1
|
Simon-Szabó L, Lizák B, Sturm G, Somogyi A, Takács I, Németh Z. Molecular Aspects in the Development of Type 2 Diabetes and Possible Preventive and Complementary Therapies. Int J Mol Sci 2024; 25:9113. [PMID: 39201799 PMCID: PMC11354764 DOI: 10.3390/ijms25169113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/17/2024] [Accepted: 08/18/2024] [Indexed: 09/03/2024] Open
Abstract
The incidence of diabetes, including type 2 diabetes (T2DM), is increasing sharply worldwide. To reverse this, more effective approaches in prevention and treatment are needed. In our review, we sought to summarize normal insulin action and the pathways that primarily influence the development of T2DM. Normal insulin action involves mitogenic and metabolic pathways, as both are important in normal metabolic processes, regeneration, etc. However, through excess energy, both can be hyperactive or attenuated/inactive leading to disturbances in the cellular and systemic regulation with the consequence of cellular stress and systemic inflammation. In this review, we detailed the beneficial molecular changes caused by some important components of nutrition and by exercise, which act in the same molecular targets as the developed drugs, and can revert the damaged pathways. Moreover, these induce entire networks of regulatory mechanisms and proteins to restore unbalanced homeostasis, proving their effectiveness as preventive and complementary therapies. These are the main steps for success in prevention and treatment of developed diseases to rid the body of excess energy, both from stored fats and from overnutrition, while facilitating fat burning with adequate, regular exercise in healthy people, and together with necessary drug treatment as required in patients with insulin resistance and T2DM.
Collapse
Affiliation(s)
- Laura Simon-Szabó
- Department of Molecular Biology, Semmelweis University, Tuzolto u. 37-47, 1094 Budapest, Hungary; (L.S.-S.); (B.L.)
| | - Beáta Lizák
- Department of Molecular Biology, Semmelweis University, Tuzolto u. 37-47, 1094 Budapest, Hungary; (L.S.-S.); (B.L.)
| | - Gábor Sturm
- Directorate of Information Technology Basic Infrastructure and Advanced Applications, Semmelweis University, Üllői út 78/b, 1082 Budapest, Hungary;
| | - Anikó Somogyi
- Department of Internal Medicine and Hematology, Semmelweis University, Baross u., 1085 Budapest, Hungary;
| | - István Takács
- Department of Internal Medicine and Oncology, Semmelweis University, Koranyi S. u 2/a, 1083 Budapest, Hungary;
| | - Zsuzsanna Németh
- Department of Internal Medicine and Oncology, Semmelweis University, Koranyi S. u 2/a, 1083 Budapest, Hungary;
| |
Collapse
|
2
|
Szalanczy AM, Key CCC, Woods LCS. Genetic variation in satiety signaling and hypothalamic inflammation: merging fields for the study of obesity. J Nutr Biochem 2022; 101:108928. [PMID: 34936921 PMCID: PMC8959400 DOI: 10.1016/j.jnutbio.2021.108928] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 11/08/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023]
Abstract
Although obesity has been a longstanding health crisis, the genetic architecture of the disease remains poorly understood. Genome-wide association studies have identified many genomic loci associated with obesity, with genes being enriched in the brain, particularly in the hypothalamus. This points to the role of the central nervous system (CNS) in predisposition to obesity, and we emphasize here several key genes along the satiety signaling pathway involved in genetic susceptibility. Interest has also risen regarding the chronic, low-grade obesity-associated inflammation, with a growing concern toward inflammation in the hypothalamus as a precursor to obesity. Recent studies have found that genetic variation in inflammatory genes play a role in obesity susceptibility, and we highlight here several key genes. Despite the interest in the genetic variants of these pathways individually, there is a lack of research that investigates the relationship between the two. Understanding the interplay between genetic variation in obesity genes enriched in the CNS and inflammation genes will advance our understanding of obesity etiology and heterogeneity, improve genetic risk prediction analyses, and highlight new drug targets for the treatment of obesity. Additionally, this increased knowledge will assist in physician's ability to develop personalized nutrition and medication strategies for combating the obesity epidemic. Though it often seems to present universally, obesity is a highly individual disease, and there remains a need in the field to develop methods to treat at the individual level.
Collapse
|
3
|
Ullah R, Rauf N, Nabi G, Yi S, Yu-Dong Z, Fu J. Mechanistic insight into high-fat diet-induced metabolic inflammation in the arcuate nucleus of the hypothalamus. Biomed Pharmacother 2021; 142:112012. [PMID: 34388531 DOI: 10.1016/j.biopha.2021.112012] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/31/2021] [Accepted: 08/03/2021] [Indexed: 12/16/2022] Open
Abstract
A high-fat diet (HFD) is linked with cytokines production by non-neuronal cells within the hypothalamus, which mediates metabolic inflammation. These cytokines then activate different inflammatory mediators in the arcuate nucleus of the hypothalamus (ARC), a primary hypothalamic area accommodating proopiomelanocortin (POMC) and agouti-related peptide (AGRP) neurons, first-order neurons that sense and integrate peripheral metabolic signals and then respond accordingly. These mediators, such as inhibitor of κB kinase-β (IKKβ), suppression of cytokine signaling 3 (SOCS3), c-Jun N-terminal kinases (JNKs), protein kinase C (PKC), etc., cause insulin and leptin resistance in POMC and AGRP neurons and support obesity and related metabolic complications. On the other hand, inhibition of these mediators has been shown to counteract the impaired metabolism. Therefore, it is important to discuss the contribution of neuronal and non-neuronal cells in HFD-induced hypothalamic inflammation. Furthermore, understanding few other questions, such as the diets causing hypothalamic inflammation, the gender disparity in response to HFD feeding, and how hypothalamic inflammation affects ARC neurons to cause impaired metabolism, will be helpful for the development of therapeutic approaches to prevent or treat HFD-induced obesity.
Collapse
Affiliation(s)
- Rahim Ullah
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China; Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Naveed Rauf
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China
| | - Ghulam Nabi
- Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei Province, China; Department of Life Sciences, School of Science, University of Management and Technology (UMT), Lahore, Pakistan
| | - Shen Yi
- Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China.
| | - Zhou Yu-Dong
- Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China.
| | - Junfen Fu
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310052, China; National Clinical Research Center for Child Health, Hangzhou 310052, China; National Children's Regional Medical Center, Hangzhou 310052, China.
| |
Collapse
|
4
|
Ferrer B, Prince LM, Tinkov AA, Santamaria A, Bowman AB, Aschner M. Chronic exposure to methylmercury disrupts ghrelin actions in C57BL/6J mice. Food Chem Toxicol 2020; 147:111918. [PMID: 33301842 DOI: 10.1016/j.fct.2020.111918] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 01/06/2023]
Abstract
Methylmercury (MeHg) is a neurotoxic pollutant widely present in the environment. Initial symptoms of MeHg may include loss of body weight. However, the mechanisms by which MeHg induces body weight changes have yet to be fully elucidated. Body weight is regulated by multiple mechanisms. Whereas multiple peripheral peptides lead to food intake cessation, ghrelin is the only recognized peripheral hormone that stimulates food intake. It exerts its action on Neuropeptide Y/Agouti-related peptide neurons in the hypothalamus. To test if MeHg affects ghrelin signaling C57BL/6J mice (males and females) were exposed to 5 ppm MeHg via drinking water during a month. On days 15 and 30 of MeHg exposure ghrelin was administered intraperitoneally and changes in body weight and food intake were recorded. In addition, changes in ghrelin-induced signaling pathways in hypothalamus were also analyzed. Here, we show that in males, MeHg enhanced ghrelin-induced body weight gain by activating the AMP-activated Kinase (AMPK)/Uncoupled protein 2 (UCP2) signaling pathway. In contrast, in females, MeHg inhibited ghrelin-induced mTOR signaling activation and decreased Npy mRNA expression, thus mitigating the ghrelin-induced weight gain. Combined, our novel results demonstrate, for the first time, that MeHg disrupts the physiological functions of ghrelin differently in males and females.
