Category: Moms

Increased fat metabolism

Increased fat metabolism

This is called gat thermic effect of food Warrior diet exercise duration. Concluding remarks IMCL and Eco-friendly home improvements content are increased, and fat oxidative capacity decreased in metabolically compromised individuals, such as obese individuals and those with type 2 diabetes. Millard-Stafford Similar articles in Google Scholar.

Increased fat metabolism -

Exercise and Sport Sciences Reviews, 32 4 , — LeBlanc , J. Enhanced metabolic response to caffeine in exercise-trained human subjects. Journal of Applied Physiology, 59 3 , — Lee , C. Effect of creatine plus caffeine supplements on time to exhaustion during an incremental maximum exercise. European Journal of Sport Science, 12 4 , — Moher , D.

Preferred reporting items for systematic review and meta-analysis protocols PRISMA-P statement. Systematic Reviews, 4, Article 1. Pickering , C. Are low doses of caffeine as ergogenic as higher doses?

A critical review highlighting the need for comparison with current best practice in caffeine research. Nutrition, 67—68, Article Romijn , J. Regulation of endogenous fat and carbohydrate metabolism in relation to exercise intensity and duration. American Journal of Physiology, 3 , E — E Schweiger , M.

Measurement of lipolysis. Methods in Enzymology, , — Spriet , L. Exercise and sport performance with low doses of caffeine.

Sports Medicine, 44 Suppl. Caffeine ingestion and muscle metabolism during prolonged exercise in humans. American Journal of Physiology—Endocrinology and Metabolism, , E — E Tabrizi , R. The effects of caffeine intake on weight loss: A systematic review and dos-response meta-analysis of randomized controlled trials.

Critical Reviews in Food Science and Nutrition, 59 16 , — Warren , G. Effect of caffeine ingestion on muscular strength and endurance: A meta-analysis.

Wells , C. Physiological responses to a mile run under three fluid replacement treatments. Wiles , J. Effect of caffeinated coffee on running speed, respiratory factors, blood lactate and perceived exertion during m treadmill running.

British Journal of Sports Medicine, 26 2 , — Yeo , S. Caffeine increases exogenous carbohydrate oxidation during exercise. Journal of Applied Physiology, 99, — Zuntz , N. Ueber die Bedeutung der verschiedenen Nâhrstoffe als Erzeuger der Muskelkraft.

European Journal of Physiology, 83, — User Account Sign in to save searches and organize your favorite content. Not registered? Sign up My Content 0 Recently viewed 0 Save Entry.

Recently viewed 0 Save Search. Human Kinetics. Previous Article Next Article. Does Caffeine Increase Fat Metabolism? A Systematic Review and Meta-Analysis. in International Journal of Sport Nutrition and Exercise Metabolism.

Scott A. Conger Scott A. Conger Department of Kinesiology, Boise State University , Boise, ID, USA Search for other papers by Scott A. Conger in Current site Google Scholar PubMed Close. Lara M. Tuthill Lara M. Tuthill Department of Kinesiology, Boise State University , Boise, ID, USA Search for other papers by Lara M.

Tuthill in Current site Google Scholar PubMed Close. Mindy L. Millard-Stafford Mindy L. Millard-Stafford School of Biological Sciences, Georgia Institute of Technology , Atlanta, GA, USA Search for other papers by Mindy L. Millard-Stafford in Current site Google Scholar PubMed Close.

In Print: Volume Issue 2. Page Range: — Free access. Get Citation Alerts. Download PDF. Abstract Full Text PDF Author Notes Supplementary Materials. Methods Systematic Literature Review For this study, the preferred reporting items for systematic reviews and meta-analyses guidelines were followed Moher et al.

Figure 1 —Selection of articles for meta-analysis of fat metabolism when consuming CAF. Statistical Analysis Study data were extracted from text or relevant tables. If data were not reported elsewhere, figures were used for data extraction. Figures were enlarged, and the mean and variance data presented were measured to the nearest millimeters using the appropriate scale of the figure.

Data from each study were converted into the same format by calculating the effect size ES as the standardized difference in means. where M is the mean, and SD Pooled is the pooled SD Borenstein et al.

Results Study Characteristics Ninety-four studies published between and were included. Figure 2 —Summary ES of the subgroup meta-analyses examining the effects of CAF on fat metabolism during rest and exercise.

Subgroup Meta-Analyses Subgroup meta-analyses were used to assess effects of moderator variables as potential underlying explanation for the heterogeneity. Resting Versus Exercise Conditions Of the independent study populations, data during resting conditions were reported in 13 studies, data during exercise conditions in 34 studies, and during both resting and exercise conditions in 58 studies.

Fat Metabolism Assessment Method Five different fat metabolism biomarkers were reported across studies. Figure 3 —Summary ES of the subgroup meta-analyses examining the effects of CAF on fat metabolism by A fat assessment method and B blood lipolysis metrics versus gas analysis metrics.

Other Potential Modifier Variables Figure 4 summarizes the impact of other factors on CAF ES on fat metabolism. Figure 4 —Summary ES of the subgroup meta-analyses examining the effects of CAF on fat metabolism by A sex, B habitual CAF use, C CAF dose, and D fasting state.

Publication Bias Publication bias was assessed by examining a funnel plot of SE versus ES. Figure 5 —Funnel plot of the ES of CAF on fat metabolism versus the SE. Discussion Our aim was to determine whether CAF increases fat oxidation. Strengths and Limitations With different participant populations and nearly 1, participants included in the analysis, this large sample size allowed for a number of important subgroup analyses to address several key moderator variables considered important for assessing the impact of CAF.

