Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Redox Biol ; 54: 102353, 2022 08.
Article in English | MEDLINE | ID: mdl-35777200

ABSTRACT

Metabolic plasticity is the ability of a biological system to adapt its metabolic phenotype to different environmental stressors. We used a whole-body and tissue-specific phenotypic, functional, proteomic, metabolomic and transcriptomic approach to systematically assess metabolic plasticity in diet-induced obese mice after a combined nutritional and exercise intervention. Although most obesity and overnutrition-related pathological features were successfully reverted, we observed a high degree of metabolic dysfunction in visceral white adipose tissue, characterized by abnormal mitochondrial morphology and functionality. Despite two sequential therapeutic interventions and an apparent global healthy phenotype, obesity triggered a cascade of events in visceral adipose tissue progressing from mitochondrial metabolic and proteostatic alterations to widespread cellular stress, which compromises its biosynthetic and recycling capacity. In humans, weight loss after bariatric surgery showed a transcriptional signature in visceral adipose tissue similar to our mouse model of obesity reversion. Overall, our data indicate that obesity prompts a lasting metabolic fingerprint that leads to a progressive breakdown of metabolic plasticity in visceral adipose tissue.


Subject(s)
Insulin Resistance , Adipose Tissue/metabolism , Animals , Homeostasis , Intra-Abdominal Fat/metabolism , Mice , Obesity/genetics , Obesity/metabolism , Proteomics
2.
Elife ; 112022 05 31.
Article in English | MEDLINE | ID: mdl-35638262

ABSTRACT

Exercise is an effective strategy in the prevention and treatment of metabolic diseases. Alterations in the skeletal muscle proteome, including post-translational modifications, regulate its metabolic adaptations to exercise. Here, we examined the effect of high-intensity interval training (HIIT) on the proteome and acetylome of human skeletal muscle, revealing the response of 3168 proteins and 1263 lysine acetyl-sites on 464 acetylated proteins. We identified global protein adaptations to exercise training involved in metabolism, excitation-contraction coupling, and myofibrillar calcium sensitivity. Furthermore, HIIT increased the acetylation of mitochondrial proteins, particularly those of complex V. We also highlight the regulation of exercise-responsive histone acetyl-sites. These data demonstrate the plasticity of the skeletal muscle proteome and acetylome, providing insight into the regulation of contractile, metabolic and transcriptional processes within skeletal muscle. Herein, we provide a substantial hypothesis-generating resource to stimulate further mechanistic research investigating how exercise improves metabolic health.


Subject(s)
High-Intensity Interval Training , Adaptation, Physiological/physiology , Humans , Muscle Contraction/physiology , Muscle, Skeletal/metabolism , Proteome/metabolism
3.
iScience ; 25(2): 103836, 2022 Feb 18.
Article in English | MEDLINE | ID: mdl-35198892

ABSTRACT

Proteomics analysis of skeletal muscle has recently progressed from whole muscle tissue to single myofibers. Here, we further focus on a specific myofiber domain crucial for force transmission from muscle to tendon, the myotendinous junction (MTJ). To overcome the anatomical constraints preventing the isolation of pure MTJs, we performed in-depth analysis of the MTJ by progressive removal of the muscle component in semitendinosus muscle-tendon samples. Using detergents with increasing stringency, we quantified >3000 proteins across all samples, and identified 112 significantly enriched MTJ proteins, including 24 known MTJ-enriched proteins. Of the 88 novel MTJ markers, immunofluorescence analysis confirmed the presence of tetraspanin-24 (CD151), kindlin-2 (FERMT2), cartilage intermediate layer protein 1 (CILP), and integrin-alpha10 (ITGA10), at the human MTJ. Together, these human data constitute the first detailed MTJ proteomics resource that will contribute to advance understanding of the biology of the MTJ and its failure in pathological conditions.

