71 results on '"Gerhart-Hines Z"'
Search Results
2. Metabolic control of muscle mitochondrial function and fatty acid oxidation through SIRT1/PGC-1 alpha
- Author
-
Gerhart-Hines Z, Rodgers JT, Bare O, Lerin C, Kim SH, Mostoslavsky R, Alt FW, Wu Z, and Puigserver P
- Published
- 2007
3. Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1 alpha
- Author
-
Lagouge M, Argmann C, Gerhart-Hines Z, Meziane H, Lerin-Martinez C, Daussin F, Messadeq N, Milne J, Lambert P, Elliott P, Geny B, Laakso M, Puigserver P, and Auwerx J
- Published
- 2006
4. Nutrient-Dependent Acetylation Controls Basic Regulatory Metabolic Switches and Cellular Reprogramming
- Author
-
Dominy, J. E., primary, Gerhart-Hines, Z., additional, and Puigserver, P., additional
- Published
- 2011
- Full Text
- View/download PDF
5. Analysis of the Nr1d1 cistrome in mouse brown adipocytes
- Author
-
Gerhart-Hines, Z, primary
- Full Text
- View/download PDF
6. NK2R control of energy expenditure and feeding to treat metabolic diseases.
- Author
-
Sass F, Ma T, Ekberg JH, Kirigiti M, Ureña MG, Dollet L, Brown JM, Basse AL, Yacawych WT, Burm HB, Andersen MK, Nielsen TS, Tomlinson AJ, Dmytiyeva O, Christensen DP, Bader L, Vo CT, Wang Y, Rausch DM, Kristensen CK, Gestal-Mato M, In Het Panhuis W, Sjøberg KA, Kernodle S, Petersen JE, Pavlovskyi A, Sandhu M, Moltke I, Jørgensen ME, Albrechtsen A, Grarup N, Babu MM, Rensen PCN, Kooijman S, Seeley RJ, Worthmann A, Heeren J, Pers TH, Hansen T, Gustafsson MBF, Tang-Christensen M, Kilpeläinen TO, Myers MG Jr, Kievit P, Schwartz TW, Hansen JB, and Gerhart-Hines Z
- Abstract
The combination of decreasing food intake and increasing energy expenditure represents a powerful strategy for counteracting cardiometabolic diseases such as obesity and type 2 diabetes
1 . Yet current pharmacological approaches require conjugation of multiple receptor agonists to achieve both effects2-4 , and so far, no safe energy-expending option has reached the clinic. Here we show that activation of neurokinin 2 receptor (NK2R) is sufficient to suppress appetite centrally and increase energy expenditure peripherally. We focused on NK2R after revealing its genetic links to obesity and glucose control. However, therapeutically exploiting NK2R signalling has previously been unattainable because its endogenous ligand, neurokinin A, is short-lived and lacks receptor specificity5,6 . Therefore, we developed selective, long-acting NK2R agonists with potential for once-weekly administration in humans. In mice, these agonists elicit weight loss by inducing energy expenditure and non-aversive appetite suppression that circumvents canonical leptin signalling. Additionally, a hyperinsulinaemic-euglycaemic clamp reveals that NK2R agonism acutely enhances insulin sensitization. In diabetic, obese macaques, NK2R activation significantly decreases body weight, blood glucose, triglycerides and cholesterol, and ameliorates insulin resistance. These findings identify a single receptor target that leverages both energy-expending and appetite-suppressing programmes to improve energy homeostasis and reverse cardiometabolic dysfunction across species., Competing Interests: Competing interests T.M., J.H.E., J.B.H., D.P.C., M.B.F.G., M.T.-C., T.W.S. and Z.G.-H. work or have worked in some capacity for Embark Laboratories ApS, a company developing therapeutics for the treatment of T2D and obesity. P.K. is a consultant for Embark Laboratories ApS. The use of and chemical composition for NK2R agonists are patented by the University of Copenhagen and Embark Laboratories ApS, respectively. R.J.S. has received research support from Novo Nordisk, Fractyl, AstraZeneca, Congruence Therapeutics, Eli Lilly, Bullfrog AI, Glycsend Therapeutics and Amgen. R.J.S. has served as a paid consultant for Novo Nordisk, Eli Lilly, CinRx, Fractyl, Structure Therapeutics, Crinetics and Congruence Therapeutics. R.J.S. has equity in Calibrate, Rewind and Levator Therapeutics. The other authors declare no competing interests., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
7. Exposing mice to extremely hypertonic treatments: a recurring problem in lactate research.
- Author
-
Lund J, Gerhart-Hines Z, and Clemmensen C
- Published
- 2024
- Full Text
- View/download PDF
8. Cardiolipin deficiency disrupts CoQ redox state and induces steatohepatitis.
- Author
-
Brothwell MJ, Cao 曹国燊 G, Maschek JA, Poss AM, Peterlin AD, Wang 汪立平 L, Baker TB, Shahtout JL, Siripoksup P, Pearce QJ, Johnson JM, Finger FM, Prola A, Pellizzari SA, Hale GL, Manuel AM, Watanabe 渡邉真也 S, Miranda ER, Affolter KE, Tippetts TS, Nikolova LS, Choi 崔蘭煕 RH, Decker ST, Patil M, Catrow JL, Holland WL, Nowinski SM, Lark DS, Fisher-Wellman KH, Mimche PN, Evason KJ, Cox JE, Summers SA, Gerhart-Hines Z, and Funai 船井勝彦 K
- Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a progressive disorder marked by lipid accumulation, leading to steatohepatitis (MASH). A key feature of the transition to MASH involves oxidative stress resulting from defects in mitochondrial oxidative phosphorylation (OXPHOS). Here, we show that pathological alterations in the lipid composition of the inner mitochondrial membrane (IMM) directly instigate electron transfer inefficiency to promote oxidative stress. Specifically, cardiolipin (CL) was downregulated across four mouse models of MASLD. Hepatocyte-specific CL synthase knockout (CLS-LKO) led to spontaneous MASH with elevated mitochondrial electron leak. Loss of CL interfered with the ability of coenzyme Q (CoQ) to transfer electrons, promoting leak primarily at sites II
F and IIIQ0 . Data from human liver biopsies revealed a highly robust correlation between mitochondrial CL and CoQ, co-downregulated with MASH. Thus, reduction in mitochondrial CL promotes oxidative stress and contributes to pathogenesis of MASH.- Published
- 2024
- Full Text
- View/download PDF
9. Mild Cold Stress at Ambient Temperature Elevates Muscle Calcium Cycling and Exercise Adaptations in Obese Female Mice.
- Author
-
Raun SH, Braun JL, Karavaeva I, Henriquez-Olguín C, Ali MS, Møller LLV, Gerhart-Hines Z, Fajardo VA, Richter EA, and Sylow L
- Subjects
- Animals, Female, Mice, Mice, Inbred C57BL, Cold-Shock Response physiology, Mice, Obese, Cold Temperature, Adiposity physiology, Physical Conditioning, Animal physiology, Muscle, Skeletal metabolism, Obesity metabolism, Obesity physiopathology, Calcium metabolism, Adaptation, Physiological physiology
- Abstract
Context: Housing temperature is a critical regulator of mouse metabolism and thermoneutral housing can improve model translation to humans. However, the impact of housing temperature on the ability of wheel running exercise training to rescue the detrimental effect of diet-induced obese mice is currently not fully understood., Objective: To investigate how housing temperature affects muscle metabolism in obese mice with regard to calcium handling and exercise training (ET) adaptations in skeletal muscle, and benefits of ET on adiposity and glucometabolic parameters., Methods: Lean or obese female mice were housed at standard ambient temperature (22 °C) or thermoneutrality (30 °C) with/without access to running wheels. The metabolic phenotype was investigated using glucose tolerance tests, indirect calorimetry, and body composition. Molecular muscle adaptations were measured using immunoblotting, qPCR, and spectrophotometric/fluorescent assays., Results: Obese female mice housed at 22 °C showed lower adiposity, lower circulating insulin levels, improved glucose tolerance, and elevated basal metabolic rate compared to 30 °C housing. Mice exposed to voluntary wheel running exhibited a larger fat loss and higher metabolic rate at 22 °C housing compared to thermoneutrality. In obese female mice, glucose tolerance improved after ET independent of housing temperature. Independent of diet and training, 22 °C housing increased skeletal muscle sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA) activity. Additionally, housing at 22 °C elevated the induction of training-responsive muscle proteins in obese mice., Conclusion: Our findings highlight that housing temperature significantly influences adiposity, insulin sensitivity, muscle physiology, and exercise adaptations in diet-induced obese female mice., (© The Author(s) 2024. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For commercial re-use, please contact reprints@oup.com for reprints and translation rights for reprints. All other permissions can be obtained through our RightsLink service via the Permissions link on the article page on our site—for further information please contact journals.permissions@oup.com. See the journal About page for additional terms.)
- Published
- 2024
- Full Text
- View/download PDF
10. MCT1 helps brown fat suck up succinate.
- Author
-
Lund J, Isidor MS, and Gerhart-Hines Z
- Subjects
- Succinates, Adipose Tissue, Brown, Succinic Acid
- Published
- 2024
- Full Text
- View/download PDF
11. ROS-induced ribosome impairment underlies ZAKα-mediated metabolic decline in obesity and aging.
- Author
-
Snieckute G, Ryder L, Vind AC, Wu Z, Arendrup FS, Stoneley M, Chamois S, Martinez-Val A, Leleu M, Dreos R, Russell A, Gay DM, Genzor AV, Choi BS, Basse AL, Sass F, Dall M, Dollet LCM, Blasius M, Willis AE, Lund AH, Treebak JT, Olsen JV, Poulsen SS, Pownall ME, Jensen BAH, Clemmensen C, Gerhart-Hines Z, Gatfield D, and Bekker-Jensen S
- Subjects
- Animals, Mice, Protein Biosynthesis, Zebrafish, Mice, Knockout, Aging metabolism, MAP Kinase Kinase Kinase 3 genetics, MAP Kinase Kinase Kinase 3 metabolism, Obesity metabolism, Reactive Oxygen Species metabolism, Ribosomes metabolism, Stress, Physiological
- Abstract
The ribotoxic stress response (RSR) is a signaling pathway in which the p38- and c-Jun N-terminal kinase (JNK)-activating mitogen-activated protein kinase kinase kinase (MAP3K) ZAKα senses stalling and/or collision of ribosomes. Here, we show that reactive oxygen species (ROS)-generating agents trigger ribosomal impairment and ZAKα activation. Conversely, zebrafish larvae deficient for ZAKα are protected from ROS-induced pathology. Livers of mice fed a ROS-generating diet exhibit ZAKα-activating changes in ribosomal elongation dynamics. Highlighting a role for the RSR in metabolic regulation, ZAK-knockout mice are protected from developing high-fat high-sugar (HFHS) diet-induced blood glucose intolerance and liver steatosis. Finally, ZAK ablation slows animals from developing the hallmarks of metabolic aging. Our work highlights ROS-induced ribosomal impairment as a physiological activation signal for ZAKα that underlies metabolic adaptation in obesity and aging.
