Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Clin Sci (Lond) ; 138(4): 173-187, 2024 02 21.
Article in English | MEDLINE | ID: mdl-38315575

ABSTRACT

Semaglutide is an anti-diabetes and weight loss drug that decreases food intake, slows gastric emptying, and increases insulin secretion. Patients begin treatment with low-dose semaglutide and increase dosage over time as efficacy plateaus. With increasing dosage, there is also greater incidence of gastrointestinal side effects. One reason for the plateau in semaglutide efficacy despite continued low food intake is due to compensatory actions whereby the body becomes more metabolically efficient to defend against further weight loss. Mitochondrial uncoupler drugs decrease metabolic efficiency, therefore we sought to investigate the combination therapy of semaglutide with the mitochondrial uncoupler BAM15 in diet-induced obese mice. Mice were fed high-fat western diet (WD) and stratified into six treatment groups including WD control, BAM15, low-dose semaglutide without or with BAM15, and high-dose semaglutide without or with BAM15. Combining BAM15 with either semaglutide dose decreased body fat and liver triglycerides, which was not achieved by any monotherapy, while high-dose semaglutide with BAM15 had the greatest effect on glucose homeostasis. This study demonstrates a novel approach to improve weight loss without loss of lean mass and improve glucose control by simultaneously targeting energy intake and energy efficiency. Such a combination may decrease the need for semaglutide dose escalation and hence minimize potential gastrointestinal side effects.


Subject(s)
Energy Intake , Weight Loss , Humans , Animals , Mice , Mice, Obese , Diet, High-Fat/adverse effects , Adipose Tissue
2.
Biochim Biophys Acta Mol Basis Dis ; 1870(1): 166908, 2024 01.
Article in English | MEDLINE | ID: mdl-37793464

ABSTRACT

Metabolic disorders such as type 2 diabetes, fatty liver disease, hyperlipidemia, and obesity commonly co-occur but clinical treatment options do not effectively target all disorders. Calorie restriction, semaglutide, rosiglitazone, and mitochondrial uncouplers have all demonstrated efficacy against one or more obesity-related metabolic disorders, but it currently remains unclear which therapeutic strategy best targets the combination of hyperglycaemia, liver fat, hypertriglyceridemia, and adiposity. Herein we performed a head-to-head comparison of 5 treatment interventions in the female db/db mouse model of severe metabolic disease. Treatments included Ć¢ĀˆĀ¼60Ā % calorie restriction (CR), semaglutide, rosiglitazone, BAM15, and niclosamide ethanolamine (NEN). Results showed that BAM15 and CR improved body weight and liver steatosis to levels superior to semaglutide, NEN, and rosiglitazone, while BAM15, semaglutide, and rosiglitazone improved glucose tolerance better than CR and NEN. BAM15, CR, semaglutide, and rosiglitazone all had efficacy against hypertriglyceridaemia. These data provide a comprehensive head-to-head comparison of several key treatment strategies for metabolic disease and highlight the efficacy of mitochondrial uncoupling to correct multiple facets of the metabolic disease milieu in female db/db mice.


Subject(s)
Diabetes Mellitus, Type 2 , Non-alcoholic Fatty Liver Disease , Mice , Animals , Female , Niclosamide/therapeutic use , Rosiglitazone/pharmacology , Rosiglitazone/therapeutic use , Ethanolamine/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Caloric Restriction , Ethanolamines/therapeutic use , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/drug therapy , Obesity/metabolism
3.
Diabetes ; 73(3): 374-384, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-37870907

ABSTRACT

Excess body fat is a risk factor for metabolic diseases and is a leading preventable cause of morbidity and mortality worldwide. There is a strong need to find new treatments that decrease the burden of obesity and lower the risk of obesity-related comorbidities, including cardiovascular disease and type 2 diabetes. Pharmacologic mitochondrial uncouplers represent a potential treatment for obesity through their ability to increase nutrient oxidation. Herein, we report the inĀ vitro and inĀ vivo characterization of compound SHD865, the first compound to be studied inĀ vivo in a newly discovered class of imidazolopyrazine mitochondrial uncouplers. SHD865 is a derivative of the furazanopyrazine uncoupler BAM15. SHD865 is a milder mitochondrial uncoupler than BAM15 that results in a lower maximal respiration rate. In a mouse model of diet-induced adiposity, 6-week treatment with SHD865 completely restored normal body composition and glucose tolerance to levels like those of chow-fed controls, without altering food intake. SHD865 treatment also corrected liver steatosis and plasma hyperlipidemia to normal levels comparable with chow-fed controls. SHD865 has maximal oral bioavailability in rats and slow clearance in human microsomes and hepatocytes. Collectively, these data identify the potential of imidazolopyrazine mitochondrial uncouplers as drug candidates for the treatment of obesity-related disorders.