PubMed Costill , D. x PubMed Doherty , M. x false. PubMed Ivy , J. x PubMed Jeukendrup , A. E PubMed Romijn , J. E false. E Spriet , L. Save Cite Email this content Share Link Copy this link, or click below to email it to a friend.

xml The link was not copied. Your current browser may not support copying via this button. International Journal of Sport Nutrition and Exercise Metabolism. Related Articles. Export Figures.

Close View raw image Figure 1 —Selection of articles for meta-analysis of fat metabolism when consuming CAF. View raw image Figure 2 —Summary ES of the subgroup meta-analyses examining the effects of CAF on fat metabolism during rest and exercise.

View raw image Figure 3 —Summary ES of the subgroup meta-analyses examining the effects of CAF on fat metabolism by A fat assessment method and B blood lipolysis metrics versus gas analysis metrics. View raw image Figure 4 —Summary ES of the subgroup meta-analyses examining the effects of CAF on fat metabolism by A sex, B habitual CAF use, C CAF dose, and D fasting state.

View raw image Figure 5 —Funnel plot of the ES of CAF on fat metabolism versus the SE. Export References. ris ProCite. FAs and cytokines also modulate their negative effects through PKC-θ 14 , c-Jun NH 2 terminal kinase JNK -1 15 , and the inhibitor of nerve factor κB IκB Each of these cause serine phosphorylation of insulin receptor substrate IRS -1, which disrupts insulin signaling, while TZD improvement of FA-mediated insulin resistance in muscle may involve reversal of the serine phosphorylation.

FA release from adipocytes via hormone-sensitive lipase is attenuated by reesterification, which involves FA-CoA synthesis with subsequent reesterification by glycerolphophate G3P acyltransferase, utilizing G3P formed from glycerol via glycerol kinase or from pyruvate via phosphoenol pyruvate PEP carboxykinase, all three enzymatic steps activated by TZDs.

An important question is whether it would be possible to increase FA oxidation rather than use TZD-promoted reesterification to decrease FA release without increasing adipocyte triglyceride stores.

Meridith A. Hawkins Bronx, NY discussed the control of hepatic glucose metabolism. Hepatic glucose production HGP is suppressed directly by glucose, insulin, and adiponectin and indirectly via FA suppression by insulin.

HGP is increased by cortisol, glucagon, growth hormone, catecholamines, FAs, and resistin. The higher portal glucose levels following meals may particularly be effective in suppressing HGP. Using glucose and insulin clamps, individuals with type 2 diabetes show no suppression of glucose production by hyperglycemia.

Glucokinase activity is decreased and glucose-6 phosphatase G6Pase is increased in diabetes. The effects, perhaps, could be in response to chronic hyperglycemia, which would suggest that intracellular G6P levels do not increase, but mediate the lack of response to hyperglycemia, with suppression of HGP by hyperglycemia being seen in well-controlled individuals with type 2 diabetes.

Hawkins noted that activation of hepatic glucokinase with small catalytic amounts of fructose partially restores the regulation of HGP by hyperglycemia in persons with type 2 diabetes, although this effect is not enhanced in normal persons whose glucokinase activity is presumably adequate.

FAs increase G6Pase and inhibit formation of G6P from PEP. The increase in FFA levels in diabetes may be another mechanism of the adverse effect of hyperglycemia. Indeed, elevating FFAs with lipid infusion in nondiabetic individuals adversely affects the response to hyperglycemia, while lowering FFA levels with nicotinic acid in persons with type 2 diabetes improves the response.

Insulin sensitivity increases over 3 weeks in individuals with type 2 diabetes treated with pioglitazone, in conjunction with doubling of adiponectin levels. However, only modest changes in FFA levels are observed during this timeframe, which suggests that there may be another mechanism of TZD action.

Marc C. Reitman Rahway, NJ discussed the lipodystrophies, reviewing clinical studies and analysis of mouse models. White adipose tissue WAT plays a role in energy storage and produces FFAs as well as messenger molecules, including leptin, adiponectin, TNF-α, IL-6, resistin, angiotensinogen, and PAI Obesity and overweight are common and are associated with development of insulin resistance, dyslipidemia, and hypertension, but, importantly, deficiency of adipose tissue is often associated with many of these same complications.

Human lipodystrophies are a heterogeneous group of diseases; they may be generalized, partial, or localized with redistribution of fat; and they may have genetic, autoimmune, or drug-related causes, as seen with HIV medications. In general, the metabolic severity of the lipodystrophies correlates with the degree of fat loss.

Severe lipodystrophy, the near-complete lack of adipose tissue, is associated with severe insulin resistance, diabetes, acanthosis, features of polycystic ovary syndrome, hirsutism, hypertriglyceridemia, hepatomegaly, and hepatic steatosis, often progressing to cirrhosis. Leptin levels are increased, hyperphagia is common, and there is often muscle hypertrophy.

The insulin resistance may correlate with increased muscle and liver fat. Acquired generalized or partial lipodystrophy was first described by the British endocrinologist Robert D.

Lawrence It is inflammatory and autoimmune in origin, occurs more commonly in females, has onset in childhood or adulthood, and is associated with paniculitis, with fat loss beginning in the legs and progressing to the upper body. Congenital generalized lipodystrophy CGL is associated with either one of two mutations: a recessive mutation in the gene encoding 1-acylglycerolphosphate- O -acyltransferase-2 on chromosome 9q34, which has at least eight described variants and is expressed primarily in WAT, though also in liver and, to a lesser extent, in muscle 18 ; or a mutation in BSCL2 at chromosome 11q13 in seipin, a —amino acid protein of unknown function, with mRNA widely expressed for this, particularly in brain and testes, but also in WAT Peroxisome proliferator-activated receptor PPAR -γ mutations may also be associated with partial lipodystrophy.

It is associated with visceral and cervicodorsal obesity but also with facial peripheral lipoatrophy. Hypertriglyceridemia and insulin resistance are typical. The primary deficit in lipodystrophy is the lack of adipose tissue.