Collapse
Affiliation(s)
- Beatriz Ferrer
- Department of Molecular Pharmacology, Neuroscience, and Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, 10461, Bronx, NY, USA.
| | - Lisa M Prince
- School of Health Sciences, Purdue University, West Lafayette, IN, USA.
| | - Alexey A Tinkov
- IM Sechenov First Moscow State Medical University, Moscow, Russia; Institute of Cellular and Intracellular Symbiosis, Russian Academy of Sciences, Orenburg, Russia; Yaroslavl State University, Yaroslavl, Russia; Federal Research Centre of Biological Systems and Agro-technologies of the Russian Academy of Sciences, Orenburg, Russia.
| | - Abel Santamaria
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía, 14269, Mexico City, Mexico.
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, USA.
| | - Michael Aschner
- Department of Molecular Pharmacology, Neuroscience, and Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, 10461, Bronx, NY, USA; IM Sechenov First Moscow State Medical University, Moscow, Russia; Institute of Cellular and Intracellular Symbiosis, Russian Academy of Sciences, Orenburg, Russia.
| |
Collapse
|
5
|
Contreras-Chavez GG, Estrada JA, Contreras I. Changes in Appetite Regulation-Related Signaling Pathways in the Brain of Mice Supplemented with Non-nutritive Sweeteners. J Mol Neurosci 2020; 71:1144-1155. [PMID: 33128194 DOI: 10.1007/s12031-020-01737-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/19/2020] [Indexed: 10/23/2022]
Abstract
Non-nutritive sweeteners (NNSs) are commonly used to prevent weight gain and development of metabolic diseases associated with consumption of high-energy diets. Recent studies have demonstrated that these compounds may have unwanted detrimental effects under specific circumstances in vivo. In particular, an association between NNS consumption and changes in signaling pathways involved in the hunger-satiety system in the brain has been reported. Nonetheless, the extent of alterations in brain signaling pathways associated with consumption of these compounds has not been determined. The objective of this study was to determine the effect of frequent consumption of NNSs on the expression of proteins involved in signaling pathways related to appetite control in the brain in vivo. Eight-week-old BALB/c mice were supplemented with sucrose, sucralose, or steviol glycosides in their daily drinking water for 6 weeks. Subsequently, total brain protein extracts were used to analyze the expression of total and phosphorylated JAK2, STAT5, ERK 1/2, JNK, as well as SHP-2 and POMC, by western blot. Serum concentrations of leptin and α-MSH were quantified by ELISA. Results show that consumption of NNSs promotes significant changes in these signaling pathways, reducing the expression of pSTAT5/STAT5, pERK 1/2, SHP-2, and pJNK/JNK in male mice supplemented with steviol glycosides. Furthermore, consumption of steviol glycosides induced a decrease of α-MSH in male mice. In contrast, steviol glycosides induced overexpression of pSTAT5, pERK, and SHP-2 in females. These data suggest that chronic consumption of NNSs promotes sex-specific changes in signaling pathways related to the central hunger-satiety system in vivo.
Collapse
Affiliation(s)
- Gerson G Contreras-Chavez
- Laboratorio de Neuroquímica, Facultad de Medicina, Universidad Autónoma del Estado de México, Paseo Tollocan S/N Esq. Jesús Carranza, Colonia Moderna de La Cruz, Estado de México, 50180, Toluca, Mexico
| | - José A Estrada
- Laboratorio de Neuroquímica, Facultad de Medicina, Universidad Autónoma del Estado de México, Paseo Tollocan S/N Esq. Jesús Carranza, Colonia Moderna de La Cruz, Estado de México, 50180, Toluca, Mexico
| | - Irazú Contreras
- Laboratorio de Neuroquímica, Facultad de Medicina, Universidad Autónoma del Estado de México, Paseo Tollocan S/N Esq. Jesús Carranza, Colonia Moderna de La Cruz, Estado de México, 50180, Toluca, Mexico.
| |
Collapse
|
6
|
Podyma B, Johnson DA, Sipe L, Remcho TP, Battin K, Liu Y, Yoon SO, Deppmann CD, Güler AD. The p75 neurotrophin receptor in AgRP neurons is necessary for homeostatic feeding and food anticipation. eLife 2020; 9:e52623. [PMID: 31995032 PMCID: PMC7056271 DOI: 10.7554/elife.52623] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/27/2020] [Indexed: 12/31/2022] Open
Abstract
Networks of neurons control feeding and activity patterns by integrating internal metabolic signals of energy balance with external environmental cues such as time-of-day. Proper circadian alignment of feeding behavior is necessary to prevent metabolic disease, and thus it is imperative that molecular players that maintain neuronal coordination of energy homeostasis are identified. Here, we demonstrate that mice lacking the p75 neurotrophin receptor, p75NTR, decrease their feeding and food anticipatory behavior (FAA) in response to daytime, but not nighttime, restricted feeding. These effects lead to increased weight loss, but do not require p75NTR during development. Instead, p75NTR is required for fasting-induced activation of neurons within the arcuate hypothalamus. Indeed, p75NTR specifically in AgRP neurons is required for FAA in response to daytime restricted feeding. These findings establish p75NTR as a novel regulator gating behavioral response to food scarcity and time-of-day dependence of circadian food anticipation.
Collapse
Affiliation(s)
- Brandon Podyma
- Department of BiologyUniversity of VirginiaCharlottesvilleUnited States
| | - Dove-Anna Johnson
- Department of BiologyUniversity of VirginiaCharlottesvilleUnited States
| | - Laura Sipe
- Department of BiologyUniversity of VirginiaCharlottesvilleUnited States
| | | | - Katherine Battin
- Department of BiologyUniversity of VirginiaCharlottesvilleUnited States
| | - Yuxi Liu
- Department of Biological Chemistry and PharmacologyThe Ohio State University College of MedicineColumbusUnited States
| | - Sung Ok Yoon
- Department of Biological Chemistry and PharmacologyThe Ohio State University College of MedicineColumbusUnited States
| | | | - Ali Deniz Güler
- Department of BiologyUniversity of VirginiaCharlottesvilleUnited States
| |
Collapse
|
7
|
El-Badawy RE, Ibrahim KA, Hassan NS, El-Sayed WM. Pterocarpus santalinus ameliorates streptozotocin-induced diabetes mellitus via anti-inflammatory pathways and enhancement of insulin function. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019; 22:932-939. [PMID: 31579450 PMCID: PMC6760478 DOI: 10.22038/ijbms.2019.34998.8325] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 02/13/2019] [Indexed: 11/17/2022]
Abstract
OBJECTIVES Morbidity and mortality due to diabetes mellitus (DM) result in exorbitant psycho-economical costs, so there is a strong need to create new strategies and drugs for controlling DM. The aim of the current study was to investigate the anti-diabetic effect of the aqueous extract of Pterocarpus santalinus on streptozotocin (STZ)-induced DM as compared to glustin. MATERIALS AND METHODS Thirty male rats were divided into five groups of six rats each as follows: control; the second group, received the aqueous plant extract (250 mg/kg) orally and daily for three weeks; the third group, was intraperitoneally injected with a single dose of 65 mg/kg of STZ and sacrificed after four weeks; the fourth and fifth groups, were injected with STZ, then after one week these were treated orally with either plant extract or with 3 mg/kg of glustin for three weeks, then sacrificed. RESULTS HPLC analysis of the plant aqueous extract showed that it contains many polyphenols and flavonoids. Treatment with STZ resulted in significant reductions in body weight, insulin level, and the expression of Fetuin-A and IRS-1. It also caused significant elevations in glucose, HOMA-IR, glycated hemoglobin, urea, and the expression of JNK and SIRT-1. STZ also caused an extensive β-cell degranulation and decreased cellular density. The aqueous extract of red sandalwood was able to abrogate the deleterious effects caused by STZ and improved the histological architecture of pancreas. CONCLUSION The aqueous extract of P. santalinus ameliorates diabetes mellitus via anti-inflammatory pathways and enhancement of insulin function.