Subject(s)
Diabetes Mellitus, Type 2 , Glucose Intolerance , Mice , Rats , Humans , Animals , Adiposity , Glucose Intolerance/drug therapy , Glucose Intolerance/metabolism , Diabetes Mellitus, Type 2/metabolism , Obesity/etiology , Liver/metabolism , Diet, High-Fat/adverse effects , Mice, Inbred C57BL
4.
Mol Metab ; 69: 101684, 2023 03.
Article in English | MEDLINE | ID: mdl-36731653

ABSTRACT

OBJECTIVE: Calorie restriction is a first-line treatment for overweight individuals with metabolic impairments. However, few patients can adhere to long-term calorie restriction. An alternative approach to calorie restriction that also causes negative energy balance is mitochondrial uncoupling, which decreases the amount of energy that can be extracted from food. Herein we compare the metabolic effects of calorie restriction with the mitochondrial uncoupler BAM15 in the db/db mouse model of severe hyperglycemia, obesity, hypertriglyceridemia, and fatty liver. METHODS: Male db/db mice were treated with Ć¢ĀˆĀ¼50% calorie restriction, BAM15 at two doses of 0.1% and 0.2% (w/w) admixed in diet, or 0.2% BAM15 with time-restricted feeding from 5 weeks of age. Mice were metabolically phenotyped over 4 weeks with assessment of key readouts including body weight, glucose tolerance, and liver steatosis. At termination, liver tissues were analysed by metabolomics and qPCR. RESULTS: Calorie restriction and high-dose 0.2% BAM15 decreased body weight to a similar extent, but mice treated with BAM15 had far better improvement in glucose control. High-dose BAM15 treatment completely normalized fasting glucose and glucose tolerance to levels similar to lean db/+ control mice. Low-dose 0.1% BAM15 did not affect body mass but partially improved glucose tolerance to a similar degree as 50% calorie restriction. Both calorie restriction and high-dose BAM15 significantly improved hyperglucagonemia and liver and serum triglyceride levels. Combining high-dose BAM15 with time-restricted feeding to match the time that calorie restricted mice were fed resulted in the best metabolic phenotype most similar to lean db/+ controls. BAM15-mediated improvements in glucose control were associated with decreased glucagon levels and decreased expression of enzymes involved in hepatic gluconeogenesis. CONCLUSIONS: BAM15 and calorie restriction treatments improved most metabolic disease phenotypes in db/db mice. However, mice fed BAM15 had superior effects on glucose control compared to the calorie restricted group that consumed half as much food. Submaximal dosing with BAM15 demonstrated that its beneficial effects on glucose control are independent of weight loss. These data highlight the potential for mitochondrial uncoupler pharmacotherapies in the treatment of metabolic disease.


Subject(s)
Fatty Liver , Metabolic Diseases , Male , Mice , Animals , Caloric Restriction , Blood Glucose/analysis , Body Weight , Glucose , Mice, Inbred Strains
5.
Cancer Metab ; 9(1): 36, 2021 Oct 09.
Article in English | MEDLINE | ID: mdl-34627389

