, and fibrogenesis are dysregulated among insulin-resistant (IR) compared with insulin-sensitive (IS) adolescents with obesity. METHODS: Seven IS and seven IR adolescents with obesity participated in a 3-h oral glucose tolerance test and a multi-section magnetic resonance imaging scan of the abdominal region to examine body fat distribution patterns and liver fat content. An 8-week 70% deuterated water (2 H2 O) labeling protocol examined adipocyte turnover, lipid dynamics, and fibrogenesis in vivo from biopsied abdominal and gluteal fat. RESULTS: Abdominal and gluteal subcutaneous adipose tissue (SAT) turnover rates of lipid components were similar among IS and IR adolescents with obesity. However, the insoluble collagen (type I, subunit α2) isoform measured from abdominal, but not gluteal, SAT was elevated in IR compared with IS individuals. In addition, abdominal insoluble collagen Iα2 was associated with ratios of visceral-to-total (visceral adipose tissue + SAT) abdominal fat and whole-body and adipose tissue insulin signaling, and it trended toward a positive association with liver fat content. CONCLUSIONS: Altered extracellular matrix dynamics, but not expandability, potentially decreases abdominal SAT lipid storage capacity, contributing to the pathophysiological pathways linking adipose tissue and whole-body IR with altered ectopic storage of lipids within the liver among IR adolescents with obesity.


Subject(s)
Insulin Resistance , Pediatric Obesity , Child , Humans , Adolescent , Insulin Resistance/physiology , Pediatric Obesity/metabolism , Insulin/metabolism , Subcutaneous Fat/diagnostic imaging , Subcutaneous Fat/metabolism , Intra-Abdominal Fat/metabolism , Lipids , Extracellular Matrix , Collagen/metabolism
2.
Dev Cell ; 59(7): 830-840.e4, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38377991

ABSTRACT

Tissue repair requires a highly coordinated cellular response to injury. In the lung, alveolar type 2 cells (AT2s) act as stem cells to replenish both themselves and alveolar type 1 cells (AT1s); however, the complex orchestration of stem cell activity after injury is poorly understood. Here, we establish longitudinal imaging of AT2s in murine intact tissues ex vivo and in vivo in order to track their dynamic behavior over time. We discover that a large fraction of AT2s become motile following injury and provide direct evidence for their migration between alveolar units. High-resolution morphokinetic mapping of AT2s further uncovers the emergence of distinct motile phenotypes. Inhibition of AT2 migration via genetic depletion of ArpC3 leads to impaired regeneration of AT2s and AT1s in vivo. Together, our results establish a requirement for stem cell migration between alveolar units and identify properties of stem cell motility at high cellular resolution.


Subject(s)
Alveolar Epithelial Cells , Lung , Mice , Animals , Lung/physiology , Alveolar Epithelial Cells/metabolism , Stem Cells/metabolism , Cell Movement , Cell Differentiation/physiology
3.
Diabetes ; 2023 Oct 23.
Article in English | MEDLINE | ID: mdl-37870826

ABSTRACT

Excessive insulin secretion independent of insulin resistance, defined as primary hypersecretion, is associated with obesity and an unfavorable metabolic phenotype. We examined the characteristics of the adipose tissue in youths with primary insulin hypersecretion and the longitudinal metabolic alterations influenced by the complex adipo-insular interplay. In a multiethnic cohort of non-diabetic adolescents with obesity, primary insulin hypersecretors had enhanced model-derived ß-cell glucose sensitivity and rate sensitivity, but worse glucose tolerance, despite similar demographics, adiposity, and insulin resistance measured by both OGTT and euglycemic-hyperinsulinemic clamp. Hypersecretors had greater intrahepatic and visceral fat depots at abdominal MRI, hypertrophic abdominal subcutaneous adipocytes, higher FFA and leptin serum levels per fat mass, and faster in vivo lipid turnover assessed by a long-term 2H2O labeling protocol. At 2-year follow up, hypersecretors had greater fat accrual and 3-fold higher risk for abnormal glucose tolerance, while individuals with hypertrophic adipocytes or higher leptin levels showed enhanced ß-cell glucose sensitivity. Primary insulin hypersecretion is associated with marked alterations in adipose tissue distribution, cellularity, and lipid dynamics, independent of whole-body adiposity and insulin resistance. Pathogenetic insight into the metabolic crosstalk between ß-cell and adipocyte may help identify individuals at risk for chronic hyperinsulinemia, body weight gain, and glucose intolerance.