- Published
- 2023
- Full Text
- View/download PDF
12. Editorial overview: Fat tissue in focus: Assembled deeply insightful perspectives on state-of-the-art explorations.
- Author
-
Gerhart-Hines Z and Loos RJ
- Subjects
- Adipose Tissue
- Abstract
Competing Interests: Conflict of interest statement ZGH is a co-founder and Chief Scientifc Officer of Embark Biotech, which was acquired by Novo Nordisk in August 2023, and is a co-founder and Chief Technology Officer of Embark Laboratories, which formed a partnership with Novo Nordisk in September 2023. The focus of both Embark Biotech and Embark Laboratories is to develop novel treatments for obesity and type 2 diabetes.
- Published
- 2023
- Full Text
- View/download PDF
13. Is lactate a driver of skin burn-induced adipose browning?
- Author
-
Lund J, Johansen VBI, Clemmensen C, and Gerhart-Hines Z
- Subjects
- Humans, Adiposity, Obesity, Lactic Acid, Burns
- Published
- 2023
- Full Text
- View/download PDF
14. Effect of Simvastatin Treatment on Mitochondrial Function and Inflammatory Status of Human White Adipose Tissue.
- Author
-
Christensen IB, Blom I, Dohlmann TL, Finger F, Helge JW, Gerhart-Hines Z, Dela F, and Larsen S
- Subjects
- Male, Female, Humans, Interleukin-6 metabolism, Cross-Sectional Studies, Mitochondria metabolism, Adipose Tissue, White metabolism, Cholesterol metabolism, Adipose Tissue metabolism, Simvastatin adverse effects, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology
- Abstract
Background: Statin therapy has shown pleiotropic effects affecting both mitochondrial function and inflammatory status. However, few studies have investigated the concurrent effects of statin exposure on mitochondrial function and inflammatory status in human subcutaneous white adipose tissue., Objectives: In a cross-sectional study, we investigated the effects of simvastatin on mitochondrial function and inflammatory status in subcutaneous white adipose tissue of 55 human participants: 38 patients (19 females/19 males) in primary prevention with simvastatin (> 40 mg/d, > 3 mo) and 17 controls (9 females/8 males) with elevated plasma cholesterol. The 2 groups were matched on age, body mass index, and maximal oxygen consumption., Methods: Anthropometrics and fasting biochemical characteristics were measured. Mitochondrial respiratory capacity was assessed in white adipose tissue by high-resolution respirometry. Subcutaneous white adipose tissue expression of the inflammatory markers IL-6, chemokine (C-C motif) ligand 2 (CCL2), CCL-5, tumor necrosis factor-α, IL-10, and IL-4 was analyzed by quantitative PCR., Results: Simvastatin-treated patients showed lower plasma cholesterol (P < .0001), low-density lipoprotein (P < .0001), and triglyceride levels (P = .0116) than controls. Simvastatin-treated patients had a lower oxidative phosphorylation capacity of mitochondrial complex II (P = .0001 when normalized to wet weight, P < .0001 when normalized to citrate synthase activity [intrinsic]), and a lower intrinsic mitochondrial electron transport system capacity (P = .0004). Simvastatin-treated patients showed higher IL-6 expression than controls (P = .0202)., Conclusion: Simvastatin treatment was linked to mitochondrial respiratory capacity in human subcutaneous white adipose tissue, but no clear link was found between statin exposure, respiratory changes, and inflammatory status of adipose tissue., (© The Author(s) 2023. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
15. Acute Deletion of the Glucocorticoid Receptor in Hepatocytes Disrupts Postprandial Lipid Metabolism in Male Mice.
- Author
-
Correia CM, Præstholm SM, Havelund JF, Pedersen FB, Siersbæk MS, Ebbesen MF, Gerhart-Hines Z, Heeren J, Brewer J, Larsen S, Blagoev B, Færgeman NJ, and Grøntved L
- Subjects
- Male, Animals, Mice, Hepatocytes, Liver, Adipogenesis, Lipid Metabolism genetics, Receptors, Glucocorticoid genetics
- Abstract
Hepatic lipid metabolism is highly dynamic, and disruption of several circadian transcriptional regulators results in hepatic steatosis. This includes genetic disruption of the glucocorticoid receptor (GR) as the liver develops. To address the functional role of GR in the adult liver, we used an acute hepatocyte-specific GR knockout model to study temporal hepatic lipid metabolism governed by GR at several preprandial and postprandial circadian timepoints. Lipidomics analysis revealed significant temporal lipid metabolism, where GR disruption results in impaired regulation of specific triglycerides, nonesterified fatty acids, and sphingolipids. This correlates with increased number and size of lipid droplets and mildly reduced mitochondrial respiration, most noticeably in the postprandial phase. Proteomics and transcriptomics analyses suggest that dysregulated lipid metabolism originates from pronounced induced expression of enzymes involved in fatty acid synthesis, β-oxidation, and sphingolipid metabolism. Integration of GR cistromic data suggests that induced gene expression is a result of regulatory actions secondary to direct GR effects on gene transcription., (© The Author(s) 2023. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
16. TNIK is a conserved regulator of glucose and lipid metabolism in obesity.
- Author
-
Pham TCP, Dollet L, Ali MS, Raun SH, Møller LLV, Jafari A, Ditzel N, Andersen NR, Fritzen AM, Gerhart-Hines Z, Kiens B, Suomalainen A, Simpson SJ, Salling Olsen M, Kieser A, Schjerling P, Nieminen AI, Richter EA, Havula E, and Sylow L
- Subjects
- Animals, Mice, Diabetes Mellitus, Type 2 genetics, Glucose metabolism, Lipids, Liver metabolism, Mice, Inbred C57BL, Mice, Knockout, Insulin Resistance, Lipid Metabolism, Obesity genetics, Obesity metabolism, Protein Serine-Threonine Kinases metabolism
- Abstract
Obesity and type 2 diabetes (T2D) are growing health challenges with unmet treatment needs. Traf2- and NCK-interacting protein kinase (TNIK) is a recently identified obesity- and T2D-associated gene with unknown functions. We show that TNIK governs lipid and glucose homeostasis in Drosophila and mice. Loss of the Drosophila ortholog of TNIK , misshapen , altered the metabolite profiles and impaired de novo lipogenesis in high sugar-fed larvae. Tnik knockout mice exhibited hyperlocomotor activity and were protected against diet-induced fat expansion, insulin resistance, and hepatic steatosis. The improved lipid profile of Tnik knockout mice was accompanied by enhanced skeletal muscle and adipose tissue insulin-stimulated glucose uptake and glucose and lipid handling. Using the T2D Knowledge Portal and the UK Biobank, we observed associations of TNIK variants with blood glucose, HbA1c, body mass index, body fat percentage, and feeding behavior. These results define an untapped paradigm of TNIK-controlled glucose and lipid metabolism.
- Published
- 2023
- Full Text
- View/download PDF
17. PICK1-Deficient Mice Maintain Their Glucose Tolerance During Diet-Induced Obesity.
- Author
-
Backe MB, Andersen RC, Jensen M, Jin C, Hundahl C, Dmytriyeva O, Treebak JT, Hansen JB, Gerhart-Hines Z, Madsen KL, and Holst B
- Abstract
Context: Metabolic disorders such as obesity represent a major health challenge. Obesity alone has reached epidemic proportions, with at least 2.8 million people worldwide dying annually from diseases caused by overweight or obesity. The brain-metabolic axis is central to maintain homeostasis under metabolic stress via an intricate signaling network of hormones. Protein interacting with C kinase 1 (PICK1) is important for the biogenesis of various secretory vesicles, and we have previously shown that PICK1-deficient mice have impaired secretion of insulin and growth hormone., Objective: The aim was to investigate how global PICK1-deficient mice respond to high-fat diet (HFD) and assess its role in insulin secretion in diet-induced obesity., Methods: We characterized the metabolic phenotype through assessment of body weight, composition, glucose tolerance, islet morphology insulin secretion in vivo, and glucose-stimulated insulin secretion ex vivo., Results: PICK1-deficient mice displayed similar weight gain and body composition as wild-type (WT) mice following HFD. While HFD impaired glucose tolerance of WT mice, PICK1-deficient mice were resistant to further deterioration of their glucose tolerance compared with already glucose-impaired chow-fed PICK1-deficient mice. Surprisingly, mice with β-cell-specific knockdown of PICK1 showed impaired glucose tolerance both on chow and HFD similar to WT mice., Conclusion: Our findings support the importance of PICK1 in overall hormone regulation. However, importantly, this effect is independent of the PICK1 expression in the β-cell, whereby global PICK1-deficient mice resist further deterioration of their glucose tolerance following diet-induced obesity., (© The Author(s) 2023. Published by Oxford University Press on behalf of the Endocrine Society.)
- Published
- 2023
- Full Text
- View/download PDF
18. Full activation of thermogenesis in brown adipocytes requires Basigin action.
- Author
-
Rupar K, Isidor MS, Argemi-Muntadas L, Agueda-Oyarzabal M, Plucińska K, Brown EL, Mattanovich M, Bossi S, Tozzi M, Tandio D, Petersen PSS, Henriksen TI, Trošt K, Hansen JB, Gerhart-Hines Z, Nielsen S, Moritz T, and Emanuelli B
- Subjects
- Mice, Animals, Basigin metabolism, Lipolysis, Obesity metabolism, Thermogenesis genetics, Uncoupling Protein 1 genetics, Uncoupling Protein 1 metabolism, Adipocytes, Brown metabolism, Adipose Tissue, Brown metabolism
- Abstract
Exploring mechanisms responsible for brown adipose tissue's (BAT) high metabolic activity is crucial to exploit its energy-dissipating ability for therapeutic purposes. Basigin (Bsg), a multifunctional highly glycosylated transmembrane protein, was recently proposed as one of the 98 critical markers allowing to distinguish 'white' and 'brown' adipocytes, yet its function in thermogenic brown adipocytes is unknown. Here, we report that Bsg is negatively associated with obesity in mice. By contrast, Bsg expression increased in the mature adipocyte fraction of BAT upon cold acclimation. Additionally, Bsg levels were highly induced during brown adipocyte maturation in vitro and were further increased upon β-adrenergic stimulation in a HIF-1α-dependent manner. siRNA-mediated Bsg gene silencing in cultured brown adipocytes did not impact adipogenesis nor mitochondrial function. However, a significant decrease in mitochondrial respiration, lipolysis and Ucp1 transcription was observed in adipocytes lacking Bsg, when activated by norepinephrine. Furthermore, using gas chromatography/mass spectrometry-time-of-flight analysis to assess the composition of cellular metabolites, we demonstrate that brown adipocytes lacking Bsg have lower levels of intracellular lactate and acetoacetate. Bsg was additionally required to regulate intracellular AcAc and tricarboxylic acid cycle intermediate levels in NE-stimulated adipocytes. Our study highlights the critical role of Bsg in active brown adipocytes, possibly by controlling cellular metabolism., (© 2023 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.)