ABSTRACT

BACKGROUND: Enhanced metabolic plasticity and diversification of energy production is a hallmark of highly proliferative breast cancers. This contributes to poor pharmacotherapy efficacy, recurrence, and metastases. We have previously identified a mitochondrial-targeted furazano[3,4-b]pyrazine named BAM15 that selectively reduces bioenergetic coupling efficiency and is orally available. Here, we evaluated the antineoplastic properties of uncoupling oxidative phosphorylation from ATP production in breast cancer using BAM15. METHODS: The anticancer effects of BAM15 were evaluated in human triple-negative MDA-MB-231 and murine luminal B, ERα-negative EO771 cells as well as in an orthotopic allograft model of highly proliferative mammary cancer in mice fed a standard or high fat diet (HFD). Untargeted transcriptomic profiling of MDA-MB-231 cells was conducted after 16-h exposure to BAM15. Additionally, oxidative phosphorylation and electron transfer capacity was determined in permeabilized cells and excised tumor homogenates after treatment with BAM15. RESULTS: BAM15 increased proton leak and over time, diminished cell proliferation, migration, and ATP production in both MDA-MB-231 and EO771 cells. Additionally, BAM15 decreased mitochondrial membrane potential, while inducing apoptosis and reactive oxygen species accumulation in MDA-MB-231 and EO771 cells. Untargeted transcriptomic profiling of MDA-MB-231 cells further revealed inhibition of signatures associated with cell survival and energy production by BAM15. In lean mice, BAM15 lowered body weight independent of food intake and slowed tumor progression compared to vehicle-treated controls. In HFD mice, BAM15 reduced tumor growth relative to vehicle and calorie-restricted weight-matched controls mediated in part by impaired cell proliferation, mitochondrial respiratory function, and ATP production. LC-MS/MS profiling of plasma and tissues from BAM15-treated animals revealed distribution of BAM15 in adipose, liver, and tumor tissue with low abundance in skeletal muscle. CONCLUSIONS: Collectively, these data indicate that mitochondrial uncoupling may be an effective strategy to limit proliferation of aggressive forms of breast cancer. More broadly, these findings highlight the metabolic vulnerabilities of highly proliferative breast cancers which may be leveraged in overcoming poor responsiveness to existing therapies.

6.
Metabolism ; 117: 154724, 2021 04.
Article in English | MEDLINE | ID: mdl-33548253

ABSTRACT

AIMS: Mitochondrial uncouplers decrease caloric efficiency and have potential therapeutic benefits for the treatment of obesity and related metabolic disorders. Herein we investigate the metabolic and physiologic effects of a recently identified small molecule mitochondrial uncoupler named SHC517 in a mouse model of diet-induced obesity. METHODS: SHC517 was administered as an admixture in food. The effect of SHC517 on in vivo energy expenditure and respiratory quotient was determined by indirect calorimetry. A dose-finding obesity prevention study was performed by starting SHC517 treatment concomitant with high fat diet for a period of 12Ć¢Ā€ĀÆdays. An obesity reversal study was performed by feeding mice western diet for 4Ć¢Ā€ĀÆweeks prior to SHC517 treatment for 7Ć¢Ā€ĀÆweeks. Biochemical assays were used to determine changes in glucose, insulin, triglycerides, and cholesterol. SHC517 concentrations were determined by mass spectrometry. RESULTS: SHC517 increased lipid oxidation without affecting body temperature. SHC517 prevented diet-induced obesity when administered at 0.05% and 0.1% w/w in high fat diet and reversed established obesity when tested at the 0.05% dose. In the obesity reversal model, SHC517 restored adiposity to levels similar to chow-fed control mice without affecting food intake or lean body mass. SHC517 improved glucose tolerance and fasting glucose levels when administered in both the obesity prevention and obesity reversal modes. CONCLUSIONS: SHC517 is a mitochondrial uncoupler with potent anti-obesity and insulin sensitizing effects in mice. SHC517 reversed obesity without altering food intake or compromising lean mass, effects that are highly sought-after in anti-obesity therapeutics.


Subject(s)
Eating/drug effects , Mitochondria/drug effects , Obesity/drug therapy , Small Molecule Libraries/pharmacology , Adiposity/drug effects , Animals , Body Weight/drug effects , Calorimetry, Indirect/methods , Diet, High-Fat/adverse effects , Diet, Western/adverse effects , Energy Metabolism/drug effects , Glucose/metabolism , Insulin/metabolism , Insulin Resistance/physiology , Lipid Metabolism/drug effects , Male , Metabolic Diseases/drug therapy , Metabolic Diseases/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Obesity/metabolism
7.
Sci Rep ; 10(1): 8182, 2020 05 18.
Article in English | MEDLINE | ID: mdl-32424263