4.
J Transl Med ; 20(1): 127, 2022 03 14.
Article in English | MEDLINE | ID: mdl-35287685

ABSTRACT

90% of esophageal cancer are esophageal squamous cell carcinoma (ESCC) and ESCC has a very poor prognosis and high mortality. Nevertheless, the key metabolic pathways associated with ESCC progression haven't been revealed yet. Metabolomics has become a new platform for biomarker discovery over recent years. We aim to elucidate dominantly metabolic pathway in all ESCC tumor/node/metastasis (TNM) stages and adjacent cancerous tissues. We collected 60 postoperative esophageal tissues and 15 normal tissues adjacent to the tumor, then performed Liquid Chromatography with tandem mass spectrometry (LC-MS/MS) analyses. The metabolites data was analyzed with metabolites differential and correlational expression heatmap according to stage I vs. con., stage I vs. stage II, stage II vs. stage III, and stage III vs. stage IV respectively. Metabolic pathways were acquired by Kyoto Encyclopedia of Genes and Genomes. (KEGG) pathway database. The metabolic pathway related genes were obtained via Gene Set Enrichment Analysis (GSEA). mRNA expression of ESCC metabolic pathway genes was detected by two public datasets: gene expression data series (GSE)23400 and The Cancer Genome Atlas (TCGA). Receiver operating characteristic curve (ROC) analysis is applied to metabolic pathway genes. 712 metabolites were identified in total. Glycerophospholipid metabolism was significantly distinct in ESCC progression. 16 genes of 77 genes of glycerophospholipid metabolism mRNA expression has differential significance between ESCC and normal controls. Phosphatidylserine synthase 1 (PTDSS1) and Lysophosphatidylcholine Acyltransferase1 (LPCAT1) had a good diagnostic value with Area under the ROC Curve (AUC) > 0.9 using ROC analysis. In this study, we identified glycerophospholipid metabolism was associated with the ESCC tumorigenesis and progression. Glycerophospholipid metabolism could be a potential therapeutic target of ESCC progression.


Subject(s)
Carcinoma, Squamous Cell , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Carcinoma, Squamous Cell/pathology , Chromatography, Liquid , Epithelial Cells/pathology , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , Humans , Metabolomics , Tandem Mass Spectrometry
5.
Nat Commun ; 13(1): 494, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35078977

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a leading cause of death worldwide, however our understanding of cell specific mechanisms underlying COPD pathobiology remains incomplete. Here, we analyze single-cell RNA sequencing profiles of explanted lung tissue from subjects with advanced COPD or control lungs, and we validate findings using single-cell RNA sequencing of lungs from mice exposed to 10 months of cigarette smoke, RNA sequencing of isolated human alveolar epithelial cells, functional in vitro models, and in situ hybridization and immunostaining of human lung tissue samples. We identify a subpopulation of alveolar epithelial type II cells with transcriptional evidence for aberrant cellular metabolism and reduced cellular stress tolerance in COPD. Using transcriptomic network analyses, we predict capillary endothelial cells are inflamed in COPD, particularly through increased CXCL-motif chemokine signaling. Finally, we detect a high-metallothionein expressing macrophage subpopulation enriched in advanced COPD. Collectively, these findings highlight cell-specific mechanisms involved in the pathobiology of advanced COPD.


Subject(s)
Alveolar Epithelial Cells/metabolism , Lung/metabolism , Pulmonary Disease, Chronic Obstructive/genetics , RNA-Seq/methods , Single-Cell Analysis/methods , A549 Cells , Alveolar Epithelial Cells/classification , Animals , Cells, Cultured , Cluster Analysis , Epithelial Cells/metabolism , Female , Gene Expression Profiling/methods , Gene Regulatory Networks , Humans , Lung/cytology , Male , Mice, Inbred C57BL , Mice, Transgenic , Pulmonary Disease, Chronic Obstructive/pathology , Signal Transduction/genetics
6.
J Clin Invest ; 131(23)2021 12 01.
Article in English | MEDLINE | ID: mdl-34850744

ABSTRACT

Excessive inflammation drives the progression from sepsis to septic shock. Macrophage migration inhibitory factor (MIF) is of interest because MIF promoter polymorphisms predict mortality in different infections, and anti-MIF antibody improves survival in experimental models when administered 8 hours after infectious insult. The recent description of a second MIF superfamily member, D-dopachrome tautomerase (D-DT/MIF-2), prompted closer investigation of MIF-dependent responses. We subjected Mif-/- and Mif-2-/- mice to polymicrobial sepsis and observed a survival benefit with Mif but not Mif-2 deficiency. Survival was associated with reduced numbers of small peritoneal macrophages (SPMs) that, in contrast to large peritoneal macrophages (LPMs), were recruited into the peritoneal cavity. LPMs produced higher quantities of MIF than SPMs, but SPMs expressed higher levels of inflammatory cytokines and the MIF receptors CD74 and CXCR2. Adoptive transfer of WT SPMs into Mif-/- hosts reduced the protective effect of Mif deficiency in polymicrobial sepsis. Notably, MIF-2 lacks the pseudo-(E)LR motif present in MIF that mediates CXCR2 engagement and SPM migration, supporting a specific role for MIF in the recruitment and accumulation of inflammatory SPMs.