- Published
- 2023
- Full Text
- View/download PDF
19. NAMPT-dependent NAD + biosynthesis controls circadian metabolism in a tissue-specific manner.
- Author
-
Basse AL, Nielsen KN, Karavaeva I, Ingerslev LR, Ma T, Havelund JF, Nielsen TS, Frost M, Peics J, Dalbram E, Dall M, Zierath JR, Barrès R, Færgeman NJ, Treebak JT, and Gerhart-Hines Z
- Subjects
- Animals, Circadian Rhythm physiology, Adipose Tissue, Brown metabolism, Obesity metabolism, Cytokines metabolism, NAD metabolism, Nicotinamide Phosphoribosyltransferase genetics, Nicotinamide Phosphoribosyltransferase metabolism
- Abstract
Molecular clocks in the periphery coordinate tissue-specific daily biorhythms by integrating input from the hypothalamic master clock and intracellular metabolic signals. One such key metabolic signal is the cellular concentration of NAD
+ , which oscillates along with its biosynthetic enzyme, nicotinamide phosphoribosyltransferase (NAMPT). NAD+ levels feed back into the clock to influence rhythmicity of biological functions, yet whether this metabolic fine-tuning occurs ubiquitously across cell types and is a core clock feature is unknown. Here, we show that NAMPT-dependent control over the molecular clock varies substantially between tissues. Brown adipose tissue (BAT) requires NAMPT to sustain the amplitude of the core clock, whereas rhythmicity in white adipose tissue (WAT) is only moderately dependent on NAD+ biosynthesis, and the skeletal muscle clock is completely refractory to loss of NAMPT. In BAT and WAT, NAMPT differentially orchestrates oscillation of clock-controlled gene networks and the diurnality of metabolite levels. NAMPT coordinates the rhythmicity of TCA cycle intermediates in BAT, but not in WAT, and loss of NAD+ abolishes these oscillations similarly to high-fat diet-induced circadian disruption. Moreover, adipose NAMPT depletion improved the ability of animals to defend body temperature during cold stress but in a time-of-day-independent manner. Thus, our findings reveal that peripheral molecular clocks and metabolic biorhythms are shaped in a highly tissue-specific manner by NAMPT-dependent NAD+ synthesis.- Published
- 2023
- Full Text
- View/download PDF
20. Oral supplementation of nicotinamide riboside alters intestinal microbial composition in rats and mice, but not humans.
- Author
-
Peluso AA, Lundgaard AT, Babaei P, Mousovich-Neto F, Rocha AL, Damgaard MV, Bak EG, Gnanasekaran T, Dollerup OL, Trammell SAJ, Nielsen TS, Kern T, Abild CB, Sulek K, Ma T, Gerhart-Hines Z, Gillum MP, Arumugam M, Ørskov C, McCloskey D, Jessen N, Herrgård MJ, Mori MAS, and Treebak JT
- Abstract
The gut microbiota impacts systemic levels of multiple metabolites including NAD
+ precursors through diverse pathways. Nicotinamide riboside (NR) is an NAD+ precursor capable of regulating mammalian cellular metabolism. Some bacterial families express the NR-specific transporter, PnuC. We hypothesized that dietary NR supplementation would modify the gut microbiota across intestinal sections. We determined the effects of 12 weeks of NR supplementation on the microbiota composition of intestinal segments of high-fat diet-fed (HFD) rats. We also explored the effects of 12 weeks of NR supplementation on the gut microbiota in humans and mice. In rats, NR reduced fat mass and tended to decrease body weight. Interestingly, NR increased fat and energy absorption but only in HFD-fed rats. Moreover, 16S rRNA gene sequencing analysis of intestinal and fecal samples revealed an increased abundance of species within Erysipelotrichaceae and Ruminococcaceae families in response to NR. PnuC-positive bacterial strains within these families showed an increased growth rate when supplemented with NR. The abundance of species within the Lachnospiraceae family decreased in response to HFD irrespective of NR. Alpha and beta diversity and bacterial composition of the human fecal microbiota were unaltered by NR, but in mice, the fecal abundance of species within Lachnospiraceae increased while abundances of Parasutterella and Bacteroides dorei species decreased in response to NR. In conclusion, oral NR altered the gut microbiota in rats and mice, but not in humans. In addition, NR attenuated body fat mass gain in rats, and increased fat and energy absorption in the HFD context., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
21. The anorectic and thermogenic effects of pharmacological lactate in male mice are confounded by treatment osmolarity and co-administered counterions.
- Author
-
Lund J, Breum AW, Gil C, Falk S, Sass F, Isidor MS, Dmytriyeva O, Ranea-Robles P, Mathiesen CV, Basse AL, Johansen OS, Fadahunsi N, Lund C, Nicolaisen TS, Klein AB, Ma T, Emanuelli B, Kleinert M, Sørensen CM, Gerhart-Hines Z, and Clemmensen C
- Subjects
- Mice, Male, Animals, Lactic Acid, Thermogenesis physiology, Sodium, Osmolar Concentration, Appetite Depressants pharmacology
- Abstract
Lactate is a circulating metabolite and a signalling molecule with pleiotropic physiological effects. Studies suggest that lactate modulates energy balance by lowering food intake, inducing adipose browning and increasing whole-body thermogenesis. Yet, like many other metabolites, lactate is often commercially produced as a counterion-bound salt and typically administered in vivo through hypertonic aqueous solutions of sodium L-lactate. Most studies have not controlled for injection osmolarity and the co-injected sodium ions. Here, we show that the anorectic and thermogenic effects of exogenous sodium L-lactate in male mice are confounded by the hypertonicity of the injected solutions. Our data reveal that this is in contrast to the antiobesity effect of orally administered disodium succinate, which is uncoupled from these confounders. Further, our studies with other counterions indicate that counterions can have confounding effects beyond lactate pharmacology. Together, these findings underscore the importance of controlling for osmotic load and counterions in metabolite research., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
22. Mitochondrial phosphatidylethanolamine modulates UCP1 to promote brown adipose thermogenesis.
- Author
-
Johnson JM, Peterlin AD, Balderas E, Sustarsic EG, Maschek JA, Lang MJ, Jara-Ramos A, Panic V, Morgan JT, Villanueva CJ, Sanchez A, Rutter J, Lodhi IJ, Cox JE, Fisher-Wellman KH, Chaudhuri D, Gerhart-Hines Z, and Funai K
- Subjects
- Mice, Animals, Uncoupling Protein 1 metabolism, Mitochondria metabolism, Thermogenesis, Obesity metabolism, Adenosine Triphosphate metabolism, Mice, Knockout, Phosphatidylethanolamines metabolism, Protons
- Abstract
Thermogenesis by uncoupling protein 1 (UCP1) is one of the primary mechanisms by which brown adipose tissue (BAT) increases energy expenditure. UCP1 resides in the inner mitochondrial membrane (IMM), where it dissipates membrane potential independent of adenosine triphosphate (ATP) synthase. Here, we provide evidence that phosphatidylethanolamine (PE) modulates UCP1-dependent proton conductance across the IMM to modulate thermogenesis. Mitochondrial lipidomic analyses revealed PE as a signature molecule whose abundance bidirectionally responds to changes in thermogenic burden. Reduction in mitochondrial PE by deletion of phosphatidylserine decarboxylase (PSD) made mice cold intolerant and insensitive to β3 adrenergic receptor agonist-induced increase in whole-body oxygen consumption. High-resolution respirometry and fluorometry of BAT mitochondria showed that loss of mitochondrial PE specifically lowers UCP1-dependent respiration without compromising electron transfer efficiency or ATP synthesis. These findings were confirmed by a reduction in UCP1 proton current in PE-deficient mitoplasts. Thus, PE performs a previously unknown role as a temperature-responsive rheostat that regulates UCP1-dependent thermogenesis.
- Published
- 2023
- Full Text
- View/download PDF
23. Ribosome stalling is a signal for metabolic regulation by the ribotoxic stress response.
- Author
-
Snieckute G, Genzor AV, Vind AC, Ryder L, Stoneley M, Chamois S, Dreos R, Nordgaard C, Sass F, Blasius M, López AR, Brynjólfsdóttir SH, Andersen KL, Willis AE, Frankel LB, Poulsen SS, Gatfield D, Gerhart-Hines Z, Clemmensen C, and Bekker-Jensen S
- Subjects
- Animals, Male, Mice, Ribosomes, MAP Kinase Kinase Kinases metabolism, Protein Biosynthesis, Stress, Physiological
- Abstract
Impairment of translation can lead to collisions of ribosomes, which constitute an activation platform for several ribosomal stress-surveillance pathways. Among these is the ribotoxic stress response (RSR), where ribosomal sensing by the MAP3K ZAKα leads to activation of p38 and JNK kinases. Despite these insights, the physiological ramifications of ribosomal impairment and downstream RSR signaling remain elusive. Here, we show that stalling of ribosomes is sufficient to activate ZAKα. In response to amino acid deprivation and full nutrient starvation, RSR impacts on the ensuing metabolic responses in cells, nematodes, and mice. The RSR-regulated responses in these model systems include regulation of AMPK and mTOR signaling, survival under starvation conditions, stress hormone production, and regulation of blood sugar control. In addition, ZAK
-/- male mice present a lean phenotype. Our work highlights impaired ribosomes as metabolic signals and demonstrates a role for RSR signaling in metabolic regulation., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
24. ADRA1A-Gα q signalling potentiates adipocyte thermogenesis through CKB and TNAP.
- Author
-
Rahbani JF, Scholtes C, Lagarde DM, Hussain MF, Roesler A, Dykstra CB, Bunk J, Samborska B, O'Brien SL, Tripp E, Pacis A, Angueira AR, Johansen OS, Cinkornpumin J, Hossain I, Lynes MD, Zhang Y, White AP, Pastor WA, Chondronikola M, Sidossis L, Klein S, Kralli A, Cypess AM, Pedersen SB, Jessen N, Tseng YH, Gerhart-Hines Z, Seale P, Calebiro D, Giguère V, and Kazak L
- Subjects
- Adipocytes metabolism, Energy Metabolism genetics, Creatine Kinase metabolism, Creatine metabolism, Thermogenesis genetics
- Abstract
Noradrenaline (NA) regulates cold-stimulated adipocyte thermogenesis
1 . Aside from cAMP signalling downstream of β-adrenergic receptor activation, how NA promotes thermogenic output is still not fully understood. Here, we show that coordinated α1 -adrenergic receptor (AR) and β3 -AR signalling induces the expression of thermogenic genes of the futile creatine cycle2,3 , and that early B cell factors, oestrogen-related receptors and PGC1α are required for this response in vivo. NA triggers physical and functional coupling between the α1 -AR subtype (ADRA1A) and Gαq to promote adipocyte thermogenesis in a manner that is dependent on the effector proteins of the futile creatine cycle, creatine kinase B and tissue-non-specific alkaline phosphatase. Combined Gαq and Gαs signalling selectively in adipocytes promotes a continual rise in whole-body energy expenditure, and creatine kinase B is required for this effect. Thus, the ADRA1A-Gαq -futile creatine cycle axis is a key regulator of facultative and adaptive thermogenesis., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