ABSTRACT

The only enzyme in the glycosphingolipid (GSL) metabolic pathway, which produces glucosylceramide (GlcCer) de novo is UDP-glucose ceramide glucosyltransferase (UGCG). UGCG is linked to pro-cancerous processes such as multidrug resistance development and increased proliferation in several cancer types. Previously, we showed an UGCG-dependent glutamine metabolism adaption to nutrient-poor environment of breast cancer cells. This adaption includes reinforced oxidative stress response and fueling the tricarboxylic acid (TCA) cycle by increased glutamine oxidation. In the current study, we investigated glycolytic and oxidative metabolic phenotypes following UGCG overexpression (OE). UGCG overexpressing MCF-7 cells underwent a metabolic shift from quiescent/aerobic to energetic metabolism by increasing both glycolysis and oxidative glucose metabolism. The energetic metabolic phenotype was not associated with increased mitochondrial mass, however, markers of mitochondrial turnover were increased. UGCG OE altered sphingolipid composition of the endoplasmic reticulum (ER)/mitochondria fractions that may contribute to increased mitochondrial turnover and increased cell metabolism. Our data indicate that GSL are closely connected to cell energy metabolism and this finding might contribute to development of novel therapeutic strategies for cancer treatment.


Subject(s)
Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Glucosyltransferases/metabolism , Glycolysis , Oxidative Phosphorylation , Endoplasmic Reticulum/metabolism , Energy Metabolism , Glucosyltransferases/genetics , Humans , MCF-7 Cells , Mitochondria/metabolism
8.
J Med Chem ; 63(11): 6203-6224, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32392051

ABSTRACT

Small molecule mitochondrial uncouplers have recently garnered great interest for their potential in treating nonalcoholic steatohepatitis (NASH). In this study, we report the structure-activity relationship profiling of a 6-amino[1,2,5]oxadiazolo[3,4-b]pyrazin-5-ol core, which utilizes the hydroxy moiety as the proton transporter across the mitochondrial inner membrane. We demonstrate that a wide array of substituents is tolerated with this novel scaffold that increased cellular metabolic rates in vitro using changes in oxygen consumption rate as a readout. In particular, compound SHS4121705 (12i) displayed an EC50 of 4.3 ĀµM in L6 myoblast cells and excellent oral bioavailability and liver exposure in mice. In the STAM mouse model of NASH, administration of 12i at 25 mg kg-1 day-1 lowered liver triglyceride levels and improved liver markers such as alanine aminotransferase, NAFLD activity score, and fibrosis. Importantly, no changes in body temperature or food intake were observed. As potential treatment of NASH, mitochondrial uncouplers show promise for future development.


Subject(s)
Pyrazines/chemistry , Alanine Transaminase/metabolism , Aniline Compounds/chemistry , Animals , Cell Line , Diet, High-Fat , Disease Models, Animal , Half-Life , Liver/drug effects , Liver/metabolism , Mice , Mitochondria/metabolism , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/pathology , Oxygen Consumption/drug effects , Pyrazines/pharmacokinetics , Pyrazines/pharmacology , Pyrazines/therapeutic use , Rats , Structure-Activity Relationship , Triglycerides/metabolism , Uncoupling Protein 1/chemistry , Uncoupling Protein 1/metabolism
9.
J Med Chem ; 63(5): 2511-2526, 2020 03 12.
Article in English | MEDLINE | ID: mdl-32017849

ABSTRACT

Small molecule mitochondrial uncouplers are emerging as a new class of molecules for the treatment of nonalcoholic steatohepatitis. We utilized BAM15, a potent protonophore that uncouples the mitochondria without depolarizing the plasma membrane, as a lead compound for structure-activity profiling. Using oxygen consumption rate as an assay for determining uncoupling activity, changes on the 5- and 6-position of the oxadiazolopyrazine core were introduced. Our studies suggest that unsymmetrical aniline derivatives bearing electron withdrawing groups are preferred compared to the symmetrical counterparts. In addition, alkyl substituents are not tolerated, and the N-H proton of the aniline ring is responsible for the protonophore activity. In particular, compound 10b had an EC50 value of 190 nM in L6 myoblast cells. In an in vivo model of NASH, 10b decreased liver triglyceride levels and showed improvement in fibrosis, inflammation, and plasma ALT. Taken together, our studies indicate that mitochondrial uncouplers have potential for the treatment of NASH.