Collapse
Affiliation(s)
| | - Khairy A Ibrahim
- Mammalian Toxicology Department, Central Agriculture Pesticides Lab, Agriculture Research Center, Dokki, Giza, 12618, Egypt
| | - Nahla S Hassan
- University of Ain Shams, Faculty of Science, Department of Biochemistry, Cairo, Egypt
| | - Wael M El-Sayed
- University of Ain Shams, Faculty of Science, Department of Zoology, Abbassia 11566, Cairo, Egypt
| |
Collapse
|
8
|
LPS-Induced Low-Grade Inflammation Increases Hypothalamic JNK Expression and Causes Central Insulin Resistance Irrespective of Body Weight Changes. Int J Mol Sci 2017; 18:ijms18071431. [PMID: 28677618 PMCID: PMC5535922 DOI: 10.3390/ijms18071431] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 06/22/2017] [Accepted: 06/27/2017] [Indexed: 01/26/2023] Open
Abstract
Metabolic endotoxemia contributes to low-grade inflammation in obesity, which causes insulin resistance due to the activation of intracellular proinflammatory pathways, such as the c-Jun N-terminal Kinase (JNK) cascade in the hypothalamus and other tissues. However, it remains unclear whether the proinflammatory process precedes insulin resistance or it appears because of the development of obesity. Hypothalamic low-grade inflammation was induced by prolonged lipopolysaccharide (LPS) exposure to investigate if central insulin resistance is induced by an inflammatory stimulus regardless of obesity. Male Wistar rats were treated with single (1 LPS) or repeated injections (6 LPS) of LPS (100 μg/kg, IP) to evaluate the phosphorylation of the insulin receptor substrate-1 (IRS1), Protein kinase B (AKT), and JNK in the hypothalamus. Single LPS increased the expression of pIRS1, pAKT, and pJNK, whereas the repeated LPS treatment failed to recruit pIRS1 and pAKT. The 6 LPS treated rats showed increased total JNK and pJNK. The 6 LPS rats became unresponsive to the hypophagic effect induced by central insulin administration (12 μM/5 μL, ICV). Prolonged exposure to LPS (24 h) impaired the insulin-induced AKT phosphorylation and the translocation of the transcription factor forkhead box protein O1 (FoxO1) from the nucleus to the cytoplasm of the cultured hypothalamic GT1-7 cells. Central administration of the JNK inhibitor (20 μM/5 μL, ICV) restored the ability of insulin to phosphorylate IRS1 and AKT in 6 LPS rats. The present data suggest that an increased JNK activity in the hypothalamus underlies the development of insulin resistance during prolonged exposure to endotoxins. Our study reveals that weight gain is not mandatory for the development of hypothalamic insulin resistance and the blockade of proinflammatory pathways could be useful for restoring the insulin signaling during prolonged low-grade inflammation as seen in obesity.
Collapse
|
9
|
Pharmacological Inhibition of c-Jun N-terminal Kinase Reduces Food Intake and Sensitizes Leptin's Anorectic Signaling Actions. Sci Rep 2017; 7:41795. [PMID: 28165482 PMCID: PMC5292945 DOI: 10.1038/srep41795] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 12/28/2016] [Indexed: 01/09/2023] Open
Abstract
The role for c-Jun N-terminal Kinase (JNK) in the control of feeding and energy balance is not well understood. Here, by use of novel and highly selective JNK inhibitors, we investigated the actions of JNK in the control of feeding and body weight homeostasis. In lean mice, intraperitoneal (i.p.) or intracerebroventricular (i.c.v.) administration of SR-3306, a brain-penetrant and selective pan-JNK (JNK1/2/3) inhibitor, reduced food intake and body weight. Moreover, i.p. and i.c.v. administrations of SR11935, a brain-penetrant and JNK2/3 isoform-selective inhibitor, exerted similar anorectic effects as SR3306, which suggests JNK2 or JNK3 mediates aspect of the anorectic effect by pan-JNK inhibition. Furthermore, daily i.p. injection of SR3306 (7 days) prevented the increases in food intake and weight gain in lean mice upon high-fat diet feeding, and this injection paradigm reduced high-fat intake and obesity in diet-induced obese (DIO) mice. In the DIO mice, JNK inhibition sensitized leptin’s anorectic effect, and enhanced leptin-induced STAT3 activation in the hypothalamus. The underlying mechanisms likely involve the downregulation of SOCS3 by JNK inhibition. Collectively, our data suggest that JNK activity promotes positive energy balance, and the therapeutic intervention inhibiting JNK activities represents a promising approach to ameliorate diet-induced obesity and leptin resistance.
Collapse
|
10
|
Sahu M, Anamthathmakula P, Sahu A. Hypothalamic Phosphodiesterase 3B Pathway Mediates Anorectic and Body Weight-Reducing Effects of Insulin in Male Mice. Neuroendocrinology 2016; 104:145-156. [PMID: 27002827 PMCID: PMC5035167 DOI: 10.1159/000445523] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 03/17/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND Insulin action in the hypothalamus plays a critical role in the regulation of energy homeostasis, yet the intracellular signaling mechanisms mediating insulin action are incompletely understood. Although phosphodiesterase 3B (PDE3B) mediates insulin action in the adipose tissue and it is highly expressed in the hypothalamic areas implicated in energy homeostasis, its role, if any, in mediating insulin action in the hypothalamus is unknown. We tested the hypothesis that insulin action in the hypothalamus is mediated by PDE3B. METHODS Using enzymatic assay, we examined the effects of peripheral or central administration of insulin on hypothalamic PDE3B activity in adult mice. Western blotting and immunohistochemistry also examined p-Akt and p-STAT3 levels in the hypothalamus. Effects of leptin on these parameters were also compared. We injected cilostamide, a PDE3 inhibitor, prior to central injection of insulin and examined the 12- to 24-hour food intake and 24-hour body weight. Finally, we examined the effect of cilostamide on insulin-induced proopiomelanocortin (Pomc), neurotensin (Nt), neuropeptide Y (Npy) and agouti-related peptide (Agrp) gene expression in the hypothalamus by qPCR. RESULTS Peripheral or central injection of insulin significantly increased PDE3B activity in the hypothalamus in association with increased p-Akt levels but without any change in p-STAT3 levels. However, leptin-induced increase in PDE3B activity was associated with an increase in both p-Akt and p-STAT3 levels in the hypothalamus. Prior administration of cilostamide reversed the anorectic and body weight-reducing effects as well as stimulatory effect of insulin on hypothalamic Pomc mRNA levels. Insulin did not alter Nt, Npy and Agrp mRNA levels. CONCLUSION Insulin induction of hypothalamic PDE3B activity and the reversal of the anorectic and body weight-reducing effects and stimulatory effect of insulin on hypothalamic Pomc gene expression by cilostamide suggest that activation of PDE3B is a novel mechanism of insulin signaling in the hypothalamus.
Collapse
Affiliation(s)
- Maitrayee Sahu
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Prashanth Anamthathmakula
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Abhiram Sahu
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
11
|
Ornellas F, Souza-Mello V, Mandarim-de-Lacerda CA, Aguila MB. Combined parental obesity augments single-parent obesity effects on hypothalamus inflammation, leptin signaling (JAK/STAT), hyperphagia, and obesity in the adult mice offspring. Physiol Behav 2015; 153:47-55. [PMID: 26485293 DOI: 10.1016/j.physbeh.2015.10.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 10/07/2015] [Accepted: 10/16/2015] [Indexed: 11/29/2022]
Abstract
We aimed to evaluate the effects of maternal and/or paternal obesity on offspring body mass, leptin signaling, appetite-regulating neurotransmitters and local inflammatory markers. C57BL/6 mice received standard chow (SC, lean groups) or high-fat diet (HF, obese groups) starting from one month of age. At three months, HF mice became obese relative to SC mice. They were then mated as follows: lean mother and lean father, lean mother and obese father, obese mother and lean father, and obese mother and obese father. The offspring received the SC diet from weaning until three months of age, when they were sacrificed. In the offspring, paternal obesity did not lead to changes in the Janus kinase (JAK)/signal transducer and activation of the transcription (STAT) pathway or feeding behavior but did induce hypothalamic inflammation. On the other hand, maternal obesity resulted in increased weight gain, hyperleptinemia, decreased leptin OBRb receptor expression, JAK/STAT pathway impairment, and increased SOCS3 signaling in the offspring. In addition, maternal obesity elevated inflammatory markers and altered NPY and POMC expression in the hypothalamus. Interestingly, combined parental obesity exacerbated the deleterious outcomes compared to single-parent obesity. In conclusion, while maternal obesity is known to program metabolic changes and obesity in offspring, the current study demonstrated that obese fathers induce hypothalamus inflammation in offspring, which may contribute to the development of metabolic syndromes in adulthood.