Subject(s)
Inflammation/metabolism , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Sepsis/metabolism , Sepsis/microbiology , Animals , Cytokines/metabolism , Disease Models, Animal , Female , Flow Cytometry , Gene Expression Profiling , Intramolecular Oxidoreductases/metabolism , Leukocyte Count , Macrophage Migration-Inhibitory Factors/metabolism , Macrophages/metabolism , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peritoneal Lavage , Phenotype , Protein Binding , RNA-Seq , Sepsis/physiopathology , Signal Transduction
7.
Obesity (Silver Spring) ; 29(12): 2068-2080, 2021 12.
Article in English | MEDLINE | ID: mdl-34672413

ABSTRACT

OBJECTIVE: This study investigated whether variations in cell death-inducing DNA fragmentation factor alpha subunit-like effector A (CIDEA) mRNA expression and protein levels are modulated by the pattern of abdominal fat distribution in adolescent girls with obesity. METHODS: This study recruited 35 adolescent girls with obesity and characterized their abdominal fat distribution by magnetic resonance imaging. Participants had only a periumbilical/abdominal (n = 14) or a paired abdominal and gluteal subcutaneous adipose tissue (SAT) biopsy (n = 21). CIDEA expression was determined by reverse transcription-polymerase chain reaction, CIDEA protein level by Western blot, and the turnover of adipose lipids and adipocytes by 2 H2 O labeling. In six girls, a second abdominal SAT biopsy was performed (after ~34.2 months) to explore the weight gain effect on CIDEA expression in abdominal SAT. RESULTS: CIDEA expression decreased in abdominal SAT from participants with high visceral adipose tissue (VAT)/(VAT+SAT); CIDEA inversely correlated with number of small adipocytes, with the increase in preadipocyte proliferation, and with adipogenesis. A strong inverse correlation was found between CIDEA protein level with the newly synthetized glycerol (r = -0.839, p = 0.0047). Following weight gain, an increase in adipocytes' cell diameter with a decrease in CIDEA expression and RNA-sequencing transcriptomic profile typical of adipocyte dysfunction was observed. CONCLUSIONS: Reduced expression of CIDEA in girls with high VAT/(VAT+SAT) is associated with adipocyte hypertrophy and insulin resistance.


Subject(s)
Apoptosis Regulatory Proteins , Obesity , Subcutaneous Fat , Abdominal Fat/metabolism , Adolescent , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Female , Humans , Intra-Abdominal Fat/metabolism , Obesity/metabolism , Subcutaneous Fat/metabolism , Subcutaneous Fat, Abdominal/metabolism
8.
JCI Insight ; 6(2)2021 01 25.
Article in English | MEDLINE | ID: mdl-33290275

ABSTRACT

The pathogenesis of chronic obstructive pulmonary disease (COPD) involves aberrant responses to cellular stress caused by chronic cigarette smoke (CS) exposure. However, not all smokers develop COPD and the critical mechanisms that regulate cellular stress responses to increase COPD susceptibility are not understood. Because microRNAs are well-known regulators of cellular stress responses, we evaluated microRNA expression arrays performed on distal parenchymal lung tissue samples from 172 subjects with and without COPD. We identified miR-24-3p as the microRNA that best correlated with radiographic emphysema and validated this finding in multiple cohorts. In a CS exposure mouse model, inhibition of miR-24-3p increased susceptibility to apoptosis, including alveolar type II epithelial cell apoptosis, and emphysema severity. In lung epithelial cells, miR-24-3p suppressed apoptosis through the BH3-only protein BIM and suppressed homology-directed DNA repair and the DNA repair protein BRCA1. Finally, we found BIM and BRCA1 were increased in COPD lung tissue, and BIM and BRCA1 expression inversely correlated with miR-24-3p. We concluded that miR-24-3p, a regulator of the cellular response to DNA damage, is decreased in COPD, and decreased miR-24-3p increases susceptibility to emphysema through increased BIM and apoptosis.