Subject(s)
Diamines/therapeutic use , Mitochondria, Liver/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Pyrazines/therapeutic use , Uncoupling Agents/therapeutic use , Animals , Diamines/chemistry , Diamines/pharmacology , Liver/drug effects , Liver/metabolism , Male , Mice, Inbred C57BL , Mitochondria, Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Oxadiazoles/chemistry , Oxadiazoles/pharmacology , Oxadiazoles/therapeutic use , Oxygen Consumption/drug effects , Pyrazines/chemistry , Pyrazines/pharmacology , Uncoupling Agents/chemistry , Uncoupling Agents/pharmacology
10.
Nat Commun ; 11(1): 2922, 2020 06 10.
Article in English | MEDLINE | ID: mdl-32523103

ABSTRACT

The conversion of white adipocytes to thermogenic beige adipocytes represents a potential mechanism to treat obesity and related metabolic disorders. However, the mechanisms involved in converting white to beige adipose tissue remain incompletely understood. Here we show profound beiging in a genetic mouse model lacking the transcriptional repressor KrĆ¼ppel-like factor 3 (KLF3). Bone marrow transplants from these animals confer the beige phenotype on wild type recipients. Analysis of the cellular and molecular changes reveal an accumulation of eosinophils in adipose tissue. We examine the transcriptomic profile of adipose-resident eosinophils and posit that KLF3 regulates adipose tissue function via transcriptional control of secreted molecules linked to beiging. Furthermore, we provide evidence that eosinophils may directly act on adipocytes to drive beiging and highlight the critical role of these little-understood immune cells in thermogenesis.


Subject(s)
Adipose Tissue/metabolism , Eosinophils/metabolism , Kruppel-Like Transcription Factors/metabolism , Signal Transduction/physiology , Adiposity/genetics , Adiposity/physiology , Animals , COS Cells , Chlorocebus aethiops , Chromatin Immunoprecipitation , Flow Cytometry , Kruppel-Like Transcription Factors/genetics , Male , Mice , Obesity/metabolism , Signal Transduction/genetics , Software
11.
EMBO Mol Med ; 12(7): e12088, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32519812

ABSTRACT

Obesity is a leading cause of preventable death worldwide. Despite this, current strategies for the treatment of obesity remain ineffective at achieving long-term weight control. This is due, in part, to difficulties in identifying tolerable and efficacious small molecules or biologics capable of regulating systemic nutrient homeostasis. Here, we demonstrate that BAM15, a mitochondrially targeted small molecule protonophore, stimulates energy expenditure and glucose and lipid metabolism to protect against diet-induced obesity. Exposure to BAM15 inĀ vitro enhanced mitochondrial respiratory kinetics, improved insulin action, and stimulated nutrient uptake by sustained activation of AMPK. C57BL/6J mice treated with BAM15 were resistant to weight gain. Furthermore, BAM15-treated mice exhibited improved body composition and glycemic control independent of weight loss, effects attributable to drug targeting of lipid-rich tissues. We provide the first phenotypic characterization and demonstration of pre-clinical efficacy for BAM15 as a pharmacological approach for the treatment of obesity and related diseases.


Subject(s)
Glucose/metabolism , Glycemic Control , Mitochondria/drug effects , Mitochondria/metabolism , Obesity/metabolism , Obesity/prevention & control , Uncoupling Agents/pharmacology , Animals , Diet, High-Fat/adverse effects , Energy Metabolism/drug effects , Glycemic Control/methods , Insulin Resistance , Male , Mice , Mice, Inbred C57BL
12.
Redox Biol ; 28: 101374, 2020 01.
Article in English | MEDLINE | ID: mdl-31743887

ABSTRACT

A hallmark of cancer cells is their ability to reprogram nutrient metabolism. Thus, disruption to this phenotype is a potential avenue for anti-cancer therapy. Herein we used a phenotypic chemical library screening approach to identify molecules that disrupted nutrient metabolism (by increasing cellular oxygen consumption rate) and were toxic to cancer cells. From this screen we discovered a 1,4-Naphthoquinone (referred to as BH10) that is toxic to a broad range of cancer cell types. BH10 has improved cancer-selective toxicity compared to doxorubicin, 17-AAG, vitamin K3, and other known anti-cancer quinones. BH10 increases glucose oxidation via both mitochondrial and pentose phosphate pathways, decreases glycolysis, lowers GSH:GSSG and NAPDH/NAPD+ ratios exclusively in cancer cells, and induces necrosis. BH10 targets mitochondrial redox defence as evidenced by increased mitochondrial peroxiredoxin 3 oxidation and decreased mitochondrial aconitase activity, without changes in markers of cytosolic or nuclear damage. Over-expression of mitochondria-targeted catalase protects cells from BH10-mediated toxicity, while the thioredoxin reductase inhibitor auranofin synergistically enhances BH10-induced peroxiredoxin 3 oxidation and cytotoxicity. Overall, BH10 represents a 1,4-Naphthoquinone with an improved cancer-selective cytotoxicity profile via its mitochondrial specificity.


