851
|
Abstract
The proper functioning of the pathways that are involved in the sensing and management of nutrients is central to metabolic homeostasis and is therefore among the most fundamental requirements for survival. Metabolic systems are integrated with pathogen-sensing and immune responses, and these pathways are evolutionarily conserved. This close functional and molecular integration of the immune and metabolic systems is emerging as a crucial homeostatic mechanism, the dysfunction of which underlies many chronic metabolic diseases, including type 2 diabetes and atherosclerosis. In this Review we provide an overview of several important networks that sense and manage nutrients and discuss how they integrate with immune and inflammatory pathways to influence the physiological and pathological metabolic states in the body.
Collapse
Affiliation(s)
- Gökhan S Hotamisligil
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts 02115, USA.
| | | |
Collapse
|
852
|
Gillum MP, Zhang D, Zhang XM, Erion DM, Jamison RA, Choi C, Dong J, Shanabrough M, Duenas HR, Frederick DW, Hsiao JJ, Horvath TL, Lo CM, Tso P, Cline GW, Shulman GI. N-acylphosphatidylethanolamine, a gut- derived circulating factor induced by fat ingestion, inhibits food intake. Cell 2008; 135:813-24. [PMID: 19041747 PMCID: PMC2643061 DOI: 10.1016/j.cell.2008.10.043] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2008] [Revised: 08/20/2008] [Accepted: 10/28/2008] [Indexed: 12/12/2022]
Abstract
N-acylphosphatidylethanolamines (NAPEs) are a relatively abundant group of plasma lipids of unknown physiological significance. Here, we show that NAPEs are secreted into circulation from the small intestine in response to ingested fat and that systemic administration of the most abundant circulating NAPE, at physiologic doses, decreases food intake in rats without causing conditioned taste aversion. Furthermore, (14)C-radiolabeled NAPE enters the brain and is particularly concentrated in the hypothalamus, and intracerebroventricular infusions of nanomolar amounts of NAPE reduce food intake, collectively suggesting that its effects may be mediated through direct interactions with the central nervous system. Finally, chronic NAPE infusion results in a reduction of both food intake and body weight, suggesting that NAPE and long-acting NAPE analogs may be novel therapeutic targets for the treatment of obesity.
Collapse
Affiliation(s)
- Matthew P. Gillum
- Howard Hughes Medical Institute, Yale University School of Medicine New Haven, Connecticut 06520, USA
- Department of Internal Medicine, Yale University School of Medicine New Haven, Connecticut 06520, USA
- Department of Cellular & Molecular Physiology, Yale University School of Medicine New Haven, Connecticut 06520, USA
| | - Dongyan Zhang
- Howard Hughes Medical Institute, Yale University School of Medicine New Haven, Connecticut 06520, USA
| | - Xian-Man Zhang
- Department of Internal Medicine, Yale University School of Medicine New Haven, Connecticut 06520, USA
| | - Derek M. Erion
- Howard Hughes Medical Institute, Yale University School of Medicine New Haven, Connecticut 06520, USA
- Department of Internal Medicine, Yale University School of Medicine New Haven, Connecticut 06520, USA
- Department of Cellular & Molecular Physiology, Yale University School of Medicine New Haven, Connecticut 06520, USA
| | - Rachel A. Jamison
- Howard Hughes Medical Institute, Yale University School of Medicine New Haven, Connecticut 06520, USA
- Department of Internal Medicine, Yale University School of Medicine New Haven, Connecticut 06520, USA
| | - Cheolsoo Choi
- Department of Internal Medicine, Yale University School of Medicine New Haven, Connecticut 06520, USA
| | - Jianying Dong
- Howard Hughes Medical Institute, Yale University School of Medicine New Haven, Connecticut 06520, USA
| | - Marya Shanabrough
- Department of Comparative Medicine, Yale University School of Medicine New Haven, Connecticut 06520, USA
| | - Hillary R. Duenas
- Department of Comparative Medicine, Yale University School of Medicine New Haven, Connecticut 06520, USA
| | - David W. Frederick
- Department of Internal Medicine, Yale University School of Medicine New Haven, Connecticut 06520, USA
| | - Jennifer J. Hsiao
- Department of Internal Medicine, Yale University School of Medicine New Haven, Connecticut 06520, USA
| | - Tamas L. Horvath
- Department of Comparative Medicine, Yale University School of Medicine New Haven, Connecticut 06520, USA
| | - Chun Min Lo
- Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA, 45627
| | - Pat Tso
- Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA, 45627
| | - Gary W. Cline
- Department of Internal Medicine, Yale University School of Medicine New Haven, Connecticut 06520, USA
| | - Gerald I. Shulman
- Howard Hughes Medical Institute, Yale University School of Medicine New Haven, Connecticut 06520, USA
- Department of Internal Medicine, Yale University School of Medicine New Haven, Connecticut 06520, USA
- Department of Cellular & Molecular Physiology, Yale University School of Medicine New Haven, Connecticut 06520, USA
| |
Collapse
|
853
|
Acute effects of breakfasts containing alpha-lactalbumin, or gelatin with or without added tryptophan, on hunger, 'satiety' hormones and amino acid profiles. Br J Nutr 2008; 101:1859-66. [PMID: 19017422 DOI: 10.1017/s0007114508131774] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Proteins are the most satiating macronutrients. Tryptophan (TRP) may contribute to the satiating effect, as it serves as a precursor for the anorexigenic neurotransmitter serotonin. To address the role of TRP in the satiating properties of dietary protein, we compared three different breakfasts, containing either alpha-lactalbumin (high in TRP), gelatin (low in TRP) or gelatin with added TRP (gelatin+TRP, high in TRP), on appetite. Twenty-four subjects (22-29 kg/m2; aged 19-37 years) received a subject-specific breakfast at t = 0 with 10, 55 and 35 % energy from protein, carbohydrate and fat respectively in a randomised, single-blind design. Hunger, glucagon-like peptide (GLP)-1, ghrelin, amino acid concentrations and energy intake during a subsequent lunch were determined. Suppression of hunger was stronger 240 min after the breakfast with alpha-lactalbumin compared with gelatin and gelatin+TRP. Total plasma amino acid concentrations were lower with alpha-lactalbumin compared with gelatin with or without TRP (from t = 180-240 min). TRP concentrations were higher after alpha-lactalbumin than after gelatin with or without TRP from t = 0-100 min, whereas from t = 100-240 min, TRP concentrations were lower after gelatin than after alpha-lactalbumin and gelatin+TRP. The plasma ratio of TRP to other large neutral amino acids (LNAA) was, only at t = 100 min, lower after gelatin+TRP than after the other breakfasts. Plasma amino acid responses, TRP concentrations and TRP:LNAA ratios were not correlated with hunger. GLP-1 and ghrelin concentrations were similar for all diets. Energy intake during a subsequent lunch was similar for all diets. Summarised, an alpha-lactalbumin breakfast suppresses hunger more than a gelatin or gelatin+TRP breakfast. This cannot be explained by (possible) differences found in TRP concentrations and TRP:LNAA ratios in the breakfasts and in plasma, as well as in circulating total amino acids, GLP-1 and ghrelin.
Collapse
|
854
|
Leptin-enhanced neointimal hyperplasia is reduced by mTOR and PI3K inhibitors. Proc Natl Acad Sci U S A 2008; 105:19006-11. [PMID: 19020099 DOI: 10.1073/pnas.0809743105] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Despite the use of the sirolimus (rapamycin) drug-eluting coronary stent, diabetics are at increased risk of developing in-stent restenosis for unclear reasons. Hyperleptinemia, which often coexists with diabetes and metabolic syndrome, is an independent risk factor for progression of coronary artery disease. It has not been determined whether elevated circulating leptin decreases the efficacy of the sirolimus drug-eluting stent in inhibiting neointimal hyperplasia, the process underlying restenosis after stenting. Here we show that leptin activates the mammalian target of rapamycin (mTOR) signaling pathway in primary murine vascular smooth muscle cells (VSMC) and stimulates VSMC proliferation in a PI3K-dependent fashion. Exogenous leptin, administered at levels comparable to those found in obese humans, promotes neointimal VSMC hyperplasia in a murine femoral artery wire injury model. Leptin significantly increases the dose of the mTOR inhibitor sirolimus that is required for effective inhibition of neointimal formation. Combination therapy with LY294002, a PI3K inhibitor, and sirolimus effectively inhibits leptin-enhanced neointimal hyperplasia. These data show that, in the setting of hyperleptinemia, higher doses of an mTOR inhibitor, or combination therapy with mTOR and PI3K inhibitors, inhibits neointimal hyperplasia after arterial injury. These studies may explain the higher rates of restenosis observed in diabetics treated with a sirolimus-eluting coronary stent and suggest a potential novel therapeutic approach for inhibiting in-stent restenosis in such patients.
Collapse
|
855
|
Established maternal obesity in the rat reprograms hypothalamic appetite regulators and leptin signaling at birth. Int J Obes (Lond) 2008; 33:115-22. [PMID: 18982008 DOI: 10.1038/ijo.2008.213] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Key appetite regulators and their receptors are already present in the fetal hypothalamus, and may respond to hormones such as leptin. Intrauterine food restriction or hyperglycemia can reprogram these circuits, possibly predisposing individuals to adverse health outcomes in adulthood. Given the global obesity epidemic, maternal overweight and obesity is becoming more prevalent. Earlier, we observed rapid growth of pups from obese dams during the suckling period. However, it is unclear whether this is because of alterations in leptin and hypothalamic appetite regulators at birth. DESIGN Female Sprague-Dawley rats were fed palatable high-fat diet (HFD) or chow for 5 weeks to induce obesity before mating. The same diet continued during gestation. At day 1, after birth, plasma and hypothalamus were collected from male and female pups. MEASUREMENTS Body weight and organ mass were recorded. Leptin and insulin levels were measured in the plasma by radioimmunoassay. Hypothalamic mRNA expression of neuropeptide-Y (NPY), pro-opiomelanocortin, leptin receptor and its downstream signal, STAT3 (signal transducer and activator of transcription 3), were measured using real-time PCR. RESULTS Body and organ weights of pups from obese dams were similar to those from lean dams, across both genders. However, plasma leptin levels were significantly lower in offspring from obese dams (male: 0.53+/-0.13 vs 1.05+/-0.21 ng ml(-1); female: 0.33+/-0.09 vs 2.12+/-0.57 ng ml(-1), respectively; both P<0.05). Hypothalamic mRNA expression of NPY, pro-opiomelanocortin, leptin receptor and STAT3 were also significantly lower in pups from obese dams. CONCLUSION Long-term maternal obesity, together with lower leptin levels in pups from obese dams may contribute to the lower expression of key appetite regulators on day 1 of life, suggesting altered intrauterine neuron development in response to intrauterine overnutrition, which may contribute to eating disorders later in life.
Collapse
|
856
|
Langhans W. Fatty acid oxidation in the energostatic control of eating—A new idea. Appetite 2008; 51:446-51. [DOI: 10.1016/j.appet.2008.06.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2008] [Revised: 06/29/2008] [Accepted: 06/30/2008] [Indexed: 11/25/2022]
|
857
|
Kim DH, Sandoval D, Reed JA, Matter EK, Tolod EG, Woods SC, Seeley RJ. The role of GM-CSF in adipose tissue inflammation. Am J Physiol Endocrinol Metab 2008; 295:E1038-46. [PMID: 18765677 PMCID: PMC2584818 DOI: 10.1152/ajpendo.00061.2008] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a proinflammatory cytokine that has a central action to reduce food intake and body weight. Consistent with this, GM-CSF knockout mice are more obese and hyperphagic than wild-type mice. However, in lung, GM-CSF is an important determinant of macrophage infiltration. Consequently, we sought to determine if GM-CSF might contribute to adipose tissue macrophage accumulation, insulin resistance, and low-grade inflammation that occurs when animals gain weight on a high-fat diet (HFD). We therefore determined how targeted genetic disruption of GM-CSF can affect adipose tissue macrophage and cytokine gene expression as well as glucose homeostasis by performing hyperinsulinemic-euglycemic clamps. The number of macrophages and CCR2 gene expression in adipose tissue of GM-CSF knockout mice was decreased relative to those in wild-type mice, and the adipocyte size of mesenteric fat was increased in GM-CSF knockout mice on a HFD compared with wild-type mice. The level of mRNA of the proinflammatory cytokines interleukin-1beta, tumor necrosis factor-alpha, and macrophage inflammatory protein-1alpha was significantly lower in mesenteric fat of GM-CSF knockout mice on the HFD than in wild-type mice. Using the hyperinsulinemic-euglycemic clamp technique, GM-CSF knockout mice had greater overall insulin sensitivity. This increase was due to enhanced peripheral uptake and utilization of glucose rather than to increased hepatic insulin sensitivity. Collectively, the data suggest that the GM-CSF knockout mutation ameliorates peripheral insulin resistance in spite of increased adiposity by reducing inflammation in adipose tissue in response to a HFD.
Collapse
Affiliation(s)
- Dong-Hoon Kim
- Department of Psychiartry, University of Cincinnati, Cincinnati, OH, USA
| | | | | | | | | | | | | |
Collapse
|
858
|
Chen H, Simar D, Lambert K, Mercier J, Morris MJ. Maternal and postnatal overnutrition differentially impact appetite regulators and fuel metabolism. Endocrinology 2008; 149:5348-56. [PMID: 18635655 DOI: 10.1210/en.2008-0582] [Citation(s) in RCA: 189] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Maternal obesity is increasing, and it is known that the intrauterine experience programs fetal and newborn metabolism. However, the relative contributions of pre- or postnatal factors are unknown. We hypothesized that maternal overnutrition caused by long-term maternal obesity would exert a stronger detrimental impact than postnatal overnutrition on offspring metabolic homeostasis, with additional postnatal overnutrition exaggerating these alterations. Female Sprague Dawley rats were exposed to chow or high-fat cafeteria diet for 5 wk before mating and throughout gestation and lactation. On postnatal d 1, litters were adjusted to three per litter to induce postnatal overnutrition (vs. 12 in control). Hypothalamic appetite regulators neuropeptide Y and proopiomelanocortin, glucose transporter 4, and lipid metabolic markers were measured. At postnatal d 20, male pups born of obese dams, or those overnourished postnatally, were 42% heavier than controls; combining both interventions led to 80% greater body weight. Maternal obesity increased pup adiposity and led to glucose intolerance in offspring; these were exaggerated by additional postnatal overnutrition during lactation. Maternal obesity was also linked to hyperlipidemia in offspring and reduced hypothalamic neuropeptide Y and increased proopiomelanocortin mRNA expression. Postnatal overnutrition of offspring from obese dams amplified these hypothalamic changes. Both maternal and postnatal overnutrition reduced muscle glucose transporter 4. Adipose carnitine palmitoyl-transferase-1 and adipose triglyceride lipase mRNA was up-regulated only by postnatal overnutrition. Maternal overnutrition appears to alter central appetite circuits and promotes early-onset obesity; postnatal overnutrition interacted to cause peripheral lipid and glucose metabolic disorders, supporting the critical message to reduce early-life adverse nutritional impact.
Collapse
Affiliation(s)
- Hui Chen
- Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, New South Wales 2052, Australia
| | | | | | | | | |
Collapse
|
859
|
Abstract
Evidence for the regulation of body energy is reviewed from the homeostatic perspective of Claude Bernard and Walter Cannon. The complementary roles of food intake and energy expenditure in the maintenance and defense of energy balance are considered. Particular attention is paid to the roles adjustments in energy expenditure play in this process and to recent investigations identifying their metabolic underpinnings. This is followed by a consideration of the many newly identified signals of body energy status and the pathways and feedback loops they utilize to inform the central regulating system. Finally, various naturally occurring and experimentally induced alterations in the regulated level of body energy are described and discussed. It is concluded that, though early investigators did not expressly consider energy a regulated feature of the milieu interieur, more recent research has provided a sound basis for judging the regulation of body energy to be another homeostatic process.
Collapse
|
860
|
Figlewicz DP, Benoit SC. Insulin, leptin, and food reward: update 2008. Am J Physiol Regul Integr Comp Physiol 2008; 296:R9-R19. [PMID: 18945945 DOI: 10.1152/ajpregu.90725.2008] [Citation(s) in RCA: 149] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The hormones insulin and leptin have been demonstrated to act in the central nervous system (CNS) as regulators of energy homeostasis at medial hypothalamic sites. In a previous review, we described new research demonstrating that, in addition to these direct homeostatic actions at the hypothalamus, CNS circuitry that subserves reward and motivation is also a direct and an indirect target for insulin and leptin action. Specifically, insulin and leptin can decrease food reward behaviors and modulate the function of neurotransmitter systems and neural circuitry that mediate food reward, i.e., midbrain dopamine and opioidergic pathways. Here we summarize new behavioral, systems, and cellular evidence in support of this hypothesis and in the context of research into the homeostatic roles of both hormones in the CNS. We discuss some current issues in the field that should provide additional insight into this hypothetical model. The understanding of neuroendocrine modulation of food reward, as well as food reward modulation by diet and obesity, may point to new directions for therapeutic approaches to overeating or eating disorders.
Collapse
Affiliation(s)
- Dianne P Figlewicz
- Metabolism/Endocrinology (151) VA Puget Sound Health Care System, 1660 So. Columbian Way, Seattle, WA 98108, USA.
| | | |
Collapse
|
861
|
Woods SC, Seeley RJ, Cota D. Regulation of food intake through hypothalamic signaling networks involving mTOR. Annu Rev Nutr 2008; 28:295-311. [PMID: 18429698 DOI: 10.1146/annurev.nutr.28.061807.155505] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
To maintain normal activity, single cells must assure that their energy needs and utilization are continuously matched. Likewise, multicellular organisms must constantly coordinate energy intake and expenditure to maintain energy homeostasis. The brain, and the hypothalamus in particular, plays a critical role in integrating and coordinating several types of signals, including hormones and nutrients, to guarantee such homeostasis. Like single cells, the hypothalamus also profits from intracellular pathways known to work as fuel sensors to maintain energy balance. One such pathway is the mammalian target of rapamycin (mTOR). mTOR integrates different sensory inputs to regulate protein synthesis rates in individual cells, and it has recently been implicated in the central nervous system to regulate food intake and body weight as well. This review provides an overview of the role of hypothalamic intracellular fuel sensors in the overall control of energy balance and discusses the potential contribution of these fuel-sensing mechanisms to the metabolic dysregulation associated with obesity.