Collapse
Affiliation(s)
- Fernanda Ornellas
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Carlos Alberto Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
12
|
Parafati M, Lascala A, Morittu VM, Trimboli F, Rizzuto A, Brunelli E, Coscarelli F, Costa N, Britti D, Ehrlich J, Isidoro C, Mollace V, Janda E. Bergamot polyphenol fraction prevents nonalcoholic fatty liver disease via stimulation of lipophagy in cafeteria diet-induced rat model of metabolic syndrome. J Nutr Biochem 2015; 26:938-48. [PMID: 26025327 DOI: 10.1016/j.jnutbio.2015.03.008] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 03/24/2015] [Accepted: 03/31/2015] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease in industrialized countries. Defective autophagy of lipid droplets (LDs) in hepatocytes, also known as lipophagy, has recently been identified as a possible pathophysiological mechanism of NAFLD. Experimental and epidemiological evidence suggests that dietary polyphenols may prevent NAFLD. To address this hypothesis and analyze the underlying mechanisms, we supplemented bergamot polyphenol fraction (BPF) to cafeteria (CAF) diet-fed rats, a good model for pediatric metabolic syndrome and NAFLD. BPF treatment (50 mg/kg/day supplemented with drinking water, 3 months) potently counteracted the pathogenic increase of serum triglycerides and had moderate effects on blood glucose and obesity in this animal model. Importantly, BPF strongly reduced hepatic steatosis as documented by a significant decrease in total lipid content (-41.3% ± 12% S.E.M.), ultrasound examination and histological analysis of liver sections. The morphometric analysis of oil-red stained sections confirmed a dramatic reduction in LDs parameters such as total LD area (48.5% ± 15% S.E.M.) in hepatocytes from CAF+BPF rats. BPF-treated livers showed increased levels of LC3 and Beclin 1 and reduction of SQSTM1/p62, suggesting autophagy stimulation. Consistent with BPF stimulation of lipophagy, higher levels of LC3II were found in the LD subcellular fractions of BPF-expose livers. This study demonstrates that the liver and its lipid metabolism are the main targets of bergamot flavonoids, supporting the concept that supplementation of BPF is an effective strategy to prevent NAFLD.
Collapse
Affiliation(s)
- Maddalena Parafati
- Department of Health Sciences, Magna Graecia University, Campus Germaneto, Catanzaro, Italy; Interregional Research Center for Food Safety and Health, Catanzaro, Italy
| | - Antonella Lascala
- Department of Health Sciences, Magna Graecia University, Campus Germaneto, Catanzaro, Italy
| | - Valeria Maria Morittu
- Department of Health Sciences, Magna Graecia University, Campus Germaneto, Catanzaro, Italy
| | - Francesca Trimboli
- Department of Health Sciences, Magna Graecia University, Campus Germaneto, Catanzaro, Italy
| | - Antonia Rizzuto
- Department of Experimental and Clinical Medicine, Magna Graecia University, Campus Germaneto, Catanzaro, Italy
| | - Elvira Brunelli
- Department of Ecology, University of Calabria, Rende, Cosenza, Italy
| | | | - Nicola Costa
- Department of Health Sciences, Magna Graecia University, Campus Germaneto, Catanzaro, Italy
| | - Domenico Britti
- Department of Health Sciences, Magna Graecia University, Campus Germaneto, Catanzaro, Italy
| | | | - Ciro Isidoro
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Vincenzo Mollace
- Department of Health Sciences, Magna Graecia University, Campus Germaneto, Catanzaro, Italy; Interregional Research Center for Food Safety and Health, Catanzaro, Italy
| | - Elzbieta Janda
- Department of Health Sciences, Magna Graecia University, Campus Germaneto, Catanzaro, Italy; Interregional Research Center for Food Safety and Health, Catanzaro, Italy
| |
Collapse
|
13
|
Portovedo M, Ignacio-Souza LM, Bombassaro B, Coope A, Reginato A, Razolli DS, Torsoni MA, Torsoni AS, Leal RF, Velloso LA, Milanski M. Saturated fatty acids modulate autophagy's proteins in the hypothalamus. PLoS One 2015; 10:e0119850. [PMID: 25786112 PMCID: PMC4364755 DOI: 10.1371/journal.pone.0119850] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 01/19/2015] [Indexed: 11/18/2022] Open
Abstract
Autophagy is an important process that regulates cellular homeostasis by degrading dysfunctional proteins, organelles and lipids. In this study, the hypothesis that obesity could lead to impairment in hypothalamic autophagy in mice was evaluated by examining the hypothalamic distribution and content of autophagic proteins in animal with obesity induced by 8 or 16 weeks high fat diet to induce obesity and in response to intracerebroventricular injections of palmitic acid. The results showed that chronic exposure to a high fat diet leads to an increased expression of inflammatory markers and downregulation of autophagic proteins. In obese mice, autophagic induction leads to the downregulation of proteins, such as JNK and Bax, which are involved in the stress pathways. In neuron cell- line, palmitate has a direct effect on autophagy even without inflammatory activity. Understanding the cellular and molecular bases of overnutrition is essential for identifying new diagnostic and therapeutic targets for obesity.
Collapse
Affiliation(s)
- Mariana Portovedo
- Faculty of Applied Science, University of Campinas, UNICAMP, Limeira, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | | | - Bruna Bombassaro
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Andressa Coope
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Andressa Reginato
- Faculty of Applied Science, University of Campinas, UNICAMP, Limeira, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Daniela S. Razolli
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Márcio A. Torsoni
- Faculty of Applied Science, University of Campinas, UNICAMP, Limeira, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Adriana S. Torsoni
- Faculty of Applied Science, University of Campinas, UNICAMP, Limeira, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Raquel F. Leal
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Licio A. Velloso
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Marciane Milanski
- Faculty of Applied Science, University of Campinas, UNICAMP, Limeira, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
- * E-mail:
| |
Collapse
|
14
|
Tsaousidou E, Paeger L, Belgardt BF, Pal M, Wunderlich CM, Brönneke H, Collienne U, Hampel B, Wunderlich FT, Schmidt-Supprian M, Kloppenburg P, Brüning JC. Distinct Roles for JNK and IKK Activation in Agouti-Related Peptide Neurons in the Development of Obesity and Insulin Resistance. Cell Rep 2014; 9:1495-506. [PMID: 25456138 DOI: 10.1016/j.celrep.2014.10.045] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 07/10/2014] [Accepted: 10/16/2014] [Indexed: 01/01/2023] Open
Abstract
Activation of c-Jun N-terminal kinase 1 (JNK1)- and inhibitor of nuclear factor kappa-B kinase 2 (IKK2)-dependent signaling plays a crucial role in the development of obesity-associated insulin and leptin resistance not only in peripheral tissues but also in the CNS. Here, we demonstrate that constitutive JNK activation in agouti-related peptide (AgRP)-expressing neurons of the hypothalamus is sufficient to induce weight gain and adiposity in mice as a consequence of hyperphagia. JNK activation increases spontaneous action potential firing of AgRP cells and causes both neuronal and systemic leptin resistance. Similarly, activation of IKK2 signaling in AgRP neurons also increases firing of these cells but fails to cause obesity and leptin resistance. In contrast to JNK activation, IKK2 activation blunts insulin signaling in AgRP neurons and impairs systemic glucose homeostasis. Collectively, these experiments reveal both overlapping and nonredundant effects of JNK- and IKK-dependent signaling in AgRP neurons, which cooperate in the manifestation of the metabolic syndrome.