Subject(s)
Mitochondria/metabolism , Naphthoquinones/pharmacology , Oxidative Stress/drug effects , Oxygen Consumption/drug effects , Benzoquinones/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/pharmacology , Glycolysis/drug effects , Humans , Lactams, Macrocyclic/pharmacology , Mitochondria/drug effects , Phenotype , Small Molecule Libraries/pharmacology , Vitamin K 3/pharmacology
13.
Nat Commun ; 11(1): 2397, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32409697

ABSTRACT

Obesity is a health problem affecting more than 40% of US adults and 13% of the global population. Anti-obesity treatments including diet, exercise, surgery and pharmacotherapies have so far failed to reverse obesity incidence. Herein, we target obesity with a pharmacotherapeutic approach that decreases caloric efficiency by mitochondrial uncoupling. We show that a recently identified mitochondrial uncoupler BAM15 is orally bioavailable, increases nutrient oxidation, and decreases body fat mass without altering food intake, lean body mass, body temperature, or biochemical and haematological markers of toxicity. BAM15 decreases hepatic fat, decreases inflammatory lipids, and has strong antioxidant effects. Hyperinsulinemic-euglycemic clamp studies show that BAM15 improves insulin sensitivity in multiple tissue types. Collectively, these data demonstrate that pharmacologic mitochondrial uncoupling with BAM15 has powerful anti-obesity and insulin sensitizing effects without compromising lean mass or affecting food intake.


Subject(s)
Diamines/administration & dosage , Insulin Resistance , Mitochondria/drug effects , Obesity/drug therapy , Oxadiazoles/administration & dosage , Pyrazines/administration & dosage , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Administration, Oral , Animals , Blood Glucose/analysis , Body Temperature/drug effects , Body Weight/drug effects , Diamines/adverse effects , Diet, Western/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , Glucose Clamp Technique , Humans , Liver/drug effects , Liver/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/metabolism , Obesity/blood , Obesity/etiology , Obesity/metabolism , Oxadiazoles/adverse effects , Oxidative Stress/drug effects , Pyrazines/adverse effects
14.
J Med Chem ; 61(11): 4641-4655, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29156129

ABSTRACT

Small molecule mitochondrial uncouplers transport protons from the mitochondrial inner membrane space into the mitochondrial matrix independent of ATP synthase, uncoupling nutrient metabolism from ATP generation. The therapeutic potential of mitochondrial uncouplers has been investigated for the treatment of metabolic diseases such as obesity and type 2 diabetes (T2D), ischemia-reperfusion injury, and neurodegenerative diseases. This communication will review the small molecule mitochondrial uncouplers reported to date and explore their potential as therapeutics.


Subject(s)
Mitochondria/drug effects , Mitochondria/metabolism , Small Molecule Libraries/pharmacology , Animals , Biological Products/pharmacology , Biological Products/therapeutic use , Cell Respiration/drug effects , Drug Synergism , Humans , Small Molecule Libraries/therapeutic use
15.
Chem Phys Lipids ; 199: 170-178, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26993747

ABSTRACT

Cholesterol is vital in mammals, but toxic in excess. Consequently, elaborate molecular mechanisms have evolved to maintain this sterol within narrow limits. How cells sense excess cholesterol is an intriguing area of research. Cells sense cholesterol, and other related sterols such as oxysterols or cholesterol synthesis intermediates, and respond to changing levels through several elegant mechanisms of feedback regulation. Cholesterol sensing involves both direct binding of sterols to the homeostatic machinery located in the endoplasmic reticulum (ER), and indirect effects elicited by sterol-dependent alteration of the physical properties of membranes. Here, we examine the mechanisms employed by cells to maintain cholesterol homeostasis.


Subject(s)
Cells/metabolism , Homeostasis , Cell Membrane/metabolism , Cells/cytology , Cholesterol/metabolism , Endoplasmic Reticulum/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL