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










Publication year range
1.
Redox Biol ; 70: 103052, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38290384

ABSTRACT

Acute liver failure caused by alcoholic hepatitis (AH) is only effectively treated with liver transplantation. Livers of patients with AH show a unique molecular signature characterized by defective hepatocellular redox metabolism, concurrent to hepatic infiltration of neutrophils that express myeloperoxidase (MPO) and form neutrophil extracellular traps (NETs). Exacerbated NET formation and MPO activity contribute to liver damage in mice with AH and predicts poor prognosis in AH patients. The identification of pathways that maladaptively exacerbate neutrophilic activity in liver could inform of novel therapeutic approaches to treat AH. Whether the redox defects of hepatocytes in AH directly exacerbate neutrophilic inflammation and NET formation is unclear. Here we identify that the protein content of the mitochondrial biliverdin exporter ABCB10, which increases hepatocyte-autonomous synthesis of the ROS-scavenger bilirubin, is decreased in livers from humans and mice with AH. Increasing ABCB10 expression selectively in hepatocytes of mice with AH is sufficient to decrease MPO gene expression and histone H3 citrullination, a specific marker of NET formation. These anti-inflammatory effects can be explained by ABCB10 function reducing ROS-mediated actions in liver. Accordingly, ABCB10 gain-of-function selectively increased the mitochondrial GSH/GSSG ratio and decreased hepatic 4-HNE protein adducts, without elevating mitochondrial fat expenditure capacity, nor mitigating steatosis and hepatocyte death. Thus, our study supports that ABCB10 function regulating ROS-mediated actions within surviving hepatocytes mitigates the maladaptive activation of infiltrated neutrophils in AH. Consequently, ABCB10 gain-of-function in human hepatocytes could potentially decrease acute liver failure by decreasing the inflammatory flare caused by excessive neutrophil activity.


Subject(s)
Hepatitis, Alcoholic , Liver Failure, Acute , Humans , Animals , Mice , Hepatitis, Alcoholic/genetics , Hepatitis, Alcoholic/metabolism , Biliverdine/metabolism , Reactive Oxygen Species/metabolism , Hepatocytes/metabolism , Liver/metabolism , Inflammation/genetics , Inflammation/metabolism , Histones/metabolism , Liver Failure, Acute/metabolism , ATP-Binding Cassette Transporters/metabolism
2.
STAR Protoc ; 4(3): 102408, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37393613

ABSTRACT

Assessing the physiological role of H2O2 requires sensitive techniques to quantify H2O2 and antioxidants in live cells. Here, we present a protocol to assess the mitochondrial redox state and unconjugated bilirubin levels in intact live primary hepatocytes from obese mice. We described steps to quantify H2O2, GSSG/GSH, and bilirubin content in the mitochondrial matrix and the cytosol using the fluorescent reporters roGFP2-ORP1, GRX1-roGFP2, and UnaG, respectively. We detail hepatocyte isolation, plating, and transduction and live-cell imaging using a high-content imaging reader. For complete details on the use and execution of this protocol, please refer to Shum et al.1.


Subject(s)
Bilirubin , Hydrogen Peroxide , Animals , Mice , Mice, Obese , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Oxidation-Reduction , Coloring Agents
3.
Nat Commun ; 14(1): 2573, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37142604

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is the most common liver disease in the world. High levels of free fatty acids in the liver impair hepatic lysosomal acidification and reduce autophagic flux. We investigate whether restoration of lysosomal function in NAFLD recovers autophagic flux, mitochondrial function, and insulin sensitivity. Here, we report the synthesis of novel biodegradable acid-activated acidifying nanoparticles (acNPs) as a lysosome targeting treatment to restore lysosomal acidity and autophagy. The acNPs, composed of fluorinated polyesters, remain inactive at plasma pH, and only become activated in lysosomes after endocytosis. Specifically, they degrade at pH of ~6 characteristic of dysfunctional lysosomes, to further acidify and enhance the function of lysosomes. In established in vivo high fat diet mouse models of NAFLD, re-acidification of lysosomes via acNP treatment restores autophagy and mitochondria function to lean, healthy levels. This restoration, concurrent with reversal of fasting hyperglycemia and hepatic steatosis, indicates the potential use of acNPs as a first-in-kind therapeutic for NAFLD.