25. Lipolysis regulates major transcriptional programs in brown adipocytes.
- Author
-
Markussen LK, Rondini EA, Johansen OS, Madsen JGS, Sustarsic EG, Marcher AB, Hansen JB, Gerhart-Hines Z, Granneman JG, and Mandrup S
- Subjects
- Adipose Tissue, Brown metabolism, Adrenergic Agents pharmacology, Peroxisome Proliferator-Activated Receptors metabolism, Thermogenesis physiology, Adipocytes, Brown metabolism, Lipolysis genetics
- Abstract
β-Adrenergic signaling is a core regulator of brown adipocyte function stimulating both lipolysis and transcription of thermogenic genes, thereby expanding the capacity for oxidative metabolism. We have used pharmacological inhibitors and a direct activator of lipolysis to acutely modulate the activity of lipases, thereby enabling us to uncover lipolysis-dependent signaling pathways downstream of β-adrenergic signaling in cultured brown adipocytes. Here we show that induction of lipolysis leads to acute induction of several gene programs and is required for transcriptional regulation by β-adrenergic signals. Using machine-learning algorithms to infer causal transcription factors, we show that PPARs are key mediators of lipolysis-induced activation of genes involved in lipid metabolism and thermogenesis. Importantly, however, lipolysis also activates the unfolded protein response and regulates the core circadian transcriptional machinery independently of PPARs. Our results demonstrate that lipolysis generates important metabolic signals that exert profound pleiotropic effects on transcription and function of cultured brown adipocytes., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
26. Leveraging GPCR signaling in thermogenic fat to counteract metabolic diseases.
- Author
-
Sveidahl Johansen O, Ma T, and Gerhart-Hines Z
- Subjects
- Adipose Tissue metabolism, Humans, Obesity metabolism, Thermogenesis, Adipocytes, Beige metabolism, Metabolic Diseases metabolism
- Abstract
Background: Thermogenic brown and beige adipocytes are recognized for their unique capacity to consume extraordinary levels of metabolites and lipids from the blood to fuel heat-producing catabolic processes [1-7]. In humans, the functions of thermogenic adipocytes are associated with cardiometabolic protection and improved glycemic control [8-13]. Consequently, engaging these macronutrient-consuming and energy-dissipating activities has gained attention as a promising therapeutic strategy for counteracting metabolic diseases, such as obesity and diabetes., Scope of Review: In this review, we highlight new advances in our understanding of the physiological role of G protein-coupled receptors (GPCRs) in controlling thermogenic adipocyte biology. We further extend our discussion to the opportunities and challenges posed by pharmacologically targeting different elements of GPCR signaling in these highly specialized fat cells., Major Conclusions: GPCRs represent appealing candidates through which to harness adipose thermogenesis. Yet safely and effectively targeting these druggable receptors on brown and beige adipocytes has thus far proven challenging. Therefore, continued interrogation across the GPCR landscape is necessary for future leaps within the field of thermogenic fat biology to unlock the therapeutic potential of adipocyte catabolism., (Copyright © 2022 The Author(s). Published by Elsevier GmbH.. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
27. Influence of NAFLD and bariatric surgery on hepatic and adipose tissue mitochondrial biogenesis and respiration.
- Author
-
Pedersen JS, Rygg MO, Chrøis K, Sustarsic EG, Gerhart-Hines Z, Wever Albrechtsen NJ, Serizawa RR, Kristiansen VB, Basse AL, Boilesen AEB, Olsen BH, Hansen T, Gluud LL, Madsbad S, Larsen S, Bendtsen F, and Dela F
- Subjects
- Adipose Tissue pathology, Humans, Liver pathology, Obesity complications, Obesity pathology, Obesity surgery, Organelle Biogenesis, Respiration, Bariatric Surgery, Non-alcoholic Fatty Liver Disease pathology, Obesity, Morbid complications, Obesity, Morbid pathology, Obesity, Morbid surgery
- Abstract
Impaired mitochondrial oxidative phosphorylation (OXPHOS) in liver tissue has been hypothesised to contribute to the development of nonalcoholic steatohepatitis in patients with nonalcoholic fatty liver disease (NAFLD). It is unknown whether OXPHOS capacities in human visceral (VAT) and subcutaneous adipose tissue (SAT) associate with NAFLD severity and how hepatic OXPHOS responds to improvement in NAFLD. In biopsies sampled from 62 patients with obesity undergoing bariatric surgery and nine control subjects without obesity we demonstrate that OXPHOS is reduced in VAT and SAT while increased in the liver in patients with obesity when compared with control subjects without obesity, but this was independent of NAFLD severity. In repeat liver biopsy sampling in 21 patients with obesity 12 months after bariatric surgery we found increased hepatic OXPHOS capacity and mitochondrial DNA/nuclear DNA content compared with baseline. In this work we show that obesity has an opposing association with mitochondrial respiration in adipose- and liver tissue with no overall association with NAFLD severity, however, bariatric surgery increases hepatic OXPHOS and mitochondrial biogenesis., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
28. Fueling the fire of adipose thermogenesis.
- Author
-
Wolfrum C and Gerhart-Hines Z
- Subjects
- Adipocytes, White metabolism, Animals, Energy Metabolism, Humans, Metabolic Diseases metabolism, Metabolic Diseases therapy, Mice, Mitochondria metabolism, Nutrients metabolism, Adipocytes, Beige metabolism, Adipocytes, Brown metabolism, Thermogenesis
- Abstract
Specialized fat tissue generates heat and holds the potential to counter metabolic diseases.
- Published
- 2022
- Full Text
- View/download PDF
29. Altered brown fat thermoregulation and enhanced cold-induced thermogenesis in young, healthy, winter-swimming men.
- Author
-
Søberg S, Löfgren J, Philipsen FE, Jensen M, Hansen AE, Ahrens E, Nystrup KB, Nielsen RD, Sølling C, Wedell-Neergaard AS, Berntsen M, Loft A, Kjær A, Gerhart-Hines Z, Johannesen HH, Pedersen BK, Karstoft K, and Scheele C
- Subjects
- Adipose Tissue, Brown diagnostic imaging, Adult, Circadian Rhythm physiology, Hormones blood, Humans, Magnetic Resonance Imaging, Male, Perception, Positron-Emission Tomography, Skin Temperature physiology, Thermography, Young Adult, Adipose Tissue, Brown physiology, Cold Temperature, Seasons, Swimming physiology, Thermogenesis physiology
- Abstract
The Scandinavian winter-swimming culture combines brief dips in cold water with hot sauna sessions, with conceivable effects on body temperature. We study thermogenic brown adipose tissue (BAT) in experienced winter-swimming men performing this activity 2-3 times per week. Our data suggest a lower thermal comfort state in the winter swimmers compared with controls, with a lower core temperature and absence of BAT activity. In response to cold, we observe greater increases in cold-induced thermogenesis and supraclavicular skin temperature in the winter swimmers, whereas BAT glucose uptake and muscle activity increase similarly to those of the controls. All subjects demonstrate nocturnal reduction in supraclavicular skin temperature, whereas a distinct peak occurs at 4:30-5:30 a.m. in the winter swimmers. Our data leverage understanding of BAT in adult human thermoregulation, suggest both heat and cold acclimation in winter swimmers, and propose winter swimming as a potential strategy for increasing energy expenditure., Competing Interests: C. Scheele is a consultant for Novo Nordisk A/S on human brown adipose tissue biology. Z.G.-H. works, in some capacity, for Embark Biotech ApS, a company developing therapeutics for the treatment of diabetes and obesity. All other authors declare no competing interests associated with this manuscript., (© 2021 The Authors.)
- Published
- 2021
- Full Text
- View/download PDF
30. Lipolysis drives expression of the constitutively active receptor GPR3 to induce adipose thermogenesis.
- Author
-
Sveidahl Johansen O, Ma T, Hansen JB, Markussen LK, Schreiber R, Reverte-Salisa L, Dong H, Christensen DP, Sun W, Gnad T, Karavaeva I, Nielsen TS, Kooijman S, Cero C, Dmytriyeva O, Shen Y, Razzoli M, O'Brien SL, Kuipers EN, Nielsen CH, Orchard W, Willemsen N, Jespersen NZ, Lundh M, Sustarsic EG, Hallgren CM, Frost M, McGonigle S, Isidor MS, Broholm C, Pedersen O, Hansen JB, Grarup N, Hansen T, Kjær A, Granneman JG, Babu MM, Calebiro D, Nielsen S, Rydén M, Soccio R, Rensen PCN, Treebak JT, Schwartz TW, Emanuelli B, Bartolomucci A, Pfeifer A, Zechner R, Scheele C, Mandrup S, and Gerhart-Hines Z
- Subjects
- Adipocytes metabolism, Animals, COS Cells, Cells, Cultured, Chlorocebus aethiops, Cold Temperature, Dietary Fats pharmacology, Humans, Mice, Inbred C57BL, Phenotype, Receptors, G-Protein-Coupled genetics, Signal Transduction, Sympathetic Nervous System metabolism, Transcription, Genetic, Mice, Adipose Tissue, Brown metabolism, Constitutive Androstane Receptor metabolism, Lipolysis, Receptors, G-Protein-Coupled metabolism, Thermogenesis
- Abstract
Thermogenic adipocytes possess a therapeutically appealing, energy-expending capacity, which is canonically cold-induced by ligand-dependent activation of β-adrenergic G protein-coupled receptors (GPCRs). Here, we uncover an alternate paradigm of GPCR-mediated adipose thermogenesis through the constitutively active receptor, GPR3. We show that the N terminus of GPR3 confers intrinsic signaling activity, resulting in continuous Gs-coupling and cAMP production without an exogenous ligand. Thus, transcriptional induction of Gpr3 represents the regulatory parallel to ligand-binding of conventional GPCRs. Consequently, increasing Gpr3 expression in thermogenic adipocytes is alone sufficient to drive energy expenditure and counteract metabolic disease in mice. Gpr3 transcription is cold-stimulated by a lipolytic signal, and dietary fat potentiates GPR3-dependent thermogenesis to amplify the response to caloric excess. Moreover, we find GPR3 to be an essential, adrenergic-independent regulator of human brown adipocytes. Taken together, our findings reveal a noncanonical mechanism of GPCR control and thermogenic activation through the lipolysis-induced expression of constitutively active GPR3., Competing Interests: Declaration of interests O.S.J., Jakob Bondo Hansen, D.P.C., T.W.S., and Z.G.-H. work or have worked, in some capacity, for Embark Biotech ApS, a company developing therapeutics for the treatment of diabetes and obesity. All other authors declare no competing interests associated with this manuscript., (Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