Subject(s)
Apoptosis/genetics , DNA Damage/genetics , MicroRNAs/genetics , Pulmonary Disease, Chronic Obstructive/genetics , Aged , Animals , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Bcl-2-Like Protein 11/genetics , Bcl-2-Like Protein 11/metabolism , Cell Line , Cigarette Smoking/adverse effects , Cohort Studies , DNA Repair , Disease Models, Animal , Disease Susceptibility , Female , Humans , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred AKR , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Middle Aged , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptome
9.
JCI Insight ; 5(6)2020 03 26.
Article in English | MEDLINE | ID: mdl-32125283

ABSTRACT

Severe obesity (SO) affects about 6% of youth in the United States, augmenting the risks for cardiovascular disease and type 2 diabetes. Herein, we obtained paired omental adipose tissue (omVAT) and abdominal subcutaneous adipose tissue (SAT) biopsies from girls with SO undergoing sleeve gastrectomy (SG), to test whether differences in cellular and transcriptomic profiles between omVAT and SAT depots affect insulin sensitivity differently. Following weight loss, these analyses were repeated in a subgroup of subjects having a second SAT biopsy. We found that omVAT displayed smaller adipocytes compared with SAT, increased lipolysis through adipose triglyceride lipase phosphorylation, reduced inflammation, and increased expression of browning/beiging markers. Contrary to omVAT, SAT adipocyte diameter correlated with insulin resistance. Following SG, both weight and insulin sensitivity improved markedly in all subjects. SAT adipocytes' size became smaller, showing increased lipolysis through perilipin 1 phosphorylation, decreased inflammation, and increased expression in browning/beiging markers. In summary, in adolescent girls with SO, both omVAT and SAT depots showed distinct cellular and transcriptomic profiles. Following weight loss, the SAT depot changed its cellular morphology and transcriptomic profiles into more favorable ones. These changes in the SAT depot may play a fundamental role in the resolution of insulin resistance.


Subject(s)
Lipolysis/physiology , Obesity, Morbid/metabolism , Obesity, Morbid/surgery , Omentum/metabolism , Subcutaneous Fat, Abdominal/metabolism , Adipose Tissue/metabolism , Adipose Tissue, Brown/metabolism , Adolescent , Female , Gastrectomy , Humans , Transcriptome , Young Adult
10.
Diabetes ; 68(6): 1168-1177, 2019 06.
Article in English | MEDLINE | ID: mdl-30936147

ABSTRACT

Patterns of abdominal fat distribution (for example, a high vs. low visceral adipose tissue [VAT]/[VAT + subcutaneous adipose tissue (SAT)] ratio), independent of obesity, during adolescence carry a high risk for insulin resistance and type 2 diabetes. Longitudinal follow-up of a cohort of obese adolescents has recently revealed that a high ratio (high VAT/[VAT + SAT]) is a major determinant of fatty liver and metabolic impairment over time, with these effects being more pronounced in girls than in boys. To unravel the underlying metabolic alterations associated with the unfavorable VAT/(VAT + SAT) phenotype, we used the 2H2O labeling method to measure the turnover of adipose lipids and cells in the subcutaneous abdominal and gluteal/femoral adipose tissue (SAT) of weight-stable obese adolescent girls with a similar level of obesity but discordant VAT/(VAT + SAT) ratios. Girls with the unfavorable (high VAT/[VAT + SAT]) phenotype exhibited higher in vivo rates of triglyceride (TG) turnover (representing both lipolysis and synthesis at steady state), without significant differences in de novo lipogenesis in both abdominal and gluteal depots, compared with obese girls with the favorable phenotype. Moreover, mature adipocytes had higher turnover, with no difference in stromal vascular cell proliferation in both depots in the metabolically unfavorable phenotype. The higher TG turnover rates were significantly correlated with higher intrahepatic fat stores. These findings are contrary to the hypothesis that impaired capacity to deposit TGs or proliferation of new mature adipocytes are potential mechanisms for ectopic fat distribution in this setting. In summary, these results suggest that increased turnover of TGs (lipolysis) and of mature adipocytes in both abdominal and gluteal SAT may contribute to metabolic impairment and the development of fatty liver, even at this very early stage of disease.


Subject(s)
Adipocytes/metabolism , Body Fat Distribution , Obesity/metabolism , Subcutaneous Fat/metabolism , Triglycerides/metabolism , Absorptiometry, Photon , Adolescent , Deuterium Oxide , Female , Humans , Intra-Abdominal Fat/diagnostic imaging , Lipid Metabolism , Lipogenesis , Magnetic Resonance Imaging , Obesity, Abdominal/diagnostic imaging , Obesity, Abdominal/metabolism , Subcutaneous Fat/diagnostic imaging , Young Adult
11.
Int J Obes (Lond) ; 43(4): 673-682, 2019 04.
Article in English | MEDLINE | ID: mdl-30337653