Subject(s)
Non-alcoholic Fatty Liver Disease , Mice , Animals , Non-alcoholic Fatty Liver Disease/metabolism , Autophagy , Liver/metabolism , Lysosomes/metabolism , Hydrogen-Ion Concentration
4.
Mol Oncol ; 17(10): 2109-2125, 2023 10.
Article in English | MEDLINE | ID: mdl-37086156

ABSTRACT

The androgen receptor (AR) is an established orchestrator of cell metabolism in prostate cancer (PCa), notably by inducing an oxidative mitochondrial program. Intriguingly, AR regulates cytoplasmic isocitrate dehydrogenase 1 (IDH1), but not its mitochondrial counterparts IDH2 and IDH3. Here, we aimed to understand the functional role of IDH1 in PCa. Mouse models, in vitro human PCa cell lines, and human patient-derived organoids (PDOs) were used to study the expression and activity of IDH enzymes in the normal prostate and PCa. Genetic and pharmacological inhibition of IDH1 was then combined with extracellular flux analyses and gas chromatography-mass spectrometry for metabolomic analyses and cancer cell proliferation in vitro and in vivo. In PCa cells, more than 90% of the total IDH activity is mediated through IDH1 rather than its mitochondrial counterparts. This profile seems to originate from the specialized prostate metabolic program, as observed using mouse prostate and PDOs. Pharmacological and genetic inhibition of IDH1 impaired mitochondrial respiration, suggesting that this cytoplasmic enzyme contributes to the mitochondrial tricarboxylic acid cycle (TCA) in PCa. Mass spectrometry-based metabolomics confirmed this hypothesis, showing that inhibition of IDH1 impairs carbon flux into the TCA cycle. Consequently, inhibition of IDH1 decreased PCa cell proliferation in vitro and in vivo. These results demonstrate that PCa cells have a hybrid cytoplasmic-mitochondrial TCA cycle that depends on IDH1. This metabolic enzyme represents a metabolic vulnerability of PCa cells and a potential new therapeutic target.


Subject(s)
Citric Acid Cycle , Prostatic Neoplasms , Male , Mice , Animals , Humans , Isocitrate Dehydrogenase/genetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Mitochondria/metabolism , Cytosol/metabolism
5.
Ther Innov Regul Sci ; 57(4): 875-885, 2023 07.
Article in English | MEDLINE | ID: mdl-37072651

ABSTRACT

Project Orbis was initiated in May 2019 by the Oncology Center of Excellence to facilitate faster patient access to innovative cancer therapies by providing a framework for concurrent submissions and review of oncology products among international partners. Since its inception, Australia's Therapeutic Goods Administration (TGA), Canada's Health Canada (HC), Singapore's Health Sciences Authority (HSA), Switzerland's Swissmedic (SMC), Brazil's National Health Surveillance Agency (ANVISA), United Kingdom's Medicines and Healthcare Products Regulatory Agency (MHRA), and most recently Israel's Ministry of Health (IMoH) Medical Technologies, Health Information, Innovation and Research (MTIIR) Directorate, have joined Project Orbis. While each country has its own expedited review pathways to bring promising therapies to patients, there are some similarities and differences in pathways and timelines. FDA's fast-track designation and MHRA's marketing authorization under exceptional circumstances (MAEC) allow non-clinical and limited clinical evidence to support approval under these programs. HC's Extraordinary Use New Drug (EUND) pathway allows granting exceptional use authorization with limited clinical evidence. ANVISA, HSA, MTIIR, and TGA do not have standard pathways that allow non-clinical evidence and limited clinical evidence. While there is no definite regulatory pathway for HSA, the current framework for approval does allow flexibility in the type of data (non-clinical or clinical) required to demonstrate the benefit-risk profile of a product. HSA may register a product if the agency is satisfied that the overall benefit outweighs the risk. All Project Orbis Partner (POP) countries have similar programs to the FDA accelerated approval program except ANVISA. Although HSA and MTIIR do not have defined pathways for accelerated approval programs, there are opportunities to request accelerated approval per these agencies. All POP countries have pathways like the FDA priority review except MHRA. Priority review timelines for new drugs range from 120 to 264 calendar days (cd). Standard review timelines for new drugs range from 180 to 365 cd.