4.
STAR Protoc ; 3(1): 101135, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35128478

ABSTRACT

The assembly of mitochondrial respiratory complexes into supercomplexes has significant implications for mitochondrial function. This protocol details mitochondrial isolation from mouse tissues and the use of blue native gel electrophoresis (BN-PAGE) to separate pre-identified mitochondrial supercomplexes into different gel bands. We then describe the excision of the individual bands, followed by in-gel protein digestion and peptide desalting for mass spectrometry (MS)-based proteomics. This protocol provides a time-efficient measurement of the abundance and distribution of proteins within known supercomplexes. For complete details on the use and execution of this profile, please refer to Gonzalez-Franquesa et al. (2021).


Subject(s)
Mitochondria , Proteomics , Animals , Electrophoresis , Mass Spectrometry , Mice , Mitochondria/chemistry , Proteomics/methods
5.
Mol Cell Proteomics ; 21(3): 100207, 2022 03.
Article in English | MEDLINE | ID: mdl-35093608

ABSTRACT

Obesity leads to the development of nonalcoholic fatty liver disease (NAFLD) and associated alterations to the plasma proteome. To elucidate the underlying changes associated with obesity, we performed liquid chromatography-tandem mass spectrometry in the liver and plasma of obese leptin-deficient ob/ob mice and integrated these data with publicly available transcriptomic and proteomic datasets of obesity and metabolic diseases in preclinical and clinical cohorts. We quantified 7173 and 555 proteins in the liver and plasma proteomes, respectively. The abundance of proteins related to fatty acid metabolism were increased, alongside peroxisomal proliferation in ob/ob liver. Putatively secreted proteins and the secretory machinery were also dysregulated in the liver, which was mirrored by a substantial alteration of the plasma proteome. Greater than 50% of the plasma proteins were differentially regulated, including NAFLD biomarkers, lipoproteins, the 20S proteasome, and the complement and coagulation cascades of the immune system. Integration of the liver and plasma proteomes identified proteins that were concomitantly regulated in the liver and plasma in obesity, suggesting that the systemic abundance of these plasma proteins is regulated by secretion from the liver. Many of these proteins are systemically regulated during type 2 diabetes and/or NAFLD in humans, indicating the clinical importance of liver-plasma cross talk and the relevance of our investigations in ob/ob mice. Together, these analyses yield a comprehensive insight into obesity and provide an extensive resource for obesity research in a prevailing model organism.


Subject(s)
Diabetes Mellitus, Type 2 , Non-alcoholic Fatty Liver Disease , Animals , Diabetes Mellitus, Type 2/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Non-alcoholic Fatty Liver Disease/metabolism , Proteomics
6.
Am J Physiol Cell Physiol ; 321(5): C770-C778, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34495765

ABSTRACT

Skeletal muscle is an endocrine organ secreting exercise-induced factors (exerkines), which play a pivotal role in interorgan cross talk. Using mass spectrometry (MS)-based proteomics, we characterized the secretome and identified thymosin ß4 (TMSB4X) as the most upregulated secreted protein in the media of contracting C2C12 myotubes. TMSB4X was also acutely increased in the plasma of exercising humans irrespective of the insulin resistance condition or exercise mode. Treatment of mice with TMSB4X did not ameliorate the metabolic disruptions associated with diet induced-obesity, nor did it enhance muscle regeneration in vivo. However, TMSB4X increased osteoblast proliferation and neurite outgrowth, consistent with its WADA classification as a prohibited growth factor. Therefore, we report TMSB4X as a human exerkine with a potential role in cellular cross talk.


Subject(s)
Cell Proliferation/drug effects , Muscle Contraction , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Neuronal Outgrowth/drug effects , Osteoblasts/drug effects , Thymosin/metabolism , Thymosin/pharmacology , Animals , Case-Control Studies , Cell Line, Tumor , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Humans , Insulin Resistance , Male , Mice, Inbred C57BL , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Diseases/metabolism , Muscular Diseases/pathology , Muscular Diseases/physiopathology , Osteoblasts/pathology , Physical Endurance , Proteomics , Signal Transduction , Tandem Mass Spectrometry
7.
Proteomes ; 9(3)2021 08 03.
Article in English | MEDLINE | ID: mdl-34449730