31. Cold-induction of afadin in brown fat supports its thermogenic capacity.
- Author
-
Lundh M, Altıntaş A, Tozzi M, Fabre O, Ma T, Shamsi F, Gerhart-Hines Z, Barrès R, Tseng YH, and Emanuelli B
- Subjects
- Animals, Kinesins genetics, Mice, Mice, Knockout, Myosins genetics, Adipocytes, Brown metabolism, Adipose Tissue, Brown metabolism, Cold Temperature, Gene Expression Regulation, Kinesins biosynthesis, Myosins biosynthesis, Thermogenesis
- Abstract
The profound energy-expending nature of brown adipose tissue (BAT) thermogenesis makes it an attractive target tissue to combat obesity-associated metabolic disorders. While cold exposure is the strongest inducer of BAT activity, the temporal mechanisms tuning BAT adaptation during this activation process are incompletely understood. Here we show that the scaffold protein Afadin is dynamically regulated by cold in BAT, and participates in cold acclimation. Cold exposure acutely increases Afadin protein levels and its phosphorylation in BAT. Knockdown of Afadin in brown pre-adipocytes does not alter adipogenesis but restricts β
3 -adrenegic induction of thermogenic genes expression and HSL phosphorylation in mature brown adipocytes. Consistent with a defect in thermogenesis, an impaired cold tolerance was observed in fat-specific Afadin knockout mice. However, while Afadin depletion led to reduced Ucp1 mRNA induction by cold, stimulation of Ucp1 protein was conserved. Transcriptomic analysis revealed that fat-specific ablation of Afadin led to decreased functional enrichment of gene sets controlling essential metabolic functions at thermoneutrality in BAT, whereas it led to an altered reprogramming in response to cold, with enhanced enrichment of different pathways related to metabolism and remodeling. Collectively, we demonstrate a role for Afadin in supporting the adrenergic response in brown adipocytes and BAT function.- Published
- 2021
- Full Text
- View/download PDF
32. Ablation of Nampt in AgRP neurons leads to neurodegeneration and impairs fasting- and ghrelin-mediated food intake.
- Author
-
de Guia RM, Hassing AS, Ma T, Plucinska K, Holst B, Gerhart-Hines Z, Emanuelli B, and Treebak JT
- Subjects
- Agouti-Related Protein genetics, Animals, Female, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Neurodegenerative Diseases etiology, Neurodegenerative Diseases metabolism, Agouti-Related Protein metabolism, Cytokines physiology, Eating, Fasting, Ghrelin pharmacology, Neurodegenerative Diseases pathology, Neurons metabolism, Nicotinamide Phosphoribosyltransferase physiology
- Abstract
Agouti-related protein (AgRP) neurons in the arcuate nucleus of the hypothalamus regulates food intake and whole-body metabolism. NAD
+ regulates multiple cellular processes controlling energy metabolism. Yet, its role in hypothalamic AgRP neurons to control food intake is poorly understood. Here, we aimed to assess whether genetic deletion of nicotinamide phosphoribosyltransferase (Nampt), a rate-limiting enzyme in NAD+ production, affects AgRP neuronal function to impact whole-body metabolism and food intake. Metabolic parameters during fed and fasted states, and upon systemic ghrelin and leptin administration were studied in AgRP-specific Nampt knockout (ARNKO) mice. We monitored neuropeptide expression levels and density of AgRP neurons in ARNKO mice from embryonic to adult age. NPY cells were used to determine effects of NAMPT inhibition on neuronal viability, energy status, and oxidative stress in vitro. In these cells, NAD+ depletion reduced ATP levels, increased oxidative stress, and promoted cell death. Agrp expression in the hypothalamus of ARNKO mice gradually decreased after weaning due to progressive AgRP neuron degeneration. Adult ARNKO mice had normal glucose and insulin tolerance, but exhibited an elevated respiratory exchange ratio (RER) when fasted. Remarkably, fasting-induced food intake was unaffected in ARNKO mice when evaluated in metabolic cages, but fasting- and ghrelin-induced feeding and body weight gain decreased in ARNKO mice when evaluated outside metabolic cages. Collectively, deletion of Nampt in AgRP neurons causes progressive neurodegeneration and impairs fasting and ghrelin responses in a context-dependent manner. Our data highlight an essential role of Nampt in AgRP neuron function and viability., (© 2021 Federation of American Societies for Experimental Biology.)- Published
- 2021
- Full Text
- View/download PDF
33. White adipose remodeling during browning in mice involves YBX1 to drive thermogenic commitment.
- Author
-
Rabiee A, Plucińska K, Isidor MS, Brown EL, Tozzi M, Sidoli S, Petersen PSS, Agueda-Oyarzabal M, Torsetnes SB, Chehabi GN, Lundh M, Altıntaş A, Barrès R, Jensen ON, Gerhart-Hines Z, and Emanuelli B
- Subjects
- Adipocytes, Brown metabolism, Adipogenesis, Adipose Tissue, Brown metabolism, Animals, Cell Differentiation, Cell Line, Cell Proliferation, Gene Expression Regulation, Jumonji Domain-Containing Histone Demethylases genetics, Jumonji Domain-Containing Histone Demethylases metabolism, Male, Mesenchymal Stem Cells, Mice, Mice, Inbred C57BL, Obesity metabolism, Proteomics, Subcutaneous Fat metabolism, Transcriptome, Up-Regulation, Adipose Tissue, White metabolism, Thermogenesis genetics, Thermogenesis physiology, Transcription Factors genetics, Transcription Factors metabolism
- Abstract
Objective: Increasing adaptive thermogenesis by stimulating browning in white adipose tissue is a promising method of improving metabolic health. However, the molecular mechanisms underlying this transition remain elusive. Our study examined the molecular determinants driving the differentiation of precursor cells into thermogenic adipocytes., Methods: In this study, we conducted temporal high-resolution proteomic analysis of subcutaneous white adipose tissue (scWAT) after cold exposure in mice. This was followed by loss- and gain-of-function experiments using siRNA-mediated knockdown and CRISPRa-mediated induction of gene expression, respectively, to evaluate the function of the transcriptional regulator Y box-binding protein 1 (YBX1) during adipogenesis of brown pre-adipocytes and mesenchymal stem cells. Transcriptomic analysis of mesenchymal stem cells following induction of endogenous Ybx1 expression was conducted to elucidate transcriptomic events controlled by YBX1 during adipogenesis., Results: Our proteomics analysis uncovered 509 proteins differentially regulated by cold in a time-dependent manner. Overall, 44 transcriptional regulators were acutely upregulated following cold exposure, among which included the cold-shock domain containing protein YBX1, peaking after 24 h. Cold-induced upregulation of YBX1 also occurred in brown adipose tissue, but not in visceral white adipose tissue, suggesting a role of YBX1 in thermogenesis. This role was confirmed by Ybx1 knockdown in brown and brite preadipocytes, which significantly impaired their thermogenic potential. Conversely, inducing Ybx1 expression in mesenchymal stem cells during adipogenesis promoted browning concurrent with an increased expression of thermogenic markers and enhanced mitochondrial respiration. At a molecular level, our transcriptomic analysis showed that YBX1 regulates a subset of genes, including the histone H3K9 demethylase Jmjd1c, to promote thermogenic adipocyte differentiation., Conclusion: Our study mapped the dynamic proteomic changes of murine scWAT during browning and identified YBX1 as a novel factor coordinating the genomic mechanisms by which preadipocytes commit to brite/beige lineage., (Copyright © 2020 The Author(s). Published by Elsevier GmbH.. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
34. Epidermal Acyl-CoA-binding protein is indispensable for systemic energy homeostasis.
- Author
-
Neess D, Kruse V, Marcher AB, Wæde MR, Vistisen J, Møller PM, Petersen R, Brewer JR, Ma T, Colleluori G, Severi I, Cinti S, Gerhart-Hines Z, Mandrup S, and Færgeman NJ
- Subjects
- Adipose Tissue, White metabolism, Animals, Body Temperature, Energy Metabolism genetics, Filaggrin Proteins, Intermediate Filament Proteins, Lipid Metabolism, Lipolysis, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Obesity metabolism, Diazepam Binding Inhibitor genetics, Diazepam Binding Inhibitor metabolism, Energy Metabolism physiology, Homeostasis, Skin metabolism
- Abstract
Objectives: The skin is the largest sensory organ of the human body and plays a fundamental role in regulating body temperature. However, adaptive alterations in skin functions and morphology have only vaguely been associated with physiological responses to cold stress or sensation of ambient temperatures. We previously found that loss of acyl-CoA-binding protein (ACBP) in keratinocytes upregulates lipolysis in white adipose tissue and alters hepatic lipid metabolism, suggesting a link between epidermal barrier functions and systemic energy metabolism., Methods: To assess the physiological responses to loss of ACBP in keratinocytes in detail, we used full-body ACBP
-/- and skin-specific ACBP-/- knockout mice to clarify how loss of ACBP affects 1) energy expenditure by indirect calorimetry, 2) response to high-fat feeding and a high oral glucose load, and 3) expression of brown-selective gene programs by quantitative PCR in inguinal WAT (iWAT). To further elucidate the role of the epidermal barrier in systemic energy metabolism, we included mice with defects in skin structural proteins (ma/ma Flgft/ft ) in these studies., Results: We show that the ACBP-/- mice and skin-specific ACBP-/- knockout mice exhibited increased energy expenditure, increased food intake, browning of the iWAT, and resistance to diet-induced obesity. The metabolic phenotype, including browning of the iWAT, was reversed by housing the mice at thermoneutrality (30 °C) or pharmacological β-adrenergic blocking. Interestingly, these findings were phenocopied in flaky tail mice (ma/ma Flgft/ft ). Taken together, we demonstrate that a compromised epidermal barrier induces a β-adrenergic response that increases energy expenditure and browning of the white adipose tissue to maintain a normal body temperature., Conclusions: Our findings show that the epidermal barrier plays a key role in maintaining systemic metabolic homeostasis. Thus, regulation of epidermal barrier functions warrants further attention to understand the regulation of systemic metabolism in further detail., (Copyright © 2020 The Author(s). Published by Elsevier GmbH.. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