Collapse
Affiliation(s)
- Stephen C Woods
- Department of Psychiatry, Genome Research Institute, University of Cincinnati, Cincinnati, Ohio 45237, USA.
| | | | | |
Collapse
|
862
|
Zhang X, Zhang G, Zhang H, Karin M, Bai H, Cai D. Hypothalamic IKKbeta/NF-kappaB and ER stress link overnutrition to energy imbalance and obesity. Cell 2008; 135:61-73. [PMID: 18854155 PMCID: PMC2586330 DOI: 10.1016/j.cell.2008.07.043] [Citation(s) in RCA: 1104] [Impact Index Per Article: 64.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2007] [Revised: 03/13/2008] [Accepted: 07/31/2008] [Indexed: 02/06/2023]
Abstract
Overnutrition is associated with chronic inflammation in metabolic tissues. Whether metabolic inflammation compromises the neural regulatory systems and therefore promotes overnutrition-associated diseases remains unexplored. Here we show that a mediator of metabolic inflammation, IKKbeta/NF-kappaB, normally remains inactive although enriched in hypothalamic neurons. Overnutrition atypically activates hypothalamic IKKbeta/NF-kappaB at least in part through elevated endoplasmic reticulum stress in the hypothalamus. While forced activation of hypothalamic IKKbeta/NF-kappaB interrupts central insulin/leptin signaling and actions, site- or cell-specific suppression of IKKbeta either broadly across the brain or locally within the mediobasal hypothalamus, or specifically in hypothalamic AGRP neurons significantly protects against obesity and glucose intolerance. The molecular mechanisms involved include regulation by IKKbeta/NF-kappaB of SOCS3, a core inhibitor of insulin and leptin signaling. Our results show that the hypothalamic IKKbeta/NF-kappaB program is a general neural mechanism for energy imbalance underlying obesity and suggest that suppressing hypothalamic IKKbeta/NF-kappaB may represent a strategy to combat obesity and related diseases.
Collapse
Affiliation(s)
- Xiaoqing Zhang
- Department of Physiology, University of Wisconsin-Madison, Madison, WI 53706
| | - Guo Zhang
- Department of Physiology, University of Wisconsin-Madison, Madison, WI 53706
| | - Hai Zhang
- Department of Physiology, University of Wisconsin-Madison, Madison, WI 53706
- Cellular & Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI 53706
| | - Michael Karin
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093
| | - Hua Bai
- Department of Physiology, University of Wisconsin-Madison, Madison, WI 53706
| | - Dongsheng Cai
- Department of Physiology, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|
863
|
Abstract
Maintenance of body temperature is achieved partly by modulating lipolysis by a network of complex regulatory mechanisms. Lipolysis is an integral part of the glycerolipid/free fatty acid (GL/FFA) cycle, which is the focus of this review, and we discuss the significance of this pathway in the regulation of many physiological processes besides thermogenesis. GL/FFA cycle is referred to as a "futile" cycle because it involves continuous formation and hydrolysis of GL with the release of heat, at the expense of ATP. However, we present evidence underscoring the "vital" cellular signaling roles of the GL/FFA cycle for many biological processes. Probably because of its importance in many cellular functions, GL/FFA cycling is under stringent control and is organized as several composite short substrate/product cycles where forward and backward reactions are catalyzed by separate enzymes. We believe that the renaissance of the GL/FFA cycle is timely, considering the emerging view that many of the neutral lipids are in fact key signaling molecules whose production is closely linked to GL/FFA cycling processes. The evidence supporting the view that alterations in GL/FFA cycling are involved in the pathogenesis of "fatal" conditions such as obesity, type 2 diabetes, and cancer is discussed. We also review the different enzymatic and transport steps that encompass the GL/FFA cycle leading to the generation of several metabolic signals possibly implicated in the regulation of biological processes ranging from energy homeostasis, insulin secretion and appetite control to aging and longevity. Finally, we present a perspective of the possible therapeutic implications of targeting this cycling.
Collapse
Affiliation(s)
- Marc Prentki
- Departments of Nutrition and Biochemistry, University of Montreal, Montreal Diabetes Research Center, CR-CHUM, Montreal, Quebec, Canada H1W 4A4.
| | | |
Collapse
|
864
|
|
865
|
Ono H, Pocai A, Wang Y, Sakoda H, Asano T, Backer JM, Schwartz GJ, Rossetti L. Activation of hypothalamic S6 kinase mediates diet-induced hepatic insulin resistance in rats. J Clin Invest 2008; 118:2959-68. [PMID: 18618016 DOI: 10.1172/jci34277] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2007] [Accepted: 05/28/2008] [Indexed: 01/08/2023] Open
Abstract
Prolonged activation of p70 S6 kinase (S6K) by insulin and nutrients leads to inhibition of insulin signaling via negative feedback input to the signaling factor IRS-1. Systemic deletion of S6K protects against diet-induced obesity and enhances insulin sensitivity in mice. Herein, we present evidence suggesting that hypothalamic S6K activation is involved in the pathogenesis of diet-induced hepatic insulin resistance. Extending previous findings that insulin suppresses hepatic glucose production (HGP) partly via its effect in the hypothalamus, we report that this effect was blunted by short-term high-fat diet (HFD) feeding, with concomitant suppression of insulin signaling and activation of S6K in the mediobasal hypothalamus (MBH). Constitutive activation of S6K in the MBH mimicked the effect of the HFD in normal chow-fed animals, while suppression of S6K by overexpression of dominant-negative S6K or dominant-negative raptor in the MBH restored the ability of MBH insulin to suppress HGP after HFD feeding. These results suggest that activation of hypothalamic S6K contributes to hepatic insulin resistance in response to short-term nutrient excess.
Collapse
Affiliation(s)
- Hiraku Ono
- Department of Medicine, Diabetes Research Center, Albert Einstein College of Medicine, New York, New York 10461, USA.
| | | | | | | | | | | | | | | |
Collapse
|
866
|
Abstract
The prevalence of obesity is steadily rising and has huge health and financial implications for society. Weight gain is due to an imbalance between dietary intake and energy expenditure and research has focused on trying to understand the complex pathways involved in controlling these aspects. This review highlights the key areas of research in the hypothalamic control of appetite. The hypothalamus consists of several nuclei that integrate peripheral signals, such as adiposity and caloric intake, to regulate important pathways within the CNS controlling food intake. The best characterized pathways are the orexigenic neuropeptide Y/Agouti-related protein and the anorexigenic pro-opiomelanocortin/cocaine- and amphetamine-related transcript neurons in the arcuate nucleus of the hypothalamus. These project from the arcuate nucleus to other key hypothalamic nuclei, such as the paraventricular, dorsomedial, ventromedial and lateral hypothalamic nuclei. There are also projections to and from the brainstem, cortical areas and reward pathways, all of which influence food intake. The challenge at present is to understand the complexity of these pathways and try to find ways of modulating them in order to find potential therapeutic targets.
Collapse
Affiliation(s)
- Katherine A Simpson
- a Department of Investigative Medicine, Imperial College, London W12 ONN, UK
| | - Niamh M Martin
- a Department of Investigative Medicine, Imperial College, London W12 ONN, UK
| | - Steve R Bloom
- b Department of Investigative Medicine, Imperial College, London W12 ONN, UK.
| |
Collapse
|
867
|
Lian J, Yan XH, Peng J, Jiang SW. The mammalian target of rapamycin pathway and its role in molecular nutrition regulation. Mol Nutr Food Res 2008; 52:393-9. [PMID: 18306429 DOI: 10.1002/mnfr.200700005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Mammalian target of rapamycin (mTOR) is a protein serine-threonine kinase that functions as a central element in signaling pathway involved in control of cell growth and proliferation. mTOR exists in at least two distinct multi-protein complexes, mTORC1 and mTORC2. mTOR kinase controls the translation machinery, in response to nutrients and growth factors, via activation of p70 ribosomal S6 kinase and inhibition of eukaryotic initiation factor-4E-binding protein. In this report, we review the mTOR signaling pathway and its interaction with food intake, insulin resistance, lifespan and adipogenic regulation during the molecular nutrition regulation.
