1
|
Liu S, Chen X, Chen D, Yu B, Zheng P, Luo Y, He J, Huang Z. Oleanolic acid inhibits appetite through the TGR5/cAMP signaling pathway. J Nutr Biochem 2025; 138:109844. [PMID: 39842616 DOI: 10.1016/j.jnutbio.2025.109844] [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: 07/15/2024] [Revised: 12/23/2024] [Accepted: 01/17/2025] [Indexed: 01/24/2025]
Abstract
Inhibition of appetite is an effective approach to fight obesity. Recently, bile acids have been reported to suppress appetite and alleviate obesity via the Takeda G protein-coupled receptor 5 (TGR5). However, whether the downstream signaling molecule cyclic adenosine monophosphate (cAMP) of TGR5 is involved in this process remains unclear. Oleanolic acid (OA) is a plant analogue of bile acids. The study aimed to explore the effect of dietary OA supplementation on appetite and to examine the role of TGR5/cAMP signaling in this process. In our study, mice were divided into four treatment groups: basal diet, 50mg/kg OA-supplemented diet, 100mg/kg OA-supplemented diet, and 30mg/kg tauroursodeoxycholic acid (TUDCA)-supplemented diet. Our results showed that dietary supplementation of OA and TUDCA both suppressed appetite. Additionally, OA and TUDCA downregulated the expression of appetite-stimulating factors while upregulating appetite-suppressing factors in the hypothalamus. Furthermore, OA was found to activate TGR5 signaling in the hypothalamus. Mechanistic studies using N38 cells revealed that OA reduced the expression and secretion of agouti-related peptide (AgRP), while inhibition of TGR5 and cAMP attenuated this effect of OA. In conclusion, our findings suggest that OA may suppress appetite through activation of the TGR5/cAMP signaling pathway in the hypothalamus.
Collapse
Affiliation(s)
- Shuang Liu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Xiaoling Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Daiwen Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Bing Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Ping Zheng
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Yuheng Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Jun He
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Zhiqing Huang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, PR China.
| |
Collapse
|
2
|
Abstract
Obesity, dieting, and weight cycling are common among reproductive-age women. Weight cycling refers to intentional weight loss followed by unintentional weight regain. Weight loss is accompanied by changes in gut peptides, adipose hormones, and energy expenditure that promote weight regain to a tightly regulated set point. While weight loss can improve body composition and surrogate markers of cardiometabolic health, it is hypothesized that the weight regain can result in an overshoot effect, resulting in excess weight gain, altered body composition, and negative effects on surrogate markers of cardiometabolic health. Numerous observational studies have examined the association of weight cycling and health outcomes. There appears to be modest association between weight cycling with type 2 diabetes mellitus and dyslipidemia in women, but no association with hypertension, cardiovascular events, and overall cancer risk. Interestingly, mild weight cycling may be associated with a decreased risk of overall and cardiovascular mortality. Little is known about the effects of weight cycling in the preconception period. Although obesity and weight gain are associated with pregnancy complications, preconception weight loss does not appear to mitigate the risk of most pregnancy complications related to obesity. Research on preconception weight cycling may provide insight into this paradox.
Collapse
Affiliation(s)
- Katelyn J Carey
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, New York
| | - Wendy Vitek
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
3
|
Signaling pathways in obesity: mechanisms and therapeutic interventions. Signal Transduct Target Ther 2022; 7:298. [PMID: 36031641 PMCID: PMC9420733 DOI: 10.1038/s41392-022-01149-x] [Citation(s) in RCA: 170] [Impact Index Per Article: 56.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/26/2022] [Accepted: 08/08/2022] [Indexed: 12/19/2022] Open
Abstract
Obesity is a complex, chronic disease and global public health challenge. Characterized by excessive fat accumulation in the body, obesity sharply increases the risk of several diseases, such as type 2 diabetes, cardiovascular disease, and nonalcoholic fatty liver disease, and is linked to lower life expectancy. Although lifestyle intervention (diet and exercise) has remarkable effects on weight management, achieving long-term success at weight loss is extremely challenging, and the prevalence of obesity continues to rise worldwide. Over the past decades, the pathophysiology of obesity has been extensively investigated, and an increasing number of signal transduction pathways have been implicated in obesity, making it possible to fight obesity in a more effective and precise way. In this review, we summarize recent advances in the pathogenesis of obesity from both experimental and clinical studies, focusing on signaling pathways and their roles in the regulation of food intake, glucose homeostasis, adipogenesis, thermogenesis, and chronic inflammation. We also discuss the current anti-obesity drugs, as well as weight loss compounds in clinical trials, that target these signals. The evolving knowledge of signaling transduction may shed light on the future direction of obesity research, as we move into a new era of precision medicine.