ABSTRACT

BACKGROUND: The relative proportion of visceral fat (VAT) to subcutaneous fat (SAT) has been described as a major determinant of insulin resistance (IR). Our study sought to evaluate the effect of body fat distribution on glucose metabolism and intrahepatic fat content over time in a multiethnic cohort of obese adolescents. SUBJECTS/METHODS: We examined markers of glucose metabolism by oral glucose tolerance test, and body fat distribution by abdominal MRI at baseline and after 19.2 ± 11.4 months in a cohort of 151 obese adolescents (88 girls, 63 boys; mean age 13.3 ± 3.4 years; mean BMI z-score 2.15 ± 0.70). Hepatic fat content was assessed by fast-gradient MRI in a subset of 93 subjects. We used the median value of VAT/(VAT + SAT) ratio within each gender at baseline to stratify our sample into high and low ratio groups (median value 0.0972 in girls and 0.118 in boys). RESULTS: Female subjects tended to remain in their VAT/(VAT + SAT) category over time (change over follow-up P = 0.14 among girls, and P = 0.04 among boys). Baseline VAT/(VAT + SAT) strongly predicted the hepatic fat content, fasting insulin, 2-h glucose, and whole-body insulin sensitivity index at follow-up among girls, but not in boys. CONCLUSIONS: The VAT/(VAT + SAT) ratio is a major determinant of impaired glucose metabolism and hepatic fat accumulation over time, and its effects are more pronounced in girls than in boys.


Subject(s)
Fatty Liver/prevention & control , Insulin Resistance/physiology , Intra-Abdominal Fat/physiology , Pediatric Obesity/physiopathology , Subcutaneous Fat/physiology , Adolescent , Body Fat Distribution , Female , Glucose Tolerance Test , Humans , Intra-Abdominal Fat/metabolism , Longitudinal Studies , Male , Pediatric Obesity/metabolism , Protective Factors , Subcutaneous Fat/metabolism
12.
Hepatology ; 68(4): 1376-1390, 2018 10.
Article in English | MEDLINE | ID: mdl-29665034

ABSTRACT

We conducted a prospective study in a large, multiethnic cohort of obese adolescents to characterize clinical and genetic features associated with pediatric nonalcoholic fatty liver (NAFL), the most common cause of chronic liver disease in youth. A total of 503 obese adolescents were enrolled, including 191 (38.0%) whites, 134 (26.6%) blacks, and 178 (35.4%) Hispanics. Participants underwent abdominal magnetic resonance imaging (MRI) to quantify hepatic fat fraction (HFF), an oral glucose tolerance test (OGTT) to assess glucose tolerance and insulin sensitivity, and the genotyping of three single-nucleotide polymorphisms (SNPs) associated with nonalcoholic fatty liver disease (NAFLD) (patatin-like phospholipase domain-containing protein 3 [PNPLA3] rs738409, glucokinase regulatory protein [GCKR] rs1260326, and transmembrane 6 superfamily member 2 [TM6SF2] rs58542926). Assessments were repeated in 133 subjects after a 2-year follow-up. Prevalence of nonalcoholic fatty liver (NAFL) was 41.6% (209 patients) and ranged widely among ethnicities, being 42.9% in whites, 15.7% in blacks, and 59.6% in Hispanics (P < 0.0001). Among adolescents with NAFL, blacks showed the highest prevalence of altered glucose homeostasis (66%; P = 0.0003). Risk factors for NAFL incidence were white or Hispanic ethnicity (P = 0.021), high fasting C-peptide levels (P = 0.0006), and weight gain (P = 0.0006), whereas baseline HFF (P = 0.004) and weight loss (P = 0.032) predicted resolution of NAFL at follow-up. Adding either gene variant to these variables improved significantly the model predictive performance. CONCLUSION: Black obese adolescents are relatively protected from liver steatosis, but are more susceptible to the deleterious effects of NAFL on glucose metabolism. The combination of ethnicity/race with markers of insulin resistance and genetic factors might help identify obese youth at risk for developing NAFL.


Subject(s)
Gene Expression Regulation , Insulin Resistance/ethnology , Non-alcoholic Fatty Liver Disease/ethnology , Non-alcoholic Fatty Liver Disease/pathology , Pediatric Obesity/ethnology , Pediatric Obesity/genetics , Adaptor Proteins, Signal Transducing/genetics , Adolescent , Biopsy, Needle , Body Mass Index , Cross-Sectional Studies , Female , Glucose Tolerance Test , Humans , Immunohistochemistry , Insulin Resistance/physiology , Magnetic Resonance Imaging/methods , Male , Membrane Proteins/genetics , Non-alcoholic Fatty Liver Disease/genetics , Pediatric Obesity/pathology , Polymerase Chain Reaction/methods , Polymorphism, Single Nucleotide , Prognosis , Prospective Studies , ROC Curve
13.
Diabetes ; 65(3): 610-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26718495

