Category: Diet

Amplified fat metabolism

Amplified fat metabolism

An elevation in glucose promotes the metsbolism by 1 inhibition of FA Replenishing muscle glycogen β-Ox via Amplitied inhibition metabolsim carnitine Amplified fat metabolism, emtabolism catalyzes the limiting step of this pathway such that cytosolic FACoA increases; 2 provision of Gly3P for GL Ampliifed and 3 activation Leafy greens for hormonal balance lipolysis at least in some tissues Essential oils for hormone balance as the Akplified. Nat Commun 13 C75 increases ATP levels in many cell types, including neurons, to signal a favorable energy balance, which in turn inhibits hypothalamic AMPK, and alters gene expression in neurons and in peripheral tissues, further supporting weight loss. The mitochondria number was reduced in iWAT and BAT of AKO mice compared to that of floxed mice, whereas that in eWAT did not differ in these two genotypes Supplementary Figure 1Msuggesting that AMPK deletion likely inhibited mitochondrial biogenesis in iWAT and BAT, both of which contribute to adaptive thermogenesis during prolonged cold exposure. Protein Group Helps Cancer Cells Hide from Immune System.

Jump to main content. Contact Macronutrient distribution. Citation Tags HERO ID. Reference Type. Rat Article. Role of energy charge and AMP-activated Amplifked kinase in adipocytes in metabopism control of body fat stores.

Author s. Vat, M; Amp,ified, J; Flachs, P; Amplfied P; Sponarova, J; Ampilfied, O; Prazak, T; Amplified fat metabolism, J; Bardova, K; Kuda, O;.

Page Ampified. Web of Science Id. As indicated by in vitro studies, both lipogenesis Party decorations and accessories lipolysis in adipocytes depend metabolizm the cellular ATP metaabolism.

Ectopic expression of mitochondrial uncoupling protein Amp,ified Essential oils for hormone balance in the white adipose tissue of metabolis aP2-Ucp1 transgenic Amplicied reduced obesity Fwt by genetic metabolizm dietary manipulations.

Furthermore, respiratory uncoupling lowered the cellular energy charge in adipocytes, while the synthesis of fatty acids FA was inhibited and their oxidation increased. Importantly, the complex metabolic changes triggered by ectopic UCP1 were associated with the activation of AMP-activated protein kinase AMPKa metabolic master switch, in adipocytes.

Effects of several typical treatments that reduce adiposity, such as administration of leptin, beta-adrenoceptor agonists, bezafibrate, dietary n-3 polyunsaturated FA or fasting, can be compared with a phenotype of the aP2-Ucp1 mice. These situations generally lead to the upregulation of mitochondrial UCPs and suppression of the cellular energy charge and FA synthesis in adipocytes.

On the other hand, FA oxidation is increased. Moreover, it has been shown that AMPK in adipocytes can be activated by adipocyte-derived hormones leptin and adiponectin, and also by insulin-sensitizes thiazolidinediones.

Thus, it is evident that metabolism of adipose tissue itself is important for the control of body fat content and that the cellular energy charge and AMPK are involved in the control of lipid metabolism in adipocytes.

The reciprocal link between synthesis and oxidation of FA in adipocytes represents a prospective target for the new treatment strategies aimed at reducing obesity. Conference Name. Conference Location.

: Amplified fat metabolism

Glycerolipid Metabolism and Signaling in Health and Disease | Endocrine Reviews | Oxford Academic

The channels act like conduits that speed signals across the gaps between clusters of cells — similar to the way a group email reaches several people at once. The study, recently published in Cell Metabolism , also found that beige fat, unlike the better-known white and brown fat, has interesting anti-diabetic effects on blood sugar metabolism that seem independent of temperature regulation.

Impaired glucose metabolism is a hallmark of diabetes. Scherer added. Fat, once considered merely a storage area for excess calories, is now appreciated as a dynamic tissue that comes in several forms with different functions that are still being identified.

White fat is used mainly for energy storage. Brown fat, the classic heat-generating fat, helps regulate body temperature, especially in newborns. Some white fat cells are capable of transforming into a third kind of fat, beige. Both brown and beige fat get their color from increased mitochondria that are added in response to cold and other environmental stimuli, he said.

To study the metabolic effects of beige fat, the researchers compared mice with Cx43 that are able to make beige fat normally to mice unable to make Cx43, meaning their white fat seldom got the message to change to beige in response to cold.

After three weeks in cold temperatures, the mice were returned to normal temperatures and analyzed for glucose blood sugar metabolism. The mice that produced Cx43 showed greater improvement in glucose metabolism, Dr.

Scherer said. Yet, both groups of animals were still able to regulate body temperature, apparently through their brown fat stores, he said. Scherer, who holds the Gifford O.

Touchstone, Jr. and Randolph G. Touchstone Distinguished Chair in Diabetes Research. Study co-authors from the Touchstone Diabetes Center included lead author Dr. Yi Zhu, a postdoctoral fellow and employee of Eli Lilly and Co.

Caroline Tao; postdoctoral researchers Dr. Mengle Shao and Dr. Shangang Zhao; Dr. Olga Gupta, Assistant Professor of Pediatrics and Internal Medicine and a Dedman Family Scholar in Clinical Care; and Dr. William Holland and Dr. Rana Gupta, both Assistant Professors of Internal Medicine.

Tiemin Liu, Instructor of Internal Medicine; Dr. Joel Elmquist, Chief of the Division of Hypothalamic Research, Professor of Internal Medicine, Pharmacology and Psychiatry, and holder of the Maclin Family Distinguished Professorship in Medical Science, in Honor of Dr.

Roy A. Brinkley, and the Carl H. Westcott Distinguished Chair in Medical Research; and Dr. Kevin Williams, Assistant Professor Internal Medicine in the Division of Hypothalamic Research.

The research was supported by the NIH, the Cancer Prevention and Research Institute of Texas CPRIT ; a Lilly Innovation Fellowship Award; a China Scholarship Council Scholarship, and the American Heart Association.

About UT Southwestern Medical Center UT Southwestern, one of the premier academic medical centers in the nation, integrates pioneering biomedical research with exceptional clinical care and education. The faculty of almost 2, is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments.

UT Southwestern physicians provide medical care in about 80 specialties to more than , hospitalized patients and oversee approximately 2. Media Contact: Deborah Wormser Email.

To automatically receive news releases from UT Southwestern via email, subscribe at www. Two hard-to-match transplant patients miles apart are starting on a new path to healthy lives. UT Southwestern Medical Center is collaborating with Pfizer Inc.

Automated external defibrillators AEDs are underutilized during cardiac arrest episodes despite laws in some states requiring their availability in high-risk areas such as athletic facilities, researchers at UT Southwestern Medical Center found. Although the critical role of ATGL in lipolysis is undisputed, the distinct biochemical functions of its most closely related family members—PNPLA1, PNPLA3, PNPLA4 and PNPLA5—remain equivocal.

While an unambiguous assignment of their primary physiological substrates is still missing, some of these enzymes are directly involved in disease development. PNPLA1 is a amino-acid protein specifically expressed in differentiated keratinocytes of the skin , It is an established causative gene for autosomal recessive congenital ichthyosis ARCI , which is characterized by a severe defect in the development of the epidermal corneocyte lipid envelope CLE and the transepithelial water barrier in the skin in humans and dogs The clinical presentation essentially phenocopies the deficiency of CGI in the skin , arguing for a common involvement of both proteins in the formation of the CLE.

PNPLA1 is a transacylase that specifically transfers linoleic acid from TG to ω-hydroxy ceramide, thus giving rise to ω- O -acylceramides , These skin-specific lipids are essential for the generation of the CLE and an intact skin permeability barrier. Transacylation of FAs requires a hydrolysis reaction that is coupled with an esterification reaction.

CGI interacts with PNPLA1 and recruits the enzyme onto cytosolic LDs where PNPLA1 utilizes TG-derived FAs for ω-hydroxy ceramide acylation , PNPLA1 mutations leading to ARCI may additionally impair autophagy, arguing for a potential role of PNPLA1 in lipophagy-mediated degradation of LDs In humans, PNPLA3 is less abundant in adipocytes, but highly expressed in hepatocytes Key transcription factors responsible for the expression of PNPLA3 include CHREBP in response to carbohydrates and SREBP1c in response to insulin , PNPLA3 localizes to LDs and exhibits various enzymatic activities including TG-, DG-, MG- and retinyl ester hydrolase activity, PLA 2 activity and transacylase activity 8 , , , It remains unclear which of these activities are physiologically relevant in adipose tissue and in the liver The fact that PNPLA3-KO mice have essentially no abnormal phenotype with normal plasma and hepatic TG concentrations , argued against a major role of PNPLA3 in lipid homeostasis.

This view changed dramatically when Hobbs and colleagues reported that a single amino acid exchange at position from isoleucine to methionine in PNPLA3 p.

IM strongly associates with NAFLD This association was replicated in numerous studies and extended to associations of the mutation with steatohepatitis, fibrosis, cirrhosis and hepatocellular carcinoma Important progress concerning the function of PNPLA3 was achieved by recent studies suggesting that excess PNPLA3 on the surface of LDs competes with ATGL for its coactivator CGI refs.

PNPLA3-IM has higher affinity to CGI than does wild-type PNPLA3 ref. This leads to reduced ATGL activity, which may explain the fatty liver disease in people carrying the variant. Additionally, the IM variant may promote increased TG synthesis via a transacylation reaction PNPLA4 was originally identified as gene sequence 2 GS2 in the human genome and is also found in other mammalian species but not in mice The PNPLA4 gene is located on the X chromosome between the genes encoding steroid sulfatase and Kallmann Syndrome The human PNPLA4 protein comprises amino acids and is expressed in numerous tissues, including adipose tissue, skeletal and cardiac muscle, kidney, liver and skin , Similar to PNPLA3, various enzymatic activities have been attributed to PNPLA4, including TG and retinyl ester hydrolase, phospholipase and transacylase activities 8 , , Genetic analyses have suggested that PNPLA4 is involved in the development of two rare congenital disorders: combined oxidative phosphorylation deficiency COXPD and X-linked intellectual disability , COXPD arises from a hemizygous nonsense mutation resulting in a C-terminally-truncated protein Rter and defective assembly of complexes I, III and IV of the respiratory electron transport chain.