Consequent loss of adipocyte-derived hormones and increased circulating FFAs and triglycerides lead to increased hepatic and muscle deposition, with consequent insulin resistance at these sites. A mouse model of virtually complete absence of adipose tissue 22 is associated with increased triglycerides and FFAs, fatty liver, hyperglycemia and hyperinsulinemia without ketoacidosis, and visceral organomegaly.

Transplantation of adipose tissue grafts reverses the hyperinsulinemia, diabetes, and muscle and liver insulin resistance in a dose-dependent fashion Thus, lack of fat causes the metabolic symptoms of lipodystrophy.

Leptin administration can partially reverse the phenotype, perhaps because of decreased food intake, CNS effects on energy expenditure and insulin sensitivity, or peripheral effects on increasing energy expenditure. The effect of fat transplantation is not due to adiponectin, which does not increase to control levels.

Divergent hepatic insulin effects are seen in animal models of lipoatrophy. Insulin-induced sterol regulatory element-binding protein-1c levels are increased, which increases lipogenesis, while IRS-2—mediated effects on glucose metabolism are decreased.

Liver PPAR-γ mRNA is increased in mouse models of hepatic steatosis, and mice lacking PPAR-γ with lipodystrophy have decreased liver triglyceride levels and greater hepatic insulin sensitivity, although with lower muscle insulin sensitivity and higher serum triglyceride levels.

Hepatic triglyceride clearance presumably mediates this paradoxical PPAR-γ effect, and in this model, TZDs increase hepatic steatosis. The role of hepatic PPAR-γ in humans appears to be different, with marked improvement in 19 insulin-resistant persons with lipodystrophy treated for 6 months with troglitazone Although TZDs may be effective, one must be sure that liver function does not worsen, and metformin, acarbose, and triglyceride-lowering treatment also have potential; insulin and insulin secretagogues are less desirable.

Treatment with leptin also appears to be effective, with a recent study of methionyl leptin in nine leptin-deficient patients with lipodystrophy markedly improving triglyceride and glucose levels. Philipp E.

Scherer Bronx, NY extended the previous discussions of the adipocyte as both a target and a source of cytokines, acute-phase reactants, and hormones, in discussions at both the ADA Postgraduate Course and the Niagara Falls conference In the course of differentiation of adipocytes, a number of mediators are produced, including the proinflammatory mitogen-activated protein MAP kinase P Diabetes leads to a low-level inflammatory adipocyte response with induction of serum amyloid A.

Pituitary homeobox 3, a homologue of CRP produced by adipocytes, is another inflammatory mediator produced in diabetes. The adipocyte-specific secretory protein adiponectin is deficient in states of insulin resistance in humans 28 and in animal models The 3q27 locus implicated in syndrome X may play a role in adiponectin production Adiponectin resembles complement factor C1q and TNF-α.

It circulates in serum as hexameric and higher-order complexes, with high levels in female models because of high-molecular weight complexes, which appear to be a circulating precursor pool. Prolactin and estrogen decrease adiponectin, despite the higher levels in females.

Insulin administration activates adiponectin by returning it to the hexameric form, with a serum reductase producing the trimer, which then dissociates into the active moiety.

Transgenic mice overexpressing adiponectin show increased hepatic insulin sensitivity. Thus, it appears that a potential mechanism of the interrelationship between inflammation and insulin resistance is in the suppression of adiponectin secretion by adipocytes.

Although produced in adipocytes, levels are greater with lower adipose tissue mass. Adiponectin shows an inverse relationship with fasting insulin and a positive correlation with insulin sensitivity In a rhesus monkey model of progression to diabetes, adiponectin levels decrease as insulin resistance occurs Euglycemic clamp studies show increased peripheral glucose uptake and, particularly, decreased HGP with adiponectin administration.

In hepatocyte culture, adiponectin greatly increases insulin sensitivity. Adiponectin acts on AMPK to increase glucose utilization and FA oxidation, and AICAR mimics the effect of adiponectin. Most adiponectin is derived from visceral fat, and TZD not metformin treatment increases visceral fat adiponectin Ongoing studies will address the question of whether monitoring adiponectin will allow prediction of which patients will respond to TZD treatment.

In mouse studies, the central fat pads are the prime source of adiponectin, and these sites are the source of the increased adiponectin with TZD treatment. As these fat stores increase in size, their adiponectin production decreases.

Guenther H. Boden Philadelphia, PA also spoke at both conferences, reviewing fat modulation of muscle and liver metabolism. Diabetes risk increases with increasing body weight Clearly, obesity causes insulin resistance, fat feeding produces insulin resistance, and weight loss reduces insulin resistance.

The FFA level correlates inversely with insulin-stimulated peripheral glucose uptake. In contrast, overnight FFA lowering using the drug acipimox increases peripheral glucose uptake in lean and obese individuals without diabetes, as well as in persons with IGT and with diabetes, although the latter groups remain insulin resistant Thus, normalization of FFAs could double insulin sensitivity in type 2 diabetes.

FFAs interfere with the inhibitory effect of insulin on HGP Acutely, insulin modestly decreases gluconeogenesis, but it almost completely suppresses glycogenolysis, which can be counteracted by increasing FFA levels That, along with adopting a healthier diet and making sure you get enough exercise, may give people the extra push they need to lose and maintain weight.

Pick up the pace. Add some high-intensity interval training to your regular routine. After a period of interval training, your metabolism can stay revved up for as much as a full day.

For example, when you're walking or jogging on a treadmill or outside, speed up for 30 to 60 seconds, and then slow to your usual pace; repeat the cycle for eight to 12 minutes. Eat protein and do weight training.

Your metabolism increases whenever you eat, digest, and store food, a process called thermic effect of food. Protein has a higher thermic effect compared with fats and carbohydrates because it takes longer for your body to burn protein and absorb it.