Subject(s)
Medicine , Neoplasms , United States , Humans , Drug Approval , United States Food and Drug Administration , Canada
6.
Mol Metab ; 55: 101403, 2022 01.
Article in English | MEDLINE | ID: mdl-34823065

ABSTRACT

OBJECTIVE: The contribution of beta-cell dysfunction to type 2 diabetes (T2D) is not restricted to insulinopenia in the late stages of the disease. Elevated fasting insulinemia in normoglycemic humans is a major factor predicting the onset of insulin resistance and T2D, demonstrating an early alteration of beta-cell function in T2D. Moreover, an early and chronic increase in fasting insulinemia contributes to insulin resistance in high-fat diet (HFD)-fed mice. However, whether there are genetic factors that promote beta-cell-initiated insulin resistance remains undefined. Human variants of the mitochondrial transporter ABCB10, which regulates redox by increasing bilirubin synthesis, have been associated with an elevated risk of T2D. The effects of T2D ABCB10 variants on ABCB10 expression and the actions of ABCB10 in beta-cells are unknown. METHODS: The expression of beta-cell ABCB10 was analyzed in published transcriptome datasets from human beta-cells carrying the T2D-risk ABCB10 variant. Insulin sensitivity, beta-cell proliferation, and secretory function were measured in beta-cell-specific ABCB10 KO mice (Ins1Cre-Abcb10flox/flox). The short-term role of beta-cell ABCB10 activity on glucose-stimulated insulin secretion (GSIS) was determined in isolated islets. RESULTS: Carrying the T2Drisk allele G of ABCB10 rs348330 variant was associated with increased ABCB10 expression in human beta-cells. Constitutive deletion of Abcb10 in beta-cells protected mice from hyperinsulinemia and insulin resistance by limiting HFD-induced beta-cell expansion. An early limitation in GSIS and H2O2-mediated signaling caused by elevated ABCB10 activity can initiate an over-compensatory expansion of beta-cell mass in response to HFD. Accordingly, increasing ABCB10 expression was sufficient to limit GSIS capacity. In health, ABCB10 protein was decreased during islet maturation, with maturation restricting beta-cell proliferation and elevating GSIS. Finally, ex-vivo and short-term deletion of ABCB10 in islets isolated from HFD-fed mice increased H2O2 and GSIS, which was reversed by bilirubin treatments. CONCLUSIONS: Beta-cell ABCB10 is required for HFD to induce insulin resistance in mice by amplifying beta-cell mass expansion to maladaptive levels that cause fasting hyperinsulinemia.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Insulin Resistance/genetics , Insulin-Secreting Cells/metabolism , ATP-Binding Cassette Transporters/genetics , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat , Female , Glucose/metabolism , Glucose Tolerance Test , Insulin/metabolism , Insulin Resistance/physiology , Insulin Secretion/drug effects , Insulin-Secreting Cells/physiology , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism
7.
J Vis Exp ; (177)2021 11 11.
Article in English | MEDLINE | ID: mdl-34842229

ABSTRACT

Energy expenditure measurements are necessary to understand how changes in metabolism can lead to obesity. Basal energy expenditure can be determined in mice by measuring whole-body oxygen consumption, CO2 production, and physical activity using metabolic cages. Thermogenic brown/beige adipocytes (BA) contribute significantly to rodent energy expenditure, particularly at low ambient temperatures. Here, measurements of basal energy expenditure and total BA capacity to expend energy in obese mice are described in two detailed protocols: the first explaining how to set up the assay to measure basal energy expenditure using analysis of covariance (ANCOVA), a necessary analysis given that energy expenditure co-varies with body mass. The second protocol describes how to measure BA energy expenditure capacity in vivo in mice. This procedure involves anesthesia, needed to limit expenditure caused by physical activity, followed by the injection of beta3-adrenergic agonist, CL-316,243, which activates energy expenditure in BA. These two protocols and their limitations are described in sufficient detail to allow a successful first experiment.


Subject(s)
Adipocytes, Brown , Thermogenesis , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Animals , Energy Metabolism , Mice , Mice, Obese , Obesity/metabolism , Thermogenesis/physiology
8.
Nat Metab ; 3(11): 1552-1568, 2021 11.
Article in English | MEDLINE | ID: mdl-34697471

ABSTRACT

We have previously suggested a central role for mitochondria in the observed sex differences in metabolic traits. However, the mechanisms by which sex differences affect adipose mitochondrial function and metabolic syndrome are unclear. Here we show that in both mice and humans, adipose mitochondrial functions are elevated in females and are strongly associated with adiposity, insulin resistance and plasma lipids. Using a panel of diverse inbred strains of mice, we identify a genetic locus on mouse chromosome 17 that controls mitochondrial mass and function in adipose tissue in a sex- and tissue-specific manner. This locus contains Ndufv2 and regulates the expression of at least 89 mitochondrial genes in females, including oxidative phosphorylation genes and those related to mitochondrial DNA content. Overexpression studies indicate that Ndufv2 mediates these effects by regulating supercomplex assembly and elevating mitochondrial reactive oxygen species production, which generates a signal that increases mitochondrial biogenesis.