ABSTRACT

Skeletal muscle is a major contributor to whole-body glucose homeostasis and is an important endocrine organ. To date, few studies have undertaken the large-scale identification of skeletal muscle-derived secreted proteins (myokines), particularly in response to stimuli that activate pathways governing energy metabolism in health and disease. Whereas the AMP-activated protein kinase (AMPK) and insulin-signaling pathways have received notable attention for their ability to independently regulate skeletal muscle substrate metabolism, little work has examined their ability to re-pattern the secretome. The present study coupled the use of high-resolution MS-based proteomics and bioinformatics analysis of conditioned media derived from 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR-an AMPK activator)- and insulin-treated differentiated C2C12 myotubes. We quantified 858 secreted proteins, including cytokines and growth factors such as fibroblast growth factor-21 (Fgf21). We identified 377 and 118 proteins that were significantly altered by insulin and AICAR treatment, respectively. Notably, the family of insulin growth factor binding-proteins (Igfbp) was differentially regulated by each treatment. Insulin- but not AICAR-induced conditioned media increased the mitochondrial respiratory capacity of myotubes, potentially via secreted factors. These findings may serve as an important resource to elucidate secondary metabolic effects of insulin and AICAR stimulation in skeletal muscle.

8.
Cell Rep ; 35(8): 109180, 2021 05 25.
Article in English | MEDLINE | ID: mdl-34038727

ABSTRACT

Mitochondrial respiratory complex subunits assemble in supercomplexes. Studies of supercomplexes have typically relied upon antibody-based quantification, often limited to a single subunit per respiratory complex. To provide a deeper insight into mitochondrial and supercomplex plasticity, we combine native electrophoresis and mass spectrometry to determine the supercomplexome of skeletal muscle from sedentary and exercise-trained mice. We quantify 422 mitochondrial proteins within 10 supercomplex bands in which we show the debated presence of complexes II and V. Exercise-induced mitochondrial biogenesis results in non-stoichiometric changes in subunits and incorporation into supercomplexes. We uncover the dynamics of supercomplex-related assembly proteins and mtDNA-encoded subunits after exercise. Furthermore, exercise affects the complexing of Lactb, an obesity-associated mitochondrial protein, and ubiquinone biosynthesis proteins. Knockdown of ubiquinone biosynthesis proteins leads to alterations in mitochondrial respiration. Our approach can be applied to broad biological systems. In this instance, comprehensively analyzing respiratory supercomplexes illuminates previously undetectable complexity in mitochondrial plasticity.


Subject(s)
Mass Spectrometry/methods , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Proteomics/methods , Animals , Female , Humans , Mice , Oxidative Phosphorylation
9.
Int J Mol Sci ; 22(7)2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33916315

ABSTRACT

Lipids are highly diverse in their composition, properties and distribution in different biological entities. We aim to establish the lipidomes of several insulin-sensitive tissues and to test their plasticity when divergent feeding regimens and lifestyles are imposed. Here, we report a proton nuclear magnetic resonance (1H-NMR) study of lipid abundance across 4 tissues of C57Bl6J male mice that includes the changes in the lipid profile after every lifestyle intervention. Every tissue analysed presented a specific lipid profile irrespective of interventions. Glycerolipids and fatty acids were most abundant in epididymal white adipose tissue (eWAT) followed by liver, whereas sterol lipids and phosphoglycerolipids were highly enriched in hypothalamus, and gastrocnemius had the lowest content in all lipid species compared to the other tissues. Both when subjected to a high-fat diet (HFD) and after a subsequent lifestyle intervention (INT), the lipidome of hypothalamus showed no changes. Gastrocnemius and liver revealed a pattern of increase in content in many lipid species after HFD followed by a regression to basal levels after INT, while eWAT lipidome was affected mainly by the fat composition of the administered diets and not their caloric density. Thus, the present study demonstrates a unique lipidome for each tissue modulated by caloric intake and dietary composition.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Lipidomics , Obesity/diet therapy , Obesity/metabolism , Adipose Tissue, White/metabolism , Animals , Caloric Restriction , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Type 2/etiology , Diet, High-Fat/adverse effects , Disease Models, Animal , Healthy Lifestyle , Hypothalamus/metabolism , Liver/metabolism , Male , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Obesity/complications , Physical Conditioning, Animal
10.
Mol Cell Proteomics ; 18(5): 1027-1035, 2019 05.
Article in English | MEDLINE | ID: mdl-30833379