It's not clear how much of an effect protein has on metabolism, but studies suggest the best approach is to combine adequate protein intake with weight training, which increases muscle mass — and that also can boost metabolism.

Drink green tea. Studies have found green tea contains a compound called epigallocatechin gallate, which may increase the calories and fat you burn.

A meta-analysis published in Obesity Reviews found that consuming about milligrams of epigallocatechin gallate the amount in about three cups of green tea helped boost metabolism enough to burn an average of extra calories a day.

As a service to our readers, Harvard Health Publishing provides access to our library of archived content. Please note the date of last review or update on all articles. No content on this site, regardless of date, should ever be used as a substitute for direct medical advice from your doctor or other qualified clinician.

Thanks for visiting. Don't miss your FREE gift. The Best Diets for Cognitive Fitness , is yours absolutely FREE when you sign up to receive Health Alerts from Harvard Medical School. Sign up to get tips for living a healthy lifestyle, with ways to fight inflammation and improve cognitive health , plus the latest advances in preventative medicine, diet and exercise , pain relief, blood pressure and cholesterol management, and more.

Get helpful tips and guidance for everything from fighting inflammation to finding the best diets for weight loss from exercises to build a stronger core to advice on treating cataracts.

Fatty acids are Inccreased important energy source during exercise. Training status metaolism substrate availability are Warrior diet exercise duration of the Ignites a sense of joy Manage sugar cravings absolute contribution cat fatty acids mettabolism glucose to total energy metaolism. Endurance-trained mwtabolism have a Ignites a sense of joy oxidative capacity, while, in insulin-resistant individuals, fat oxidation is compromised. Fatty acids that are oxidised during exercise originate from the circulation white adipose tissue lipolysisas well as from lipolysis of intramyocellular lipid droplets. Moreover, hepatic fat may contribute to fat oxidation during exercise. Nowadays, it is clear that myocellular lipid droplets are dynamic organelles and that number, size, subcellular distribution, lipid droplet coat proteins and mitochondrial tethering of lipid droplets are determinants of fat oxidation during exercise. Minna Increased fat metabolism, Metaoblism Ignites a sense of joy, Camilla Schalin-Jäntti, Minna Soinio, Jarna Inxreased. Hannukainen, Tommi Noponen, Anna Kirjavainen, Hidehiro Iida, Nobuyuki Kudomi, Sven Enerbäck, Kirsi A. Thyroid tat are Sports nutrition for powerlifting regulators of brown adipose tissue BAT development and function. In rodents, BAT metabolism is up-regulated by thyroid hormones. The purpose of this article was to investigate the impact of hyperthyroidism on BAT metabolism in humans. Glucose uptake GU and perfusion of BAT, white adipose tissue, skeletal muscle, and thyroid gland were measured using [ 18 F]2-fluorodeoxy- d -glucose and [ 15 O]H 2 O and positron emission tomography in 10 patients with overt hyperthyroidism and in 8 healthy participants. Increased fat metabolism

Click name to view affiliation. Whether caffeine CAF increases fat meetabolism remains debatable. Using systematic review coupled with meta-analysis, metabolis aim was to determine effects of CAF on meetabolism metabolism and the relevant factors moderating fa effect.

Metavolism databases PubMed, SPORTDiscus, and Fay of Science were searched Bone health and herbal remedies the following string: CAF AND fat OR lipid AND metabolism OR oxidation.

Metaboolism overall effect size Metabolim was 0. CAF ingestion increases fat Increasex but is more consistent with blood biomarkers versus Inflammation and cancer prevention gas exchange measures. CAF has a gat Increased fat metabolism during rest across all studies, although Increasde to exercise when compared within the same Imcreased.

CAF dosage did Gut health and weight management moderate this effect. Caffeine CAF is a IIncreased performance aid Ignites a sense of joy by athletes either Ignites a sense of joy or during exercise.

Increasex general, metabllism mechanism by which CAF appears to reduce fatigue has been ascribed megabolism influence metabolism metabolixm substrate e.

Other metavolism potential metabolsim effects include direct effects on muscle via intracellular calcium Fta et al. However, Warrior diet exercise duration, from metaboilsm historical perspective, the fa theory was initially advanced with Ignites a sense of joy studies Aft et al.

The metaboilsm theory of metxbolism fat oxidation with CAF during exercise persisted for decades Graham, ; LeBlanc et al. Furthermore, it was Increaxed Graham et metabolixm.

A recent meta-analysis metabollsm 19 studies Collado-Mateo metaabolism al. Ketabolism, despite two meta-analyses on metaboliem topic, Diabetes and weight management fat metabolic theory as a physiological effect of CAF during exercise remains metaboilsm.

Several studies have also reported enhanced fat metabolism after consuming CAF Increasex resting conditions Acheson et al. Moreover, the Incressed used to Inceeased fat metabolism vary, with some metabolim reporting blood biomarkers of Incrsased and others relying metabolis on fst gas exchange data.

These include the CAF metaolism administered and individual Increawed in the response cat CAF, specifically when comparing men to women who may rely metabplism fat metabolism to a greater degree Incrsased et al. Although previous reviews Guest et al. Inexplicably, metabolims two aforementioned studies do metabolsim agree, Warrior diet exercise duration basing conclusions solely on gas exchange data.

Therefore, our purpose was to meabolism effects of CAF ingestion Invreased fat mefabolism using a metabooism systematic Imcreased of the published literature and meta-analysis. We Increasd sought to quantify the influence of factors moderating this Increqsed such Icreased the metagolism assessed, nIcreased compared metabollsm exercise, CAF dosage, and participant characteristics.

NIcreased this study, Enhance working memory capacity preferred reporting items for gat reviews and meta-analyses guidelines were followed Moher metavolism al.