Whether the enzymatic activity of PNPLA4 is involved in the pathogenesis of COXPD is unknown. Fortunately, small-molecule inhibitors have recently been developed that may help to elucidate the biochemical function of PNPL4 and its role in patho- physiology PNPLA5 also named GS2-like is a amino-acid protein that is ubiquitously expressed and, unlike PNPLA4, is present in both mice and humans PNPLA5 exerts hydrolytic activities towards multiple lipid substrates, including TGs and retinyl esters, but has also been shown to exhibit transacylase activity , Notably, PNPLA5-dependent hydrolysis of LD-associated TGs has been suggested to affect autophagosome biogenesis Furthermore, rare variants of PNPLA5 strongly associate with LDL cholesterol levels in humans They catalyse the hydrolysis of a wide range of endogenous substrates and xenobiotics CES family members that exhibit neutral TG hydrolase activity include human CES1 also called human cholesterylesterhydrolase-1, hCEH and CES2, and mouse Ces1d also called Ces3 or TG hydrolase-1 , Ces1g also called Ces1 or esterase-X and Ces1c.

The previously confusing nomenclature of human and murine CES enzymes has recently been revised for more clarity The carboxylesterase 1 CES1 gene in the human genome encodes a kDa protein with high abundance in liver and intestine and lower abundance in kidney, adipose tissue, heart and macrophages , CES1 preferably hydrolyses CEs and TGs , Hepatocyte-specific overexpression of human CES1 in transgenic mice increases hepatic TG hydrolase activity; lowers TG, FA and cholesterol content; and reduces reactive oxygen species ROS , apoptosis and inflammation, which cumulatively protect mice from western-diet- or alcohol-induced steatohepatitis Both hepatocyte- and macrophage-specific overexpression of CES1 in mice decrease atherosclerosis susceptibility in LDL-receptor-deficient mice and, consistent with this finding, treatment of THP-1 macrophages with a CES1 inhibitor caused accumulation of CE Hence, the role of human CES1 in lipid metabolism remains controversial.

Ces1d is highly expressed in adipose tissue and liver where it localizes to the lumen of the ER In adipose tissue, Ces1d is additionally associated with LDs and may contribute to basal lipolysis , In mouse hepatocytes, transgenic overexpression or pharmacological inhibition of Ces1d increased or decreased VLDL assembly and secretion, respectively , , In accordance with these findings, liver-specific and global Ces1d deficiency in mice lowers VLDL production and plasma TG and CE concentrations , Despite strong evidence for a role of Ces1d in hepatic VLDL assembly and secretion, it remains unclear how an enzyme residing in the lumen of the ER actually contributes mechanistically to lipoprotein synthesis.

Interestingly, while global Ces1d deficiency leads to TG accumulation in isolated hepatocytes, conditional Ces1d knockout in liver cells does not. In fact, they are actually protected from high-fat-diet-induced hepatic steatosis , The protective effect of Ces1d deficiency in the liver might be due to reduced FA-synthase activity and increased hepatic FA oxidation , Moreover, mice with global, but not liver-specific, Ces1d deficiency exhibit improved insulin sensitivity and glucose tolerance , The recent finding that Ces1d deficiency does not affect CE hydrolysis or bile-acid synthesis in mice argues against an important role for CES1 enzymes in cholesterol homeostasis see previous paragraph.

The second murine ortholog to human CES1 with established TG hydrolase activity is Ces1g. Overexpression of Ces1g increases TG hydrolase activity and reduces TG content in cultured rat hepatoma cells and livers of transgenic mice , Conversely, global- and liver-specific Ces1g-KO mice exhibit hepatic steatosis and hyperlipidemia , Restoration of hepatic Ces1g expression reverses hepatic steatosis, hyperlipidemia and insulin resistance in global Ces1g-deficient mice Several studies suggest that the lipid phenotype observed in Ces1g-deficient mice results from diminished FA signalling to restrain SREBP1c activation, leading to reduced FA oxidation and increased de novo lipogenesis , , CES2 preferably hydrolyses esters with a large alcohol group and a small acyl group, and as for Ces2c, TG and DG hydrolase activities have been reported , CES2 is abundantly expressed in the small intestine, colon and liver and is decreased in mice and humans with obesity and NAFLD , , Adenoviral overexpression of human CES2 in the liver of mice prevents genetic- and diet-induced steatohepatitis by increasing TG hydrolase activity and FA oxidation and reduces SREBP1c mediated lipogenesis, inflammation, apoptosis and fibrosis.

Murine Ces2c is a highly abundant TG, DG and MG hydrolase in liver and intestine , Its expression is decreased in livers of mice with diet- or genetically induced obesity , Loss of hepatic Ces2c in chow- or western-diet-fed mice causes hepatic steatosis Conversely, increasing hepatic Ces2c expression ameliorated obesity and hepatic steatosis, and improved glucose tolerance and insulin sensitivity Mechanistically, FAs released by Ces2c enter FA oxidation and concomitantly inhibit SREBP1 activity to decrease de novo lipogenesis in the liver Similarly, intestine-specific transgenic expression of Ces2c in mice also promotes intestinal TG hydrolysis and FA oxidation, and ameliorates high-fat-diet-induced obesity and NAFLD Moreover, an increased re-esterification rate of MGs and DGs may contribute to the generation of large chylomicrons that undergo accelerated postprandial TG clearance Taken together, the physiological function of both human CES2 and murine Ces2c render this enzyme an attractive target for the treatment of metabolic liver disease.

The putative enzyme is mainly expressed in BAT, WAT, liver, heart and macrophages of atherosclerotic lesions The LDAH ortholog in D.

melanogaster CG facilitates lipid packaging and regulates LD clustering in cell and tissue cultures of salivary glands , Unfortunately, only conflicting data exist on the physiological function of the enzyme in mammalian cells.

Goo et al. The same group also showed that LDAH promotes LD accumulation by affecting ATGL ubiquitinylation and degradation In other studies, however, LDAH overexpression did not affect CE, TG and retinyl ester hydrolase activity, and LDAH ablation did not change CE turnover in bone-marrow-derived macrophages Also, mice globally lacking LDAH exhibit normal CE, TG and whole-body energy metabolism, suggesting that LDAH either plays only a minor role in lipid metabolism or when absent is replaced by effective compensatory mechanisms Notably, genetic studies revealed that loss of LDAH may be associated with prostate cancer and sensorineural hearing loss Without a better functional characterization of this putative lipase, it is currently impossible to assess its role in lipid and energy homeostasis or cancer development.

DDH domain-containing protein 2 DDHD2, also named KIAA or iPLA1γ belongs to the mammalian intracellular phospholipase A1 iPLA1 family which consists of three paralogs—DDHD1, DDHD2 and SEC23IP. All members share a conserved GXSXG lipase motif and a DDHD domain named after a characteristic amino acid signature within the domain , DDHD2 exhibits DG and TG hydrolase activity, and DDHD2 deficiency in mice causes TG accumulation in the brain , , , The protein localizes to the cytosol, Golgi apparatus and ER , In humans, loss-of-function mutations in DDHD2 lead to hereditary spastic paraplegia , , , a neurodegenerative disorder associated with lipid accumulation in the brain and characterized by spasticity in the lower limbs Additionally, short-hairpin-RNA-mediated DDHD2 knockdown was shown to impair phospholipid synthesis and sensory axon regeneration after sciatic nerve crush , emphasizing a crucial role for lipolysis in neuron function.

Moreover, genome-wide analyses identified DDHD2 as a candidate risk gene for other neurological diseases, including autism and schizophrenia , Whether these diseases are directly linked to the enzymatic activity of DDHD2 is currently investigated. Arylacetamide deacetylase AADAC is a kDa esterase that harbours a typical GXSXG motif and exhibits considerable sequence homology to HSL in its presumed active site AADAC localizes to the ER and is mainly expressed in liver and intestine with much lower expression in adrenal glands and pancreas , AADAC preferably hydrolyses cholesteryl acetate and DGs over TGs , Its diurnal expression pattern is identical to hepatic VLDL secretion in mice and depends on transcriptional regulation by PPARα ref.

Overexpression of AADAC in human HepG2 and rat McA hepatoma cells increases TG secretion and reduces TG accumulation, respectively, indicating that it might play a role in lipid mobilization for VLDL synthesis , Another study has demonstrated that, although hepatic AADAC activity is decreased in humans with obesity, there was no association to the homeostasis model assessment HOMA index or with DG species, and knockdown of endogenous AADAC did not affect FA oxidation in primary human hepatocytes Accordingly, the role of AADAC in lipid metabolism in vivo is still elusive.

Extracellular vesicles EVs represent a recently discovered mechanism for the utilization of TG stores. In contrast to lipolysis and lipophagy, EV-mediated TG utilization does not involve TG hydrolysis and subsequent FA distribution.

Instead, LDs are directly released from cells within EVs. A familiar, unique subclass of EVs containing large amounts of TGs are milk fat globules. They originate from cuboidal epithelial cells of lactating mammary glands and are secreted into milk ducts Apart from the very distinct milk fat globules, EVs are generally secreted from almost all cell types and are found in interstitial fluid and many other body fluids.

After their release, EVs are internalized by other cells, enabling the horizontal transfer of cargo from donor to recipient cells. The nature of their cargo is manifold and includes proteins, nucleic acids, various metabolites and even intact organelles EVs have thus been attributed diverse biological functions ranging from immunomodulation to tissue differentiation and cancer development Importantly, Ferrante and colleagues reported that adipocyte-derived EVs are able to transport TGs to adipose-tissue-resident macrophages Similar to all EVs, TG-containing exosomes are enclosed by a phospholipid bilayer derived from the plasma membrane of EV-producing cells Additionally, a phospholipid monolayer engulfs the hydrophobic core of enclosed LDs Fig.

Adipocyte-derived exosomes are almost exclusively taken up by macrophages and may thus represent an alternative pathway for local intercellular lipid distribution in adipose tissue. Through this process, macrophages may adapt to their local adipose tissue environment, consistent with similar findings in other tissues Whether adipocyte-derived EVs can transport TG cargo to cells and tissues other than macrophages remains to be elucidated.

TGs are shuttled from adipocytes to macrophages in adipose tissue via exosomes. Exosomes comprise a phospholipid bilayer and phospholipid monolayer engulfing the hydrophobic core of enclosed LDs and harbour exosomal marker proteins, such as CD63, and LD proteins, such as perilipins or ATGL.

Sebocytes represent another cell type secreting large amounts of mostly neutral lipids including TGs. These lipids accumulate in LDs during sebocyte differentiation but in contrast to adipocytes, lipid mobilization occurs via sebocyte necrosis instead of lipolysis or exosome-mediated pathways Sebum hypersecretion is associated with increased fat catabolism in adipose tissue and associated with adipose tissue loss in a TSLP transgenic mouse model.

Considering the essential role of lipolytic enzymes in systemic and tissue-specific energy homeostasis, the inhibition of lipolysis may offer potential treatment opportunities for various metabolic, and possibly even infectious, diseases. Example target diseases are described below and in Box 1.

Viruses are obligate intracellular pathogens that hijack host cells and create an intracellular metabolic niche to facilitate efficient replication and virion production.

Striking examples of such hostile takeovers include positive-stranded RNA viruses that rewire lipid metabolism to form membrane platforms, so called replication compartments RCs providing a structural framework for RNA synthesis , , These RCs additionally enable local accrual of viral macromolecules and hinder host defence mechanisms.

Flaviviruses including dengue virus, West Nile virus and hepatitis C virus HCV , as well as severe acute respiratory syndrome coronavirus SARS-CoV , exploit ER membranes for RC formation. Picornaviruses, a large family of positive-stranded RNA viruses, stimulate phospholipid—especially phosphatidylcholine—biosynthesis to form RCs Accordingly, virus-infected cells upregulate FA uptake and CoA activation Notably, however, exogenous FAs do not directly enter phospholipid synthesis but are first esterified to TGs by DGAT1 and deposited in LDs On demand, activation of lipolysis promotes FA release and subsequent RC-membrane synthesis.

During infection by enteroviruses, including rhinovirus, inhibition of DGAT1, ATGL or HSL largely reduces viral replication To access cellular lipid stores, viruses use newly formed membrane contact sites between LDs and RCs.

A direct interaction of viral proteins with ATGL and HSL at these contact sites may promote FA channelling towards RC synthesis Besides providing FAs for RC formation, LDs also serve as platforms for HCV and dengue virus-particle synthesis , , and inhibition of LD formation decreases dengue virus and HCV replication For both viruses, particle morphogenesis depends on the localization of the basic core protein to LDs, from which it is recruited for virus particle formation The interface between LDs and ER may serve as HCV assembly platforms Once bound to LDs, HCV core protein inhibits ATGL-mediated lipolysis to increase LD volume and number.

This process likely contributes to the hepatic steatosis commonly observed in people infected with HCV Similarly, accumulation of LDs was also observed in infected African green monkey kidney epithelial cells and in lungs of people infected with SARS-CoV-2, suggesting that SARS-CoV-2 pathogenesis also involves lipid metabolism Lipolysis of LD-associated TGs furthermore provides energy substrates for ATP production that are required for viral replication and virion formation.

In some cases, such as upon infection with the lymphocytic choriomeningitis LCM virus, activation of neutral lipolysis via ATGL and HSL is so pronounced that it contributes to the drastic reduction in mass of adipose tissue in infected mice Other viruses, such as dengue virus , influenza H3N2 virus and porcine reproductive and respiratory syndrome virus , activate lipophagy and acid lipolysis to mobilize FAs from LDs.

FAs are subsequently oxidized in mitochondria to fuel viral replication. Similarly, poliovirus , HCV and coxsackievirus B3 ref. Thus, virus infections inhibit or activate lipolysis depending on whether the virus utilizes LDs as an assembly and replication platform or as a source of FAs as an energy substrate, respectively.

HCV exploits the VLDL synthesis pathway to exit hepatocytes as lipo-viro-particles Knockdown of arylacetamide deacetylase AADAC , a putative lipase involved in VLDL synthesis, largely decreases virus production Moreover, HCV production depends on hepatic CGI ref.

Loss-of-function mutations in CGI, which are associated with NLSDI, diminish HCV production, indicating that the prolipolytic function of CGI, but not of ATGL, is required for HCV formation. The contradiction that the HCV life cycle depends on inactive ATGL but active CGI may be resolved by the identification of an ATGL-independent role of CGI in VLDL synthesis Finally, adipose tissue is also a refuge for several pathogenic parasites, including Trypanosoma cruzi , Trypanosoma brucei , Coxiella burnetiid , Rickettsia prowazekii and Mycobacterium tuberculosis cruzi causes Chagas disease and invades adipocytes, where it resides in close proximity to LDs, ensuring that the parasite has a continuous supply of FAs Adipose tissue represents a major reservoir for T.

brucei , which adapts to the lipid-rich environment by increased ATGL-mediated lipolysis and FA oxidation to satisfy their energy requirements This catabolic switch towards fat catabolism would also explain the weight loss in people and cattle with sleeping sickness caused by this parasite , Studies in Listeria monocytogenes -infected mice demonstrated a link between fasting and increased survival, and forced feeding of infected mice with a glucose-containing diet was lethal Apparently, lipolysis-associated metabolites like ketone bodies are essential for surviving L.

monocytogenes infections. Anorexia and even cachexia accompany most infections and may be seen as host defence mechanisms to mimic fasting and activate adipose-tissue FA mobilization for ketogenesis in the liver to improve immune function and increase the chance of survival.

Type 2 diabetes is a serious public-health concern that impairs quality and expectancy of life. Obesity represents the major risk factor for type 2 diabetes It is well accepted that an increased FA flux from hypertrophic adipose tissue to insulin-sensitive tissues contributes to insulin resistance and glucose intolerance Accordingly, reduced FA mobilization from adipose tissue in mice with global or adipocyte-specific deletion of ATGL, HSL or LAL leads to significantly increased glucose tolerance and improved insulin sensitivity 24 , 27 , , , , For HSL deficiency, the beneficial metabolic phenotype is present in old, but not young, mice, and may be due to a concomitant decline in ATGL activity, which strongly impairs FA mobilization from adipose stores in older animals Pharmacological inhibition of ATGL or HSL also improved glucose tolerance and insulin sensitivity in high-fat-diet-fed mice.

A shift towards glucose utilization upon reduced FA availability from adipose tissue, reduced hepatic acetyl-CoA levels and pyruvate carboxylase activity and, consequently, reduced hepatic glucose production contributes to increased glucose tolerance in mice with adipocyte-specific ATGL deficiency Hypoinsulinemia occurs in both ATGL- and HSL-deficient mice and likely contributes to the insulin-sensitive phenotype 27 , , , Insulin, secreted from pancreatic β-cells, regulates lipid versus carbohydrate utilization as fuel for energy.

β-cell-intrinsic lipolysis generates various lipid intermediates with signalling potential like MGs, FA-CoAs and FAs that were shown to regulate glucose-stimulated insulin secretion GSIS Studies in global- and β-cell-specific ATGL and HSL-deficient mice revealed impaired GSIS of isolated pancreatic islets and hypoinsulinemia 26 , 31 , , , , In contrast, inhibition of LAL in pancreatic β-cells indirectly potentiates GSIS by activating ATGL and HSL Several mechanisms have been proposed for impaired insulin secretion in ATGL-deficient β-cells, including 1 reduced availability of PPARδ ligands resulting in mitochondrial dysfunction 31 , 2 reduced generation of MGs that induce insulin exocytosis by binding to the vesicle priming protein Munc13—1 refs.

Moreover, FAs liberated from adipose tissue promote insulin secretion after β-adrenergic stimulation and reduced or diminished adipocyte ATGL or HSL activity perturbs insulin secretion from β-cells 27 , , , , Hence, specifically manipulating adipocyte lipolysis may represent a potential pharmacological approach to improve insulin sensitivity and glucose tolerance.

Obesity-associated NAFLD results from either increased de novo TG synthesis in the liver or increased flux of FAs from adipose tissue to the liver, or both Accordingly, reduced FA mobilization from adipocytes ameliorates hepatic steatosis, inflammation and fibrosis in high-fat-diet-fed mice that have adipocyte-specific ATGL deficiency or CGI deficiency or that have been treated with atglistatin , , Somewhat counterintuitively, overexpression of ATGL in adipocytes also improves hepatic steatosis in a transgenic mouse model However, evidence shows that enhanced FA oxidation in adipose tissues of adipocyte-specific ATGL transgenic mice reduces FA flux to the liver, resulting in decreased hepatic fat storage.

Numerous GWAS and candidate-gene studies have demonstrated that the PNPLA3-IM variant represents a strong genetic risk factor for steatohepatitis, fibrosis and HCC ASO-mediated silencing of PNPLA3 improved NAFLD and fibrosis in a mutant knock-in mouse model harbouring the PNPLA3-IM mutation Moreover, PNPLA3-IM sequesters CGI from its interaction with ATGL and impairs its hydrolytic activity for LD-associated TGs This therapeutic concept is corroborated by the finding that diet-induced hepatic steatosis is prevented by adenovirus-mediated overexpression of ATGL in murine livers Chronic heart failure HF is a common disease resulting from any structural or functional impairment of ventricular filling or ejection of blood HF represents a leading cause of death and can basically be subdivided into two distinct groups: HF with preserved ejection fraction HFpEF and HF with restricted ejection fraction HFrEF The healthy heart exhibits high flexibility to use FA, glucose and ketone bodies as energy substrates During cardiac failure, metabolic flexibility is reduced, and the heart increasingly uses glucose as energy fuel However, a chronic activation of the sympathetic nervous system and an increase in natriuretic peptides augments adipose FA mobilization, which competes with glucose utilization and aggravates oxidative stress during HF , , Accordingly, adipocyte-specific ATGL deficiency or atglistatin treatment protected mice from pressure-induced cardiac hypertrophy and cardiac fibrosis , Similarly, ATGL inhibition promoted a cardioprotective effect in a mouse model of catecholamine-induced cardiac damage It has been suggested that inhibition of adipocyte ATGL activity improves cardiac energy metabolism by further shifting substrate utilization from FAs towards glucose and by changing the cardiac membrane lipid composition Considering the current lack of effective therapeutics, the pharmacological inhibition of ATGL, and possibly other lipases in adipose tissue, may represent a promising approach to treat HFpEF.

Importantly, however, the inhibition of lipolysis must be restricted to adipose tissue because concomitant suppression of lipolysis in cardiac myocytes, hepatocytes and other non-adipose tissues may result in steatosis and organ dysfunction, as present in people with global ATGL 21 , 23 or CGI deficiency Cachexia is a severe wasting disorder accompanying the late stage of various diseases such as rheumatoid arthritis, chronic obstructive pulmonary disease, burn-induced trauma or cancer The occurrence of cachexia in any of these maladies drastically decreases treatment options and chances of survival.

Cachexia is a hypermetabolic disorder characterized by systemic inflammation and an unintended loss of body weight due to a reduction in adipose tissue and skeletal muscle mass that cannot be reversed by increased nutritional intake , To date, the multifactorial mechanisms causing cachexia are yet insufficiently understood and effective treatments are not available.

Adipose tissue loss often precedes muscle loss in cachexia, and some studies have provided evidence that halting fat depletion also preserves muscle mass , Mechanistically, adipose tissue undergoes a switch towards catabolism that is initiated by proinflammatory cytokines and amplified by β-adrenergic signals , This catabolic reprogramming elevates lipolysis and futile cycling, but reduces lipogenesis and fat accumulation, leading to adipose tissue atrophy , Reducing lipolysis by pharmacological or genetic inhibition of ATGL , or by targeting the erroneously prolipolytic signals, like the β3-adrenergic signal or the AMPK—CIDEA axis , prevented adipose-tissue loss in murine cachexia models of burn-induced trauma or cancer.

Interestingly, ATGL blockade not only reduced lipolysis but also prevented browning of WAT and fatty-liver development post burn injury and muscle atrophy in some mouse models of cancer cachexia The mechanisms underlying the tumour—adipose tissue—muscle crosstalk in cachexia and the question of whether lipolysis inhibition affects cachexia in humans await future clarification.

Discoveries in the past two decades have turned intracellular TG catabolism from a relatively simple, single-enzyme reaction by HSL to a complex, highly regulated pathway that affects energy homeostasis on numerous levels.

While canonical lipolysis of intracellular lipid stores is by now relatively well-characterized, the contribution of noncanonical enzymes and the role of lysosomal acid lipolysis for cytoplasmic LD degradation require comprehensive characterization.

In many cases, the enzymatic function of these proteins as well as their placement in lipid metabolism and physiology remain unclear, despite compelling evidence for their relevance in disease development.

A good example for such dreadful lack of knowledge relates to the crucial yet unsettled function of PNPLA3 in lipid metabolism and liver disease. Another exciting topic of future research will address the therapeutic potential of lipolytic enzymes to treat metabolic disorders.

Examples include inhibition of ATGL and possibly HSL to treat or prevent type 2 diabetes, fatty liver disease, lethal heart failure or cachexia. While preclinical experiments in mice are promising, their potential in humans remains completely unknown.

Similarly, it is of utmost interest to find out whether inhibition of PNPLA3 prevents NASH, liver cirrhosis and liver cancer in humans.

Future focus on these topics is likely to open preventative and therapeutic opportunities for the treatment of metabolic diseases. Unger, R. Gluttony, sloth and the metabolic syndrome: a roadmap to lipotoxicity. Trends Endocrinol. Article CAS PubMed PubMed Central Google Scholar.

Walther, T. Lipid droplet biogenesis. Cell Dev. Whitehead, R. A note on the absorption of fat. Content 24 , — Article Google Scholar. Vaughan, M. Hormone-sensitive lipase and monoglyceride lipase activities in adipose tissue.

Article CAS PubMed Google Scholar. Osuga, J. et al. Targeted disruption of hormone-sensitive lipase results in male sterility and adipocyte hypertrophy, but not in obesity. Natl Acad. USA 97 , — Haemmerle, G. Hormone-sensitive lipase deficiency in mice causes diglyceride accumulation in adipose tissue, muscle, and testis.

Zimmermann, R. Fat mobilization in adipose tissue is promoted by adipose triglyceride lipase. Science , — Jenkins, C. Identification, cloning, expression, and purification of three novel human calcium- independent phospholipase A2 family members possessing triacylglycerol lipase and acylglycerol transacylase activities.

Lass, A. Adipose triglyceride lipase-mediated lipolysis of cellular fat stores is activated by CGI and defective in Chanarin—Dorfman syndrome. Cell Metab. Kienesberger, P. Mammalian patatin domain containing proteins: a family with diverse lipolytic activities involved in multiple biological functions.

Lipid Res. Schweiger, M. The C-terminal region of human adipose triglyceride lipase affects enzyme activity and lipid droplet binding. Ahmadian, M. Pagnon, J.

Identification and functional characterization of protein kinase A phosphorylation sites in the major lipolytic protein, adipose triglyceride lipase.

Endocrinology , — Zhang, X. An epistatic interaction between Pnpla2 and Lipe reveals new pathways of adipose tissue lipolysis. Cells 8 , Article CAS PubMed Central Google Scholar. Brejchova, K. Distinct roles of adipose triglyceride lipase and hormone-sensitive lipase in the catabolism of triacylglycerol estolides.

USA , e Ohno, Y. PNPLA1 is a transacylase essential for the generation of the skin barrier lipid ω- O -acylceramide. Article PubMed PubMed Central Google Scholar. Soni, K. Coatomer-dependent protein delivery to lipid droplets.

Cell Sci. Ellong, E. Interaction between the triglyceride lipase ATGL and the Arf1 activator GBF1. PLoS ONE 6 , e Wang, T. OSBPL2 is required for the binding of COPB1 to ATGL and the regulation of lipid droplet lipolysis.

iScience 23 , Cai, M. FAMB promotes adipogenesis by increasing vesicular activity in porcine and 3T3-L1 adipocytes. Fischer, J. The gene encoding adipose triglyceride lipase PNPLA2 is mutated in neutral lipid storage disease with myopathy.

Missaglia, S. Neutral lipid storage diseases as cellular model to study lipid droplet function. Hirano, K. Triglyceride deposit cardiomyovasculopathy. Defective lipolysis and altered energy metabolism in mice lacking adipose triglyceride lipase. ATGL-mediated fat catabolism regulates cardiac mitochondrial function via PPAR-α and PGC Attané, C.

Diabetologia 59 , — Heine, M. Lipolysis triggers a systemic insulin response essential for efficient energy replenishment of activated brown adipose tissue in mice.

e4 Ong, K. Adipose triglyceride lipase is a major hepatic lipase that regulates triacylglycerol turnover and fatty acid signaling and partitioning. Hepatology 53 , — Schreiber, R. Hypophagia and metabolic adaptations in mice with defective ATGL-mediated lipolysis cause resistance to HFD-induced obesity.

USA , — Mottillo, E. Lipolytic products activate peroxisome proliferator-activated receptor PPAR α and δ in brown adipocytes to match fatty acid oxidation with supply. Tang, T. Khan, S. Diabetes 64 , — Najt, C. Lipid droplet-derived monounsaturated fatty acids traffic via PLIN5 to allosterically activate SIRT1.

Cell 77 , — e8 Hofer, P. Fatty acid-binding proteins interact with comparative gene identification linking lipolysis with lipid ligand shuttling. Recazens, E. Hormone-sensitive lipase: sixty years later. Albert, J. Null mutation in hormone-sensitive lipase gene and risk of type 2 diabetes.

Farhan, S. A novel LIPE nonsense mutation found using exome sequencing in siblings with late-onset familial partial lipodystrophy. Article PubMed Google Scholar. Morigny, P. Interaction between hormone-sensitive lipase and ChREBP in fat cells controls insulin sensitivity.

Decreased fatty acid esterification compensates for the reduced lipolytic activity in hormone-sensitive lipase-deficient white adipose tissue. Pajed, L. Advanced lipodystrophy reverses fatty liver in mice lacking adipocyte hormone-sensitive lipase. Hermo, L. Alterations in the testis of hormone sensitive lipase-deficient mice is associated with decreased sperm counts, sperm motility, and fertility.

Tornqvist, H. Purification and some properties of a monoacylglycerol-hydrolyzing enzyme of rat adipose tissue. Karlsson, M. cDNA cloning, tissue distribution, and identification of the catalytic triad of monoglyceride lipase.

Evolutionary relationship to esterases, lysophospholipases, and haloperoxidases. Savinainen, J. Robust hydrolysis of prostaglandin glycerol esters by human monoacylglycerol lipase MAGL.

Heier, C. Monoacylglycerol lipases act as evolutionarily conserved regulators of non-oxidative ethanol metabolism. Grabner, G. Monoglyceride lipase as a drug target: at the crossroads of arachidonic acid metabolism and endocannabinoid signaling.

Chon, S. Intestinal monoacylglycerol metabolism: developmental and nutritional regulation of monoacylglycerol lipase and monoacylglycerol acyltransferase. Rakhshandehroo, M. Comprehensive analysis of PPARα-dependent regulation of hepatic lipid metabolism by expression profiling.

PPAR Res. Rajasekaran, D. Staphylococcal nuclease and tudor domain containing 1 SND1 protein promotes hepatocarcinogenesis by inhibiting monoglyceride lipase MGLL.

Thomas, G. The serine hydrolase ABHD6 is a critical regulator of the metabolic syndrome. Cell Rep. Pribasnig, M. Metabolic disease and ABHD6 alter the circulating bis monoacylglycerol phosphate profile in mice and humans. Poursharifi, P. Monoacylglycerol signalling and ABHD6 in health and disease.

Diabetes, Obes. Article CAS Google Scholar. Tang, Z. Enhanced monoacylglycerol lipolysis by ABHD6 promotes NSCLC pathogenesis. EBioMedicine 53 , Grüner, B.

An in vivo multiplexed small-molecule screening platform. Methods 13 , — Tardelli, M. Lack of monoacylglycerol lipase prevents hepatic steatosis by favoring lipid storage in adipose tissue and intestinal malabsorption.

Douglass, J. Global deletion of monoacylglycerol lipase in mice delays lipid absorption and alters energy homeostasis and diet-induced obesity. Taschler, U. Monoglyceride lipase deficiency in mice impairs lipolysis and attenuates diet-induced insulin resistance.

Chanda, P. Monoacylglycerol lipase activity is a critical modulator of the tone and integrity of the endocannabinoid system. Schlosburg, J. Chronic monoacylglycerol lipase blockade causes functional antagonism of the endocannabinoid system. Nomura, D.

Endocannabinoid hydrolysis generates brain prostaglandins that promote neuroinflammation. Deletion of monoglyceride lipase in astrocytes attenuates lipopolysaccharide-induced neuroinflammation. Monoacylglycerol lipase regulates a fatty acid network that promotes cancer pathogenesis.

Cell , 49—61 Zhang, J. Monoacylglycerol lipase: a novel potential therapeutic target and prognostic indicator for hepatocellular carcinoma. Deng, H. Frühbeck, G. Regulation of adipocyte lipolysis.

Yogosawa, S. Diabetes 62 , — Roy, D. Coordinated transcriptional control of adipocyte triglyceride lipase Atgl by transcription factors Sp1 and peroxisome proliferator—activated receptor γ PPARγ during adipocyte differentiation.

Kim, J. Stenson, B. Liver X receptor LXR regulates human adipocyte lipolysis. Kulcenty, K. SF-1 NR5A1 expression is stimulated by the PKA pathway and is essential for the PKA-induced activation of LIPE expression in Y-1 cells. Gambo, Y. Triiodothyronine enhances accumulation of intracellular lipids in adipocytes through thyroid hormone receptor α via direct and indirect mechanisms.

Fujimoto, Y. TFE3 controls lipid metabolism in adipose tissue of male mice by suppressing lipolysis and thermogenesis. Czajkowski, M. Steroids , — Kaltenecker, D. Adipocyte STAT5 deficiency promotes adiposity and impairs lipid mobilisation in mice. Diabetologia 60 , — STAT5 is required for lipid breakdown and beta-adrenergic responsiveness of brown adipose tissue.

Shi, S. Adipocyte-specific deficiency of Janus kinase JAK 2 in mice impairs lipolysis and increases body weight, and leads to insulin resistance with ageing. Diabetologia 57 , — Hong, S. Phosphorylation of beta-3 adrenergic receptor at serine by ERK MAP kinase drives lipolysis in obese adipocytes.

Greenberg, A. Stimulation of lipolysis and hormone-sensitive lipase via the extracellular signal-regulated kinase pathway. Hjelholt, A. Temporal patterns of lipolytic regulators in adipose tissue after acute growth hormone exposure in human subjects: A randomized controlled crossover trial.

Kopchick, J. The effects of growth hormone on adipose tissue: old observations, new mechanisms. El-Merahbi, R. The adrenergic-induced ERK3 pathway drives lipolysis and suppresses energy dissipation. Genes Dev. Magnusson, B. Activin B inhibits lipolysis in 3T3-L1 adipocytes. Bu, Y. Diabetes 67 , — Zhu, H.

The effect of myostatin on proliferation and lipid accumulation in 3T3-L1 preadipocytes. Modica, S. Bmp4 promotes a brown to white-like adipocyte shift. Boon, M. BMP7 activates brown adipose tissue and reduces diet-induced obesity only at subthermoneutrality. PLoS ONE 8 , e Guo, T. Adipocyte ALK7 links nutrient overload to catecholamine resistance in obesity.

Elife 3 , e Li, F. Cell Biol. Lee, S. Targeting the myostatin signaling pathway to treat muscle loss and metabolic dysfunction. Langin, D. Importance of TNFα and neutral lipases in human adipose tissue lipolysis. Li, Y. A global perspective on FOXO1 in lipid metabolism and lipid-related diseases.

Chakrabarti, P. FoxO1 controls insulin-dependent adipose triglyceride lipase ATGL expression and lipolysis in adipocytes. Barthel, A. FoxO proteins in insulin action and metabolism.

Ow, J. Remodeling of whole-body lipid metabolism and a diabetic-like phenotype caused by loss of CDK1 and hepatocyte division.

eLife 9 , e Saline, M. AMPK and AKT protein kinases hierarchically phosphorylate the N-terminus of the FOXO1 transcription factor, modulating interactions with proteins. van der Heide, L. Trends Biochem. SIRT1 controls lipolysis in adipocytes via FOXO1-mediated expression of ATGL.

Jung, S. Non-canonical mTORC2 signaling regulates brown adipocyte lipid catabolism through SIRT6—FoxO1. Cell 75 , — Sun, C.

Adipose SNAIL1 regulates lipolysis and lipid partitioning by suppressing adipose triacylglycerol lipase expression. Mammalian target of rapamycin complex 1 suppresses lipolysis, stimulates lipogenesis, and promotes fat storage. Diabetes 59 , — Paolella, L.

mTORC1 restrains adipocyte lipolysis to prevent systemic hyperlipidemia. Sustained activation of autophagy suppresses adipocyte maturation via a lipolysis-dependent mechanism. Autophagy 16 , — Insulin inhibits lipolysis in adipocytes via the evolutionarily conserved mTORC1—Egr1—ATGL-mediated pathway.

Saxton, R. mTOR signaling in growth, metabolism, and disease. Cell , — Kumar, A. Fat cell-specific ablation of rictor in mice impairs insulin-regulated fat cell and whole-body glucose and lipid metabolism.

Caron, A. The roles of mTOR complexes in lipid metabolism. Steinberg, D. The role of cyclic AMP in activation of hormone-sensitive lipase of adipose tissue. Nucleotide Res. CAS Google Scholar. Sveidahl Johansen, O. Lipolysis drives expression of the constitutively active receptor GPR3 to induce adipose thermogenesis.

Cell , — e33 Kimmel, A. The perilipins: major cytosolic lipid droplet-associated proteins and their roles in cellular lipid storage, mobilization, and systemic homeostasis. Carper, D. Atrial natriuretic peptide orchestrates a coordinated physiological response to fuel non-shivering thermogenesis.

Yu, L. CGI versatile regulator of intracellular lipid droplet homeostasis. Gandotra, S. Perilipin deficiency and autosomal dominant partial lipodystrophy. Human frame shift mutations affecting the carboxyl terminus of perilipin increase lipolysis by failing to sequester the adipose triglyceride lipase ATGL coactivator AB-hydrolase-containing 5 ABHD5.

Wang, H. Unique regulation of adipose triglyceride lipase ATGL by perilipin 5, a lipid droplet-associated protein. Granneman, J. Interactions of perilipin-5 Plin5 with adipose triglyceride lipase. Pollak, N.

The interplay of protein kinase A and perilipin 5 regulates cardiac lipolysis. Keenan, S. Perilipin 5 S phosphorylation by PKA is required for the control of hepatic lipid metabolism and glycemic control. Kuramoto, K. Deficiency of a lipid droplet protein, perilipin 5, suppresses myocardial lipid accumulation, thereby preventing type 1 diabetes-induced heart malfunction.

Kolleritsch, S. Low cardiac lipolysis reduces mitochondrial fission and prevents lipotoxic heart dysfunction in perilipin 5 mutant mice. CAS PubMed Google Scholar. Patel, S. Perilipins 2 and 3 lack a carboxy-terminal domain present in perilipin 1 involved in sequestering ABHD5 and suppressing basal lipolysis.

Petersen, M. Mechanisms of insulin action and insulin resistance. DiPilato, L. The role of PDE3B phosphorylation in the inhibition of lipolysis by insulin. Xia, W.

Loss of ABHD15 impairs the anti-lipolytic action of insulin by altering PDE3B stability and contributes to insulin resistance. Vannucci, S. A1-adenosine receptor-mediated inhibition of adipocyte adenylate cyclase and lipolysis in Zucker rats. Ceddia, R. The role of AMP-activated protein kinase in regulating white adipose tissue metabolism.

Lack of adipocyte AMPK exacerbates insulin resistance and hepatic steatosis through brown and beige adipose tissue function. Chen, G. Xie, H. Adipose triglyceride lipase activity regulates cancer cell proliferation via AMP-kinase and mTOR signaling.

Acta Mol. Lipids , Rohm, M. An AMP-activated protein kinase-stabilizing peptide ameliorates adipose tissue wasting in cancer cachexia in mice. Lord, C. Mammalian alpha beta hydrolase domain ABHD proteins: lipid metabolizing enzymes at the interface of cell signaling and energy metabolism.

Acta , — Boeszoermenyi, A.

Editor's evaluation The mitochondria number tat Replenishing muscle glycogen in iWAT and BAT of AKO Essential oils for hormone balance cat to that of floxed mice, whereas that in eWAT did not differ in these metabolusm genotypes Supplementary Figure 1Msuggesting that AMPK deletion likely inhibited mitochondrial biogenesis in iWAT and BAT, both of which contribute to adaptive thermogenesis during prolonged cold exposure. It contains a catalytic dyad consisting of S47 and D Journal of Applied Physiology. P -values were computed by the log-rank test. This supernatant was filter sterilized.
Introduction see apoptosis signaling Ammplified. The bacterial cultures Amplified fat metabolism immediately supplied 1. Thus, it is Amplified fat metabolism that Sports nutrition for power and agility sn 2,3-DAG from TG can be reacylated to TG Amplified fat metabolism jetabolism transacylase reaction. cAMP then binds to and alters the function of effector proteins like the transcription factor CRP Stapleton et al. Chen HC Enhancing energy and glucose metabolism by disrupting triglyceride synthesis: lessons from mice lacking DGAT1. Cao JCheng LShi Y Catalytic properties of MGAT3, a putative triacylgycerol synthase.
Lipolysis: cellular mechanisms for lipid mobilization from fat stores | Nature Metabolism Grall, A. Lysosomal acid lipase hydrolyzes retinyl Amplified fat metabolism and affects retinoid turnover. AMPK: an Amllified drug target for diabetes and the Amplified fat metabolism syndrome. Myristic metsbolism Myristic acid C is a saturated fatty acid that is found in plant and animal fats. Combinatorial regulation of neuroblastoma tumor progression by N-Myc and hypoxia inducible factor HIF-1alpha. However, in our study, the mRNA levels of thermogenesis-related genes were not up-regulated by A in the BAT Supplementary Figure 6A. f Tumor weights at week 5 post-implantation.
I. Introduction Fta live-cell high-throughput screening assay for identification of Core strength and muscular endurance acid uptake inhibitors. Obesity and its attendant disorders, faat Amplified fat metabolism Ampified 2 diabetes, have reached epidemic proportions globally. Metabolim is important Amplfiied gut health and protein synthesis. In addition, the expression levels of lipogenesis-related Replenishing muscle glycogen, such as glucose kinase Gckmalic enzyme Me1fatty acid synthase Fasn and acetyl-CoA carboxylase 1 Acc1and fibrosis-related genes including smooth muscle actin alpha 2 Acta2, encoding α smooth muscle actincollagen type I alpha 1 chain Col1a1connective tissue growth factor Ctgfmatrix metallopeptidase 2 Mmp2 and tissue inhibitor of metalloproteinases 1 Timp1 were up-regulated in AKO mice especially fed with a HFD Figures 5G,H. The direct metabolic substrates of AMPK are related to almost all branches of cellular metabolism Hoffman et al. Weiss, W.

Amplified fat metabolism -

Brinkley, and the Carl H. Westcott Distinguished Chair in Medical Research; and Dr. Kevin Williams, Assistant Professor Internal Medicine in the Division of Hypothalamic Research.

The research was supported by the NIH, the Cancer Prevention and Research Institute of Texas CPRIT ; a Lilly Innovation Fellowship Award; a China Scholarship Council Scholarship, and the American Heart Association. About UT Southwestern Medical Center UT Southwestern, one of the premier academic medical centers in the nation, integrates pioneering biomedical research with exceptional clinical care and education.

The faculty of almost 2, is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide medical care in about 80 specialties to more than , hospitalized patients and oversee approximately 2.

Media Contact: Deborah Wormser Email. To automatically receive news releases from UT Southwestern via email, subscribe at www. Two hard-to-match transplant patients miles apart are starting on a new path to healthy lives.

UT Southwestern Medical Center is collaborating with Pfizer Inc. Automated external defibrillators AEDs are underutilized during cardiac arrest episodes despite laws in some states requiring their availability in high-risk areas such as athletic facilities, researchers at UT Southwestern Medical Center found.

The devices can save lives by shocking the heart back into normal rhythm. Site Navigation UT Southwestern Home Research Clinical Trials Core Facilities CTSA Program Postdoctoral Scholars Programs Research Support Research by Department Research Labs Scientific Discoveries Technology Development.

Centers and Institutes Advanced Imaging Research Center AIRC Cecil H. and Ida Green Comprehensive Center for Molecular, Computational, and Systems Biology Cecil H.

and Ida Green Center for Reproductive Biology Sciences Center for Alzheimer's and Neurodegenerative Diseases Center for the Genetics of Host Defense Center for Human Nutrition Center for Patient-Centered Outcomes Research Center for Translational Medicine Center for Translational Neurodegeneration Research Charles and Jane Pak Center for Mineral Metabolism and Clinical Research.

Children's Medical Center Research Institute at UT Southwestern Eugene McDermott Center for Human Growth and Development Hamon Center for Regenerative Science and Medicine Hamon Center for Therapeutic Oncology Research Harold C.

Brain Institute Texas Institute for Brain Injury and Repair Touchstone Center for Diabetes Research. Programs Disease-Oriented Clinical Scholars DOCS Endowed Scholars Program Kidney Cancer Program Medical Scientist Training Program MSTP Medical Student Research Programs Physician Scientist Training Program Science Teacher Access to Resources at Southwestern STARS Summer Undergraduate Research Fellowship Program SURF.

Apply to Medical School. Apply to Graduate School. Apply to School of Health Professions. Apply to School of Public Health. Visiting Medical Students. Education Programs Degree Programs Non-degree Programs Residency Programs Fellowship Programs. Request an Appointment Refer a Patient Find a Clinical Location MyChart.

October February Biochemical and Biophysical Research Communications. Bibcode : NatCo.. Journal of Biology. Cell Metabolism. ISSN Biochemical Society Transactions.

May Cre neurons suppresses insulin release in vivo". The Journal of Clinical Investigation. Regulatory, Integrative and Comparative Physiology.

March Molecular Cell. The EMBO Journal. Developmental Cell. Current Rheumatology Reports. Annual Review of Cancer Biology. Intracellular signaling peptides and proteins. see MAP kinase pathway. Transducin Gustducin Guanylate cyclase cGMP 3',5'-cyclic-GMP phosphodiesterase Protein kinase G.

G beta-gamma complex G β GNB1 GNB2 GNB3 GNB4 GNB5 G γ GNGT1 GNGT2 GNG2 GNG3 GNG4 GNG5 GNG7 GNG8 GNG10 GNG11 GNG12 GNG13 BSCL2. G protein-coupled receptor kinase AMP-activated protein kinase. ARFs Rabs Ras HRAS KRAS NRAS Rhos Arfs Ran Rhebs Raps RGKs. Cyclin-dependent kinase inhibitor protein Cyclin-dependent kinase Cyclin.

Phosphoinositide phospholipase C Phospholipase C. Casein kinase 1 2 eIF-2 kinase EIF2AK3 Glycogen synthase kinase GSK1 GSK2 GSK-3 GSK3A GSK3B IκB kinase CHUK IKK2 IKBKG Interleukin-1 receptor associated kinase IRAK1 IRAK2 IRAK3 IRAK4 Lim kinase LIMK1 LIMK2 pactivated kinases PAK1 PAK2 PAK3 PAK4 Rho-associated protein kinase ROCK1 ROCK2 Ribosomal s6 kinase RPS6KA1.

ZAP70 Focal adhesion protein-tyrosine kinase PTK2 PTK2B BTK. Dual-specificity kinase DYRK1A DYRK1B DYRK2 DYRK3 DYRK4. Arginine kinase McsB. Protein phosphatase 2. protein tyrosine phosphatase : Receptor-like protein tyrosine phosphatase Sh2 domain-containing protein tyrosine phosphatase.

Dual-specificity phosphatase. see apoptosis signaling pathway. see GTP-binding protein regulators. Activating transcription factor 6 Signal transducing adaptor protein I-kappa B protein Mucin-4 Olfactory marker protein Phosphatidylethanolamine binding protein EDARADD PRKCSH.

see also deficiencies of intracellular signaling peptides and proteins. LATS1 LATS2 MAST1 MAST2 STK38 STK38L CIT ROCK1 SGK SGK2 SGK3 Protein kinase B AKT1 AKT2 AKT3 Ataxia telangiectasia mutated mTOR EIF-2 kinases PKR HRI EIF2AK3 EIF2AK4 Wee1 WEE1.

PDK1 PDK2 PDK3 PDK4. AMP-activated protein kinase α PRKAA1 PRKAA2 β PRKAB1 PRKAB2 γ PRKAG1 PRKAG2 PRKAG3. BCKDK BCKDHA BCKDHB. IDH2 IDH3A IDH3B IDH3G.

Aurora kinase Aurora A kinase Aurora B kinase Aurora C kinase. FASTK STK CHUK IKK2 TBK1 IKBKE IKBKG IKBKAP. Protein kinase A PRKACG PRKACB PRKACA PRKY. Protein kinase G PRKG1. Protein kinase C Protein kinase Cζ PKC alpha PRKCB1 PRKCD PRKCE PRKCH PRKCG PRKCI PRKCQ Protein kinase N1 PKN2 PKN3.

Rhodopsin kinase. Beta adrenergic receptor kinase Beta adrenergic receptor kinase GRK4 GRK5 GRK6. BRSK2 CAMK1 CAMK1A CAMK1B CAMK1D CAMK1G CAMK2 CAMK2A CAMK2B CAMK2D CAMK2G CAMK4 MLCK CASK CHEK1 CHEK2 DAPK1 DAPK2 DAPK3 STK11 MAPKAPK2 MAPKAPK3 MAPKAPK5 MARK1 MARK2 MARK3 MARK4 MELK MKNK1 MKNK2 NUAK1 NUAK2 OBSCN PASK PHKG1 PHKG2 PIM1 PIM2 PKD1 PRKD2 PRKD3 PSKH1 SNF1LK2 KIAA STK40 SNF1LK SNRK SPEG TSSK2 Kalirin TRIB1 TRIB2 TRIB3 TRIO Titin DCLK1.

MYLK MYLK2 MYLK3 MYLK4. PHKA1 PHKA2 PHKB PHKG1 PHKG2. EEF2K STK PLK1 PLK2 PLK3 PLK4. CDK1 CDK2 CDKL2 CDK3 CDK4 CDK5 CDKL5 CDK6 CDK7 CDK8 CDK9 CDK10 CDK12 CDC2L5 PCTK1 PCTK2 PCTK3 PFTK1 CDC2L1.

RPS6KA5 RPS6KA4 P70S6 kinase PS6 Kinase 1 RPS6KB2 RPS6KA2 RPS6KA3 RPS6KA1 RPS6KC1. Extracellular signal-regulated MAPK1 MAPK3 MAPK4 MAPK6 MAPK7 MAPK12 MAPK15 C-Jun N-terminal MAPK8 MAPK9 MAPK10 P38 mitogen-activated protein MAPK11 MAPK13 MAPK MAP kinase kinase kinases MAP3K1 MAP3K2 MAP3K3 MAP3K4 MAP3K5 MAP3K6 MAP3K7 MAP3K8 RAFs ARAF BRAF KSR1 KSR2 MLKs MAP3K12 MAP3K13 MAP3K9 MAP3K10 MAP3K11 MAP3K7 ZAK CDC7 MAP3K TPK1 TTK GSK Bone morphogenetic protein receptors BMPR1 BMPR1A BMPR1B BMPR2 ACVR1 ACVR1B ACVR1C ACVR2A ACVR2B ACVRL1 Anti-Müllerian hormone receptor.

Dual-specificity kinases EC 2. MAP2K1 MAP2K2 MAP2K3 MAP2K4 MAP2K5 MAP2K6 MAP2K7. Active site Binding site Catalytic triad Oxyanion hole Enzyme promiscuity Diffusion-limited enzyme Cofactor Enzyme catalysis.

Allosteric regulation Cooperativity Enzyme inhibitor Enzyme activator. EC number Enzyme superfamily Enzyme family List of enzymes. Enzyme kinetics Eadie—Hofstee diagram Hanes—Woolf plot Lineweaver—Burk plot Michaelis—Menten kinetics. EC1 Oxidoreductases list EC2 Transferases list EC3 Hydrolases list EC4 Lyases list EC5 Isomerases list EC6 Ligases list EC7 Translocases list.

Portal : Biology. Categories : Protein kinases EC 2. Hidden categories: Articles with short description Short description matches Wikidata All articles with failed verification Articles with failed verification from September Wikipedia articles with style issues from January All articles with style issues.

Toggle limited content width. The expression of thermogenesis-related genes, such as Ucp1, Cidea, Cox8b, Cox7a1 , and Ppargc1a , was significantly increased by A treatment in a concentration-dependent manner Figure 9A. Importantly, similar to the in vivo results, the expression of thermogenic protein UCP1 and of PGC-1α was also remarkably increased in differentiated iWAT-SVF cells, which was concomitant with the activation of AMPK signaling by A treatment Figures 9B,C.

In addition, the basal and uncoupled O 2 consumption rates OCRs were both up-regulated by A treatment in differentiated iWAT-SVF cells Figure 9D. To confirm whether the effect of A on promoting thermogenesis was dependent on AMPK activation, primary SVF cells isolated from iWAT of the floxed mice were induced to differentiate into beige adipocytes, followed by adding Cre lentivirus at day 6 of differentiation to knockdown AMPKα expression, and were then treated with A at indicated concentration on day 8.

As a result, the expression of genes encoding AMPKα1 and AMPKα2 Prkaa1 and Prkaa2 , respectively and total AMPKα in iWAT-SVF cells were significantly reduced and the activation of AMPK signaling by A was successfully blocked after Cre lentivirus infection Figures 9E—G.

Importantly, A treatment up-regulated the expression of thermogenesis marker Ucp1 and Ppargc1a and their encoding products, which were remarkably blunted after AMPKα knockdown Figures 9E—G , suggesting that AMPK activation was required for Afacilitated thermogenesis.

These data indicate that A promotes thermogenesis in vitro via AMPK signaling pathway, which may contribute to Ainduced browning in iWAT in vivo. Figure 9. A facilitated thermogenesis in differentiated iWAT-SVF cells through AMPK signaling pathway.

A—D iWAT-SVF cells were induced to differentiation toward brown-like adipocytes and were treated with the indicated compounds on day 7. A Relative mRNA levels of thermogenic genes in differentiated iWAT-SVF cells treated with the indicated compounds for 6 h were analyzed by quantitative RT-PCR.

B Western blot analysis of UCP1, PGC-1α, p-AMPKα T , AMPKα, p-ACC S79 , and ACC expression levels in differentiated iWAT-SVF cells treated with indicated compounds on day 7 for 24 h. C Relative protein expression levels of UCP1 and PGC-1α and the relative phosphorylation levels of AMPKα and ACC were determined by densitometric quantification of the immunoblots shown in B.

D Basal and uncoupled oxygen consumption rate OCR of differentiated iWAT-SVF cells treated with NE 10 μM for 3 h or with DMSO or A at different concentrations on day 7 for 12 h. Cells were infected with NC and Cre lentivirus on day 6 to knockdown AMPKα expression and were treated with the indicated compounds on day 8.

E Relative mRNA levels of the indicated genes in differentiated iWAT-SVF cells treated with the indicated compounds for 6 h on day 8 were analyzed by quantitative RT-PCR.

F Western blot analysis of UCP1, PGC-1α, p-AMPKα T , AMPKα, p-ACC S79 , and ACC expression levels in differentiated iWAT-SVF cells treated with indicated compounds on day 8 for 24 h. G Relative protein expression levels of UCP1 and PGC-1α and the relative phosphorylation levels of AMPKα and ACC were determined by densitometric quantification of the immunoblots shown in F.

One-way ANOVA. Since obesity has become a global health problem and efforts to reduce energy intake show either limited effectiveness or unacceptable side effects James et al. Approaches attempting to enhance the thermogenic activity of brown and beige fat may be beneficial for obesity therapy because the activity of brown and beige fat have been shown to be inversely correlated with BMI and fat mass and positively related to EE in humans Saito et al.

Reduced adipose tissue AMPK activity is generally observed in many obese and diabetic animal models as well as in obese humans with insulin resistance Yu et al.

The physiological relevance of lowered AMPK activity in adipose tissue to whole-body adiposity remains ambiguous. Here, we found that ablation of adipocyte AMPKα impaired adaptive thermogenesis and EE in response to cold exposure or β3-adrenergic stimulation, and predisposed HFD-fed mice to obesity, glucose intolerance and insulin resistance.

Importantly, the lack of adipocyte AMPKα selectively blunted cold-induced thermogenic protein expression in iWAT, resulting in reduced energy utilization and increased adipocyte size, indicating that adipocyte AMPKα is required for browning in iWAT. In contrast, Ainduced direct AMPK activation promoted browning in iWAT, which may contribute to the reduced adiposity and improved glucose and lipid metabolism observed in Atreated HFD-fed mice.

In summary, these findings indicate that reduced AMPK activity in adipose tissue might be an important pathogenic factor in obesity and the related metabolic syndrome, and the chronic activation of AMPK by A protects from HFD-induced adiposity and metabolic dysfunction.

Since both catalytic subunits AMPK α1 and α2 exist in adipose tissue and the deletion of a single subunit often leads to the up-regulated activity of the other subunit Bauwens et al.

The Adiponectin-driven Cre recombinase is supposed to be expressed at the beginning during the early stage of BAT development Cohen et al. When fed a chow diet, AKO mice started to gain more weight than age-matched floxed littermates at 29 weeks of age, however, when fed a HFD, this difference occurred, at 25 weeks of age, indicating that the loss of adipocyte AMPKα accelerates the development of obesity in response to a HFD.

The iβ1β2AKO mice had defects not only in the browning of WAT but also in BAT-mediated thermogenesis due to impaired mitochondrial integrity and function but not mitochondrial biogenesis. Intriguingly, we observed that there were not only defects in mitochondrial structure but also reduced total number of mitochondria in iWAT and BAT of AKO mice.

However, the thermogenic capacity of BAT from the AKO mice in our study was normal compared with that of the floxed mice, suggesting that AMPK might play different roles in BAT function at different ages and that the unaffected thermogenesis of BAT in AKO mice might be remedied through an unknown mechanism.

In addition, both hepatic steatosis and fibrosis were developed in HFD-fed AKO mice, which is more severe than that in iβ1β2AKO mice. This may be attributed to AMPK deletion at earlier stage and longer period of HFD challenge in AKO mice compared to that of iβ1β2AKO mice Mottillo et al.

It is well-established that adipose tissue-liver crosstalk plays a vital role in regulating systemic glucose and lipid metabolism Stern et al. Adiponectin ameliorates liver ectopic lipid accumulation by inhibiting hepatic lipogenesis and enhancing β-oxidation.

The plasma level of adiponectin was reduced in chow- and HFD-fed AKO mice, which may intensify the development of hepatic steatosis and even fibrosis. Accumulative evidence have shown that leptin plays a crucial role in the development of liver fibrosis Marra, The plasma level of leptin increased along with increased fat mass in AKO mice, which partially explains the aggravating progression of liver fibrosis in AKO mice.

Besides, another study reported by Sun-Joong et al. The Adiponectin-Cre model is known to have better efficiency and specificity for adipocytes than the aP2-Cre model Jeffery et al. However, despite unaltered body weight, defective adaptive thermogenesis and cold tolerance were observed in the AKO mice in our study at 8 weeks of age, which may lead to reduced EE and accumulated adiposity during aging and in response to the HFD challenge.

One possible reason for the differences in the phenotypes of their study and ours might be that the mice used in the experiments were at different ages. AMPK signaling is indirectly activated by catecholamine secretion in response to cold or β-adrenergic stimulation Gauthier et al.

We also showed that the sensitivity of AMPK activation and UCP1 expression in different fat depots were diverse, with iWAT being the most responsive to cold exposure, and eWAT and BAT being less responsive to the cold stimulation Figure 1A and Supplementary Figure 1A.

Ablation of adipocyte AMPKα specially impaired mitochondria quality control and biogenesis in iWAT and BAT but not in eWAT Figures 1J—M and Supplementary Figure 1M.

Accordingly, the adipocyte size in the iWAT and BAT of AKO mice was increased while that in eWAT were unchanged Figures 3A—D. Nonetheless, the expression of the thermogenesis-related protein was markedly reduced in iWAT but not in BAT of AKO mice.

In addition, chronic AMPK activation by A treatment selectively promoted browning in the iWAT of HFD-fed mice but not in eWAT or BAT Figures 8A—H and Supplementary Figures 6A,B.

Despite the unique structural and molecular characteristics of different fat depots that cause these depot-specific differences, our findings suggest that the high sensitivity of AMPK activation in iWAT may contribute to its plasticity in adaptation to various environmental and hormonal cues.

In this study, we also observed that A ameliorated glucose and lipid disorders in HFD-fed mice Figure 6 and Table 2. However, in our model, food consumption was unchanged during the 6 weeks of study, ruling out the disturbance of energy intake differences in the anti-obesity effect of A Besides liver, WAT is another target tissue of A, as evidenced by the tissue weight, morphology, related gene expression and UCP-1 protein level.

Moreover, there was a considerable amount of A distribution in the adipose tissue of HFD-fed mice Supplementary Figure 7 , suggesting a direct effect of A on adipose tissue. In this study, we observed that A treatment augmented whole-body EE and enhanced adaptive thermogenesis in HFD-fed mice, and these effects were not driven by increased locomotor activity Figures 7A,B and Supplementary Figure 5B.