ABSTRACT

Universal proteomics sample preparation is challenging because of the high heterogeneity of biological samples. Here we describe a novel mechanism that exploits the inherent instability of denatured proteins for nonspecific immobilization on microparticles by protein aggregation capture. To demonstrate the general applicability of this mechanism, we analyzed phosphoproteomes, tissue proteomes, and interaction proteomes as well as dilute secretomes. The findings present a practical, sensitive and cost-effective proteomics sample preparation method.


Subject(s)
Cell-Derived Microparticles/metabolism , Protein Aggregates , Proteomics/methods , Animals , Cell Line, Tumor , Humans , Mice , Protein Processing, Post-Translational , RAW 264.7 Cells
11.
FASEB J ; 33(2): 2343-2358, 2019 02.
Article in English | MEDLINE | ID: mdl-30277821

ABSTRACT

Calorie restriction (CR) exerts remarkable, beneficial effects on glucose homeostasis by mechanisms that are not fully understood. Given the relevance of white adipose tissue (WAT) in glucose homeostasis, we aimed at identifying the main cellular processes regulated in WAT in response to CR in a pathologic context of obesity. For this, a gene-expression profiling study was first conducted in mice fed ad libitum or subjected to 40% CR. We found that the gene network related to mitochondria was the most highly upregulated in WAT by CR. To study the role that increased mitochondrial biogenesis plays on glucose homeostasis following CR, we generated a mouse model devoid of the coactivators peroxisome proliferator-activated receptor γ coactivator 1 (PGC-1)α and PGC-1ß specifically in adipocytes. Our results show that mice lacking PGC-1s in adipocytes are unable to increase mitochondrial biogenesis in WAT upon CR. Despite a blunted induction of mitochondrial biogenesis in response to calorie deprivation, mice lacking adipose PGC-1s still respond to CR by improving their glucose homeostasis. Our study demonstrates that PGC-1 coactivators are major regulators of CR-induced mitochondrial biogenesis in WAT and that increased mitochondrial biogenesis and oxidative function in adipose tissue are not required for the improvement of glucose homeostasis mediated by CR.-Pardo, R., Vilà, M., Cervela, L., de Marco, M., Gama-Pérez, P., González-Franquesa, A., Statuto, L., Vilallonga, R., Simó, R., Garcia-Roves, P. M., Villena, J. A. Calorie restriction prevents diet-induced insulin resistance independently of PGC-1-driven mitochondrial biogenesis in white adipose tissue.


Subject(s)
Adipose Tissue, White/physiopathology , Caloric Restriction , Diet/adverse effects , Glucose Intolerance/prevention & control , Insulin Resistance , Organelle Biogenesis , Transcription Factors/physiology , Animals , Gene Expression Profiling , Glucose Intolerance/etiology , Glucose Intolerance/metabolism , Homeostasis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
12.
Trends Endocrinol Metab ; 29(4): 201-202, 2018 04.
Article in English | MEDLINE | ID: mdl-29409712

ABSTRACT

Merging transcriptomics or metabolomics data remains insufficient for metabolic flux estimation. Ramirez et al. integrate a genome-scale metabolic model with extracellular flux data to predict and validate metabolic differences between white and brown adipose tissue. This method allows both metabolic phenotyping and the identification of potential therapeutic targets for obesity.