Subject(s)
Adipose Tissue/metabolism , Biomarkers , Gene Expression Regulation , Metabolic Syndrome/etiology , Metabolic Syndrome/metabolism , Mitochondria/genetics , Mitochondria/metabolism , NADH Dehydrogenase/genetics , Adiposity/genetics , Animals , Cell Respiration/genetics , Chromosomes, Human, Pair 17 , Disease Models, Animal , Disease Susceptibility , Female , Gene Expression Profiling , Genetic Association Studies , Humans , Male , Metabolic Syndrome/diagnosis , Mice , NADH Dehydrogenase/metabolism , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Quantitative Trait, Heritable , Reactive Oxygen Species/metabolism , Sex Factors
9.
Sci Transl Med ; 13(594)2021 05 19.
Article in English | MEDLINE | ID: mdl-34011630

ABSTRACT

Although the role of hydrophilic antioxidants in the development of hepatic insulin resistance and nonalcoholic fatty liver disease has been well studied, the role of lipophilic antioxidants remains poorly characterized. A known lipophilic hydrogen peroxide scavenger is bilirubin, which can be oxidized to biliverdin and then reduced back to bilirubin by cytosolic biliverdin reductase. Oxidation of bilirubin to biliverdin inside mitochondria must be followed by the export of biliverdin to the cytosol, where biliverdin is reduced back to bilirubin. Thus, the putative mitochondrial exporter of biliverdin is expected to be a major determinant of bilirubin regeneration and intracellular hydrogen peroxide scavenging. Here, we identified ABCB10 as a mitochondrial biliverdin exporter. ABCB10 reconstituted into liposomes transported biliverdin, and ABCB10 deletion caused accumulation of biliverdin inside mitochondria. Obesity with insulin resistance up-regulated hepatic ABCB10 expression in mice and elevated cytosolic and mitochondrial bilirubin content in an ABCB10-dependent manner. Revealing a maladaptive role of ABCB10-driven bilirubin synthesis, hepatic ABCB10 deletion protected diet-induced obese mice from steatosis and hyperglycemia, improving insulin-mediated suppression of glucose production and decreasing lipogenic SREBP-1c expression. Protection was concurrent with enhanced mitochondrial function and increased inactivation of PTP1B, a phosphatase disrupting insulin signaling and elevating SREBP-1c expression. Restoration of cellular bilirubin content in ABCB10 KO hepatocytes reversed the improvements in mitochondrial function and PTP1B inactivation, demonstrating that bilirubin was the maladaptive effector linked to ABCB10 function. Thus, we identified a fundamental transport process that amplifies intracellular bilirubin redox actions, which can exacerbate insulin resistance and steatosis in obesity.


Subject(s)
Biliverdine , Mitochondria , Animals , Antioxidants , Bilirubin , Liver , Mice , Obesity
10.
STAR Protoc ; 2(1): 100243, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33458705

ABSTRACT

Mitochondria play a central role in lipid metabolism and can bind to lipid droplets. However, the role and functional specialization of the population of peridroplet mitochondria (PDMs) remain unclear, as methods to isolate functional PDMs were not developed until recently. Here, we describe an approach to isolate intact PDMs from murine brown adipose tissue based on their adherence to lipid droplets. PDMs isolated using our approach can be used to study their specialized function by respirometry. For complete information on the use and execution of this protocol, please refer to Benador et al. (2018).


Subject(s)
Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Lipid Droplets/metabolism , Lipid Metabolism , Mitochondria/metabolism , Adipocytes, Brown/cytology , Adipose Tissue, Brown/cytology , Animals , Mice
11.
Mol Metab ; 50: 101134, 2021 08.
Article in English | MEDLINE | ID: mdl-33276146