Collapse
Affiliation(s)
- Eva Tsaousidou
- Department of Mouse Genetics and Metabolism, Institute for Genetics, Cologne 50674, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne 50937, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany
| | - Lars Paeger
- Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Institute for Zoology, University of Cologne, Cologne 50674, Germany
| | - Bengt F Belgardt
- Department of Mouse Genetics and Metabolism, Institute for Genetics, Cologne 50674, Germany
| | - Martin Pal
- Baker IDI Heart and Diabetes Institute, Cellular and Molecular Metabolism Laboratory, Melbourne, VIC 3004, Australia
| | - Claudia M Wunderlich
- Department of Mouse Genetics and Metabolism, Institute for Genetics, Cologne 50674, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne 50937, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany
| | - Hella Brönneke
- Mouse Phenotyping Core Facility, Cologne Excellence Cluster on Cellular Stress Responses, Cologne 50931, Germany
| | - Ursel Collienne
- Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Institute for Zoology, University of Cologne, Cologne 50674, Germany
| | - Brigitte Hampel
- Department of Mouse Genetics and Metabolism, Institute for Genetics, Cologne 50674, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne 50937, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany
| | - F Thomas Wunderlich
- Department of Mouse Genetics and Metabolism, Institute for Genetics, Cologne 50674, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne 50937, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany
| | - Marc Schmidt-Supprian
- Department of Hematology and Oncology, Klinikum Rechts der Isar, Technical University of Munich, Munich 81675, Germany
| | - Peter Kloppenburg
- Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Institute for Zoology, University of Cologne, Cologne 50674, Germany
| | - Jens C Brüning
- Department of Mouse Genetics and Metabolism, Institute for Genetics, Cologne 50674, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), Cologne 50931, Germany; Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne 50937, Germany; Max Planck Institute for Metabolism Research, Cologne 50931, Germany.
| |
Collapse
|
15
|
Marissal-Arvy N, Diane A, Moisan MP, Larue-Achagiotis C, Tridon C, Tome D, Fromentin G, Mormède P. QTLs influencing carbohydrate and fat choice in a LOU/CxFischer 344 F2 rat population. Obesity (Silver Spring) 2014; 22:565-75. [PMID: 23596094 DOI: 10.1002/oby.20485] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 03/19/2013] [Indexed: 01/27/2023]
Abstract
OBJECTIVE Individual differences in macronutrient selection, particularly fat and carbohydrate, and associated body weight gain are partly inherited as polygenic traits, but the potential genetic pathways are unknown. To give an overview of the Quantitative Trait Loci (QTLs) and candidate gene pathways influencing these differences in rat was aimed in this study. DESIGN AND METHODS To that end, F2 rats obtained from the crossbreeding between LOU/C and Fischer 344 rat strains to diet self-selection during 3 weeks were submitted. A genome scan was conducted with microsatellite markers covering evenly the whole genome. Genotypes and phenotypes were analyzed separately in male and female F2 rats by multiple interval mapping. Then, lists of candidate genes were treated by the Ingenuity Pathway software to propose gene pathways involved in our phenotypes. RESULTS Among numerous others, a QTL on chromosome 12 that influences body weight gain, and fat and carbohydrate choices in the LOU/C x Fischer 344 F2 rat population was found. This locus contains notably the acyl-co-A dehydrogenase gene. CONCLUSION A strong genetic determinism and complex pathways involving numerous candidate genes and processes, notably in accordance with the metabolic theory of feeding behavior control were found.
Collapse
Affiliation(s)
- Nathalie Marissal-Arvy
- INRA, Laboratory of Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux Cedex, France; Laboratory of Nutrition and Integrative Neurobiology, University of Bordeaux, UMR 1286, 33076 Bordeaux Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Hagimoto S, Arima H, Adachi K, Ito Y, Suga H, Sugimura Y, Goto M, Banno R, Oiso Y. Expression of neuropeptide Y and agouti-related protein mRNA stimulated by glucocorticoids is attenuated via NF-κB p65 under ER stress in mouse hypothalamic cultures. Neurosci Lett 2013; 553:165-9. [DOI: 10.1016/j.neulet.2013.08.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Revised: 08/07/2013] [Accepted: 08/20/2013] [Indexed: 11/27/2022]
|
17
|
Castro G, C. Areias MF, Weissmann L, Quaresma PG, Katashima CK, Saad MJ, Prada PO. Diet-induced obesity induces endoplasmic reticulum stress and insulin resistance in the amygdala of rats. FEBS Open Bio 2013; 3:443-9. [PMID: 24251109 PMCID: PMC3829990 DOI: 10.1016/j.fob.2013.09.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 09/03/2013] [Accepted: 09/05/2013] [Indexed: 12/25/2022] Open
Abstract
Insulin acts in the hypothalamus, decreasing food intake (FI) by the IR/PI3K/Akt pathway. This pathway is impaired in obese animals and endoplasmic reticulum (ER) stress and low-grade inflammation are possible mechanisms involved in this impairment. Here, we highlighted the amygdala as an important brain region for FI regulation in response to insulin. This regulation was dependent on PI3K/AKT pathway similar to the hypothalamus. Insulin was able to decrease neuropeptide Y (NPY) and increase oxytocin mRNA levels in the amygdala via PI3K, which may contribute to hypophagia. Additionally, obese rats did not reduce FI in response to insulin and AKT phosphorylation was decreased in the amygdala, suggesting insulin resistance. Insulin resistance was associated with ER stress and low-grade inflammation in this brain region. The inhibition of ER stress with PBA reverses insulin action/signaling, decreases NPY and increases oxytocin mRNA levels in the amygdala from obese rats, suggesting that ER stress is probably one of the mechanisms that induce insulin resistance in the amygdala.
Collapse
Key Words
- AGRP, agouti-related peptide
- AMY, amygdala
- Amygdala
- BW, body weight
- CNS, central nervous system
- CRH, corticotrophin-releasing hormone
- ER, endoplasmic reticulum
- Endoplasmic reticulum stress
- FI, food intake
- FKBP51, FK506 binding protein 51
- HFD, high-fat diet
- HPRT, hypoxanthine phosphoribosyl transferase
- IKKβ, I kappa B kinase
- IR, insulin receptor
- IRE1α, inositol-requiring kinase alpha
- IRS-1, insulin substrate 1
- Inflammation
- Insulin
- JNK, c-Jun N-terminal kinase
- LGI, low-grade inflammation
- NPY
- NPY, neuropeptide Y
- Obesity
- Oxytocin
- PBA, 4-phenyl butyric acid
- PERK, RNA-activated protein kinase-like ER resident kinase
- PI3K, phosphoinositide 3-kinase
- PKB or Akt, protein kinase B
- Phosphatidylinositol 3-kinase
Collapse
Affiliation(s)
- Gisele Castro
- School of Applied Sciences, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | | | - Lais Weissmann
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Paula G.F. Quaresma
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Carlos K. Katashima
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Mario J.A. Saad
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Patricia O. Prada
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil
- School of Applied Sciences, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| |
Collapse
|
18
|
Benzler J, Ganjam GK, Legler K, Stöhr S, Krüger M, Steger J, Tups A. Acute inhibition of central c-Jun N-terminal kinase restores hypothalamic insulin signalling and alleviates glucose intolerance in diabetic mice. J Neuroendocrinol 2013; 25:446-54. [PMID: 23301857 DOI: 10.1111/jne.12018] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 12/10/2012] [Accepted: 12/26/2012] [Indexed: 12/31/2022]
Abstract
The hypothalamus has been identified as a main insulin target tissue for regulating normal body weight and glucose metabolism. Recent observations suggest that c-Jun-N-terminal kinase (JNK)-signalling plays a crucial role in the development of obesity and insulin resistance because neuronal JNK-1 ablation in the mouse prevented high-fat diet-induced obesity (DIO) and increased energy expenditure, as well as insulin sensitivity. In the present study, we investigated whether central JNK inhibition is associated with sensitisation of hypothalamic insulin signalling in mice fed a high-fat diet for 3 weeks and in leptin-deficient mice. We determined whether i.c.v. injection of a pharmacological JNK-inhibitor (SP600125) improved impaired glucose homeostasis. By immunohistochemistry, we first observed that JNK activity was increased in the arcuate nucleus (ARC) and the ventromedial hypothalamus (VMH) in both mouse models, relative to normoglycaemic controls. This suggests that up-regulation of JNK in these regions is associated with glucose intolerance and obesity, independent of leptin levels. Acute i.c.v. injection of SP600125 ameliorated glucose tolerance within 30 min in both leptin-deficient and DIO mice. Given the acute nature of i.c.v. injections, these effects cannot be attributed to changes in food intake or energy balance. In a hypothalamic cell line, and in the ARC and VMH of leptin-deficient mice, JNK inhibition by SP600125 consistently improved impaired insulin signalling. This was determined by a reduction of phospho-insulin receptor substrate-1 [IRS-1(Ser612)] protein in a hypothalamic cell line and a decline in the number of pIRS-1(Ser612) immunoreactive cells in the ARC and VMH. Serine 612 phosphorylation of IRS-1 is assumed to negatively regulate insulin signalling. In leptin-deficient mice, in both nuclei, central inhibition of JNK increased the number of cells immunoreactive for phospho-Akt (Ser473) and phospho-GSK-3β (Ser9), which are important markers of insulin signalling. Collectively, our data suggest that the acute inhibition of central JNK improves impaired glucose homeostasis and is associated with sensitisation of hypothalamic insulin signalling.