Subject(s)
Adipocytes, White , Adipose Tissue, Brown , Humans , Metabolomics , Obesity
14.
Adv Exp Med Biol ; 982: 465-520, 2017.
Article in English | MEDLINE | ID: mdl-28551803

ABSTRACT

Insulin resistance precedes and predicts the onset of type 2 diabetes (T2D) in susceptible humans, underscoring its important role in the complex pathogenesis of this disease. Insulin resistance contributes to multiple tissue defects characteristic of T2D, including reduced insulin-stimulated glucose uptake in insulin-sensitive tissues, increased hepatic glucose production, increased lipolysis in adipose tissue, and altered insulin secretion. Studies of individuals with insulin resistance, both with established T2D and high-risk individuals, have consistently demonstrated a diverse array of defects in mitochondrial function (i.e., bioenergetics, biogenesis and dynamics). However, it remains uncertain whether mitochondrial dysfunction is primary (critical initiating defect) or secondary to the subtle derangements in glucose metabolism, insulin resistance, and defective insulin secretion present early in the course of disease development. In this chapter, we will present the evidence linking mitochondrial dysfunction and insulin resistance, and review the potential for mitochondrial targets as a therapeutic approach for T2D.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Energy Metabolism , Insulin Resistance , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Adipose Tissue/physiopathology , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/physiopathology , Energy Metabolism/drug effects , Humans , Hypoglycemic Agents/therapeutic use , Insulin/blood , Lipid Metabolism , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/pathology , Mitochondrial Dynamics , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Oxidative Stress , Risk Factors , Signal Transduction
15.
Curr Diabetes Rev ; 13(4): 370-385, 2017.
Article in English | MEDLINE | ID: mdl-26456359

ABSTRACT

INTRODUCTION: Mitochondria form an interconnected and dynamic web that undergoes continuous cycles of fusion and fission events. This phenomenon, known as mitochondrial dynamics, represents a key quality control system to maintain a healthy mitochondrial population but also a mechanism to bioenergetically adapt to the cellular and tissue energetic demands. Consequently, mitochondria can be viewed not only as energy supply organelles but also as energy sensors. It is therefore not surprising that disrupted mitochondrial bioenergetics, concomitantly with alterations in mitochondrial architecture, has been associated with several diseases including metabolic disorders. CONCLUSION: Here, we review current evidences connecting mitochondrial dynamics and bioenergetic alterations with the development of obesity and diabetes-related phenotypes, and how current strategies to alleviate such phenotypes impact on mitochondrial network and function.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Energy Metabolism , Mitochondria/metabolism , Mitochondrial Dynamics , Adipose Tissue/metabolism , Adipose Tissue/pathology , Adipose Tissue/physiopathology , Animals , Autophagy , Biomarkers/blood , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Homeostasis , Humans , Insulin Resistance , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Liver/metabolism , Liver/pathology , Liver/physiopathology , Mitochondria/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Obesity/blood , Obesity/pathology , Obesity/physiopathology , Signal Transduction
16.
Redox Biol ; 11: 60-72, 2017 04.
Article in English | MEDLINE | ID: mdl-27888692

ABSTRACT

Niemann Pick type C (NPC) disease is a progressive lysosomal storage disorder caused by mutations in genes encoding NPC1/NPC2 proteins, characterized by neurological defects, hepatosplenomegaly and premature death. While the primary biochemical feature of NPC disease is the intracellular accumulation of cholesterol and gangliosides, predominantly in endolysosomes, mitochondrial cholesterol accumulation has also been reported. As accumulation of cholesterol in mitochondria is known to impair the transport of GSH into mitochondria, resulting in mitochondrial GSH (mGSH) depletion, we investigated the impact of mGSH recovery in NPC disease. We show that GSH ethyl ester (GSH-EE), but not N-acetylcysteine (NAC), restored the mGSH pool in liver and brain of Npc1-/- mice and in fibroblasts from NPC patients, while both GSH-EE and NAC increased total GSH levels. GSH-EE but not NAC increased the median survival and maximal life span of Npc1-/- mice. Moreover, intraperitoneal therapy with GSH-EE protected against oxidative stress and oxidant-induced cell death, restored calbindin levels in cerebellar Purkinje cells and reversed locomotor impairment in Npc1-/- mice. High-resolution respirometry analyses revealed that GSH-EE improved oxidative phosphorylation, coupled respiration and maximal electron transfer in cerebellum of Npc1-/- mice. Lipidomic analyses showed that GSH-EE treatment had not effect in the profile of most sphingolipids in liver and brain, except for some particular species in brain of Npc1-/- mice. These findings indicate that the specific replenishment of mGSH may be a potential promising therapy for NPC disease, worth exploring alone or in combination with other options.