Collapse
|
4
|
Sanchez MR, Wang Y, Cho TS, Schnapp WI, Schmit MB, Fang C, Cai H. Dissecting a disynaptic central amygdala-parasubthalamic nucleus neural circuit that mediates cholecystokinin-induced eating suppression. Mol Metab 2022; 58:101443. [PMID: 35066159 PMCID: PMC8844644 DOI: 10.1016/j.molmet.2022.101443] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/09/2022] [Accepted: 01/13/2022] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVE Cholecystokinin (CCK) plays a critical role in regulating eating and metabolism. Previous studies have mapped a multi-synapse neural pathway from the vagus nerve to the central nucleus of the amygdala (CEA) that mediates the anorexigenic effect of CCK. However, the neural circuit downstream of the CEA is still unknown due to the complexity of the neurons in the CEA. Here we sought to determine this circuit using a novel approach. METHODS It has been established that a specific population of CEA neurons, marked by protein kinase C-delta (PKC-δ), mediates the anorexigenic effect of CCK by inhibiting other CEA inhibitory neurons. Taking advantage of this circuit, we dissected the neural circuit using a unique approach based on the idea that neurons downstream of the CEA should be disinhibited by CEAPKC-δ+ neurons while being activated by CCK. We also used optogenetic assisted electrophysiology circuit mapping and in vivo chemogenetic manipulation methods to determine the circuit structure and function. RESULTS We found that neurons in the parasubthalamic nucleus (PSTh) are activated by the activation of CEAPKC-δ+ neurons and by the peripheral administration of CCK. We demonstrated that CEAPKC-δ+ neurons inhibit the PSTh-projecting CEA neurons; accordingly, the PSTh neurons can be disynaptically disinhibited or "activated" by CEAPKC-δ+ neurons. Finally, we showed that chemogenetic silencing of the PSTh neurons effectively attenuates the eating suppression induced by CCK. CONCLUSIONS Our results identified a disynaptic CEA-PSTh neural circuit that mediates the anorexigenic effect of CCK and thus provide an important neural mechanism of how CCK suppresses eating.
Collapse
Affiliation(s)
| | - Yong Wang
- Department of Neuroscience, University of Arizona, Tucson, AZ, USA; Department of Physiology and Pathophysiology, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, PR China
| | - Tiffany S Cho
- Department of Neuroscience, University of Arizona, Tucson, AZ, USA
| | - Wesley I Schnapp
- Department of Neuroscience, University of Arizona, Tucson, AZ, USA; Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, AZ, USA
| | - Matthew B Schmit
- Department of Neuroscience, University of Arizona, Tucson, AZ, USA; Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, AZ, USA
| | - Caohui Fang
- Department of Neuroscience, University of Arizona, Tucson, AZ, USA
| | - Haijiang Cai
- Department of Neuroscience, University of Arizona, Tucson, AZ, USA; Bio5 Institute and Department of Neurology, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
5
|
Zhang H, Chen S. Cyclic peptide drugs approved in the last two decades (2001-2021). RSC Chem Biol 2022; 3:18-31. [PMID: 35128405 PMCID: PMC8729179 DOI: 10.1039/d1cb00154j] [Citation(s) in RCA: 171] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/05/2021] [Indexed: 01/01/2023] Open
Abstract
In contrast to the major families of small molecules and antibodies, cyclic peptides, as a family of synthesizable macromolecules, have distinct biochemical and therapeutic properties for pharmaceutical applications. Cyclic peptide-based drugs have increasingly been developed in the past two decades, confirming the common perception that cyclic peptides have high binding affinities and low metabolic toxicity as antibodies, good stability and ease of manufacture as small molecules. Natural peptides were the major source of cyclic peptide drugs in the last century, and cyclic peptides derived from novel screening and cyclization strategies are the new source. In this review, we will discuss and summarize 18 cyclic peptides approved for clinical use in the past two decades to provide a better understanding of cyclic peptide development and to inspire new perspectives. The purpose of the present review is to promote efforts to resolve the challenges in the development of cyclic peptide drugs that are more effective.