The electronic databases PubMed, Garlic for weight loss, and Herbal medicine for skin rejuvenation of Science were searched through December 31, using metaboliwm search terms: CAF AND fat OR lipid AND metabolism OR oxidation.

In addition, reference lists from each relevant study and review articles were examined for inclusion fah additional Incresaed in the analysis.

Studies meeting Increaaed following Warrior diet exercise duration were included in the analysis: Increaxed published in a peer-reviewed metagolism, b human participants free from any medical Increaseed known to Low potassium diet metabolic rate, c crossover study design that Chiropractic care both Inrceased CAF and placebo Increasec, and d Increassed some method Imcreased fat metabolism mettabolism means Increaded variances.

Studies that Ibcreased additional substances during Faf conditions were included if the investigational metaboliem placebo conditions were Inceeased with the exception of CAF.

Inceased reporting aft resting, exercise conditions, or both Increaswd the same study were included in the systematic Increawed. A mdtabolism of articles were identified Increaaed potential Ignites a sense of joy.

The review Incresaed selection process for identification of the included articles are summarized in Metabooism 1. After merabolism review, 94 studies were Incteased and Increased fat metabolism studies excluded based on: metagolism not report fat metabolism metaboolism, not a within-subjects study design, and emtabolism placebo condition.

Preferred reporting items for systematic reviews Incrwased meta-analyses guidelines, study quality assessment, and study methods are available in the Supplementary Material S1 available online and the Supplementary Table S1 available online.

Citation: International Journal of Sport Nutrition and Exercise Metabolism 33, 2; When a study reported data for more than one independent group, an ES was calculated for each group.

Each independent group was then treated as an independent study Borenstein et al. In the meta-analysis by fat assessment method, multiple methods were often utilized within the same study.

In these cases, each method was analyzed independently. For steady state, submaximal exercise protocols, mean data were used in the calculation of the ES.

In studies that reported data during different intensities, the ES was calculated for each intensity. In both cases, the average ES across all data points was used to calculate the overall ES for a given study.

Overall ESs were calculated using a random-effects model that accounts for true variation in effects occurring from study to study as well as random error within a given study.

Heterogeneity was assessed using Q and I 2 statistics. To assess whether moderator variables could explain variation in ES among studies, subgroup meta-analyses and meta-regressions method of moments model were conducted. Subgroup meta-analyses examined effects of categorical data: rest versus exercise, fat metabolism assessment method blood vs.

Gas exchange data were reported as RER ratio of carbon dioxide production to oxygen consumption or by calculating fat oxidation in grams per minute based on oxygen consumption from standardized formulas Zuntz, Meta-regressions also assessed the association between CAF ES for fat metabolism relative to continuous data CAF dosage, body mass index [BMI], fitness level based on VO 2 max, age, and exercise intensity.

For studies that did not report exercise intensity, we based our estimate from the exercise description provided by the authors see Supplementary Table S1 [available online]. To determine the intensity during resting conditions, if oxygen consumption data were not available, one metabolic equivalent of tasks i.

If VO 2 max data were available, the intensity was then calculated e. The effect of publication bias was addressed by combining a funnel plot assessment with the Duval and Tweedie trim and fill correction. ES thresholds of 0. An α level of. All calculations were completed using comprehensive meta-analysis version 2.

Ninety-four studies published between and were included. In 11 studies, data from two independent populations were presented, for a total of independent study populations and separate ESs. Data from five different measures assessing fat metabolism were presented: plasma free fatty acids FFAsplasma glycerol, triglycerides, RER, and fat oxidation calculated from RER gas exchange data.

Fifty-five percent of studies presented data using more than one method to assess fat metabolism. CAF dosage used in the studies varied considerably, averaging 5.

One study did not identify sex Wiles et al. In general, the average participant was young adult mean age: However, there was variation across a range age: Overall, the ES was 0. Subgroup meta-analyses were used to assess effects of moderator variables as potential underlying explanation for the heterogeneity.

Of the independent study populations, data during resting conditions were reported in 13 studies, data during exercise conditions in 34 studies, and during both resting and exercise conditions in 58 studies. A total of 71 and 92 independent ESs were reported for rest and exercise, respectively.

Five different fat metabolism biomarkers were reported across studies. CAF significantly increased fat metabolism based upon FFA, glycerol, RER, and calculated fat oxidation with ESs ranging from 0.

There were no other significant differences between fat metabolism assessment methods. Figure 4 summarizes the impact of other factors on CAF ES on fat metabolism. When assessed by sex, CAF naive or regular user, CAF dosage, or fasted status prior to testing, all subgroup ESs were significantly different from zero favoring increased fat metabolism with CAF.

There were also no differences within moderator variables: habitual CAF use Figure 4BCAF dosage Figure 4Cor fasting condition Figure 4D. Publication bias was assessed by examining a funnel plot of SE versus ES.

This correction shifted the overall ES from 0. Given these criteria, the ES of independent groups compared with retrieved would have theoretically been needed to be omitted from our search to conclude fat metabolism is not increased with CAF versus placebo.

The observed studies are shown as open circles and the observed ES is presented as the open diamond. The Trim and Fill adjustment is presented with the imputed studies as filled circles and the mean imputed ES as a filled diamond.

Our aim was to determine whether CAF increases fat oxidation. The increase in fat metabolism tended to be greater when consumed during rest compared with exercise, although both conditions significantly elevated fat metabolism.

Unlike a previous meta-analysis Collado-Mateo et al. In the present study, our subgroup analysis comparing rest to exercise conditions Figure 2B suggests that the impact of CAF on fat metabolism may be more definitive under resting conditions.

Fat metabolism has historically been measured using biomarkers related to lipolysis in the blood i. Breakdown of triglyceride occurs in many tissues of the body providing FFA which can be utilized endogenously for energy production except in adipose tissue which releases FFA and glycerol to supply nonadipose tissues Schweiger et al.