35. Autocrine negative feedback regulation of lipolysis through sensing of NEFAs by FFAR4/GPR120 in WAT.
- Author
-
Husted AS, Ekberg JH, Tripp E, Nissen TAD, Meijnikman S, O'Brien SL, Ulven T, Acherman Y, Bruin SC, Nieuwdorp M, Gerhart-Hines Z, Calebiro D, Dragsted LO, and Schwartz TW
- Subjects
- 3T3-L1 Cells, Adipocytes metabolism, Adipocytes, White metabolism, Adipose Tissue metabolism, Adipose Tissue, White metabolism, Animals, Autocrine Communication physiology, Chromatography, Liquid methods, Culture Media, Conditioned pharmacology, Fatty Acids metabolism, Feedback, Physiological physiology, Female, Humans, Lipolysis physiology, Male, Mass Spectrometry methods, Mice, Mice, Inbred C57BL, Receptors, G-Protein-Coupled physiology, Fatty Acids, Nonesterified metabolism, Receptors, G-Protein-Coupled metabolism
- Abstract
Objectives: Long-chain fatty acids (LCFAs) released from adipocytes inhibit lipolysis through an unclear mechanism. We hypothesized that the LCFA receptor, FFAR4 (GPR120), which is highly expressed in adipocytes, may be involved in this feedback regulation., Methods and Results: Liquid chromatography mass spectrometry (LC-MS) analysis of conditioned media from isoproterenol-stimulated primary cultures of murine and human adipocytes demonstrated that most of the released non-esterified free fatty acids (NEFAs) are known agonists for FFAR4. In agreement with this, conditioned medium from isoproterenol-treated adipocytes stimulated signaling strongly in FFAR4 transfected COS-7 cells as opposed to non-transfected control cells. In transfected 3T3-L1 cells, FFAR4 agonism stimulated Gi- and Go-mini G protein binding more strongly than Gq, effects which were blocked by the selective FFAR4 antagonist AH7614. In primary cultures of murine white adipocytes, the synthetic, selective FFAR4 agonist CpdA inhibited isoproterenol-induced intracellular cAMP accumulation in a manner similar to the antilipolytic control agent nicotinic acid acting through another receptor, HCAR2. In vivo, oral gavage with the synthetic, specific FFAR4 agonist CpdB decreased the level of circulating NEFAs in fasting lean mice to a similar degree as nicotinic acid. In agreement with the identified anti-lipolytic effect of FFAR4, plasma NEFAs and glycerol were increased in FFAR4-deficient mice as compared to littermate controls despite having elevated insulin levels, and cAMP accumulation in primary adipocyte cultures was augmented by treatment with the FFAR4 antagonist conceivably by blocking the stimulatory tone of endogenous NEFAs on FFAR4., Conclusions: In white adipocytes, FFAR4 functions as an NEFA-activated, autocrine, negative feedback regulator of lipolysis by decreasing cAMP though Gi-mediated signaling., (Copyright © 2020 The Authors. Published by Elsevier GmbH.. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
36. Thyroid hormone receptor α in skeletal muscle is essential for T3-mediated increase in energy expenditure.
- Author
-
Nicolaisen TS, Klein AB, Dmytriyeva O, Lund J, Ingerslev LR, Fritzen AM, Carl CS, Lundsgaard AM, Frost M, Ma T, Schjerling P, Gerhart-Hines Z, Flamant F, Gauthier K, Larsen S, Richter EA, Kiens B, and Clemmensen C
- Subjects
- Animals, Male, Mice, Mice, Knockout, Muscle Fibers, Fast-Twitch cytology, Muscle Fibers, Fast-Twitch drug effects, Muscle Fibers, Slow-Twitch cytology, Muscle Fibers, Slow-Twitch drug effects, Muscle, Skeletal cytology, Muscle, Skeletal drug effects, Physical Conditioning, Animal, Transcriptome, Energy Metabolism drug effects, Muscle Fibers, Fast-Twitch physiology, Muscle Fibers, Slow-Twitch physiology, Muscle, Skeletal physiology, Thyroid Hormone Receptors alpha physiology, Thyroid Hormones pharmacology
- Abstract
Thyroid hormones are important for homeostatic control of energy metabolism and body temperature. Although skeletal muscle is considered a key site for thyroid action, the contribution of thyroid hormone receptor signaling in muscle to whole-body energy metabolism and body temperature has not been resolved. Here, we show that T3-induced increase in energy expenditure requires thyroid hormone receptor alpha 1 (TRα
1 ) in skeletal muscle, but that T3-mediated elevation in body temperature is achieved in the absence of muscle-TRα1 . In slow-twitch soleus muscle, loss-of-function of TRα1 (TRαHSACre ) alters the fiber-type composition toward a more oxidative phenotype. The change in fiber-type composition, however, does not influence the running capacity or motivation to run. RNA-sequencing of soleus muscle from WT mice and TRαHSACre mice revealed differentiated transcriptional regulation of genes associated with muscle thermogenesis, such as sarcolipin and UCP3, providing molecular clues pertaining to the mechanistic underpinnings of TRα1 -linked control of whole-body metabolic rate. Together, this work establishes a fundamental role for skeletal muscle in T3-stimulated increase in whole-body energy expenditure., (© 2020 The Authors. The FASEB Journal published by Wiley Periodicals LLC on behalf of Federation of American Societies for Experimental Biology.)- Published
- 2020
- Full Text
- View/download PDF
37. Role of Energy Excretion in Human Body Weight Regulation.
- Author
-
Lund J, Gerhart-Hines Z, and Clemmensen C
- Subjects
- Humans, Body Constitution physiology, Energy Intake physiology, Energy Metabolism physiology, Intestinal Elimination physiology, Weight Gain physiology
- Abstract
Food intake and energy expenditure are the typical determinants of body weight. Yet, recent observations underscore that a third and often-neglected factor, fecal energy loss, can influence energy balance. Here, we explore how macronutrient excretion modulates human energy homeostasis and highlight its potential impact on the propensity to gain weight., (Copyright © 2020 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
38. Cytoplasmic Citrate Flux Modulates the Immune Stimulatory NKG2D Ligand MICA in Cancer Cells.
- Author
-
Møller SH, Mellergaard M, Madsen M, Bermejo AV, Jepsen SD, Hansen MH, Høgh RI, Aldana BI, Desler C, Rasmussen LJ, Sustarsic EG, Gerhart-Hines Z, Daskalaki E, Wheelock CE, Hiron TK, Lin D, O'Callaghan CA, Wandall HH, Andresen L, and Skov S
- Subjects
- Cell Line, Tumor, Chromatin Assembly and Disassembly, Female, Gene Editing, Gene Expression Regulation, Glycolysis, HEK293 Cells, Histocompatibility Antigens Class I genetics, Humans, Ligands, Lymphocyte Activation, Lymphocytes immunology, Lymphocytes metabolism, Mitochondria genetics, Mitochondria metabolism, Models, Biological, N-Acetylglucosaminyltransferases genetics, N-Acetylglucosaminyltransferases metabolism, Protein Binding, Transcription Initiation Site, Citric Acid metabolism, Cytoplasm metabolism, Histocompatibility Antigens Class I metabolism, Immunomodulation, NK Cell Lectin-Like Receptor Subfamily K metabolism, Neoplasms immunology, Neoplasms metabolism
- Abstract
Immune surveillance of cancer cells is facilitated by the Natural Killer Group 2D (NKG2D) receptor expressed by different lymphocyte subsets. It recognizes NKG2D ligands that are rarely expressed on healthy cells, but upregulated by tumorigenesis, presenting a target for immunological clearance. The molecular mechanisms responsible for NKG2D ligand regulation remain complex. Here we report that cancer cell metabolism supports constitutive surface expression of the NKG2D ligand MHC class I chain-related proteins A (MICA). Knockout of the N -glycosylation gene N -acetylglucosaminyltransferase V (MGAT5) in HEK293 cells induced altered metabolism and continuous high MICA surface expression. MGAT5 knockout cells were used to examine the association of cell metabolism and MICA expression through genetic, pharmacological and metabolic assays. Findings were verified in cancer cell lines. Cells with constitutive high MICA expression showed enhanced spare respiratory capacity and elevated mitochondrial efflux of citrate, determined by extracellular flux analysis and metabolomics. MICA expression was reduced by inhibitors of mitochondrial function, FCCP and etomoxir e.g., and depended on conversion of citrate to acetyl-CoA and oxaloacetate by ATP citrate lyase, which was also observed in several cancer cell types. Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq) analysis revealed that upregulated MICA transcription was associated with an open chromatin structure at the MICA transcription start site. We identify mitochondria and cytoplasmic citrate as key regulators of constitutive MICA expression and we propose that metabolic reprogramming of certain cancer cells facilitates MICA expression and NKG2D-mediated immune recognition., (Copyright © 2020 Møller, Mellergaard, Madsen, Bermejo, Jepsen, Hansen, Høgh, Aldana, Desler, Rasmussen, Sustarsic, Gerhart-Hines, Daskalaki, Wheelock, Hiron, Lin, O’Callaghan, Wandall, Andresen and Skov.)
- Published
- 2020
- Full Text
- View/download PDF
39. Lactate released by inflammatory bone marrow neutrophils induces their mobilization via endothelial GPR81 signaling.
- Author
-
Khatib-Massalha E, Bhattacharya S, Massalha H, Biram A, Golan K, Kollet O, Kumari A, Avemaria F, Petrovich-Kopitman E, Gur-Cohen S, Itkin T, Brandenburger I, Spiegel A, Shulman Z, Gerhart-Hines Z, Itzkovitz S, Gunzer M, Offermanns S, Alon R, Ariel A, and Lapidot T
- Subjects
- Animals, Bone Marrow blood supply, Bone Marrow Cells metabolism, Disease Models, Animal, Endothelium, Vascular metabolism, Female, Humans, Lipopolysaccharides immunology, Male, Mice, Mice, Knockout, Neutrophils metabolism, Receptors, G-Protein-Coupled genetics, Salmonella Infections microbiology, Salmonella typhimurium immunology, Signal Transduction immunology, Bone Marrow Cells immunology, Lactic Acid metabolism, Neutrophils immunology, Receptors, G-Protein-Coupled metabolism, Salmonella Infections immunology
- Abstract
Neutrophils provide first line of host defense against bacterial infections utilizing glycolysis for their effector functions. How glycolysis and its major byproduct lactate are triggered in bone marrow (BM) neutrophils and their contribution to neutrophil mobilization in acute inflammation is not clear. Here we report that bacterial lipopolysaccharides (LPS) or Salmonella Typhimurium triggers lactate release by increasing glycolysis, NADPH-oxidase-mediated reactive oxygen species and HIF-1α levels in BM neutrophils. Increased release of BM lactate preferentially promotes neutrophil mobilization by reducing endothelial VE-Cadherin expression, increasing BM vascular permeability via endothelial lactate-receptor GPR81 signaling. GPR81
-/- mice mobilize reduced levels of neutrophils in response to LPS, unless rescued by VE-Cadherin disrupting antibodies. Lactate administration also induces release of the BM neutrophil mobilizers G-CSF, CXCL1 and CXCL2, indicating that this metabolite drives neutrophil mobilization via multiple pathways. Our study reveals a metabolic crosstalk between lactate-producing neutrophils and BM endothelium, which controls neutrophil mobilization under bacterial infection.- Published