Subject(s)
Glutathione/metabolism , Mitochondria/metabolism , Niemann-Pick Disease, Type C/metabolism , Proteins/genetics , Vesicular Transport Proteins/genetics , Acetylcysteine/metabolism , Animals , Cerebellum/metabolism , Cerebellum/pathology , Cholesterol/metabolism , Glutathione/pharmacology , Humans , Intracellular Signaling Peptides and Proteins , Lysosomes/genetics , Lysosomes/metabolism , Mice , Mice, Knockout , Mitochondria/pathology , Mutation , Niemann-Pick C1 Protein , Niemann-Pick Disease, Type C/genetics , Niemann-Pick Disease, Type C/pathology , Oxidative Phosphorylation , Proteins/metabolism , Purkinje Cells/metabolism , Vesicular Transport Proteins/metabolism
17.
Curr Diab Rep ; 16(8): 74, 2016 08.
Article in English | MEDLINE | ID: mdl-27319324

ABSTRACT

Type 2 diabetes (T2D) is increasing worldwide, making identification of biomarkers for detection, staging, and effective prevention strategies an especially critical scientific and medical goal. Fortunately, advances in metabolomics techniques, together with improvements in bioinformatics and mathematical modeling approaches, have provided the scientific community with new tools to describe the T2D metabolome. The metabolomics signatures associated with T2D and obesity include increased levels of lactate, glycolytic intermediates, branched-chain and aromatic amino acids, and long-chain fatty acids. Conversely, tricarboxylic acid cycle intermediates, betaine, and other metabolites decrease. Future studies will be required to fully integrate these and other findings into our understanding of diabetes pathophysiology and to identify biomarkers of disease risk, stage, and responsiveness to specific treatments.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Metabolomics/methods , Diabetes Mellitus, Type 2/genetics , Environment , Genome , Humans , Metabolome/genetics , Risk Factors
18.
Diabetes ; 65(4): 902-12, 2016 04.
Article in English | MEDLINE | ID: mdl-26858359

ABSTRACT

Identifying markers of human insulin resistance may permit development of new approaches for treatment and prevention of type 2 diabetes. To this end, we analyzed the fasting plasma metabolome in metabolically characterized human volunteers across a spectrum of insulin resistance. We demonstrate that plasma betaine levels are reduced in insulin-resistant humans and correlate closely with insulin sensitivity. Moreover, betaine administration to mice with diet-induced obesity prevents the development of impaired glucose homeostasis, reduces hepatic lipid accumulation, increases white adipose oxidative capacity, and enhances whole-body energy expenditure. In parallel with these beneficial metabolic effects, betaine supplementation robustly increased hepatic and circulating fibroblast growth factor (Fgf)21 levels. Betaine administration failed to improve glucose homeostasis and liver fat content in Fgf21(-/-) mice, demonstrating that Fgf21 is necessary for betaine's beneficial effects. Together, these data indicate that dietary betaine increases Fgf21 levels to improve metabolic health in mice and suggest that betaine supplementation merits further investigation as a supplement for treatment or prevention of type 2 diabetes in humans.


Subject(s)
Betaine/pharmacology , Fibroblast Growth Factors/blood , Glucose/metabolism , Lipid Metabolism/drug effects , Liver/drug effects , Adult , Animals , Cells, Cultured , Dietary Supplements , Fatty Liver/chemically induced , Fatty Liver/drug therapy , Fatty Liver/genetics , Fatty Liver/metabolism , Female , Fibroblast Growth Factors/genetics , Glucose Intolerance/blood , Glucose Intolerance/drug therapy , Homeostasis/drug effects , Homeostasis/genetics , Humans , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
19.
J Neuroinflammation ; 12: 152, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26310930