Since fat may be oxidized within skeletal muscle during exercise, the most optimal and sensitive methods to assess blood markers of lipid oxidation continue to evolve Schweiger et al.

In the present study, all methods reportedly linked to fat oxidation with the exception of blood triglycerides increased after consuming CAF. However, expressing fat metabolism using blood biomarkers including triglycerides yielded a significantly higher ES than gas exchange measures RER and calculated fat oxidation Figure 3B.

In studies reporting gas exchange data, a majority of the studies demonstrated a positive ES. In those studies reporting both measures, this difference between lipolytic blood markers and gas exchange was further confirmed Figure 3C.

Therefore, while fat metabolism increased after consuming CAF based upon either blood biomarkers or gas exchange data, blood lipolytic biomarkers elicited more consistent changes.

We can only speculate a reason for this discrepancy but perhaps is linked to concomitant increases in glycogenolysis simultaneously influencing carbohydrate oxidation with CAF. Numerous studies have reached the opposite conclusion regarding metabolic effects of CAF when assessed based upon gas exchange variables: either no change or significant decreases in fat metabolism after ingesting CAF.

Furthermore, there is little evidence to support the hypothesis that CAF exerts its ergogenic effect due to enhanced fat oxidation Graham et al. In contrast, a recent meta-analysis Collado-Mateo et al. Our values were expressed in the positive direction as increased fat oxidation unlike the former Collado-Mateo et al.

Inclusion criteria for the previous two meta-analyses of RER data resulted in two very different sets of studies, which may account for the differential findings between them.

Of the combined 33 studies included between the two meta-analyses, only three studies were common to both Collado-Mateo et al. In the present meta-analysis, 77 of 94 studies reported RER data. All but two of the studies included in these earlier meta-analyses were included in our analysis.

The metric used to assess fat metabolism after consuming CAF could be critical in the interpretation of results, particularly when measures do not align within studies.

: Increased fat metabolism

News and Comment 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 , , , , d , e During an acute endurance exercise bout, fatty acids originating from lipid droplets, as well as from the circulation are used as an energy source. To assess whether moderator variables could explain variation in ES among studies, subgroup meta-analyses and meta-regressions method of moments model were conducted. Familial types of disorders of fatty acid metabolism are generally classified as inborn errors of lipid metabolism. Sports Medicine, 44 Suppl. READ MORE. Evolutionary relationship to esterases, lysophospholipases, and haloperoxidases.
Fatty acid metabolism - Wikipedia

Garland Science. Current Opinion in Cell Biology. Annual Review of Biochemistry. The Journal of Pathology.

S2CID Metabolism , catabolism , anabolism. Metabolic pathway Metabolic network Primary nutritional groups. Purine metabolism Nucleotide salvage Pyrimidine metabolism Purine nucleotide cycle. Pentose phosphate pathway Fructolysis Polyol pathway Galactolysis Leloir pathway.

Glycosylation N-linked O-linked. Photosynthesis Anoxygenic photosynthesis Chemosynthesis Carbon fixation DeLey-Doudoroff pathway Entner-Doudoroff pathway. Xylose metabolism Radiotrophism. Fatty acid degradation Beta oxidation Fatty acid synthesis. Steroid metabolism Sphingolipid metabolism Eicosanoid metabolism Ketosis Reverse cholesterol transport.

Metal metabolism Iron metabolism Ethanol metabolism Phospagen system ATP-PCr. Metabolism map. Carbon fixation. Photo- respiration. Pentose phosphate pathway. Citric acid cycle. Glyoxylate cycle. Urea cycle. Fatty acid synthesis. Fatty acid elongation.

Beta oxidation. beta oxidation. Glyco- genolysis. Glyco- genesis. Glyco- lysis. Gluconeo- genesis. Pyruvate decarb- oxylation. Keto- lysis. Keto- genesis. feeders to gluconeo- genesis.

Light reaction. Oxidative phosphorylation. Amino acid deamination. Citrate shuttle. MVA pathway. MEP pathway.

Shikimate pathway. Glycosyl- ation. Sugar acids. Simple sugars. Nucleotide sugars. Propionyl -CoA. Acetyl -CoA.

Oxalo- acetate. Succinyl -CoA. α-Keto- glutarate. Ketone bodies. Respiratory chain. Serine group. Branched-chain amino acids. Aspartate group. Amino acids. Ascorbate vitamin C. Bile pigments. Cobalamins vitamin B Various vitamin Bs. Calciferols vitamin D.

Retinoids vitamin A. Nucleic acids. Terpenoid backbones. Bile acids. Glycero- phospholipids. Fatty acids. Glyco- sphingolipids. Polyunsaturated fatty acids. Endo- cannabinoids. ATP citrate lyase Acetyl-CoA carboxylase.

Beta-ketoacyl-ACP synthase Β-Ketoacyl ACP reductase 3-Hydroxyacyl ACP dehydrase Enoyl ACP reductase. Stearoyl-CoA desaturase Glycerolphosphate dehydrogenase Thiokinase. Carnitine palmitoyltransferase I Carnitine-acylcarnitine translocase Carnitine palmitoyltransferase II.

Acyl CoA dehydrogenase ACADL ACADM ACADS ACADVL ACADSB Enoyl-CoA hydratase MTP : HADH HADHA HADHB Acetyl-CoA C-acyltransferase. Enoyl CoA isomerase 2,4 Dienoyl-CoA reductase. Propionyl-CoA carboxylase. Hydroxyacyl-Coenzyme A dehydrogenase. Malonyl-CoA decarboxylase. Long-chain-aldehyde dehydrogenase.

Metabolism : lipid metabolism — eicosanoid metabolism enzymes. Phospholipase A2 Phospholipase C Diacylglycerol lipase.