Collapse
Affiliation(s)
- Jun Lian
- Laboratory of Animal Molecular Nutrition, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, P.R. China
| | | | | | | |
Collapse
|
868
|
Robertson SA, Leinninger GM, Myers MG. Molecular and neural mediators of leptin action. Physiol Behav 2008; 94:637-42. [PMID: 18501391 PMCID: PMC2516921 DOI: 10.1016/j.physbeh.2008.04.005] [Citation(s) in RCA: 128] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2008] [Accepted: 04/02/2008] [Indexed: 01/17/2023]
Abstract
The adipose tissue-derived hormone, leptin, acts via its receptor (LepRb) in the brain to regulate energy balance and neuroendocrine function. Parsing the biology of leptin requires understanding LepRb signaling and the roles for specific signaling pathways in neural and physiological leptin action. Since the leptin acts via a broadly distributed network of LepRb-expressing neurons, understanding the function of each of these LepRb neural populations will also be crucial. Here, we review the status of knowledge regarding the molecular mediators of leptin action and the neural substrate via which leptin acts to regulate physiologic processes.
Collapse
Affiliation(s)
- Scott A. Robertson
- Division of Metabolism, Endocrinology and Diabetes, Department of Medicine and Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gina M. Leinninger
- Division of Metabolism, Endocrinology and Diabetes, Department of Medicine and Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Martin G. Myers
- Division of Metabolism, Endocrinology and Diabetes, Department of Medicine and Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
869
|
Abstract
The central nervous system regulates food intake (FI) and body weight (BW), but the associated mechanisms remain to be elucidated. Here we report that central injections of lactate reduced FI and BW in rodents. Inhibition of central lactate metabolism to pyruvate with the lactate dehydrogenase inhibitor oxamate abolished the central effects of lactate on FI and BW. Conversely, central injections of pyruvate recapitulated the effects of lactate. Finally, inhibition of central lactate metabolism prevented the ability of circulating lactate to lower FI and BW. Together, the data indicate that activation of central lactate metabolism lowers FI and BW.
Collapse
Affiliation(s)
- Carol K L Lam
- Department of Physiology, University of Toronto, 101 College Street, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
870
|
Arsic D, Guerin PM. Nutrient content of diet affects the signaling activity of the insulin/target of rapamycin/p70 S6 kinase pathway in the African malaria mosquito Anopheles gambiae. JOURNAL OF INSECT PHYSIOLOGY 2008; 54:1226-1235. [PMID: 18634792 DOI: 10.1016/j.jinsphys.2008.06.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Revised: 06/17/2008] [Accepted: 06/18/2008] [Indexed: 05/26/2023]
Abstract
Regulation of female mosquito feeding and reproduction plays a central role in their disease-vector competence. In this study we show that Anopheles gambiae mosquitoes engorged on albumin, amino acid and saline meals the same way as on blood, whereas sucrose evoked a typical plant nectar feeding response. Among the artificial diets, only the albumin-containing ones allowed follicular development. The target of rapamycin (TOR)/p70 S6 kinase (S6K) pathway has been identified as an essential nutrient-sensing tool controlling egg development in mosquitoes under the control of regulating inputs from the insulin pathway. We assayed the early response of TOR, S6K, tuberous sclerosis (TSC2), insulin receptor (INR) and two insulin-like peptides (ILPs) by quantitative real-time PCR assessment of mRNA levels and immunoblotting of phosphorylated active TOR and S6K in An. gambiae ovary and brain 3 h after engorgement. We show that transcript levels of s6k and members of the insulin pathway are readily affected by nutrients (especially one ILP in the head) and that the TOR/S6K phosphorylation is able to react quickly to a meal to an extent which depends on the true nutritive value.
Collapse
Affiliation(s)
- Dany Arsic
- Laboratory of Animal Physiology, Institute of Biology, University of Neuchâtel, Emile-Argand 11, CP 158, 2009 Neuchâtel, Switzerland.
| | | |
Collapse
|
871
|
Cota D, Matter EK, Woods SC, Seeley RJ. The role of hypothalamic mammalian target of rapamycin complex 1 signaling in diet-induced obesity. J Neurosci 2008; 28:7202-8. [PMID: 18614690 PMCID: PMC2597379 DOI: 10.1523/jneurosci.1389-08.2008] [Citation(s) in RCA: 150] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2008] [Revised: 05/12/2008] [Accepted: 05/28/2008] [Indexed: 02/06/2023] Open
Abstract
The mammalian target of rapamycin (mTOR) kinase is a key regulator of several cellular functions, including cell growth and differentiation. Because hypothalamic mTOR complex 1 (mTORC1) signaling has been implicated as a target of leptin in the regulation of energy balance, we investigated its role in obesity-induced leptin resistance. In contrast to rats maintained on a low-fat (LF) diet for 3 weeks, rats maintained on a high-fat (HF)-diet had no anorexic response to intracerebroventricular leptin. Western blot analysis revealed that leptin was unable to modulate hypothalamic mTORC1 signaling in the HF group, whereas it significantly induced phosphorylation of both S6 kinase 1 (S6K1) and S6 ribosomal protein (S6) in the LF group. Similar to leptin, the cytokine ciliary neurotrophic factor (CNTF) induces hypophagia and increases signal transduction activator of transcription 3 phosphorylation. However, CNTF and its analog CNTF(Ax15) activate leptin-like pathways in the hypothalamus, even in leptin-resistant states, including diet-induced obesity. Intracerebroventricular CNTF(Ax15) decreased 24 h food intake and body weight in rats on HF or LF diets and increased the phosphorylation of hypothalamic S6K1 and S6 in a comparable way in both diets. Importantly, mice lacking the expression of S6K1 (S6K1(-/-)) did not respond to the anorectic action of either leptin or CNTF(Ax15), implying a crucial role for S6K1 in modulating the actions of these two cytokines. Finally, exposure to HF diet decreased mTORC1 signaling within the hypothalamus. Overall, these findings point strongly to the possibility that reduced hypothalamic mTORC1 signaling contributes to the development of hyperphagia, weight gain, and leptin resistance during diet-induced obesity.
Collapse
Affiliation(s)
- Daniela Cota
- Department of Psychiatry, University of Cincinnati, Genome Research Institute, Cincinnati, Ohio 45237, USA.
| | | | | | | |
Collapse
|
872
|
Cao R, Lee B, Cho HY, Saklayen S, Obrietan K. Photic regulation of the mTOR signaling pathway in the suprachiasmatic circadian clock. Mol Cell Neurosci 2008; 38:312-24. [PMID: 18468454 PMCID: PMC2590753 DOI: 10.1016/j.mcn.2008.03.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2007] [Revised: 02/18/2008] [Accepted: 03/13/2008] [Indexed: 01/02/2023] Open
Abstract
Here we analyzed the light-responsiveness of the mammalian target of rapamycin (mTOR) cascade, a key regulator of inducible translation, in the suprachiasmatic nuclei (SCN), the locus of the master circadian clock. Brief light exposure during the subjective night, but not during the subjective day, triggered rapid phosphorylation (a marker of catalytic activity) of the mTOR translation effectors p70 S6K, ribosomal S6 protein (S6) and 4E-BP1. In the absence of photic stimulation, marked S6 and 4E-BP1 phosphorylation was detected, indicating tonic mTOR activity in the SCN. Light stimulated the colocalized activation of p70 S6K and extracellular signal-regulated protein kinase (ERK), and pharmacological disruption of ERK signaling abolished light-induced mTOR activity, revealing that the MAPK cascade is an essential intermediate that couples light to mTOR. Together these data identify a light-responsive mTOR cascade in the SCN, and thus, raise the possibility that inducible translation contributes to the clock entrainment process.
Collapse
Affiliation(s)
- Ruifeng Cao
- Department of Neuroscience, Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | |
Collapse
|
873
|
Abstract
AMP-activated protein kinase is a key enzyme in the regulation of energy metabolism. Its activation has pleiotropic effects in multiple tissues, including increased fatty acid oxidation, glucose uptake and glycolysis, as well as the inhibition of fatty acid and glycogen synthesis and gluconeogenesis, and stimulation of mitochondrial biogenesis. Recently, the AMP-activated protein kinase (AMPK) has also emerged as a regulator of appetite, contributing to the control of energy metabolism at both cell and the whole body levels. Pharmacological and genetic activation or inhibition of hypothalamic AMPK lead to increased or reduced food intake, respectively. AMPK appears to play a role in hypothalamic glucose and nutrient sensing and numerous studies have suggested a role for AMPK in mediating the orexigenic or anorexigenic effects of various endogenous and exogenous substances.