ABSTRACT

The innate immune cell sensor leucine-rich-containing family, pyrin domain containing 3 (NLRP3) inflammasome controls the activation of caspase-1, and the release of proinflammatory cytokines interleukin (IL)-1ß and IL-18. The NLRP3 inflammasome is implicated in adipose tissue inflammation and the pathogenesis of insulin resistance. Herein, we tested the hypothesis that adipose tissue inflammation and NLRP3 inflammasome are linked to the downregulation of subcutaneous adipose tissue (SAT) adipogenesis/lipogenesis in obese adolescents with altered abdominal fat partitioning. We performed abdominal SAT biopsies on 58 obese adolescents and grouped them by MRI-derived visceral fat to visceral adipose tissue (VAT) plus SAT (VAT/VAT+SAT) ratio (cutoff 0.11). Adolescents with a high VAT/VAT+SAT ratio showed higher SAT macrophage infiltration and higher expression of the NLRP3 inflammasome-related genes (i.e., TLR4, NLRP3, IL1B, and CASP1). The increase in inflammation markers was paralleled by a decrease in genes related to insulin sensitivity (ADIPOQ, GLUT4, PPARG2, and SIRT1) and lipogenesis (SREBP1c, ACC, LPL, and FASN). Furthermore, SAT ceramide concentrations correlated with the expression of CASP1 and IL1B. Infiltration of macrophages and upregulation of the NLRP3 inflammasome together with the associated high ceramide content in the plasma and SAT of obese adolescents with a high VAT/VAT+SAT may contribute to the limited expansion of the subcutaneous abdominal adipose depot and the development of insulin resistance.


Subject(s)
Adipogenesis/genetics , Carrier Proteins/genetics , Intra-Abdominal Fat/metabolism , Lipogenesis/genetics , Obesity/metabolism , Subcutaneous Fat/metabolism , Abdomen , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/metabolism , Adiponectin/genetics , Adiponectin/metabolism , Adolescent , Carrier Proteins/immunology , Caspase 1/genetics , Caspase 1/metabolism , Child , Down-Regulation , Fatty Acid Synthase, Type I/genetics , Fatty Acid Synthase, Type I/metabolism , Female , Gene Expression Profiling , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Humans , Inflammasomes , Insulin Resistance , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Intra-Abdominal Fat/pathology , Leptin/metabolism , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Macrophages/immunology , Magnetic Resonance Imaging , Male , NLR Family, Pyrin Domain-Containing 3 Protein , Obesity/immunology , PPAR gamma/genetics , PPAR gamma/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Subcutaneous Fat/immunology , Subcutaneous Fat/pathology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
14.
J Biol Chem ; 291(2): 989-97, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26586915

ABSTRACT

To translate the 13 mtDNA-encoded mRNAs involved in oxidative phosphorylation (OXPHOS), mammalian mitochondria contain a dedicated set of ribosomes comprising rRNAs encoded by the mitochondrial genome and mitochondrial ribosomal proteins (MRPs) that are encoded by nuclear genes and imported into the matrix. In addition to their role in the ribosome, several MRPs have auxiliary functions or have been implicated in other cellular processes like cell cycle regulation and apoptosis. For example, we have shown that human MRPL12 binds and activates mitochondrial RNA polymerase (POLRMT), and hence has distinct functions in the ribosome and mtDNA transcription. Here we provide concrete evidence that there are two mature forms of mammalian MRPL12 that are generated by a two-step cleavage during import, involving efficient cleavage by mitochondrial processing protease and a second inefficient or regulated cleavage by mitochondrial intermediate protease. We also show that knock-down of MRPL12 by RNAi results in instability of POLRMT, but not other primary mitochondrial transcription components, and a corresponding decrease in mitochondrial transcription rates. Knock-down of MRPL10, the binding partner of MRPL12 in the ribosome, results in selective degradation of the mature long form of MRPL12, but has no effect on POLRMT. We propose that the two forms of MRPL12 are involved in homeostatic regulation of mitochondrial transcription and ribosome biogenesis that likely contribute to cell cycle, growth regulation, and longevity pathways to which MRPL12 has been linked.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Mitochondrial Proteins/metabolism , Proteolysis , Ribosomal Proteins/metabolism , Alternative Splicing/genetics , Amino Acid Sequence , Animals , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Mice , Mitochondria/metabolism , Molecular Sequence Data , Protein Isoforms/metabolism , Protein Stability , Protein Transport , Ribosomal Proteins/chemistry , Ribosomes/metabolism , Transcription, Genetic
15.
Am J Pathol ; 185(12): 3132-40, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26552864