Collapse
Affiliation(s)
- J Benzler
- Department of Animal Physiology, Faculty of Biology, Philipps University Marburg, Germany
| | | | | | | | | | | | | |
Collapse
|
19
|
Affiliation(s)
- Anthony W Ferrante
- Department of Medicine, Naomi Berrie Diabetes Center, Columbia University, 1150 Saint Nicholas Avenue, New York, NY 10032, USA.
| |
Collapse
|
20
|
Abstract
Inflammation is a biological response mounted by the immune system against dangerous assaults that threaten the integrity and normal physiology of an organism. During the past decades, cross-disciplinary research from immunology and endocrinology has much broadened this knowledge by demonstrating that chronic conditions of nutritional excess constitute an independent category of inflammatory activators, and the resulting chronic and low-grade inflammation is an important characteristic of overnutrition-induced diseases. A large body of research has demonstrated that these diseases are pathogenically associated with the local, negative actions of inflammation in peripheral tissues predominantly including the liver, muscle, and fat. In this research background, more recent research has advanced to a new level, with the important discoveries showing that overnutrition-induced inflammation occurs in the brain and thus plays a broad and leadership role in overnutrition-induced diseases. While much more research establishments are expected in this emerging and quickly expanding research, the appreciated understandings have been mainly based on proinflammatory IKKβ/NF-κB pathway and related molecules in the hypothalamus. In this chapter, the author focuses on describing IKKβ/NF-κB-induced neural inflammation in the context of overnutrition-induced metabolic inflammation and especially the central roles of this neural inflammation in the development of a spectrum of overnutrition-related diseases.
Collapse
Affiliation(s)
- Dongsheng Cai
- Department of Molecular Pharmacology, Institute of Aging, Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York, USA.
| |
Collapse
|
21
|
Newton AJ, Hess S, Paeger L, Vogt MC, Fleming Lascano J, Nillni EA, Brüning JC, Kloppenburg P, Xu AW. AgRP innervation onto POMC neurons increases with age and is accelerated with chronic high-fat feeding in male mice. Endocrinology 2013; 154:172-83. [PMID: 23161869 PMCID: PMC3529372 DOI: 10.1210/en.2012-1643] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
In many mammals, body weight increases continuously throughout adulthood until late middle age. The hormone leptin is necessary for maintaining body weight, in that high levels of leptin promote negative energy balance. As animals age, however, their increase in body weight is accompanied by a steady rise in circulating leptin levels, indicating the progressive development of counterregulatory mechanisms to antagonize leptin's anorexigenic effects. Hypothalamic neurons coexpressing agouti-related peptide (AgRP) and neuropeptide Y are direct leptin targets. These neurons promote positive energy balance, and they inhibit anorexigenic proopiomelanocortin (POMC) neurons via direct neuropeptide action and release of γ-aminobutyric acid. We show here that AgRP and neuropeptide Y innvervation onto POMC neurons increases dramatically with age in male mice. This is associated with progressive increase of inhibitory postsynaptic currents and decrease of POMC firing rate with age. Neuronal activity is significantly attenuated in POMC neurons that receive a high density of AgRP puncta. These high-density AgRP inputs correlate with leptin levels in normal mice and are nearly absent in mice lacking leptin. The progression of increased AgRP innervation onto POMC somas is accelerated in hyperleptinemic, diet-induced obese mice. Together our study suggests that modulation of hypothalamic AgRP innervation constitutes one mechanism to counter the effects of the age-associated rise in leptin levels, thus sustaining body weight and fat mass at an elevated level in adulthood.
Collapse
Affiliation(s)
- A Jamila Newton
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
A growing number of studies have shown that a diet high in long chain SFA and/or obesity cause profound changes to the energy balance centres of the hypothalamus which results in the loss of central leptin and insulin sensitivity. Insensitivity to these important anorexigenic messengers of nutritional status perpetuates the development of both obesity and peripheral insulin insensitivity. A high-fat diet induces changes in the hypothalamus that include an increase in markers of oxidative stress, inflammation, endoplasmic reticulum (ER) stress, autophagy defect and changes in the rate of apoptosis and neuronal regeneration. In addition, a number of mechanisms have recently come to light that are important in the hypothalamic control of energy balance, which could play a role in perpetuating the effect of a high-fat diet on hypothalamic dysfunction. These include: reactive oxygen species as an important second messenger, lipid metabolism, autophagy and neuronal and synaptic plasticity. The importance of nutritional activation of the Toll-like receptor 4 and the inhibitor of NF-κB kinase subunit β/NK-κB and c-Jun amino-terminal kinase 1 inflammatory pathways in linking a high-fat diet to obesity and insulin insensitivity via the hypothalamus is now widely recognised. All of the hypothalamic changes induced by a high-fat diet appear to be causally linked and inhibitors of inflammation, ER stress and autophagy defect can prevent or reverse the development of obesity pointing to potential drug targets in the prevention of obesity and metabolic dysfunction.
Collapse
|
23
|
Clarke M, Pentz R, Bobyn J, Hayley S. Stressor-like effects of c-Jun N-terminal kinase (JNK) inhibition. PLoS One 2012; 7:e44073. [PMID: 22952879 PMCID: PMC3430637 DOI: 10.1371/journal.pone.0044073] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 07/30/2012] [Indexed: 11/18/2022] Open
Abstract
There is an urgent need for novel treatment strategies for stressor related disorders, particularly depression and anxiety disorders. Indeed, existing drug treatments are only clinically successful in a subset of patients and relapse is common. This likely stems from the fact that stressor disorders are heterogeneous with multiple biological pathways being affected. To this end, the present investigation sought to assess in mice the contribution of the c-Jun N terminal kinase (JNK) pathway to the behavioral, hormonal and neurochemical effects of an acute stressor. Indeed, although JNK has been shown to modulate glucocorticoid receptors in vitro, virtually nothing is known of the role for JNK in affecting stressor induced pathology. We presently found that the JNK antagonist, SP600125, (but not the p38 antagonist, SB203580) increased plasma corticosterone levels under resting conditions and in the context of an acute stressor (wet bedding + restraint). SP600125 also reduced exploration in an open field arena, but prevented the stressor induced increase in open arm exploration in an elevated plus maze. Finally, SP600125 affected noradrenergic activity in the central amygdala and locus coruleus under resting condition, but prevented the noradrenergic effects within the paraventricular nucleus of the hypothalamus that were induced by the acute stressor exposure. These data suggest inhibiting endogenous JNK can have stressor-like corticoid, behavioral and central monoamine effects under basal conditions, but can actually reverse some behavioral and neurochemical effects of an acute stressor. Thus, endogenous JNK appears to affect stress relevant processes in a context-dependent manner.