Collapse
Affiliation(s)
- B Kola
- Centre for Endocrinology, Barts and London School of Medicine and Dentistry, University of London, London, UK.
| |
Collapse
|
874
|
|
875
|
Dixit VD. Adipose-immune interactions during obesity and caloric restriction: reciprocal mechanisms regulating immunity and health span. J Leukoc Biol 2008; 84:882-92. [PMID: 18579754 DOI: 10.1189/jlb.0108028] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Increasing evidence suggests a tight coupling of metabolic and immune systems. This cross-talk mediated by neuroendocrine peptides as well as numerous cytokines and chemokines is believed to be responsible for integrating energy balance to immune function. These neuroendocrine-immune interactions are heightened during the state of chronic positive energy balance, as seen during obesity, and negative energy balance caused by caloric restriction (CR). Emerging evidence suggests that obesity may be associated with an immunodeficient state and chronic inflammation, which contribute to an increased risk of premature death. The direct interactions between expanded leukocyte populations within the adipose tissue during obesity and an increased number of adipocytes within an aging lymphoid microenvironment may constitute an important adaptive or pathological response as a result of change in energy balance. In stark contrast to obesity, CR causes negative energy balance and robustly prolongs a healthy lifespan in all of the species studied to date. Therefore, the endogenous neuroendocrine-metabolic sensors elevated or suppressed as a result of changes in energy balance may offer an important mechanism in understanding the antiaging and potential immune-enhancing nature of CR. Ghrelin, one such sensor of negative energy balance, is reduced during obesity and increased by CR. Ghrelin also regulates immune function by reducing proinflammatory cytokines and promotes thymopoiesis during aging and thus, may be a new CR mimetic target. The identification of immune effects and molecular pathways used by such orexigenic metabolic factors could offer potentially novel approaches to enhance immunity and increase healthy lifespan.
Collapse
Affiliation(s)
- Vishwa Deep Dixit
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA.
| |
Collapse
|
876
|
White UA, Stewart WC, Mynatt RL, Stephens JM. Neuropoietin attenuates adipogenesis and induces insulin resistance in adipocytes. J Biol Chem 2008; 283:22505-12. [PMID: 18562323 DOI: 10.1074/jbc.m710462200] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Recent findings have implicated gp130 receptor ligands, particularly ciliary neurotrophic factor (CNTF), as potential anti-obesity therapeutics. Neuropoietin (NP) is a recently discovered cytokine in the gp130 family that shares functional and structural features with CNTF and signals via the CNTF receptor tripartite complex comprised of CNTFRalpha, LIF receptor, and gp130. NP plays a role in the development of the nervous system, but the effects of NP on adipocytes have not been previously examined. Because CNTF exerts anti-obesogenic effects in adipocytes and NP shares the same receptor complex, we investigated the effects of NP on adipocyte development and insulin action. Using cultured 3T3-L1 adipocytes, we observed that NP has the ability to block adipogenesis in a dose- and time-dependent manner. We also observed that cultured adipocytes, as well as murine adipose tissue, are highly responsive to acute NP treatment. Rodents injected with NP had a substantial increase in STAT3 tyrosine phosphorylation and ERK 1 and 2 activation. We also observed the induction of SOCS-3 mRNA in 3T3-L1 adipocytes following NP treatment. Unlike CNTF, our studies have revealed that NP also substantially attenuates insulin-stimulated glucose uptake in 3T3-L1 adipocytes. In addition, NP blocks insulin action in adipose tissue in vivo. These observations are supported by data demonstrating that NP impairs insulin signaling via decreased activation of both IRS-1 and Akt. In summary, we have observed that both adipocytes in vitro and in vivo are highly responsive to NP, and this cytokine has the ability to affect insulin signaling in fat cells. These novel observations suggest that NP, unlike CNTF, may not be a viable obesity therapeutic.
Collapse
Affiliation(s)
- Ursula A White
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana 70803, USA
| | | | | | | |
Collapse
|
877
|
Buettner C, Muse ED, Cheng A, Chen L, Scherer T, Pocai A, Su K, Cheng B, Li X, Harvey-White J, Schwartz GJ, Kunos G, Rossetti L, Buettner C. Leptin controls adipose tissue lipogenesis via central, STAT3-independent mechanisms. Nat Med 2008; 14:667-75. [PMID: 18516053 PMCID: PMC2671848 DOI: 10.1038/nm1775] [Citation(s) in RCA: 262] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2008] [Accepted: 04/18/2008] [Indexed: 12/14/2022]
Abstract
Leptin (encoded by Lep) controls body weight by regulating food intake and fuel partitioning. Obesity is characterized by leptin resistance and increased endocannabinoid tone. Here we show that leptin infused into the mediobasal hypothalamus (MBH) of rats inhibits white adipose tissue (WAT) lipogenesis, which occurs independently of signal transducer and activator of transcription-3 (STAT3) signaling. Correspondingly, transgenic inactivation of STAT3 signaling by mutation of the leptin receptor (s/s mice) leads to reduced adipose mass compared to db/db mice (complete abrogation of leptin receptor signaling). Conversely, the ability of hypothalamic leptin to suppress WAT lipogenesis in rats is lost when hypothalamic phosphoinositide 3-kinase signaling is prevented or when sympathetic denervation of adipose tissue is performed. MBH leptin suppresses the endocannabinoid anandamide in WAT, and, when this suppression of endocannabinoid tone is prevented by systemic CB1 receptor activation, MBH leptin fails to suppress WAT lipogenesis. These data suggest that the increased endocannabinoid tone observed in obesity is linked to a failure of central leptin signaling to restrain peripheral endocannabinoids.
Collapse
Affiliation(s)
- Christoph Buettner
- Department of Medicine, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1005, New York, New York 10029, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
878
|
Morgan TM, Pitts TE, Gross TS, Poliachik SL, Vessella RL, Corey E. RAD001 (Everolimus) inhibits growth of prostate cancer in the bone and the inhibitory effects are increased by combination with docetaxel and zoledronic acid. Prostate 2008; 68:861-71. [PMID: 18361409 PMCID: PMC3162313 DOI: 10.1002/pros.20752] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
INTRODUCTION mTOR activity is increased in advanced prostate cancer (CaP) as a result of a high rate of PTEN mutations. RAD001 (Everolimus) is a new orally available mTOR inhibitor. The objective of our study was to evaluate the effects of RAD001 on the growth of CaP in the bone, both alone and in combination with docetaxel and zoledronic acid. METHODS C4-2 CaP cells were injected into tibiae of mice and the animals were treated with RAD001, docetaxel, and zoledronic acid alone or in combination. Histomorphometrical analysis, serum PSA measurements, bone mineral density (BMD), and microCT were used to determine the effects of treatment on tumor and bone. RESULTS All three agents alone decreased tumor volume, and RAD001 and docetaxel also decreased levels of serum PSA by 68% and 65%, respectively (both P < 0.01). Combinations of the agents were more effective in decreasing tumor volume than single agents. Three-drug treatment showed the greatest effect: 64% inhibition versus control (P < 0.01). Treatment with RAD001 interfered with the weight loss associated with growth of this tumor in the bone (non-RAD001 groups: 4.0% decrease in body weight, P = 0.0014; RAD001 groups: increase of 3.6% in body weight, P = 0.0037). CONCLUSIONS RAD001 inhibited growth of C4-2 cells in bone, an effect augmented by addition of docetaxel and zoledronic acid. Moreover RAD001 had a significant impact on maintenance of body weight. RAD001 may hold promise for its effects on both metastatic CaP and the important syndrome of tumor cachexia.
Collapse
Affiliation(s)
- Todd M. Morgan
- Department of Urology, University of Washington School of Medicine, Seattle, Washington
| | - Tiffany E.M. Pitts
- Department of Urology, University of Washington School of Medicine, Seattle, Washington
| | - Ted S. Gross
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Sandra L. Poliachik
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Robert L. Vessella
- Department of Urology, University of Washington School of Medicine, Seattle, Washington
| | - Eva Corey
- Department of Urology, University of Washington School of Medicine, Seattle, Washington
| |
Collapse
|
879
|
Faipoux R, Tomé D, Gougis S, Darcel N, Fromentin G. Proteins activate satiety-related neuronal pathways in the brainstem and hypothalamus of rats. J Nutr 2008; 138:1172-8. [PMID: 18492852 DOI: 10.1093/jn/138.6.1172] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Our objective was to study the relationship between the satiety induced by high-protein meals and the activation of brain areas involved in the onset of satiety. In rats, we used immunohistochemistry to monitor brain centers activated by a meal by receiving information from the gastrointestinal tract or via humoral pathways. In the nucleus of the solitary tract (NTS), the acute or chronic intake of high-protein meals led to increased activation of the noradrenergic/adrenergic neurons involved in cholecystokinin-induced satiety. In the arcuate nucleus of the hypothalamus, the melanocortin pathway was also more strongly activated after the acute or chronic intake of high-protein meals. Moreover, the glucagon-like peptide 1 pathway arising from the NTS, which is triggered, among other behaviors, during nonphysiological anorexia, was not activated by high-protein meals, supporting the lack of aversive behavior associated with this diet. Taken together, these results show that the ability of high-protein meals to inhibit food intake occurs alongside the activation, in nutrient-sensitive brain areas, of several specific neuronal populations involved in satiety.