ABSTRACT

The A1555G mutation in the 12S rRNA gene of human mitochondrial DNA causes maternally inherited, nonsyndromic deafness, an extreme case of tissue-specific mitochondrial pathology. A transgenic mouse strain that robustly overexpresses the mitochondrial 12S ribosomal RNA methyltransferase TFB1M (Tg-mtTFB1 mice) exhibits progressive hearing loss that we proposed models aspects of A1555G-related pathology in humans. Although our previous studies of Tg-mtTFB1 mice implicated apoptosis in the spiral ganglion and stria vascularis because of mitochondrial reactive oxygen species-mediated activation of AMP kinase (AMPK) and the nuclear transcription factor E2F1, detailed auditory pathology was not delineated. Herein, we show that Tg-mtTFB1 mice have reduced endocochlear potential, indicative of significant stria vascularis dysfunction, but without obvious signs of strial atrophy. We also observed decreased auditory brainstem response peak 1 amplitude and prolonged wave I latency, consistent with apoptosis of spiral ganglion neurons. Although no major loss of hair cells was observed, there was a mild impairment of voltage-dependent electromotility of outer hair cells. On the basis of these results, we propose that these events conspire to produce the progressive hearing loss phenotype in Tg-mtTFB1 mice. Finally, genetically reducing AMPK α1 rescues hearing loss in Tg-mtTFB1 mice, confirming that aberrant up-regulation of AMPK signaling promotes the observed auditory pathology. The relevance of these findings to human A1555G patients and the potential therapeutic value of reducing AMPK activity are discussed.


Subject(s)
Deafness/pathology , Mitochondrial Diseases/pathology , AMP-Activated Protein Kinases/deficiency , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/physiology , Animals , Apoptosis/physiology , DNA, Mitochondrial/genetics , Deafness/genetics , Deafness/physiopathology , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/physiology , Hair Cells, Auditory, Inner/pathology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitochondrial Diseases/genetics , Mitochondrial Diseases/physiopathology , Mutation , Organ of Corti/pathology , Reaction Time/physiology , Signal Transduction/physiology , Spiral Ganglion/pathology , Spiral Ganglion/physiopathology , Stria Vascularis/pathology , Transcription Factors/genetics
16.
Cell ; 158(3): 477-8, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-25083863

ABSTRACT

Regulation of gene expression in mammalian mitochondria by microRNAs is reported by Zhang et al. During muscle cell differentiation, localization of a miRNA is increased within mitochondria, where it interacts with Ago2 to selectively activate translation of mtDNA-encoded mRNAs. The findings represent a new mitochondrial regulatory pathway and a potentially powerful means to purposefully manipulate mtDNA expression.


Subject(s)
Cell Differentiation , MicroRNAs/metabolism , Mitochondria/metabolism , Myoblasts/metabolism , Myocytes, Cardiac/metabolism , Protein Biosynthesis , Animals
17.
Hum Mol Genet ; 23(5): 1311-9, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24158852

ABSTRACT

Oxidative phosphorylation and fatty acid oxidation are two major metabolic pathways in mitochondria. Acyl-CoA dehydrogenase 9 (ACAD9), an enzyme assumed to play a role in fatty acid oxidation, was recently identified as a factor involved in complex I biogenesis. Here we further investigated the role of ACAD9's enzymatic activity in fatty acid oxidation and complex I biogenesis. We provide evidence indicating that ACAD9 displays enzyme activity in vivo. Knockdown experiments in very-long-chain acyl-CoA dehydrogenase (VLCAD)-deficient fibroblasts revealed that ACAD9 is responsible for the production of C14:1-carnitine from oleate and C12-carnitine from palmitate. These results explain the origin of these obscure acylcarnitines that are used to diagnose VLCAD deficiency in humans. Knockdown of ACAD9 in control fibroblasts did not reveal changes in the acylcarnitine profiles upon fatty acid loading. Next, we investigated whether catalytic activity of ACAD9 was necessary for complex I biogenesis. Catalytically inactive ACAD9 gave partial-to-complete rescue of complex I biogenesis in ACAD9-deficient cells and was incorporated in high-molecular-weight assembly intermediates. Our results underscore the importance of the ACAD9 protein in complex I assembly and suggest that the enzymatic activity is a rudiment of the duplication event.