ABSTRACT

BACKGROUND: Mitochondrial oxidative function plays a key role in the development of non-alcoholic fatty liver disease (NAFLD) and insulin resistance (IR). Recent studies reported that fatty liver might not be a result of decreased mitochondrial fat oxidation caused by mitochondrial damage. Rather, NAFLD and IR induce an elevation in mitochondrial function that covers the increased demand for carbon intermediates and ATP caused by elevated lipogenesis and gluconeogenesis. Furthermore, mitochondria play a role in regulating hepatic insulin sensitivity and lipogenesis by modulating redox-sensitive signaling pathways. SCOPE OF REVIEW: We review the contradictory studies indicating that NAFLD and hyperglycemia can either increase or decrease mitochondrial oxidative capacity in the liver. We summarize mechanisms regulating mitochondrial heterogeneity inside the same cell and discuss how these mechanisms may determine the role of mitochondria in NAFLD. We further discuss the role of endogenous antioxidants in controlling mitochondrial H2O2 release and redox-mediated signaling. We describe the emerging concept that the subcellular location of cellular antioxidants is a key determinant of their effects on NAFLD. MAJOR CONCLUSIONS: The balance of fat oxidation versus accumulation depends on mitochondrial fuel preference rather than ATP-synthesizing respiration. As such, therapies targeting fuel preference might be more suitable for treating NAFLD. Similarly, suppressing maladaptive antioxidants, rather than interfering with physiological mitochondrial H2O2-mediated signaling, may allow the maintenance of intact hepatic insulin signaling in NAFLD. Exploration of the subcellular compartmentalization of different antioxidant systems and the unique functions of specific mitochondrial subpopulations may offer new intervention points to treat NAFLD.


Subject(s)
Hyperglycemia/complications , Hypoglycemic Agents/therapeutic use , Liver/pathology , Mitochondria/pathology , Non-alcoholic Fatty Liver Disease/pathology , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Clinical Trials as Topic , Disease Models, Animal , Gluconeogenesis/drug effects , Humans , Hydrogen Peroxide/metabolism , Hyperglycemia/blood , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Lipogenesis/drug effects , Liver/cytology , Liver/drug effects , Mitochondria/drug effects , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Signal Transduction/drug effects , Treatment Outcome
12.
Nat Commun ; 11(1): 6378, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33311503

ABSTRACT

Homeotherms maintain a stable internal body temperature despite changing environments. During energy deficiency, some species can cease to defend their body temperature and enter a hypothermic and hypometabolic state known as torpor. Recent advances have revealed the medial preoptic area (MPA) as a key site for the regulation of torpor in mice. The MPA is estrogen-sensitive and estrogens also have potent effects on both temperature and metabolism. Here, we demonstrate that estrogen-sensitive neurons in the MPA can coordinate hypothermia and hypometabolism in mice. Selectively activating estrogen-sensitive MPA neurons was sufficient to drive a coordinated depression of metabolic rate and body temperature similar to torpor, as measured by body temperature, physical activity, indirect calorimetry, heart rate, and brain activity. Inducing torpor with a prolonged fast revealed larger and more variable calcium transients from estrogen-sensitive MPA neurons during bouts of hypothermia. Finally, whereas selective ablation of estrogen-sensitive MPA neurons demonstrated that these neurons are required for the full expression of fasting-induced torpor in both female and male mice, their effects on thermoregulation and torpor bout initiation exhibit differences across sex. Together, these findings suggest a role for estrogen-sensitive MPA neurons in directing the thermoregulatory and metabolic responses to energy deficiency.


Subject(s)
Body Temperature/physiology , Estrogens/metabolism , Neurons/physiology , Preoptic Area/metabolism , Torpor/physiology , Animals , Body Temperature/genetics , Body Temperature Regulation/physiology , Energy Metabolism/physiology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Fasting , Female , Hypothermia/genetics , Hypothermia/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
13.
Clin Cancer Res ; 26(24): 6412-6416, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33037016

ABSTRACT

In 2019, the FDA Oncology Center of Excellence launched Project Orbis, a global collaborative review program to facilitate faster patient access to innovative cancer therapies across multiple countries. Project Orbis aims for concurrent submission, review, and regulatory action for high-impact clinically significant marketing applications among the participating partner countries. Current Project Orbis partners (POP) include the regulatory health authorities (RHA) of Australia, Brazil, Canada, Singapore, and Switzerland. Project Orbis leverages the existing scientific and regulatory partnerships between the various RHA under mutual confidentiality agreements. While FDA serves as the primary coordinator for application selection and review, each country remains fully independent on their final regulatory decision. In the first year of Project Orbis (June 2019 to June 2020), a total of 60 oncology marketing applications were received, representing 16 unique projects, and resulting in 38 approvals. New molecular entities, also known as new active substances, comprised 28% of the received marketing applications. The median time gap between FDA and Orbis submission dates was 0.6 months with a range of -0.8 to 9.0 months. Across the program, the median time-to-approval was similar between FDA (4.2 months, range 0.9-6.9, N = 18) and the POP (4.4 months, range 1.7-6.8, N = 20). Participating countries have signified a strong commitment for continuation and growth of the program. Project Orbis expansion considerations include the addition of more countries and management of more complex applications.