Collapse
Affiliation(s)
- Rodolphe Faipoux
- UMR914 Nutrition Physiology and Ingestive Behavior, INRA, AgroParisTech, CRNH-IdF, F-75005 Paris, France
| | | | | | | | | |
Collapse
|
880
|
Abstract
The adipose tissue-derived hormone leptin acts via its receptor (LRb) in the brain to regulate energy balance and neuroendocrine function. LRb signaling via STAT3 and a number of other pathways is required for the totality of leptin action. The failure of elevated leptin levels to suppress feeding and mediate weight loss in common forms of obesity defines a state of so-called leptin resistance. A number of mechanisms, including the leptin-stimulated phosphorylation of Tyr(985) on LRb and the suppressor of cytokine signaling 3, attenuate leptin signaling and promote a cellular leptin resistance in obesity. Several unique features of the arcuate nucleus of the hypothalamus may contribute to the severity of cellular leptin resistance in this region. Other mechanisms that govern feeding behavior and food reward may also underlie the inception of obesity.
Collapse
Affiliation(s)
- Martin G Myers
- Division of Metabolism, Endocrinology and Diabetes, Department of Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | | | | |
Collapse
|
881
|
Sandoval D, Cota D, Seeley RJ. The integrative role of CNS fuel-sensing mechanisms in energy balance and glucose regulation. Annu Rev Physiol 2008; 70:513-35. [PMID: 17988209 DOI: 10.1146/annurev.physiol.70.120806.095256] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The incidences of both obesity and type 2 diabetes mellitus are rising at epidemic proportions. Despite this, the balance between caloric intake and expenditure is tremendously accurate under most circumstances. Growing evidence suggests that nutrient and hormonal signals converge and directly act on brain centers, leading to changes in fuel metabolism and, thus, stable body weight over time. Growing evidence also suggests that these same signals act on the central nervous system (CNS) to regulate glucose metabolism independently. We propose that this is not coincidental and that the CNS responds to peripheral signals to orchestrate changes in both energy and glucose homeostasis. In this way the CNS ensures that the nutrient demands of peripheral tissues (and likely of the brain itself) are being met. Consequently, dysfunction of the ability of the CNS to integrate fuel-sensing signals may underlie the etiology of metabolic diseases such as obesity and diabetes.
Collapse
Affiliation(s)
- Darleen Sandoval
- Department of Psychiatry, Genome Research Institute, University of Cincinnati, Cincinnati, OH 45237, USA
| | | | | |
Collapse
|
882
|
Abstract
Food intake and energy expenditure are controlled by complex, redundant, and distributed neural systems that reflect the fundamental biological importance of adequate nutrient supply and energy balance. Much progress has been made in identifying the various hormonal and neural mechanisms by which the brain informs itself about availability of ingested and stored nutrients and, in turn, generates behavioral, autonomic, and endocrine output. While hypothalamus and caudal brainstem play crucial roles in this homeostatic function, areas in the cortex and limbic system are important for processing information regarding prior experience with food, reward, and emotion, as well as social and environmental context. Most vertebrates can store a considerable amount of energy as fat for later use, and this ability has now become one of the major health risks for many human populations. The predisposition to develop obesity can theoretically result from any pathological malfunction or lack of adaptation to changing environments of this highly complex system.
Collapse
Affiliation(s)
- Hans-Rudolf Berthoud
- Neurobiology of Nutrition Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA.
| | | |
Collapse
|
883
|
Upper intestinal lipids trigger a gut-brain-liver axis to regulate glucose production. Nature 2008; 452:1012-6. [PMID: 18401341 DOI: 10.1038/nature06852] [Citation(s) in RCA: 220] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Accepted: 02/22/2008] [Indexed: 12/21/2022]
Abstract
Energy and glucose homeostasis are regulated by food intake and liver glucose production, respectively. The upper intestine has a critical role in nutrient digestion and absorption. However, studies indicate that upper intestinal lipids inhibit food intake as well in rodents and humans by the activation of an intestine-brain axis. In parallel, a brain-liver axis has recently been proposed to detect blood lipids to inhibit glucose production in rodents. Thus, we tested the hypothesis that upper intestinal lipids activate an intestine-brain-liver neural axis to regulate glucose homeostasis. Here we demonstrate that direct administration of lipids into the upper intestine increased upper intestinal long-chain fatty acyl-coenzyme A (LCFA-CoA) levels and suppressed glucose production. Co-infusion of the acyl-CoA synthase inhibitor triacsin C or the anaesthetic tetracaine with duodenal lipids abolished the inhibition of glucose production, indicating that upper intestinal LCFA-CoAs regulate glucose production in the preabsorptive state. Subdiaphragmatic vagotomy or gut vagal deafferentation interrupts the neural connection between the gut and the brain, and blocks the ability of upper intestinal lipids to inhibit glucose production. Direct administration of the N-methyl-d-aspartate ion channel blocker MK-801 into the fourth ventricle or the nucleus of the solitary tract where gut sensory fibres terminate abolished the upper-intestinal-lipid-induced inhibition of glucose production. Finally, hepatic vagotomy negated the inhibitory effects of upper intestinal lipids on glucose production. These findings indicate that upper intestinal lipids activate an intestine-brain-liver neural axis to inhibit glucose production, and thereby reveal a previously unappreciated pathway that regulates glucose homeostasis.
Collapse
|
884
|
Yang X, Yang C, Farberman A, Rideout TC, de Lange CFM, France J, Fan MZ. The mammalian target of rapamycin-signaling pathway in regulating metabolism and growth1,2. J Anim Sci 2008; 86:E36-50. [DOI: 10.2527/jas.2007-0567] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
885
|
Zheng H, Berthoud HR. Neural Systems Controlling the Drive to Eat: Mind Versus Metabolism. Physiology (Bethesda) 2008; 23:75-83. [DOI: 10.1152/physiol.00047.2007] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
With the bleak outlook that 75% of Americans will be overweight or obese in 10 years, it is essential to find efficient help very soon. Knowledge of the powerful and complex neural systems conferring the basic drive to eat is a prerequisite for designing efficient therapies. Recent studies suggest that the cross talk between brain areas involved in cognitive, emotional, and metabolic-regulatory functions may explain why energy homeostasis breaks down for many predisposed individuals in our modern environment.
Collapse
Affiliation(s)
- Huiyuan Zheng
- Neurobiology of Nutrition Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Hans-Rudi Berthoud
- Neurobiology of Nutrition Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| |
Collapse
|
886
|
Wolfgang MJ, Lane MD. Hypothalamic malonyl-coenzyme A and the control of energy balance. Mol Endocrinol 2008; 22:2012-20. [PMID: 18356287 DOI: 10.1210/me.2007-0538] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
An intermediate in the fatty acid biosynthetic pathway, malonyl-coenzyme A (CoA), has emerged as a major regulator of energy homeostasis not only in peripheral metabolic tissues but also in regions of the central nervous system that control satiety and energy expenditure. Fluctuations in hypothalamic malonyl-CoA lead to changes in food intake and peripheral energy expenditure in a manner consistent with an anorexigenic signaling intermediate. Hypothalamic malonyl-CoA is regulated by nutritional and endocrine cues including glucose and leptin, respectively. That malonyl-CoA is an essential component in the energy homeostatic signaling system of the hypothalamus is supported by convergence of physiological, pharmacological, and genetic evidence. This review will focus on evidence implicating malonyl-CoA as a central player in the control of body weight and adiposity as well as clues to the molecular mechanism by which carbon flux through the fatty acid biosynthetic pathway is linked to the neural control of energy balance.
Collapse
Affiliation(s)
- Michael J Wolfgang
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, 512 WBSB, 725 North Wolfe Street, Baltimore, Maryland 21205, USA
| | | |
Collapse
|
887
|
Ropelle ER, Pauli JR, Fernandes MFA, Rocco SA, Marin RM, Morari J, Souza KK, Dias MM, Gomes-Marcondes MC, Gontijo JAR, Franchini KG, Velloso LA, Saad MJA, Carvalheira JBC. A central role for neuronal AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR) in high-protein diet-induced weight loss. Diabetes 2008; 57:594-605. [PMID: 18057094 DOI: 10.2337/db07-0573] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE A high-protein diet (HPD) is known to promote the reduction of body fat, but the mechanisms underlying this change are unclear. AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR) function as majors regulators of cellular metabolism that respond to changes in energy status, and recent data demonstrated that they also play a critical role in systemic energy balance. Here, we sought to determine whether the response of the AMPK and mTOR pathways could contribute to the molecular effects of an HPD. RESEARCH DESIGN AND METHODS Western blotting, confocal microscopy, chromatography, light microscopy, and RT-PCR assays were combined to explore the anorexigenic effects of an HPD. RESULTS An HPD reduced food intake and induced weight loss in both normal rats and ob/ob mice. The intracerebroventricular administration of leucine reduced food intake, and the magnitude of weight loss and reduction of food intake in a leucine-supplemented diet are similar to that achieved by HPD in normal rats and in ob/ob mice, suggesting that leucine is a major component of the effects of an HPD. Leucine and HPD decrease AMPK and increase mTOR activity in the hypothalamus, leading to inhibition of neuropeptide Y and stimulation of pro-opiomelanocortin expression. Consistent with a cross-regulation between AMPK and mTOR to control food intake, our data show that the activation of these enzymes occurs in the same specific neuronal subtypes. CONCLUSIONS These findings provide support for the hypothesis that AMPK and mTOR interact in the hypothalamus to regulate feeding during HPD in a leucine-dependent manner.