- 2020
- Full Text
- View/download PDF
40. Fasting- and ghrelin-induced food intake is regulated by NAMPT in the hypothalamus.
- Author
-
de Guia RM, Hassing AS, Skov LJ, Ratner C, Plucińska K, Madsen S, Diep TA, Dela Cruz GV, Trammell SAJ, Sustarsic EG, Emanuelli B, Gillum MP, Gerhart-Hines Z, Holst B, and Treebak JT
- Subjects
- Acrylamides administration & dosage, Agouti-Related Protein genetics, Agouti-Related Protein metabolism, Animals, Cell Line, Eating, Female, Male, Mice, Mice, Inbred C57BL, Phosphotransferases (Alcohol Group Acceptor) metabolism, Piperidines administration & dosage, Pro-Opiomelanocortin genetics, Pro-Opiomelanocortin metabolism, Fasting metabolism, Ghrelin metabolism, Hypothalamus metabolism, Nicotinamide Phosphoribosyltransferase metabolism
- Abstract
Aim: Neurons in the arcuate nucleus of the hypothalamus are involved in regulation of food intake and energy expenditure, and dysregulation of signalling in these neurons promotes development of obesity. The role of the rate-limiting enzyme in the NAD
+ salvage pathway, nicotinamide phosphoribosyltransferase (NAMPT), for regulation energy homeostasis by the hypothalamus has not been extensively studied., Methods: We determined whether Nampt mRNA or protein levels in the hypothalamus of mice were affected by diet-induced obesity, by fasting and re-feeding, and by leptin and ghrelin treatment. Primary hypothalamic neurons were treated with FK866, a selective inhibitor of NAMPT, or rAAV carrying shRNA directed against Nampt, and levels of reactive oxygen species (ROS) and mitochondrial respiration were assessed. Fasting and ghrelin-induced food intake was measured in mice in metabolic cages after intracerebroventricular (ICV)-mediated FK866 administration., Results: NAMPT levels in the hypothalamus were elevated by administration of ghrelin and leptin. In diet-induced obese mice, both protein and mRNA levels of NAMPT decreased in the hypothalamus. NAMPT inhibition in primary hypothalamic neurons significantly reduced levels of NAD+ , increased levels of ROS, and affected the expression of Agrp, Pomc and genes related to mitochondrial function. Finally, ICV-induced NAMPT inhibition by FK866 did not cause malaise or anhedonia, but completely ablated fasting- and ghrelin-induced increases in food intake., Conclusion: Our findings indicate that regulation of NAMPT levels in hypothalamic neurons is important for the control of fasting- and ghrelin-induced food intake., (© 2020 Scandinavian Physiological Society. Published by John Wiley & Sons Ltd.)- Published
- 2020
- Full Text
- View/download PDF
41. Housing temperature influences exercise training adaptations in mice.
- Author
-
Raun SH, Henriquez-Olguín C, Karavaeva I, Ali M, Møller LLV, Kot W, Castro-Mejía JL, Nielsen DS, Gerhart-Hines Z, Richter EA, and Sylow L
- Subjects
- Adipose Tissue drug effects, Animals, Body Composition, Energy Metabolism, Female, Gastrointestinal Microbiome, Insulin pharmacology, Mice, Mice, Inbred C57BL, Muscle, Skeletal drug effects, Stress, Physiological, Temperature, Adaptation, Physiological physiology, Housing, Animal, Physical Conditioning, Animal physiology
- Abstract
Exercise training is a powerful means to combat metabolic diseases. Mice are extensively used to investigate the benefits of exercise, but mild cold stress induced by ambient housing temperatures may confound translation to humans. Thermoneutral housing is a strategy to make mice more metabolically similar to humans but its effects on exercise adaptations are unknown. Here we show that thermoneutral housing blunts exercise-induced improvements in insulin action in muscle and adipose tissue and reduces the effects of training on energy expenditure, body composition, and muscle and adipose tissue protein expressions. Thus, many reported effects of exercise training in mice are likely secondary to metabolic stress of ambient housing temperature, making it challenging to translate to humans. We conclude that adaptations to exercise training in mice critically depend upon housing temperature. Our findings underscore housing temperature as a critical parameter in the design and interpretation of murine exercise training studies.
- Published
- 2020
- Full Text
- View/download PDF
42. Proteomics-Based Comparative Mapping of the Secretomes of Human Brown and White Adipocytes Reveals EPDR1 as a Novel Batokine.
- Author
-
Deshmukh AS, Peijs L, Beaudry JL, Jespersen NZ, Nielsen CH, Ma T, Brunner AD, Larsen TJ, Bayarri-Olmos R, Prabhakar BS, Helgstrand C, Severinsen MCK, Holst B, Kjaer A, Tang-Christensen M, Sanfridson A, Garred P, Privé GG, Pedersen BK, Gerhart-Hines Z, Nielsen S, Drucker DJ, Mann M, and Scheele C
- Subjects
- Adult, Aged, Animals, Cohort Studies, Female, Gene Knockdown Techniques, Goiter blood, Goiter pathology, Goiter surgery, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Middle Aged, Neoplasm Proteins genetics, Nerve Tissue Proteins, Secretory Pathway genetics, Signal Transduction genetics, Transfection, Young Adult, Adipocytes, Brown metabolism, Adipocytes, White metabolism, Adipose Tissue, Brown metabolism, Neoplasm Proteins blood, Proteomics methods
- Abstract
Adipokines secreted from white adipose tissue play a role in metabolic crosstalk and homeostasis, whereas the brown adipose secretome is less explored. We performed high-sensitivity mass-spectrometry-based proteomics on the cell media of human adipocytes derived from the supraclavicular brown adipose and from the subcutaneous white adipose depots of adult humans. We identified 471 potentially secreted proteins covering interesting categories such as hormones, growth factors, extracellular matrix proteins, and proteins of the complement system, which were differentially regulated between brown and white adipocytes. A total of 101 proteins were exclusively quantified in brown adipocytes, and among these was ependymin-related protein 1 (EPDR1). EPDR1 was detected in human plasma, and functional studies suggested a role for EPDR1 in thermogenic determination during adipogenesis. In conclusion, we report substantial differences between the secretomes of brown and white human adipocytes and identify novel candidate batokines that can be important regulators of human metabolism., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
43. Afadin is a scaffold protein repressing insulin action via HDAC6 in adipose tissue.
- Author
-
Lundh M, Petersen PS, Isidor MS, Kazoka-Sørensen DN, Plucińska K, Shamsi F, Ørskov C, Tozzi M, Brown EL, Andersen E, Ma T, Müller U, Barrès R, Kristiansen VB, Gerhart-Hines Z, Tseng YH, and Emanuelli B
- Subjects
- 3T3-L1 Cells, Adipocytes metabolism, Adipocytes pathology, Adipose Tissue pathology, Animals, Antigens, CD metabolism, Diet, High-Fat adverse effects, Glucose metabolism, Histone Deacetylase 6 metabolism, Homeostasis genetics, Humans, Insulin metabolism, Insulin pharmacology, Insulin Resistance, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Microfilament Proteins metabolism, Obesity etiology, Obesity metabolism, Obesity pathology, Phosphorylation, Primary Cell Culture, Receptor, Insulin metabolism, Adipose Tissue metabolism, Antigens, CD genetics, Histone Deacetylase 6 genetics, Insulin genetics, Microfilament Proteins genetics, Obesity genetics, Protein Processing, Post-Translational, Receptor, Insulin genetics
- Abstract
Insulin orchestrates metabolic homeostasis through a complex signaling network for which the precise mechanisms controlling its fine-tuning are not completely understood. Here, we report that Afadin, a scaffold protein, is phosphorylated on S1795 (S1718 in humans) in response to insulin in adipocytes, and this phosphorylation is impaired with obesity and insulin resistance. In turn, loss of Afadin enhances the response to insulin in adipose tissues via upregulation of the insulin receptor protein levels. This happens in a cell-autonomous and phosphorylation-dependent manner. Insulin-stimulated Afadin-S1795 phosphorylation modulates Afadin binding with interaction partners in adipocytes, among which HDAC6 preferentially interacts with phosphorylated Afadin and acts as a key intermediate to suppress insulin receptor protein levels. Adipose tissue-specific Afadin depletion protects against insulin resistance and improves glucose homeostasis in diet-induced obese mice, independently of adiposity. Altogether, we uncover a novel insulin-induced cellular feedback mechanism governed by the interaction of Afadin with HDAC6 to negatively control insulin action in adipocytes, which may offer new strategies to alleviate insulin resistance., (© 2019 The Authors.)
- Published
- 2019
- Full Text
- View/download PDF
44. Skeletal Muscle Insulin Sensitivity Show Circadian Rhythmicity Which Is Independent of Exercise Training Status.
- Author
-
Basse AL, Dalbram E, Larsson L, Gerhart-Hines Z, Zierath JR, and Treebak JT
- Abstract
Circadian rhythms can be perturbed by shift work, travel across time zones, many occupational tasks, or genetic mutations. Perturbed circadian rhythms are associated with the increasing problem of obesity, metabolic dysfunction, and insulin resistance. We hypothesized that insulin sensitivity in skeletal muscle follows a circadian pattern and that this pattern is important for overall metabolic function. This hypothesis was verified using mice as a model system. We observed circadian rhythmicity in whole body insulin tolerance, as well as in signaling pathways regulating insulin- and exercise-induced glucose uptake in skeletal muscle, including AKT, 5'-adenosine monophosphate-activated protein kinase (AMPK) and TBC1 domain family member 4 (TBC1D4) phosphorylation. Basal and insulin-stimulated glucose uptake in skeletal muscle and adipose tissues in vivo also differed between day- and nighttime. However, the rhythmicity of glucose uptake differed from the rhythm of whole-body insulin tolerance. These results indicate that neither skeletal muscle nor adipose tissue play a major role for the circadian rhythmicity in whole-body insulin tolerance. To study the circadian pattern of insulin sensitivity directly in skeletal muscle, we determined glucose uptake under basal and submaximal insulin-stimulated conditions ex vivo every sixth hour. Both insulin sensitivity and signaling of isolated skeletal muscle peaked during the dark period. We next examined the effect of exercise training on the circadian rhythmicity of insulin sensitivity. As expected, voluntary exercise training enhanced glucose uptake in skeletal muscle. Nevertheless, exercise training did not affect the circadian rhythmicity of skeletal muscle insulin sensitivity. Taken together, our results provide evidence that skeletal muscle insulin sensitivity exhibits circadian rhythmicity.