Subject(s)
Disease , Drug Approval/legislation & jurisprudence , Drug Discovery/organization & administration , Global Health , Government Agencies/legislation & jurisprudence , Intersectoral Collaboration , Product Surveillance, Postmarketing/statistics & numerical data , Humans
14.
Nat Commun ; 11(1): 3347, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32620768

ABSTRACT

A sharp increase in mitochondrial Ca2+ marks the activation of brown adipose tissue (BAT) thermogenesis, yet the mechanisms preventing Ca2+ deleterious effects are poorly understood. Here, we show that adrenergic stimulation of BAT activates a PKA-dependent mitochondrial Ca2+ extrusion via the mitochondrial Na+/Ca2+ exchanger, NCLX. Adrenergic stimulation of NCLX-null brown adipocytes (BA) induces a profound mitochondrial Ca2+ overload and impaired uncoupled respiration. Core body temperature, PET imaging of glucose uptake and VO2 measurements confirm a thermogenic defect in NCLX-null mice. We show that Ca2+ overload induced by adrenergic stimulation of NCLX-null BAT, triggers the mitochondrial permeability transition pore (mPTP) opening, leading to a remarkable mitochondrial swelling and cell death. Treatment with mPTP inhibitors rescue mitochondrial function and thermogenesis in NCLX-null BAT, while calcium overload persists. Our findings identify a key pathway through which BA evade apoptosis during adrenergic stimulation of uncoupling. NCLX deletion transforms the adrenergic pathway responsible for thermogenesis activation into a death pathway.


Subject(s)
Adipocytes, Brown/pathology , Adipose Tissue, Brown/metabolism , Norepinephrine/metabolism , Sodium-Calcium Exchanger/metabolism , Thermogenesis/physiology , Adipocytes, Brown/cytology , Adipocytes, Brown/drug effects , Adipose Tissue, Brown/cytology , Adrenergic Agents/pharmacology , Animals , Apoptosis/drug effects , Calcium/metabolism , Cells, Cultured , Cold Temperature/adverse effects , Cyclosporine/pharmacology , Energy Metabolism/drug effects , Energy Metabolism/physiology , Female , Intravital Microscopy , Male , Mice , Mice, Knockout , Microscopy, Fluorescence , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Primary Cell Culture , Signal Transduction , Sodium-Calcium Exchanger/genetics , Thermogenesis/drug effects
15.
Nat Metab ; 2(4): 351-363, 2020 04.
Article in English | MEDLINE | ID: mdl-32377634

ABSTRACT

Estrogen receptor a (ERa) signaling in the ventromedial hypothalamus (VMH) contributes to energy homeostasis by modulating physical activity and thermogenesis. However, the precise neuronal populations involved remain undefined. Here, we describe six neuronal populations in the mouse VMH by using single-cell RNA transcriptomics and in situ hybridization. ERa is enriched in populations showing sex biased expression of reprimo (Rprm), tachykinin 1 (Tac1), and prodynorphin (Pdyn). Female biased expression of Tac1 and Rprm is patterned by ERa-dependent repression during male development, whereas male biased expression of Pdyn is maintained by circulating testicular hormone in adulthood. Chemogenetic activation of ERa positive VMH neurons stimulates heat generation and movement in both sexes. However, silencing Rprm gene function increases core temperature selectively in females and ectopic Rprm expression in males is associated with reduced core temperature. Together these findings reveal a role for Rprm in temperature regulation and ERa in the masculinization of neuron populations that underlie energy expenditure.


Subject(s)
Energy Metabolism , Estrogen Receptor alpha/metabolism , Hypothalamus/metabolism , Sex Characteristics , Animals , Female , Fluorescent Dyes/chemistry , Genetic Markers , Hypothalamus/cytology , Male , Mice , Neurons/metabolism
16.
Mol Metab ; 30: 30-47, 2019 12.
Article in English | MEDLINE | ID: mdl-31767179