Collapse
Affiliation(s)
- Eduardo R Ropelle
- Department of Internal Medicine, State University of Campinas (UNICAMP), 13083-970, Campinas, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
888
|
Kohno D, Nakata M, Maejima Y, Shimizu H, Sedbazar U, Yoshida N, Dezaki K, Onaka T, Mori M, Yada T. Nesfatin-1 neurons in paraventricular and supraoptic nuclei of the rat hypothalamus coexpress oxytocin and vasopressin and are activated by refeeding. Endocrinology 2008; 149:1295-301. [PMID: 18048495 DOI: 10.1210/en.2007-1276] [Citation(s) in RCA: 205] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nesfatin-1, a newly discovered satiety molecule, is located in the hypothalamic nuclei, including the paraventricular nucleus (PVN) and supraoptic nucleus (SON). In this study, fine localization and regulation of nesfatin-1 neurons in the PVN and SON were investigated by immunohistochemistry of neuropeptides and c-Fos. In the PVN, 24% of nesfatin-1 neurons overlapped with oxytocin, 18% with vasopressin, 13% with CRH, and 12% with TRH neurons. In the SON, 35% of nesfatin-1 neurons overlapped with oxytocin and 28% with vasopressin. After a 48-h fast, refeeding for 2 h dramatically increased the number of nesfatin-1 neurons expressing c-Fos immunoreactivity by approximately 10 times in the PVN and 30 times in the SON, compared with the fasting controls. In the SON, refeeding also significantly increased the number of nesfatin-1-immunoreactive neurons and NUCB2 mRNA expression, compared with fasting. These results indicate that nesfatin-1 neurons in the PVN and SON highly overlap with oxytocin and vasopressin neurons and that they are activated markedly by refeeding. Feeding-activated nesfatin-1 neurons in the PVN and SON could play a role in the postprandial regulation of feeding behavior and energy homeostasis.
Collapse
Affiliation(s)
- Daisuke Kohno
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University, School of Medicine, Tochigi 329-0498, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
889
|
Atkinson TJ. Central and peripheral neuroendocrine peptides and signalling in appetite regulation: considerations for obesity pharmacotherapy. Obes Rev 2008; 9:108-20. [PMID: 18257752 DOI: 10.1111/j.1467-789x.2007.00412.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Appetite and satiety are mediated by complex neuroendocrine signalling pathways involving over 40 hormones, neuropeptides, enzymes, other chemical messengers and their receptors. Research efforts continue to expand understanding of the role of signalling molecules between central hypothalamic nuclei and peripheral enteroendocrine cells; and discoveries of novel networks and messengers provide new biological insights on how to manipulate appetite-satiety pathways. Despite the vast array of peptides that are potentially useful for anti-obesity drug development, only four classes of agents are approved: (i) catecholamine stimulants; (ii) serotonin and noradrenaline reuptake inhibitors; (iii) lipase inhibitors; and (iv) more recently cannabinoid-1 receptor antagonists. Clinical effects of these drugs confer modest improvements, and side effects negatively impact long-term treatment course. This paper suggests single target pharmacological interventions are possibly hampered by the myriad of alternate orexigenic peptidic signals that drive hyperphagia, hence a multiple target model or combination treatment approach is proposed to offer greater therapeutic potential in modulating appetite and managing weight.
Collapse
|
890
|
Abstract
A key phenotype associated with type 2 diabetes in humans is impaired mitochondrial oxidative metabolism in skeletal muscle, a pattern potentially contributing to increased lipid accumulation and impaired metabolic flexibility-in turn, central features of both insulin resistance and diabetes. Since thyroid hormone regulates mitochondrial gene expression and function in skeletal muscle, reductions in T3-mediated transcription may contribute to diabetes-related impairments in oxidative metabolism. We review the evidence for relationships between thyroid hormone action and diabetes risk, and discuss potential mechanisms linking intracellular thyroid hormone availability, thyroid receptor action, and the transcriptional coactivator PGC1 in regulating oxidative metabolism.
Collapse
Affiliation(s)
- Sarah Crunkhorn
- Research Division, Joslin Diabetes Center, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
891
|
Abstract
PURPOSE OF REVIEW Obesity has reached epidemic proportions throughout the world and poses significant health and economic burdens to both developed and developing societies. Most recent data from the NHANES study (2003-2004) report that 17.1% of US children are overweight and 32.2% of adults are obese, a significant increase compared with data obtained only 6 years earlier. RECENT FINDINGS The neurohormonal control of appetite, body composition, and glucose homeostasis is mediated by hormones secreted from adipose tissue, endocrine glands, and enteroendocrine cells, which converge at the vagus nerve, brainstem and hypothalamus to modulate complex interactions of neurotransmitters and central appetite-regulating peptides. These hormonal signals are tightly regulated to maintain body weight/adiposity within a narrow, individually defined range that may be further impacted by variables such as ingested calories, meal composition, and lifestyle. SUMMARY Clinical manifestations of obesity, the metabolic syndrome and impaired glucose tolerance reflect biochemical alterations in a complex hormonal milieu. Elucidation of these hormonal perturbations in obese patients has already provided novel pharmacologic treatments to improve weight management and address the metabolic sequelae of obesity. The remarkable redundancy of these hormones, however, and their interactions make a monopharmaceutical approach unlikely to be successful.
Collapse
Affiliation(s)
- Anne Lenz
- University of South Florida College of Medicine, Tampa, Florida, USA
| | | |
Collapse
|
892
|
Bibliography. Current world literature. Growth and development. Curr Opin Endocrinol Diabetes Obes 2008; 15:79-101. [PMID: 18185067 DOI: 10.1097/med.0b013e3282f4f084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
893
|
Cruzado JM. Nonimmunosuppressive effects of mammalian target of rapamycin inhibitors. Transplant Rev (Orlando) 2008; 22:73-81. [DOI: 10.1016/j.trre.2007.09.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
894
|
Leinninger GM, Myers MG. LRb signals act within a distributed network of leptin-responsive neurones to mediate leptin action. Acta Physiol (Oxf) 2008; 192:49-59. [PMID: 18171429 DOI: 10.1111/j.1748-1716.2007.01784.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The adipose tissue-derived hormone, leptin, acts via its receptor (LRb) in the brain to regulate energy balance and neuroendocrine function. In order to understand leptin action we have explored the physiological function of LRb signalling pathways, defining important roles for signal transducer and activator of transcription-3 (STAT3) in positive signalling and for LRbTyr(985)-mediated feedback inhibition in leptin signal attenuation. As the cells on which leptin acts are not homogeneous, but rather represent a broadly distributed network of neurones with divergent projections and functions, it is also crucial to consider how each of these populations responds to LRb signals to contribute to leptin action. While well-known LRb-expressing neurones within the arcuate nucleus of the hypothalamus mediate crucial effects on satiety and energy expenditure, other populations of LRb-expressing neurones in the ventral tegmental area and elsewhere likely control the mesolimbic dopamine system. Additional populations of LRb-expressing neurones likely contribute to other aspects of neuroendocrine regulation. It will be important to define the molecular mechanisms by which leptin acts to regulate neurophysiology in each of these LRb-expressing neural populations in order to understand the totality of leptin action.
Collapse
Affiliation(s)
- G M Leinninger
- Division of Metabolism, Endocrinology and Diabetes, Department of Medicine, University of Michigan Medical School, Ann Arbor, MI 48109-0678, USA
| | | |
Collapse
|
895
|
Swiech L, Perycz M, Malik A, Jaworski J. Role of mTOR in physiology and pathology of the nervous system. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2008; 1784:116-32. [PMID: 17913600 DOI: 10.1016/j.bbapap.2007.08.015] [Citation(s) in RCA: 269] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2007] [Revised: 08/09/2007] [Accepted: 08/10/2007] [Indexed: 01/04/2023]
Abstract
Mammalian target of rapamycin (mTOR) is a serine-threonine protein kinase that regulates several intracellular processes in response to extracellular signals, nutrient availability, energy status of the cell and stress. mTOR regulates survival, differentiation and development of neurons. Axon growth and navigation, dendritic arborization, as well as synaptogenesis, depend on proper mTOR activity. In adult brain mTOR is crucial for synaptic plasticity, learning and memory formation, and brain control of food uptake. Recent studies reveal that mTOR activity is modified in various pathologic states of the nervous system, including brain tumors, tuberous sclerosis, cortical displasia and neurodegenerative disorders such as Alzheimer's, Parkinson's and Huntington's diseases. This review presents current knowledge about the role of mTOR in the physiology and pathology of the nervous system, with special focus on molecular targets acting downstream of mTOR that potentially contribute to neuronal development, plasticity and neuropathology.