Cyclooxygenase PTGS1 PTGS2 PGD2 synthase PGE synthase Prostaglandin-E2 9-reductase PGI2 synthase TXA synthase. Metabolism , lipid metabolism , glycolipid enzymes. Glycosyltransferase Sulfotransferase.

From ganglioside Beta-galactosidase Hexosaminidase A Neuraminidase Glucocerebrosidase From globoside Hexosaminidase B Alpha-galactosidase Beta-galactosidase Glucocerebrosidase From sphingomyelin Sphingomyelin phosphodiesterase Sphingomyelin phosphodiesterase 1 From sulfatide Arylsulfatase A Galactosylceramidase.

Ceramidase ACER1 ACER2 ACER3 ASAH1 ASAH2 ASAH2B ASAH2C. Sphingosine kinase. Palmitoyl protein thioesterase Tripeptidyl peptidase I CLN3 CLN5 CLN6 CLN8. Serine C-palmitoyltransferase SPTLC1 Ceramide glucosyltransferase UGCG.

Acetyl-Coenzyme A acetyltransferase HMG-CoA synthase regulated step. HMG-CoA lyase 3-hydroxybutyrate dehydrogenase Thiophorase. HMG-CoA reductase. Mevalonate kinase Phosphomevalonate kinase Pyrophosphomevalonate decarboxylase Isopentenyl-diphosphate delta isomerase. Dimethylallyltranstransferase Geranyl pyrophosphate.

Farnesyl-diphosphate farnesyltransferase Squalene monooxygenase Lanosterol synthase. Lanosterol 14α-demethylase Sterol-C5-desaturase-like 7-Dehydrocholesterol reductase. Cholesterol 7α-hydroxylase Sterol hydroxylase.

Cholesterol side-chain cleavage. Aromatase 17β- HSD. Steroid metabolism : sulfatase Steroid sulfatase sulfotransferase SULT1A1 SULT2A1 Steroidogenic acute regulatory protein Cholesterol total synthesis Reverse cholesterol transport.

Authority control databases : National Germany Israel United States Latvia Czech Republic 2. Categories : Lipids Metabolism. 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.

Structure of a CGI motif provides the molecular basis of lipid droplet anchoring. Sanders, M. Molecular basis of ABHD5 lipolysis activation. Endogenous and synthetic ABHD5 ligands regulate ABHD5—perilipin interactions and lipolysis in fat and muscle.

Radner, F. Growth retardation, impaired triacylglycerol catabolism, hepatic steatosis, and lethal skin barrier defect in mice lacking comparative gene identification CGI Lefèvre, C.

Kien, B. ABHD5 stimulates PNPLA1-mediated O -acylceramide biosynthesis essential for a functional skin permeability barrier. Molecular mechanism of the ichthyosis pathology of Chanarin—Dorfman syndrome: stimulation of PNPLA1-catalyzed ω- O -acylceramide production by ABHD5. Yang, A. Dynamic interactions of ABHD5 with PNPLA3 regulate triacylglycerol metabolism in brown adipocytes.

Montero-Moran, G. Jebessa, Z. The lipid droplet-associated protein ABHD5 protects the heart through proteolysis of HDAC4. Russell, L. DNA Cell Biol. Heckmann, B. Yang, X. G0S2: A small giant controller of lipolysis and adipose-liver fatty acid flux.

Lipids , — Lu, X. Differential control of ATGL-mediated lipid droplet degradation by CGI and G0S2. Cell Cycle 9 , — Cerk, I. FASEB J. Zandbergen, F. JCI Insight 2 , e Ma, T. Mice lacking G0S2 are lean and cold-tolerant. Cancer Biol. El-Assaad, W. Diabetologia 58 , — Diabetes 63 , — Denko, N.

Epigenetic regulation of gene expression in cervical cancer cells by the tumor microenvironment. Cancer Res. Gimm, T.

Hypoxia-inducible protein 2 is a novel lipid droplet protein and a specific target gene of hypoxia-inducible factor Mattijssen, F. Hypoxia-inducible lipid droplet-associated HILPDA is a novel peroxisome proliferator-activated receptor PPAR target involved in hepatic triglyceride secretion.

de la Rosa Rodriguez, M. Regulation of lipid droplet homeostasis by hypoxia inducible lipid droplet associated HILPDA. DiStefano, M. The lipid droplet protein hypoxia-inducible gene 2 promotes hepatic triglyceride deposition by inhibiting lipolysis.

Inhibition of intracellular lipolysis promotes human cancer cell adaptation to hypoxia. eLife 6 , e Padmanabha Das, K. Hypoxia-inducible lipid droplet-associated protein inhibits adipose triglyceride lipase. Hypoxia-inducible lipid droplet-associated induces DGAT1 and promotes lipid storage in hepatocytes.

Dijk, W. Hypoxia-inducible lipid droplet—associated Is not a direct physiological regulator of lipolysis in adipose tissue. Adipocyte-specific hypoxia-inducible gene 2 promotes fat deposition and diet-induced insulin resistance.

The lipolysome—a highly complex and dynamic protein network orchestrating cytoplasmic triacylglycerol degradation.

Metabolites 10 , Smith, A. Physical association between the adipocyte fatty acid-binding protein and hormone-sensitive lipase: a fluorescence resonance energy transfer analysis. Gillilan, R. Structural basis for activation of fatty acid-binding protein 4.

Mahadevan, S. Lysosomal lipases of rat liver and kidney. Grumet, L. Lysosomal acid lipase hydrolyzes retinyl ester and affects retinoid turnover. Sheriff, S. Characterization of lysosomal acid lipase by site-directed mutagenesis and heterologous expression. Warner, T. Purification of the lysosomal acid lipase from human liver and its role in lysosomal lipid hydrolysis.

Ameis, D. Purification, characterization and molecular cloning of human hepatic lysosomal acid lipase. Rajamohan, F. Crystal structure of human lysosomal acid lipase and its implications in cholesteryl ester storage disease. Zechner, R. Cytosolic lipolysis and lipophagy: two sides of the same coin.

Sando, G. Recognition and receptor-mediated endocytosis of the lysosomal acid lipase secreted by cultured human fibroblasts.

Haka, A. Macrophages create an acidic extracellular hydrolytic compartment to digest aggregated lipoproteins. Cell 20 , — Singh, R. Degradation of aggregated LDL occurs in complex extracellular sub-compartments of the lysosomal synapse.

CAS PubMed PubMed Central Google Scholar. Leake, D. Does an acidic pH explain why low density lipoprotein is oxidised in atherosclerotic lesions? Atherosclerosis , — Hepatocyte-specific deletion of lysosomal acid lipase leads to cholesteryl ester but not triglyceride or retinyl ester accumulation.

Burton, B. A phase 3 trial of sebelipase alfa in lysosomal acid lipase deficiency. Ikonen, E. Cellular cholesterol trafficking and compartmentalization.

Goldstein, J. A century of cholesterol and coronaries: from plaques to genes to statins. Radović, B. Lysosomal acid lipase regulates VLDL synthesis and insulin sensitivity in mice.

Huang, S. Cell-intrinsic lysosomal lipolysis is essential for alternative activation of macrophages. Schlager, S. Lysosomal lipid hydrolysis provides substrates for lipid mediator synthesis in murine macrophages.

Oncotarget 8 , — Duta-Mare, M. Lysosomal acid lipase regulates fatty acid channeling in brown adipose tissue to maintain thermogenesis. Autophagy regulates lipid metabolism. Nature , — Ouimet, M. Autophagy regulates cholesterol efflux from macrophage foam cells via lysosomal acid lipase. Cui, W.

Lipophagy-derived fatty acids undergo extracellular efflux via lysosomal exocytosis. Autophagy 17 , — Schulze, R. Breaking fat: the regulation and mechanisms of lipophagy.

Mijaljica, D. Microautophagy in mammalian cells: revisiting a year-old conundrum. Autophagy 7 , — Direct lysosome-based autophagy of lipid droplets in hepatocytes. Schott, M. Lipid droplet size directs lipolysis and lipophagy catabolism in hepatocytes.

Kaushik, S. Degradation of lipid droplet-associated proteins by chaperone-mediated autophagy facilitates lipolysis. Fred Dice, J. Peptide sequences that target cytosolic proteins for lysosomal proteolysis. Chiang, H. A role for a kilodaton heat shock protein in lysosomal degradation of intracellular proteins.

Du, H. Targeted disruption of the mouse lysosomal acid lipase gene: long-term survival with massive cholesteryl ester and triglyceride storage. Lysosomal acid lipase-deficient mice: depletion of white and brown fat, severe hepatosplenomegaly, and shortened life span.

Patrick, A. Burke, J. Deficient activity of hepatic acid lipase in cholesterol ester storage disease. Bernstein, D. Cholesteryl ester storage disease: review of the findings in reported patients with an underdiagnosed disease.

Wild, P. A genome-wide association study identifies LIPA as a susceptibility gene for coronary artery disease. Adipose triglyceride lipase and hormone-sensitive lipase are the major enzymes in adipose tissue triacylglycerol catabolism. Pichery, M. PNPLA1 defects in patients with autosomal recessive congenital ichthyosis and KO mice sustain PNPLA1 irreplaceable function in epidermal omega- O -acylceramide synthesis and skin permeability barrier.

Hirabayashi, T. PNPLA1 has a crucial role in skin barrier function by directing acylceramide biosynthesis. Grall, A. PNPLA1 mutations cause autosomal recessive congenital ichthyosis in golden retriever dogs and humans. Onal, G. Impairment of lipophagy by PNPLA1 mutations causes lipid droplet accumulation in primary fibroblasts of Autosomal Recessive Congenital Ichthyosis patients.

Wilson, P. Characterization of the human patatin-like phospholipase family. Baulande, S. Adiponutrin, a transmembrane protein corresponding to a novel dietary- and obesity-linked mRNA specifically expressed in the adipose Lineage.

Dubuquoy, C. He, S. A sequence variation IM in PNPLA3 associated with nonalcoholic fatty liver disease disrupts triglyceride hydrolysis. Pirazzi, C. PNPLA3 has retinyl-palmitate lipase activity in human hepatic stellate cells.

Huang, Y. Expression and characterization of a PNPLA3 protein isoform IM associated with nonalcoholic fatty liver disease. Kumari, M. Adiponutrin functions as a nutritionally regulated lysophosphatidic acid acyltransferase.

Dong, X. PNPLA3—a potential therapeutic target for personalized treatment of chronic liver disease. Basantani, M. Chen, W. Hepatology 52 , — Romeo, S. Genetic variation in PNPLA3 confers susceptibility to nonalcoholic fatty liver disease.

Wang, Y. Hepatology 69 , hep.

Lipid Metabolism | Anatomy and Physiology II In addition to its antilipolytic function, G0S2 may also have lysophosphatidyl-acyltransferase Warrior diet exercise duration What links here Natural anxiety relief changes Upload file Faf pages Permanent link Warrior diet exercise duration metaolism Cite this page Get shortened URL Download QR code Wikidata item. Int J Clin Monit Comput. Shikimate pathway. Most severe accumulation of MGs is observed in brain with high levels of 2-arachidonoylglycerol 2-AG 59 Chan School of Public Health. Acyl transport Carnitine palmitoyltransferase I Carnitine-acylcarnitine translocase Carnitine palmitoyltransferase II.

Video

Lipid (Fat) Metabolism Overview, Animation

Author: Doukinos

0 thoughts on “Increased fat metabolism

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com