ABSTRACT

OBJECTIVE: Lipocalin-2 (LCN2) is a secreted protein involved in innate immunity and has also been associated with several cardiometabolic traits in both mouse and human studies. However, the causal relationship of LCN2 to these traits is unclear, and most studies have examined only males. METHODS: Using adeno-associated viral vectors we expressed LCN2 in either adipose or liver in a tissue specific manner on the background of a whole-body Lcn2 knockout or wildtype mice. Metabolic phenotypes including body weight, body composition, plasma and liver lipids, glucose homeostasis, insulin resistance, mitochondrial phenotyping, and metabolic cage studies were monitored. RESULTS: We studied the genetics of LCN2 expression and associated clinical traits in both males and females in a panel of 100 inbred strains of mice (HMDP). The natural variation in Lcn2 expression across the HMDP exhibits high heritability, and genetic mapping suggests that it is regulated in part by Lipin1 gene variation. The correlation analyses revealed striking tissue dependent sex differences in obesity, insulin resistance, hepatic steatosis, and dyslipidemia. To understand the causal relationships, we examined the effects of expression of LCN2 selectively in liver or adipose. On a Lcn2-null background, LCN2 expression in white adipose promoted metabolic disturbances in females but not males. It acted in an autocrine/paracrine manner, resulting in mitochondrial dysfunction and an upregulation of inflammatory and fibrotic genes. On the other hand, on a null background, expression of LCN2 in liver had no discernible impact on the traits examined despite increasing the levels of circulating LCN2 more than adipose LCN2 expression. The mechanisms underlying the sex-specific action of LCN2 are unclear, but our results indicate that adipose LCN2 negatively regulates its receptor, LRP2 (or megalin), and its repressor, ERα, in a female-specific manner and that the effects of LCN2 on metabolic traits are mediated in part by LRP2. CONCLUSIONS: Following up on our population-based studies, we demonstrate that LCN2 acts in a highly sex- and tissue-specific manner in mice. Our results have important implications for human studies, emphasizing the importance of sex and the tissue source of LCN2.


Subject(s)
Adipose Tissue/metabolism , Lipocalin-2/metabolism , Adiposity , Animals , Body Composition , Body Weight , Female , Glucose/analysis , Homeostasis , Insulin Resistance , Lipids/analysis , Lipocalin-2/genetics , Lipocalin-2/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Obesity/metabolism , Sex Factors
17.
J Biol Chem ; 294(32): 12250-12260, 2019 08 09.
Article in English | MEDLINE | ID: mdl-31243102

ABSTRACT

mTOR complex 1 (mTORC1) and p70 S6 kinase (S6K1) are both involved in the development of obesity-linked insulin resistance. Recently, we showed that the S6K1 inhibitor PF-4708671 (PF) increases insulin sensitivity. However, we also reported that PF can increase glucose metabolism even in the absence of insulin in muscle and hepatic cells. Here we further explored the potential mechanisms by which PF increases glucose metabolism in muscle and liver cells independent of insulin. Time course experiments revealed that PF induces AMP-activated protein kinase (AMPK) activation before inhibiting S6K1. However, PF-induced glucose uptake was not prevented in primary muscle cells from AMPK α1/2 double KO (dKO) mice. Moreover, PF-mediated suppression of hepatic glucose production was maintained in hepatocytes derived from AMPK α1/2-dKO mice. Remarkably, PF could still reduce glucose production and activate AMPK in hepatocytes from S6K1/2 dKO mice. Mechanistically, bioenergetics experiments revealed that PF reduces mitochondrial complex I activity in both muscle and hepatic cells. The stimulatory effect of PF on glucose uptake was partially reduced by expression of the Saccharomyces cerevisiae NADH:ubiquinone oxidoreductase in L6 cells. These results indicate that PF-mediated S6K1 inhibition is not required for its effect on insulin-independent glucose metabolism and AMPK activation. We conclude that, although PF rapidly activates AMPK, its ability to acutely increase glucose uptake and suppress glucose production does not require AMPK activation. Unexpectedly, PF rapidly inhibits mitochondrial complex I activity, a mechanism that partially underlies PF's effect on glucose metabolism.


Subject(s)
Electron Transport Complex I/metabolism , Glucose/metabolism , Imidazoles/pharmacology , Mitochondria/drug effects , Piperazines/pharmacology , AMP-Activated Protein Kinases/deficiency , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/genetics , Insulin/pharmacology , Liver/cytology , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Phosphorylation/drug effects , Rats , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism
18.
Cell Metab ; 27(4): 869-885.e6, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29617645