Subject(s)
Acyl-CoA Dehydrogenases/metabolism , Fatty Acids/metabolism , Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Acyl-CoA Dehydrogenases/chemistry , Acyl-CoA Dehydrogenases/deficiency , Acyl-CoA Dehydrogenases/genetics , Carnitine/biosynthesis , Catalysis , Cell Line , Congenital Bone Marrow Failure Syndromes , Electron Transport Complex I/deficiency , Enzyme Activation , Humans , Lipid Metabolism, Inborn Errors/metabolism , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Models, Molecular , Molecular Weight , Muscular Diseases/metabolism , Mutation , Oxidation-Reduction , Oxidative Phosphorylation , Protein Conformation
18.
JIMD Rep ; 12: 37-45, 2014.
Article in English | MEDLINE | ID: mdl-23996478

ABSTRACT

Here we report a patient with a new pathogenic mutation in ACAD9. Shortly after birth she presented with respiratory insufficiency and a high lactate level. At age 7 weeks, she was diagnosed with severe hypertrophic cardiomyopathy and she suffered from muscle weakness and hypotonia. Her condition deteriorated during intercurrent illnesses and she died at 6 months of age in cardiogenic shock. Analysis of respiratory chain activities in muscle and fibroblasts revealed an isolated complex I deficiency. A genome-wide screen for homozygosity revealed several homozygous regions. Four candidate genes were found and sequencing revealed a homozygous missense mutation in ACAD9. The mutation results in an Ala220Val amino acid substitution located near the catalytic core of ACAD9. SDS and BN-PAGE analysis showed severely decreased ACAD9 and complex I protein levels, and lentiviral complementation of patient fibroblasts partially rescued the complex I deficiency. Riboflavin supplementation did not ameliorate the complex I deficiency in patient fibroblasts. More than a dozen ACAD9 patients with complex I deficiency have been identified in the last 3 years, indicating that ACAD9 is important for complex I assembly, and that ACAD9 mutations are a relatively frequent cause of complex I deficiency.

19.
J Inherit Metab Dis ; 36(6): 913-21, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23355087

ABSTRACT

Elevated urinary excretion of 3-methylglutaconic acid is considered rare in patients suspected of a metabolic disorder. In 3-methylglutaconyl-CoA hydratase deficiency (mutations in AUH), it derives from leucine degradation. In all other disorders with 3-methylglutaconic aciduria the origin is unknown, yet mitochondrial dysfunction is thought to be the common denominator. We investigate the biochemical, clinical and genetic data of 388 patients referred to our centre under suspicion of a metabolic disorder showing 3-methylglutaconic aciduria in routine metabolic screening. Furthermore, we investigate 591 patients with 50 different, genetically proven, mitochondrial disorders for the presence of 3-methylglutaconic aciduria. Three percent of all urine samples of the patients referred showed 3-methylglutaconic aciduria, often in correlation with disorders not reported earlier in association with 3-methylglutaconic aciduria (e.g. organic acidurias, urea cycle disorders, haematological and neuromuscular disorders). In the patient cohort with genetically proven mitochondrial disorders 11% presented 3-methylglutaconic aciduria. It was more frequently seen in ATPase related disorders, with mitochondrial DNA depletion or deletion, but not in patients with single respiratory chain complex deficiencies. Besides, it was a consistent feature of patients with mutations in TAZ, SERAC1, OPA3, DNAJC19 and TMEM70 accounting for mitochondrial membrane related pathology. 3-methylglutaconic aciduria is found quite frequently in patients suspected of a metabolic disorder, and mitochondrial dysfunction is indeed a common denominator. It is only a discriminative feature of patients with mutations in AUH, TAZ, SERAC1, OPA3, DNAJC19 TMEM70. These conditions should therefore be referred to as inborn errors of metabolism with 3-methylglutaconic aciduria as discriminative feature.


Subject(s)
Glutarates/urine , Metabolism, Inborn Errors/diagnosis , Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/classification , Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/epidemiology , Amino Acid Metabolism, Inborn Errors/genetics , DNA Mutational Analysis , Diagnosis, Differential , Humans , Metabolism, Inborn Errors/epidemiology , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/epidemiology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/urine , Netherlands/epidemiology , Retrospective Studies , Urinalysis/methods
20.
Brain ; 135(Pt 1): 12-22, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22036961

ABSTRACT

Complex I deficiency is the most frequent cause of oxidative phosphorylation disorders. The disease features a large diversity of clinical symptoms often leading to progressive encephalomyopathies with a fatal outcome. There is currently no cure, and although disease-causing mutations have been found in the genes encoding complex I subunits, half of the cases remain unexplained. However, in the past 5 years a new class of complex I disease genes has emerged with the finding of specific assembly factors. So far nine such genes have been described and it is believed that in the near future more will be found. In this review, we will address whether the functions of these chaperones point towards a general molecular mechanism of disease and whether this enables us to design a treatment for complex I deficiency.


Subject(s)
DNA, Mitochondrial/genetics , Mitochondrial Diseases/genetics , Molecular Chaperones/genetics , Electron Transport Complex I/deficiency , Electron Transport Complex I/genetics , Humans , Mitochondrial Diseases/therapy , Oxidative Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL
...