Collapse
Affiliation(s)
- Melanie Clarke
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Rowan Pentz
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Jessica Bobyn
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Shawn Hayley
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
- * E-mail:
| |
Collapse
|
24
|
Burmeister MA, Bracy DP, James FD, Holt RM, Ayala J, King EM, Wasserman DH, Drucker DJ, Ayala JE. Regulation of glucose kinetics during exercise by the glucagon-like peptide-1 receptor. J Physiol 2012; 590:5245-55. [PMID: 22890715 DOI: 10.1113/jphysiol.2012.234914] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In response to oral glucose, glucagon-like peptide-1 receptor (Glp1r) knockout (Glp1r−/−) mice become hyperglycaemic due to impaired insulin secretion. Exercise also induces hyperglycaemia in Glp1r−/− mice. In contrast to oral glucose, exercise decreases insulin secretion. This implies that exercise-induced hyperglycaemia in Glp1r−/− mice results from the loss of a non-insulinotropic effect mediated by the Glp1r. Muscle glucose uptake (MGU) is normal in exercising Glp1r−/− mice. Thus, we hypothesize that exercise-induced hyperglycaemia in Glp1r−/− mice is due to excessive hepatic glucose production (HGP). Wild-type (Glp1r+/+) and Glp1r−/− mice implanted with venous and arterial catheters underwent treadmill exercise or remained sedentary for 30 min. [3-3H]glucose was used to estimate rates of glucose appearance (Ra), an index of HGP, and disappearance (Rd). 2[14C]deoxyglucose was used to assess MGU. Glp1r−/− mice displayed exercise-induced hyperglycaemia due to an excessive increase in Ra but normal Rd and MGU. Exercise-induced glucagon levels were ∼2-fold higher in Glp1r−/− mice, resulting in a ∼2-fold higher glucagon:insulin ratio. Since inhibition of the central Glp1r stimulates HGP, we tested whether intracerebroventricular (ICV) infusion of the Glp1r antagonist exendin(9–39) (Ex9) in Glp1r+/+ mice would result in exercise-induced hyperglycaemia. ICV Ex9 did not enhance glucose levels or HGP during exercise, suggesting that glucoregulatory effects of Glp1 during exercise are mediated via the pancreatic Glp1r. In conclusion, functional disruption of the Glp1r results in exercise-induced hyperglycaemia associated with an excessive increase in glucagon secretion and HGP. These results suggest an essential role for basal Glp1r signalling in the suppression of alpha cell secretion during exercise.
Collapse
Affiliation(s)
- M A Burmeister
- Metabolic Signaling and Disease Program, Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute at Lake Nona, 6400 Sanger Road, Orlando, FL 32827, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
van de Sande-Lee S, Velloso LA. Disfunção hipotalâmica na obesidade. ACTA ACUST UNITED AC 2012; 56:341-50. [DOI: 10.1590/s0004-27302012000600001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2012] [Accepted: 08/09/2012] [Indexed: 12/31/2022]
Abstract
A obesidade, definida como o acúmulo excessivo ou anormal de gordura que pode causar dano à saúde do indivíduo, é considerada atualmente um dos principais problemas de saúde pública. Resulta de um desequilíbrio entre a ingestão alimentar e o gasto corporal de energia. O controle do balanço energético de animais e seres humanos é realizado pelo sistema nervoso central (SNC) por meio de conexões neuroendócrinas, em que hormônios periféricos circulantes, como a leptina e a insulina, sinalizam neurônios especializados do hipotálamo sobre os estoques de gordura do organismo e induzem respostas apropriadas para a manutenção da estabilidade desses estoques. A maioria dos casos de obesidade se associa a um quadro de resistência central à ação da leptina e da insulina. Em animais de experimentação, a dieta hiperlipídica é capaz de induzir um processo inflamatório no hipotálamo, que interfere com as vias intracelulares de sinalização por esses hormônios, resultando em hiperfagia, diminuição do gasto de energia e, por fim, obesidade. Evidências recentes obtidas por intermédio de estudos de neuroimagem e avaliação de marcadores inflamatórios no líquido cefalorraquidiano de indivíduos obesos sugerem que alterações semelhantes podem estar presentes também em seres humanos. Nesta revisão, apresentamos sumariamente os mecanismos envolvidos com a perda do controle homeostático do balanço energético em modelos animais de obesidade e as evidências atuais de disfunção hipotalâmica em humanos obesos.
Collapse
|
26
|
Abstract
The hypothalamus is one of the master regulators of various physiological processes, including energy balance and nutrient metabolism. These regulatory functions are mediated by discrete hypothalamic regions that integrate metabolic sensing with neuroendocrine and neural controls of systemic physiology. Neurons and nonneuronal cells in these hypothalamic regions act supportively to execute metabolic regulations. Under conditions of brain and hypothalamic inflammation, which may result from overnutrition-induced intracellular stresses or disease-associated systemic inflammatory factors, extracellular and intracellular environments of hypothalamic cells are disrupted, leading to central metabolic dysregulations and various diseases. Recent research has begun to elucidate the effects of hypothalamic inflammation in causing diverse components of metabolic syndrome leading to diabetes and cardiovascular disease. These new understandings have provocatively expanded previous knowledge on the cachectic roles of brain inflammatory response in diseases, such as infections and cancers. This review describes the molecular and cellular characteristics of hypothalamic inflammation in metabolic syndrome and related diseases as opposed to cachectic diseases, and also discusses concepts and potential applications of inhibiting central/hypothalamic inflammation to treat nutritional diseases.
Collapse
Affiliation(s)
- Dongsheng Cai
- Department of Molecular Pharmacology, Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York 10461, USA.
| | | |
Collapse
|
27
|
Affiliation(s)
- Dongsheng Cai
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, USA.
| |
Collapse
|
28
|
Diverse Roles of JNK and MKK Pathways in the Brain. JOURNAL OF SIGNAL TRANSDUCTION 2012; 2012:459265. [PMID: 22496975 PMCID: PMC3307000 DOI: 10.1155/2012/459265] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Accepted: 11/01/2011] [Indexed: 11/18/2022]
Abstract
The c-Jun NH(2)-terminal protein kinase (JNK) plays important roles in a broad range of physiological processes. JNK is controlled by two upstream regulators, mitogen-activated protein kinase kinase (MKK) 4 and MKK7, which are activated by various MAPKKKs. Studies employing knockout mice have demonstrated that the JNK signaling pathway is involved in diverse phenomena in the brain, regulating brain development and maintenance as well as animal metabolism and behavior. Furthermore, examination of single or combined knockout mice of Jnk1, Jnk2, and Jnk3 has revealed both functional differences and redundancy among JNK1, JNK2, and JNK3. Phenotypic differences between knockouts of MKK4 and MKK7 have also been observed, suggesting that the JNK signaling pathway in the brain has a complex nature and is intricately regulated. This paper summarizes the functional properties of the major JNK signaling components in the developing and adult brain.
Collapse
|
29
|
Abstract
The central nervous system (CNS), generally accepted to regulate energy homeostasis, has been implicated in the metabolic perturbations that either cause or are associated with obesity. Normally, the CNS receives hormonal, metabolic, and neuronal input to assure adequate energy levels and maintain stable energy homeostasis. Recent evidence also supports that the CNS uses these same inputs to regulate glucose homeostasis and this aspect of CNS regulation also becomes impaired in the face of dietary-induced obesity. This review focuses on the literature surrounding hypothalamic regulation of energy and glucose homeostasis and discusses how dysregulation of this system may contribute to obesity and T2DM.
Collapse
|
30
|
The Endocannabinoid System as Pharmacological Target Derived from Its CNS Role in Energy Homeostasis and Reward. Applications in Eating Disorders and Addiction. Pharmaceuticals (Basel) 2011; 4:1101-1136. [PMID: 32143540 PMCID: PMC4058662 DOI: 10.3390/ph4081101] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 07/18/2011] [Accepted: 07/28/2011] [Indexed: 01/26/2023] Open
Abstract
The endocannabinoid system (ECS) has been implicated in many physiological functions, including the regulation of appetite, food intake and energy balance, a crucial involvement in brain reward systems and a role in psychophysiological homeostasis (anxiety and stress responses). We first introduce this important regulatory system and chronicle what is known concerning the signal transduction pathways activated upon the binding of endogenous cannabinoid ligands to the Gi/0-coupled CB1 cannabinoid receptor, as well as its interactions with other hormones and neuromodulators which can modify endocannabinoid signaling in the brain. Anorexia nervosa (AN) and bulimia nervosa (BN) are severe and disabling psychiatric disorders, characterized by profound eating and weight alterations and body image disturbances. Since endocannabinoids modulate eating behavior, it is plausible that endocannabinoid genes may contribute to the biological vulnerability to these diseases. We present and discuss data suggesting an impaired endocannabinoid signaling in these eating disorders, including association of endocannabinoid components gene polymorphisms and altered CB1-receptor expression in AN and BN. Then we discuss recent findings that may provide new avenues for the identification of therapeutic strategies based on the endocannabinod system. In relation with its implications as a reward-related system, the endocannabinoid system is not only a target for cannabis but it also shows interactions with other drugs of abuse. On the other hand, there may be also a possibility to point to the ECS as a potential target for treatment of drug-abuse and addiction. Within this framework we will focus on enzymatic machinery involved in endocannabinoid inactivation (notably fatty acid amide hydrolase or FAAH) as a particularly interesting potential target. Since a deregulated endocannabinoid system may be also related to depression, anxiety and pain symptomatology accompanying drug-withdrawal states, this is an area of relevance to also explore adjuvant treatments for improving these adverse emotional reactions.