ABSTRACT

BACKGROUND: In brain inflammatory diseases, axonal damage is one of the most critical steps in the cascade that leads to permanent disability. Thus, identifying the initial events triggered by inflammation or oxidative stress that provoke axonal damage is critical for the development of neuroprotective therapies. Energy depletion due to mitochondrial dysfunction has been postulated as an important step in the damage of axons. This prompted us to study the effects of acute inflammation and oxidative stress on the morphology, transport, and function of mitochondria in axons. METHODS: Mouse cerebellar slice cultures were challenged with either lipopolysaccharide (LPS) or hydrogen peroxide (H2O2) ex vivo for 24 h. Axonal mitochondrial morphology was evaluated by transmission electron microscopy (TEM) and mitochondrial transportation by time-lapse imaging. In addition, mitochondrial function in the cerebellar slice cultures was analyzed through high-resolution respirometry assays and quantification of adenosine triphosphate (ATP) production. RESULTS: Both conditions promoted an increase in the size and complexity of axonal mitochondria evident in electron microscopy images, suggesting a compensatory response. Such compensation was reflected at the tissue level as increased respiratory activity of complexes I and IV and as a transient increase in ATP production in response to acute inflammation. Notably, time-lapse microscopy indicated that mitochondrial transport (mean velocity) was severely impaired in axons, increasing the proportion of stationary mitochondria in axons after LPS challenge. Indeed, the two challenges used produced different effects: inflammation mostly reducing retrograde transport and oxidative stress slightly enhancing retrograde transportation. CONCLUSIONS: Neuroinflammation acutely impairs axonal mitochondrial transportation, which would promote an inappropriate delivery of energy throughout axons and, by this way, contribute to axonal damage. Thus, preserving axonal mitochondrial transport might represent a promising avenue to exploit as a therapeutic target for neuroprotection in brain inflammatory diseases like multiple sclerosis.


Subject(s)
Axonal Transport/physiology , Axons/ultrastructure , Cerebellum/ultrastructure , Mitochondria/metabolism , Adenosine Triphosphate/metabolism , Animals , Animals, Newborn , Axonal Transport/drug effects , Axons/drug effects , Cerebellum/drug effects , Gene Expression Regulation/drug effects , Hydrogen Peroxide/toxicity , In Vitro Techniques , Lipopolysaccharides/toxicity , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Mitochondria/drug effects , Mitochondria/ultrastructure , Multienzyme Complexes/metabolism , Organ Culture Techniques , Oxidants/toxicity , Time Factors , Transfection
20.
J Clin Endocrinol Metab ; 100(3): E407-15, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25532038

ABSTRACT

CONTEXT: Diabetes is frequently diagnosed late, when the development of complications is almost inevitable, decreasing the quality of life of patients. However, early detection of affected individuals would allow the implementation of timely and effective therapies. OBJECTIVE: Here we set to describe the profile of circulating microRNAs (miRNAs) in prediabetic patients with the intention of identifying novel diagnostic and therapeutic tools. DESIGN: We used real-time RT-PCR to measure the abundance of 176 miRNAs in serum of a cohort of 92 control and prediabetic individuals with either impaired fasting glucose or impaired glucose tolerance, as well as newly diagnosed diabetic patients. We validated the results in a second cohort of control and prediabetic subjects undergoing a therapeutic exercise intervention, as well as in a mouse model of glucose intolerance. RESULTS: We identified two miRNAs, miR-192 and miR-193b, whose abundance is significantly increased in the prediabetic state but not in diabetic patients. Strikingly, these miRNAs are also increased in plasma of glucose-intolerant mice. Moreover, circulating levels of miR-192 and miR-193b return to baseline in both prediabetic humans and glucose-intolerant mice undergoing a therapeutic intervention consisting in chronic exercise, which succeeded in normalizing metabolic parameters. CONCLUSIONS: Our data show that the pattern of circulating miRNAs is modified by defects in glucose metabolism in a similar manner in mice and humans. This circulating miRNA signature for prediabetes could be used as a new diagnostic tool, as well as to monitor response to intervention.


Subject(s)
Biomarkers/blood , Exercise Therapy , MicroRNAs/blood , Prediabetic State/blood , Prediabetic State/therapy , Animals , Exercise/physiology , Female , Gene Expression Profiling , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Prediabetic State/genetics , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...