Collapse
Affiliation(s)
- Lukasz Swiech
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| | | | | | | |
Collapse
|
896
|
Abstract
The mammalian target of rapamycin (mTOR) has drawn much attention recently because of its essential role in cell growth control and its involvement in human tumorigenesis. Great endeavors have been made to elucidate the functions and regulation of mTOR in the past decade. The current prevailing view is that mTOR regulates many fundamental biological processes, such as cell growth and survival, by integrating both intracellular and extracellular signals, including growth factors, nutrients, energy levels, and cellular stress. The significance of mTOR has been highlighted most recently by the identification of mTOR-associated proteins. Amazingly, when bound to different proteins, mTOR forms distinctive complexes with very different physiological functions. These findings not only expand the roles that mTOR plays in cells but also further complicate the regulation network. Thus, it is now even more critical that we precisely understand the underlying molecular mechanisms in order to directly guide the development and usage of anti-cancer drugs targeting the mTOR signaling pathway. In this review, we will discuss different mTOR-associated proteins, the regulation of mTOR complexes, and the consequences of mTOR dysregulation under pathophysiological conditions.
Collapse
Affiliation(s)
- Qian Yang
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
897
|
Hermanussen M, Tresguerres JAF. Overweight, appetite control, and the role of glutamate and excess nutritional protein during child development. ACTA ACUST UNITED AC 2007. [DOI: 10.1002/huon.200700004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
898
|
She P, Van Horn C, Reid T, Hutson SM, Cooney RN, Lynch CJ. Obesity-related elevations in plasma leucine are associated with alterations in enzymes involved in branched-chain amino acid metabolism. Am J Physiol Endocrinol Metab 2007; 293:E1552-63. [PMID: 17925455 PMCID: PMC2767201 DOI: 10.1152/ajpendo.00134.2007] [Citation(s) in RCA: 389] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Elevations in branched-chain amino acids (BCAAs) in human obesity were first reported in the 1960s. Such reports are of interest because of the emerging role of BCAAs as potential regulators of satiety, leptin, glucose, cell signaling, adiposity, and body weight (mTOR and PKC). To explore loss of catabolic capacity as a potential contributor to the obesity-related rises in BCAAs, we assessed the first two enzymatic steps, catalyzed by mitochondrial branched chain amino acid aminotransferase (BCATm) or the branched chain alpha-keto acid dehydrogenase (BCKD E1alpha subunit) complex, in two rodent models of obesity (ob/ob mice and Zucker rats) and after surgical weight loss intervention in humans. Obese rodents exhibited hyperaminoacidemia including BCAAs. Whereas no obesity-related changes were observed in rodent skeletal muscle BCATm, pS293, or total BCKD E1alpha or BCKD kinase, in liver BCKD E1alpha was either unaltered or diminished by obesity, and pS293 (associated with the inactive state of BCKD) increased, along with BCKD kinase. In epididymal fat, obesity-related declines were observed in BCATm and BCKD E1alpha. Plasma BCAAs were diminished by an overnight fast coinciding with dissipation of the changes in adipose tissue but not in liver. BCAAs also were reduced by surgical weight loss intervention (Roux-en-Y gastric bypass) in human subjects studied longitudinally. These changes coincided with increased BCATm and BCKD E1alpha in omental and subcutaneous fat. Our results are consistent with the idea that tissue-specific alterations in BCAA metabolism, in liver and adipose tissue but not in muscle, may contribute to the rise in plasma BCAAs in obesity.
Collapse
Affiliation(s)
- Pengxiang She
- Department of Cellular and Molecular Physiology, College of Medicine, the Pennsylvania State University, Hershey, PA 17033
| | - Cynthia Van Horn
- Department of Biochemistry and Molecular Biology, Nutrition Research Center, Wake Forest University Health Sciences, Medical Center Blvd, Winston-Salem, NC 27157
| | - Tanya Reid
- Department of Biochemistry and Molecular Biology, Nutrition Research Center, Wake Forest University Health Sciences, Medical Center Blvd, Winston-Salem, NC 27157
| | - Susan M. Hutson
- Department of Biochemistry and Molecular Biology, Nutrition Research Center, Wake Forest University Health Sciences, Medical Center Blvd, Winston-Salem, NC 27157
| | - Robert N. Cooney
- Department of Cellular and Molecular Physiology, College of Medicine, the Pennsylvania State University, Hershey, PA 17033
- Department of Surgery, College of Medicine, the Pennsylvania State University, Hershey, PA 17033
| | - Christopher J. Lynch
- Department of Cellular and Molecular Physiology, College of Medicine, the Pennsylvania State University, Hershey, PA 17033
| |
Collapse
|
899
|
Yutzy B, Holznagel E, Coulibaly C, Stuke A, Hahmann U, Deslys JP, Hunsmann G, Löwer J. Time-course studies of 14-3-3 protein isoforms in cerebrospinal fluid and brain of primates after oral or intracerebral infection with bovine spongiform encephalopathy agent. J Gen Virol 2007; 88:3469-3478. [DOI: 10.1099/vir.0.83128-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Experimental transmission of bovine spongiform encephalopathy (BSE) to cynomolgus monkeys (Macaca fascicularis) is an animal model for variant Creutzfeldt–Jakob disease (vCJD). The presence of 14-3-3 proteins in cerebrospinal fluid (CSF) samples indicates neuronal destruction and is therefore used as a clinical biomarker. However, time-course studies using 14-3-3 proteins have not been performed until now in simian vCJD. The main goals of this study were to determine isoform patterns, to examine kinetics and to correlate the clinical course with the occurrence of this biomarker in simian vCJD. In monkeys dosed intracerebrally with BSE, the earliest clinical sign of illness was a drop in body weight that was detected months before the onset of mild neurological signs. Macaques dosed orally or intracerebrally with BSE developed neurological signs 4.3 (3.7–4.6) and 4.8 (2.9–6.0) years post-infection, respectively. 14-3-3β- and -γ-positive CSF samples were found around the time of onset of mild neurological signs, but not earlier. In contrast, 14-3-3ϵ and -ηisoforms were not detectable. 14-3-3 levels increased with time and were positively correlated with the degree of neurological symptoms. Post-mortem examination of brain samples revealed a positive correlation between PrPresand 14-3-3ϵ levels. Interestingly, florid plaques characteristic of human vCJD could not be detected in diseased monkeys. It was concluded that analysis of 14-3-3 proteins in CSF is a reliable tool to characterize the time course of brain degeneration in simian vCJD. However, there are differences in the clinical course between orally and intracerebrally infected animals that may influence the detection of other biomarkers.
Collapse
Affiliation(s)
| | | | | | - Andreas Stuke
- Department of Virology and Immunology, German Primate Centre, Göttingen, Germany
| | - Uwe Hahmann
- Department of Virology and Immunology, German Primate Centre, Göttingen, Germany
| | | | - Gerhard Hunsmann
- Department of Virology and Immunology, German Primate Centre, Göttingen, Germany
| | | |
Collapse
|
900
|
Maya-Monteiro CM, Almeida PE, D'Avila H, Martins AS, Rezende AP, Castro-Faria-Neto H, Bozza PT. Leptin induces macrophage lipid body formation by a phosphatidylinositol 3-kinase- and mammalian target of rapamycin-dependent mechanism. J Biol Chem 2007; 283:2203-10. [PMID: 18039669 DOI: 10.1074/jbc.m706706200] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Leptin is an adipocyte-derived hormone/cytokine that links nutritional status with neuroendocrine and immune functions. Lipid bodies (lipid droplets) are emerging as dynamic organelles with roles in lipid metabolism and inflammation. Here we investigated the roles of leptin in signaling pathways involved in cytoplasmic lipid body biogenesis and leukotriene B(4) synthesis in macrophages. Our results demonstrated that leptin directly activated macrophages and induced the formation of adipose differentiation-related protein-enriched lipid bodies. Newly formed lipid bodies were sites of 5-lipoxygenase localization and correlated with an enhanced capacity of leukotriene B(4) production. We demonstrated that leptin-induced macrophage activation was dependent on phosphatidylinositol 3-kinase (PI3K) activity, since the lipid body formation was inhibited by LY294002 and was absent in the PI3K knock-out mice. Leptin induces phosphorylation of p70(S6K) and 4EBP1 key downstream signaling intermediates of the mammalian target of rapamycin (mTOR) pathway in a rapamycin-sensitive mechanism. The mTOR inhibitor, rapamycin, inhibited leptin-induced lipid body formation, both in vivo and in vitro. In addition, rapamycin inhibited leptin-induced adipose differentiation-related protein accumulation in macrophages and lipid body-dependent leukotriene synthesis, demonstrating a key role for mTOR in lipid body biogenesis and function. Our results establish PI3K/mTOR as an important signaling pathway for leptin-induced cytoplasmic lipid body biogenesis and adipose differentiation-related protein accumulation. Furthermore, we demonstrate a previously unrecognized link between intracellular (mTOR) and systemic (leptin) nutrient sensors in macrophage lipid metabolism. Leptin-induced increased formation of cytoplasmic lipid bodies and enhanced inflammatory mediator production in macrophages may have implications for obesity-related cardiovascular diseases.
Collapse
Affiliation(s)
- Clarissa M Maya-Monteiro
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Av. Brasil 4365, Manguinhos, Rio de Janeiro, RJ21045-900 Brazil.
| | | | | | | | | | | | | |
Collapse
|