Collapse
|
31
|
Meng Q, Cai D. Defective hypothalamic autophagy directs the central pathogenesis of obesity via the IkappaB kinase beta (IKKbeta)/NF-kappaB pathway. J Biol Chem 2011; 286:32324-32. [PMID: 21784844 DOI: 10.1074/jbc.m111.254417] [Citation(s) in RCA: 203] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Autophagy has been recently demonstrated to control cell and tissue homeostasis, including the functions of various metabolic tissues. However, it remains unclear whether autophagy is critical for the central nervous system and particularly the hypothalamus for exerting metabolic regulation. Using autophagy-related protein 7 (Atg7) as an autophagic marker, this work showed that autophagy was highly active in the mediobasal hypothalamus of normal mice. In contrast, chronic development of dietary obesity was associated with autophagic decline in the mediobasal hypothalamus. To investigate the potential role of autophagy in the hypothalamic control of metabolic physiology, a mouse model was developed with autophagic inhibition in the mediobasal hypothalamus using site-specific delivery of lentiviral shRNA against Atg7. This model revealed that hypothalamic inhibition of autophagy increased energy intake and reduced energy expenditure. These metabolic changes were sufficient to increase body weight gain under normal chow feeding and exacerbate the progression of obesity and whole-body insulin resistance under high-fat diet feeding. To explore the underlying mechanism, this study found that defective hypothalamic autophagy led to hypothalamic inflammation, including the activation of proinflammatory IκB kinase β pathway. Using brain-specific IκB kinase β knockout mice, it was found that the effects of defective hypothalamic autophagy in promoting obesity were reversed by IκB kinase β inhibition in the brain. In conclusion, hypothalamic autophagy is crucial for the central control of feeding, energy, and body weight balance. Conversely, decline of hypothalamic autophagy under conditions of chronic caloric excess promotes hypothalamic inflammation and thus impairs hypothalamic control of energy balance, leading to accelerated development of obesity and comorbidities.
Collapse
Affiliation(s)
- Qingyuan Meng
- Department of Molecular Pharmacology and Diabetes Research Center, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | |
Collapse
|
32
|
Berthoud HR, Lenard NR, Shin AC. Food reward, hyperphagia, and obesity. Am J Physiol Regul Integr Comp Physiol 2011; 300:R1266-77. [PMID: 21411768 PMCID: PMC3119156 DOI: 10.1152/ajpregu.00028.2011] [Citation(s) in RCA: 158] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Accepted: 03/11/2011] [Indexed: 01/14/2023]
Abstract
Given the unabated obesity problem, there is increasing appreciation of expressions like "my eyes are bigger than my stomach," and recent studies in rodents and humans suggest that dysregulated brain reward pathways may be contributing not only to drug addiction but also to increased intake of palatable foods and ultimately obesity. After describing recent progress in revealing the neural pathways and mechanisms underlying food reward and the attribution of incentive salience by internal state signals, we analyze the potentially circular relationship between palatable food intake, hyperphagia, and obesity. Are there preexisting individual differences in reward functions at an early age, and could they be responsible for development of obesity later in life? Does repeated exposure to palatable foods set off a cascade of sensitization as in drug and alcohol addiction? Are reward functions altered by secondary effects of the obese state, such as increased signaling through inflammatory, oxidative, and mitochondrial stress pathways? Answering these questions will significantly impact prevention and treatment of obesity and its ensuing comorbidities as well as eating disorders and drug and alcohol addiction.
Collapse
Affiliation(s)
- Hans-Rudolf Berthoud
- Neurobiology of Nutrition Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, 70808, USA.
| | | | | |
Collapse
|
33
|
Abstract
Energy homeostasis involves a complex network of hypothalamic and extra-hypothalamic neurons that transduce hormonal, nutrient and neuronal signals into responses that ultimately match caloric intake to energy expenditure and thereby promote stability of body fat stores. Growing evidence suggests that rather than reflecting a failure to regulate caloric intake, common forms of obesity involve fundamental changes to this homeostatic system that favor the defense of an elevated level of body adiposity. This article reviews emerging evidence that during high-fat feeding, obesity pathogenesis involves fundamental alteration of hypothalamic systems that regulate food intake and energy expenditure.
Collapse
|
34
|
Abstract
The stress-activated c-Jun amino-terminal kinase (JNK) plays a pivotal role in metabolic conditions such as obesity, insulin resistance, and type 2 diabetes. Intricate tissue-specific tweaking of JNK activity in preclinical models of metabolic diseases reveals a complex interplay among local and systemic effects on carbohydrate and lipid metabolism. Synthesis of these entangled effects illustrates that for JNK inhibitors to have therapeutic impact, they must function in multiple cell types to modulate JNK activity.
Collapse
Affiliation(s)
- Sara N Vallerie
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, MA 02115, USA
| | | |
Collapse
|
35
|
Arruda AP, Milanski M, Velloso LA. Hypothalamic inflammation and thermogenesis: the brown adipose tissue connection. J Bioenerg Biomembr 2011; 43:53-8. [DOI: 10.1007/s10863-011-9325-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
36
|
Abstract
Activation of stress-kinase signaling has recently been recognized as an important pathophysiological mechanism in the development of diet-induced obesity, type 2 diabetes mellitus and other aging-related pathologies. Here, c-Jun N-terminal Kinase (JNK) 1 knockout mice have been shown to exhibit protection from diet-induced obesity, glucose intolerance, and insulin resistance. Nonetheless, the tissue-specific role of JNK1-activation in the development of the metabolic syndrome has been poorly defined so far. Recently, it was demonstrated that JNK1 signaling plays a crucial role in the central nervous system (CNS) and in the pituitary to control systemic glucose and lipid metabolism partially through regulation of hormones involved in growth and energy expenditure.
Collapse
Affiliation(s)
- Bengt F Belgardt
- Institute for Genetics, Department of Mouse Genetics and Metabolism, Center for Molecular Medicine, University of Cologne (CMMC), Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), 2nd Department for Internal Medicine University of Cologne 50674 Cologne, Germany
| | | | | |
Collapse
|
37
|
Belgardt BF, Brüning JC. CNS leptin and insulin action in the control of energy homeostasis. Ann N Y Acad Sci 2010; 1212:97-113. [PMID: 21070248 DOI: 10.1111/j.1749-6632.2010.05799.x] [Citation(s) in RCA: 200] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The obesity and diabetes pandemics have made it an urgent necessity to define the central nervous system (CNS) pathways controlling body weight, energy expenditure, and fuel metabolism. The pancreatic hormone insulin and the adipose tissue-derived leptin are known to act on diverse neuronal circuits in the CNS to maintain body weight and metabolism in a variety of species, including humans. Because these homeostatic circuits are disrupted during the development of obesity, the pathomechanisms leading to CNS leptin and insulin resistance are a focal point of research. In this review, we summarize the recent findings concerning the mechanisms and novel neuronal mediators of both insulin and leptin action in the CNS.
Collapse
Affiliation(s)
- Bengt F Belgardt
- Department of Mouse Genetics and Metabolism, Institute for Genetics, Center for Molecular Medicine, University of Cologne, Zülpicher Strasse 47a, Cologne, Germany
| | | |
Collapse
|