ABSTRACT

Mitochondria associate with lipid droplets (LDs) in fat-oxidizing tissues, but the functional role of these peridroplet mitochondria (PDM) is unknown. Microscopic observation of interscapular brown adipose tissue reveals that PDM have unique protein composition and cristae structure and remain adherent to the LD in the tissue homogenate. We developed an approach to isolate PDM based on their adherence to LDs. Comparison of purified PDM to cytoplasmic mitochondria reveals that (1) PDM have increased pyruvate oxidation, electron transport, and ATP synthesis capacities; (2) PDM have reduced ß-oxidation capacity and depart from LDs upon activation of brown adipose tissue thermogenesis and ß-oxidation; (3) PDM support LD expansion as Perilipin5-induced recruitment of mitochondria to LDs increases ATP synthase-dependent triacylglyceride synthesis; and (4) PDM maintain a distinct protein composition due to uniquely low fusion-fission dynamics. We conclude that PDM represent a segregated mitochondrial population with unique structure and function that supports triacylglyceride synthesis.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Lipid Droplets/metabolism , Mitochondria/metabolism , Adenosine Triphosphate/metabolism , Adipocytes/cytology , Animals , Electron Transport , Energy Metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondrial Dynamics , Mitochondrial Proteins/metabolism , Muscle Proteins/metabolism , Oxidation-Reduction , Pyruvic Acid/metabolism , Thermogenesis
19.
FASEB J ; 32(4): 1741-1777, 2018 04.
Article in English | MEDLINE | ID: mdl-29242278

ABSTRACT

Skeletal muscle possesses a remarkable ability to adapt to various physiologic conditions. AMPK is a sensor of intracellular energy status that maintains energy stores by fine-tuning anabolic and catabolic pathways. AMPK's role as an energy sensor is particularly critical in tissues displaying highly changeable energy turnover. Due to the drastic changes in energy demand that occur between the resting and exercising state, skeletal muscle is one such tissue. Here, we review the complex regulation of AMPK in skeletal muscle and its consequences on metabolism ( e.g., substrate uptake, oxidation, and storage as well as mitochondrial function of skeletal muscle fibers). We focus on the role of AMPK in skeletal muscle during exercise and in exercise recovery. We also address adaptations to exercise training, including skeletal muscle plasticity, highlighting novel concepts and future perspectives that need to be investigated. Furthermore, we discuss the possible role of AMPK as a therapeutic target as well as different AMPK activators and their potential for future drug development.-Kjøbsted, R., Hingst, J. R., Fentz, J., Foretz, M., Sanz, M.-N., Pehmøller, C., Shum, M., Marette, A., Mounier, R., Treebak, J. T., Wojtaszewski, J. F. P., Viollet, B., Lantier, L. AMPK in skeletal muscle function and metabolism.


Subject(s)
Muscle, Skeletal/metabolism , Protein Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Adaptation, Physiological , Animals , Energy Metabolism , Exercise , Humans , Muscle, Skeletal/physiology , Protein Kinases/chemistry , Protein Kinases/genetics
20.
Mol Metab ; 6(5): 447-458, 2017 05.
Article in English | MEDLINE | ID: mdl-28462079

ABSTRACT

OBJECTIVE: The mechanistic target of rapamycin (mTOR) is a serine/threonine kinase that functions into distinct protein complexes (mTORC1 and mTORC2) that regulates growth and metabolism. DEP-domain containing mTOR-interacting protein (DEPTOR) is part of these complexes and is known to reduce their activity. Whether DEPTOR loss affects metabolism and organismal growth in vivo has never been tested. METHODS: We have generated a conditional transgenic mouse allowing the tissue-specific deletion of DEPTOR. This model was crossed with CMV-cre mice or Albumin-cre mice to generate either whole-body or liver-specific DEPTOR knockout (KO) mice. RESULTS: Whole-body DEPTOR KO mice are viable, fertile, normal in size, and do not display any gross physical and metabolic abnormalities. To circumvent possible compensatory mechanisms linked to the early and systemic loss of DEPTOR, we have deleted DEPTOR specifically in the liver, a tissue in which DEPTOR protein is expressed and affected in response to mTOR activation. Liver-specific DEPTOR null mice showed a reduction in circulating glucose upon fasting versus control mice. This effect was not associated with change in hepatic gluconeogenesis potential but was linked to a sustained reduction in circulating glucose during insulin tolerance tests. In addition to the reduction in glycemia, liver-specific DEPTOR KO mice had reduced hepatic glycogen content when fasted. We showed that loss of DEPTOR cell-autonomously increased oxidative metabolism in hepatocytes, an effect associated with increased cytochrome c expression but independent of changes in mitochondrial content or in the expression of genes controlling oxidative metabolism. We found that liver-specific DEPTOR KO mice showed sustained mTORC1 activation upon fasting, and that acute treatment with rapamycin was sufficient to normalize glycemia in these mice. CONCLUSION: We propose a model in which hepatic DEPTOR accelerates the inhibition of mTORC1 during the transition to fasting to adjust metabolism to the nutritional status.


Subject(s)
Blood Glucose/metabolism , Fasting/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Liver/metabolism , Animals , Cytochromes c/metabolism , Glycogen/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...