- Published
- 2018
- Full Text
- View/download PDF
45. Cold-Activated Lipid Dynamics in Adipose Tissue Highlights a Role for Cardiolipin in Thermogenic Metabolism.
- Author
-
Lynes MD, Shamsi F, Sustarsic EG, Leiria LO, Wang CH, Su SC, Huang TL, Gao F, Narain NR, Chen EY, Cypess AM, Schulz TJ, Gerhart-Hines Z, Kiebish MA, and Tseng YH
- Subjects
- Animals, Biomarkers blood, Humans, Metabolomics, Mice, Models, Animal, Phosphatidylglycerols metabolism, Adipose Tissue, Brown metabolism, Adipose Tissue, White metabolism, Cardiolipins metabolism, Cold Temperature, Lipids blood, Thermogenesis
- Abstract
Thermogenic fat expends energy during cold for temperature homeostasis, and its activity regulates nutrient metabolism and insulin sensitivity. We measured cold-activated lipid landscapes in circulation and in adipose tissue by MS/MS
ALL shotgun lipidomics. We created an interactive online viewer to visualize the changes of specific lipid species in response to cold. In adipose tissue, among the approximately 1,600 lipid species profiled, we identified the biosynthetic pathway of the mitochondrial phospholipid cardiolipin as coordinately activated in brown and beige fat by cold in wild-type and transgenic mice with enhanced browning of white fat. Together, these data provide a comprehensive lipid bio-signature of thermogenic fat activation in circulation and tissue and suggest pathways regulated by cold exposure., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
46. Cardiolipin Synthesis in Brown and Beige Fat Mitochondria Is Essential for Systemic Energy Homeostasis.
- Author
-
Sustarsic EG, Ma T, Lynes MD, Larsen M, Karavaeva I, Havelund JF, Nielsen CH, Jedrychowski MP, Moreno-Torres M, Lundh M, Plucinska K, Jespersen NZ, Grevengoed TJ, Kramar B, Peics J, Hansen JB, Shamsi F, Forss I, Neess D, Keipert S, Wang J, Stohlmann K, Brandslund I, Christensen C, Jørgensen ME, Linneberg A, Pedersen O, Kiebish MA, Qvortrup K, Han X, Pedersen BK, Jastroch M, Mandrup S, Kjær A, Gygi SP, Hansen T, Gillum MP, Grarup N, Emanuelli B, Nielsen S, Scheele C, Tseng YH, Færgeman NJ, and Gerhart-Hines Z
- Subjects
- Animals, Cells, Cultured, Energy Metabolism, Humans, Membrane Proteins genetics, Mice, Mice, Inbred C57BL, Thermogenesis, Transferases (Other Substituted Phosphate Groups) genetics, Adipocytes metabolism, Adipose Tissue, Beige metabolism, Adipose Tissue, Brown metabolism, Cardiolipins biosynthesis, Membrane Proteins metabolism, Mitochondria metabolism, Transferases (Other Substituted Phosphate Groups) metabolism
- Abstract
Activation of energy expenditure in thermogenic fat is a promising strategy to improve metabolic health, yet the dynamic processes that evoke this response are poorly understood. Here we show that synthesis of the mitochondrial phospholipid cardiolipin is indispensable for stimulating and sustaining thermogenic fat function. Cardiolipin biosynthesis is robustly induced in brown and beige adipose upon cold exposure. Mimicking this response through overexpression of cardiolipin synthase (Crls1) enhances energy consumption in mouse and human adipocytes. Crls1 deficiency in thermogenic adipocytes diminishes inducible mitochondrial uncoupling and elicits a nuclear transcriptional response through endoplasmic reticulum stress-mediated retrograde communication. Cardiolipin depletion in brown and beige fat abolishes adipose thermogenesis and glucose uptake, which renders animals insulin resistant. We further identify a rare human CRLS1 variant associated with insulin resistance and show that adipose CRLS1 levels positively correlate with insulin sensitivity. Thus, adipose cardiolipin has a powerful impact on organismal energy homeostasis through thermogenic fat bioenergetics., (Copyright © 2018 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
47. NAMPT-mediated NAD + biosynthesis is indispensable for adipose tissue plasticity and development of obesity.
- Author
-
Nielsen KN, Peics J, Ma T, Karavaeva I, Dall M, Chubanava S, Basse AL, Dmytriyeva O, Treebak JT, and Gerhart-Hines Z
- Subjects
- Animals, Cells, Cultured, Cytokines genetics, Diet, High-Fat adverse effects, Energy Metabolism, Glucose metabolism, Loss of Function Mutation, Male, Mice, Mice, Inbred C57BL, Nicotinamide Phosphoribosyltransferase genetics, Obesity etiology, Adipose Tissue metabolism, Cytokines metabolism, NAD biosynthesis, Nicotinamide Phosphoribosyltransferase metabolism, Obesity metabolism
- Abstract
Objective: The ability of adipose tissue to expand and contract in response to fluctuations in nutrient availability is essential for the maintenance of whole-body metabolic homeostasis. Given the nutrient scarcity that mammals faced for millions of years, programs involved in this adipose plasticity were likely evolved to be highly efficient in promoting lipid storage. Ironically, this previously advantageous feature may now represent a metabolic liability given the caloric excess of modern society. We speculate that nicotinamide adenine dinucleotide (NAD
+ ) biosynthesis exemplifies this concept. Indeed NAD+ /NADH metabolism in fat tissue has been previously linked with obesity, yet whether it plays a causal role in diet-induced adiposity is unknown. Here we investigated how the NAD+ biosynthetic enzyme nicotinamide phosphoribosyltransferase (NAMPT) supports adipose plasticity and the pathological progression to obesity., Methods: We utilized a newly generated Nampt loss-of-function model to investigate the tissue-specific and systemic metabolic consequences of adipose NAD+ deficiency. Energy expenditure, glycemic control, tissue structure, and gene expression were assessed in the contexts of a high dietary fat burden as well as the transition back to normal chow diet., Results: Fat-specific Nampt knockout (FANKO) mice were completely resistant to high fat diet (HFD)-induced obesity. This was driven in part by reduced food intake. Furthermore, HFD-fed FANKO mice were unable to undergo healthy expansion of adipose tissue mass, and adipose depots were rendered fibrotic with markedly reduced mitochondrial respiratory capacity. Yet, surprisingly, HFD-fed FANKO mice exhibited improved glucose tolerance compared to control littermates. Removing the HFD burden largely reversed adipose fibrosis and dysfunction in FANKO animals whereas the improved glucose tolerance persisted., Conclusions: These findings indicate that adipose NAMPT plays an essential role in handling dietary lipid to modulate fat tissue plasticity, food intake, and systemic glucose homeostasis., (Copyright © 2018 The Authors. Published by Elsevier GmbH.. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
48. A Diurnal Rhythm in Brown Adipose Tissue Causes Rapid Clearance and Combustion of Plasma Lipids at Wakening.
- Author
-
van den Berg R, Kooijman S, Noordam R, Ramkisoensing A, Abreu-Vieira G, Tambyrajah LL, Dijk W, Ruppert P, Mol IM, Kramar B, Caputo R, Puig LS, de Ruiter EM, Kroon J, Hoekstra M, van der Sluis RJ, Meijer OC, Willems van Dijk K, van Kerkhof LWM, Christodoulides C, Karpe F, Gerhart-Hines Z, Kersten S, Meijer JH, Coomans CP, van Heemst D, Biermasz NR, and Rensen PCN
- Subjects
- Animals, Circadian Rhythm, Humans, Mice, Wakefulness, Adipose Tissue, Brown metabolism, Fatty Acids metabolism, Lipid Metabolism physiology
- Abstract
Many favorable metabolic effects have been attributed to thermogenic activity of brown adipose tissue (BAT). Yet, time of day has rarely been considered in this field of research. Here, we show that a diurnal rhythm in BAT activity regulates plasma lipid metabolism. We observed a high-amplitude rhythm in fatty acid uptake by BAT that synchronized with the light/dark cycle. Highest uptake was found at the onset of the active period, which coincided with high lipoprotein lipase expression and low angiopoietin-like 4 expression by BAT. Diurnal rhythmicity in BAT activity determined the rate at which lipids were cleared from the circulation, thereby imposing the daily rhythm in plasma lipid concentrations. In mice as well as humans, postprandial lipid excursions were nearly absent at waking. We anticipate that diurnal BAT activity is an important factor to consider when studying the therapeutic potential of promoting BAT activity., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
49. Kynurenic Acid and Gpr35 Regulate Adipose Tissue Energy Homeostasis and Inflammation.
- Author
-
Agudelo LZ, Ferreira DMS, Cervenka I, Bryzgalova G, Dadvar S, Jannig PR, Pettersson-Klein AT, Lakshmikanth T, Sustarsic EG, Porsmyr-Palmertz M, Correia JC, Izadi M, Martínez-Redondo V, Ueland PM, Midttun Ø, Gerhart-Hines Z, Brodin P, Pereira T, Berggren PO, and Ruas JL
- Subjects
- Adipocytes metabolism, Adipose Tissue, Beige metabolism, Adipose Tissue, White metabolism, Adiposity, Animals, Body Weight drug effects, Cells, Cultured, Diet, High-Fat, Epididymis metabolism, Gene Expression Profiling, Gene Expression Regulation, Glucose metabolism, Lymphocytes metabolism, Male, Mice, Inbred C57BL, Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha metabolism, Physical Conditioning, Animal, RGS Proteins metabolism, Receptors, Adrenergic, beta metabolism, Receptors, G-Protein-Coupled deficiency, Subcutaneous Fat metabolism, Transcription, Genetic, Adipose Tissue metabolism, Adipose Tissue pathology, Energy Metabolism, Homeostasis, Inflammation metabolism, Inflammation pathology, Kynurenic Acid metabolism, Receptors, G-Protein-Coupled metabolism
- Abstract
The role of tryptophan-kynurenine metabolism in psychiatric disease is well established, but remains less explored in peripheral tissues. Exercise training activates kynurenine biotransformation in skeletal muscle, which protects from neuroinflammation and leads to peripheral kynurenic acid accumulation. Here we show that kynurenic acid increases energy utilization by activating G protein-coupled receptor Gpr35, which stimulates lipid metabolism, thermogenic, and anti-inflammatory gene expression in adipose tissue. This suppresses weight gain in animals fed a high-fat diet and improves glucose tolerance. Kynurenic acid and Gpr35 enhance Pgc-1α1 expression and cellular respiration, and increase the levels of Rgs14 in adipocytes, which leads to enhanced beta-adrenergic receptor signaling. Conversely, genetic deletion of Gpr35 causes progressive weight gain and glucose intolerance, and sensitizes to the effects of high-fat diets. Finally, exercise-induced adipose tissue browning is compromised in Gpr35 knockout animals. This work uncovers kynurenine metabolism as a pathway with therapeutic potential to control energy homeostasis., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
50. Keeping fat on time: Circadian control of adipose tissue.
- Author
-
Ninel Hansen S, Peics J, and Gerhart-Hines Z
- Subjects
- Animals, Humans, Adipose Tissue physiology, Circadian Rhythm physiology
- Abstract
Circadian clocks harmonize processes ranging from intracellular biochemistry to whole-body physiology in accordance with the Earth's 24h rotation. These intrinsic oscillators are based on an interlocked transcriptional-translational feedback loop comprised from a set of core clock factors. In addition to maintaining rhythmicity in nearly every cell of the body, these clock factors also mediate tissue specific metabolic functions. In this review, we will explore how the molecular clock shapes the unique features of different adipose depots., (Copyright © 2017. Published by Elsevier Inc.)
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.