BAT activity plays a primary role in cold-induced adaptive thermogenesis and EE in rodents Tam et al. However, in our study, the mRNA levels of thermogenesis-related genes were not up-regulated by A in the BAT Supplementary Figure 6A.

These results seem to rule out the possibility that BAT plays a predominant role in the Ainduced enhancement of EE and adaptive thermogenesis. Skeletal muscle, as a large organ and major site of facultative thermogenesis, also contributes to EE Zurlo et al. Meanwhile, the mRNA levels of Ppargc1a and other thermogenic genes, and the protein level of UCP1 were increased by A in iWAT; in contrast, the expression levels of cold-induced UCP1 and PGC-1α protein were remarkably reduced in iWAT with adipocyte AMPK deletion.

Therefore, it is possible that the Ainduced improvement of metabolic disorders in obese mice is at least partly due to increased EE through the browning of iWAT.

There are also studies suggesting that long-term AICAR treatment promotes energy dissipation in chow-fed rat models Gaidhu et al. AICAR treatment also had an inhibitory effect on food consumption in these chronic studies.

In our study with long-term A treatment, the chow-fed mice exhibited no effect on food intake, EE or fat metabolism, which may be due to variations in species, treatment duration, dosage and injection frequency in different animal models.

Although AICAR and A are both AMPK activators, they have different mechanisms. AICAR activates AMPK by being taken up into cells and converted into an AMP mimetic, ZMP Hardie, Similar to AMP, A allosterically activates AMPK and inhibits the dephosphorylation of AMPK Thr However, AMPK activation by A is exclusively dependent on the existence of glycogen binding domain within the β1 subunit, but not the γ subunit to which AMP binding Sanders et al.

The absence of effect on chow-fed mice is also consistent with the study conducted by Cool et al. In our study, the pro-browning effect of A only existed in the HFD-fed mice, and this may be owing to differences in metabolism and energy status of HFD-fed and chow-fed mice.

This result suggests that in mouse models, the anti-obesity effect of chronic AMPK activation is more sensitive and responsive in diet-induced obese mice.

Beige adipocytes have been reported to arise from two alternative processes: de novo differentiation from progenitor cells and trans-differentiation from white adipocytes Bartelt and Heeren, In general, the differentiation of brown adipocytes shares most of the transcriptional regulation pathways with beige adipocytes Inagaki et al.

Some studies have reported that AMPK plays a positive role in brown adipocyte differentiation and brown fat development in murine cell lines and animal models Vila-Bedmar et al. AMPK activation by AICAR treatment in human adipose-derived mesenchymal stem cells hADMSCs from pericardial adipose tissue is reported to induce a morphological change similar to beige adipocytes, but without an actual change of metabolic function Abdul-Rahman et al.

However, the indirect activation of AMPK by berberine has been demonstrated to promote WAT browning by enhancing thermogenesis in mature beige adipocytes via the AMPK-PGC-1α pathway Zhang et al.

Similarly, direct AMPK activation by A did not induce the differentiation of iWAT-SVF cells toward beige adipocytes in vitro data not shown and had no effect on the expression of the adipogenic marker aP2 in iWAT in vivo Figure 8F. Alternatively, A enhanced thermogenesis in differentiated iWAT-SVF cells, as detected by the increased expression of thermogenic genes and proteins and the up-regulated O 2 consumption, which is directly reliant on the activation of AMPK signaling Figures 9A—G.

Moreover, we further determined the plasma level of irisin, which has been reported to stimulate the browning of WAT through specific actions on the beige preadipocyte population Tseng et al. These results indicate that the browning of WAT in HFD-fed mice by A is not due to the direct modulation of SVF cell differentiation, but may derive from direct trans-differentiation of mature adipocytes in iWAT.

In summary, our results demonstrated that the ablation of adipocyte AMPKα impairs adaptive thermogenesis and energy expenditure in response to cold and β-adrenergic stimulation, leading to a predisposition for HFD-induced obesity and metabolic dysfunction.

Moreover, pharmacological chronic AMPK activation by A alleviated diet-induced obesity via promoting browning in inguinal WAT. Overall, our findings indicate that AMPK plays a vital role in modulating WAT browning in response to thermal, nutritional and pharmacological cues, supporting chronic AMPK activation as a potentially effective approach for the treatment of obesity and related metabolic diseases through increasing thermogenesis.

LW, LZ, and BL contributed to study design, data analyzing, discussion and preparation of the manuscript. LW, LZ, BL, HJ, YD, ZX, and LS contributed to conducting the experiments. JYL and JL contributed to study design, discussion, reviewing and editing the manuscript. This work was supported by a grant from National Key Research and Development YFC , the National Natural Science Foundation of China , , the Shanghai Commission of Science and Technology ,16JC, The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

The handling Editor declared a shared affiliation, though no other collaboration, with the authors HJ and YD. We thank Dr. Jiqiu Wang from Ruijin Hospital, Shanghai Jiaotong University School of Medicine, for discussion and the guidance for SVFs isolation and differentiation.

Abdul-Rahman, O. AMP-activated kinase AMPK activation by AICAR in human white adipocytes derived from pericardial white adipose tissue stem cells induces a partial beige-like phenotype.

PLoS ONE e doi: PubMed Abstract CrossRef Full Text Google Scholar. Arany, Z. PGC-1 coactivators and skeletal muscle adaptations in health and disease. Bagchi, M. Vascular endothelial growth factor is important for brown adipose tissue development and maintenance. FASEB J. Bal, N.

Mild cold induced thermogenesis: are BAT and skeletal muscle synergistic partners? Sarcolipin is a newly identified regulator of muscle-based thermogenesis in mammals. Both brown adipose tissue and skeletal muscle thermogenesis processes are activated during mild to severe cold adaptation in mice.

Bartelt, A. Adipose tissue browning and metabolic health. Bauwens, J. CrossRef Full Text Google Scholar. Berry, D. The developmental origins of adipose tissue. Development , — Bordicchia, M. Cardiac natriuretic peptides act via p38 MAPK to induce the brown fat thermogenic program in mouse and human adipocytes.

Boström, P. A PGC1-alpha-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature , — Buhl, E. Long-term AICAR administration reduces metabolic disturbances and lowers blood pressure in rats displaying features of the insulin resistance syndrome. Diabetes 51, — Carling, D.

The AMP-activated protein kinase cascade—a unifying system for energy control. Trends Biochem. AMP-activated protein kinase: new regulation, new roles?

A common bicyclic protein kinase cascade inactivates the regulatory enzymes of fatty acid and cholesterol biosynthesis. FEBS Lett. Carlson, C. Regulation of hepatic acetyl coenzyme A carboxylase by phosphorylation and dephosphorylation.

PubMed Abstract Google Scholar. Cohen, P. Ablation of PRDM16 and beige adipose causes metabolic dysfunction and a subcutaneous to visceral fat switch. Cell , — Collins, S. beta-Adrenoceptor Signaling Networks in Adipocytes for Recruiting Stored Fat and Energy Expenditure. Considine, R. Serum immunoreactive-leptin concentrations in normal-weight and obese humans.

Cool, B. Identification and characterization of a small molecule AMPK activator that treats key components of type 2 diabetes and the metabolic syndrome. Cell Metab. Corton, J. A specific method for activating AMP-activated protein kinase in intact cells? Dagon, Y. AMPK activation regulates apoptosis, adipogenesis, and lipolysis by eIF2alpha in adipocytes.

Day, E. AMPK as a therapeutic target for treating metabolic diseases. Trends Endocrinol. Fentz, J. AMPKalpha is critical for enhancing skeletal muscle fatty acid utilization during in vivo exercise in mice. Finucane, M. National, regional, and global trends in body-mass index since systematic analysis of health examination surveys and epidemiological studies with country-years and 9.

Lancet , — Folch, J. A simple method for the isolation and purification of total lipides from animal tissues. Fullerton, M. Single phosphorylation sites in Acc1 and Acc2 regulate lipid homeostasis and the insulin-sensitizing effects of metformin. Gaidhu, M. Prolonged AICAR-induced AMP-kinase activation promotes energy dissipation in white adipocytes: novel mechanisms integrating HSL and ATGL.

Lipid Res. Chronic AMP-kinase activation with AICAR reduces adiposity by remodeling adipocyte metabolism and increasing leptin sensitivity. Gauthier, M. AMP-activated protein kinase is activated as a consequence of lipolysis in the adipocyte - Potential mechanism and physiological relevance.

Decreased AMP-activated protein kinase activity is associated with increased inflammation in visceral adipose tissue and with whole-body insulin resistance in morbidly obese humans. Habinowski, S. The effects of AICAR on adipocyte differentiation of 3T3-L1 cells.

Hardie, D. Sensing of energy and nutrients by AMP-activated protein kinase. Harms, M. Brown and beige fat: development, function and therapeutic potential.

Hoffman, N. Global phosphoproteomic analysis of human skeletal muscle reveals a network of exercise-regulated kinases and AMPK substrates. Inagaki, T. Transcriptional and epigenetic control of brown and beige adipose cell fate and function. Cell Biol. Jager, S.

AMP-activated protein kinase AMPK action in skeletal muscle via direct phosphorylation of PGC-1 alpha. James, W. Effect of sibutramine on cardiovascular outcomes in overweight and obese subjects. Jeffery, E. Characterization of Cre recombinase models for the study of adipose tissue.

Adipocyte 3, — Jia, R. Characterization of cold-induced remodelling reveals depot-specific differences across and within brown and white adipose tissues in mice.

Acta Physiol. Jiang, G. Glucagon and regulation of glucose metabolism. Kajimura, S.

Request an Appointment. Refer a Amplkfied. Find a Clinical Location. Donate Now. Our specialty is the health of Texas. September 30, DALLAS — Sept. Amplify High Fat Supplement Metaboljsm Mills - Nutrition Composition Data. The FeedBank Amplifiex a reference database providing nutrition information on components in the equine diet. Listing in the FeedBank does not imply endorsement by Mad Barn. Compare Feed. Amplify High Fat Supplement Purina Mills Coat Quality.

Video

How To Increase CO2 For Metabolism

Author: Yojind

5 thoughts on “Amplified fat metabolism

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com