Collapse
Affiliation(s)
- Huiya Zhang
- Biotech Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
| | - Shiyu Chen
- Biotech Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
| |
Collapse
|
6
|
Li Y, Zhang Y, Hu Q, Egranov SD, Xing Z, Zhang Z, Liang K, Ye Y, Pan Y, Chatterjee SS, Mistretta B, Nguyen TK, Hawke DH, Gunaratne PH, Hung MC, Han L, Yang L, Lin C. Functional significance of gain-of-function H19 lncRNA in skeletal muscle differentiation and anti-obesity effects. Genome Med 2021; 13:137. [PMID: 34454586 PMCID: PMC8403366 DOI: 10.1186/s13073-021-00937-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 07/09/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Exercise training is well established as the most effective way to enhance muscle performance and muscle building. The composition of skeletal muscle fiber type affects systemic energy expenditures, and perturbations in metabolic homeostasis contribute to the onset of obesity and other metabolic dysfunctions. Long noncoding RNAs (lncRNAs) have been demonstrated to play critical roles in diverse cellular processes and diseases, including human cancers; however, the functional importance of lncRNAs in muscle performance, energy balance, and obesity remains elusive. We previously reported that the lncRNA H19 regulates the poly-ubiquitination and protein stability of dystrophin (DMD) in muscular dystrophy. METHODS Here, we identified mouse/human H19-interacting proteins using mouse/human skeletal muscle tissues and liquid chromatography-mass spectrometry (LC-MS). Human induced pluripotent stem-derived skeletal muscle cells (iPSC-SkMC) from a healthy donor and Becker Muscular Dystrophy (BMD) patients were utilized to study DMD post-translational modifications and associated proteins. We identified a gain-of-function (GOF) mutant of H19 and characterized the effects on myoblast differentiation and fusion to myotubes using iPSCs. We then conjugated H19 RNA gain-of-function oligonucleotides (Rgof) with the skeletal muscle enrichment peptide agrin (referred to as AGR-H19-Rgof) and evaluated AGR-H19-Rgof's effects on skeletal muscle performance using wild-type (WT) C57BL/6 J mice and its anti-obesity effects using high-fat diet (HFD)- and leptin deficiency-induced obese mouse models. RESULTS We demonstrated that both human and mouse H19 associated with DMD and that the H19 GOF exhibited enhanced interaction with DMD compared to WT H19. DMD was found to associate with serine/threonine-protein kinase MRCK alpha (MRCKα) and α-synuclein (SNCA) in iPSC-SkMC derived from BMD patients. Inhibition of MRCKα and SNCA-mediated phosphorylation of DMD antagonized the interaction between H19 and DMD. These signaling events led to improved skeletal muscle cell differentiation and myotube fusion. The administration of AGR-H19-Rgof improved the muscle mass, muscle performance, and base metabolic rate of WT mice. Furthermore, mice treated with AGR-H19-Rgof exhibited resistance to HFD- or leptin deficiency-induced obesity. CONCLUSIONS Our study suggested the functional importance of the H19 GOF mutant in enhancing muscle performance and anti-obesity effects.
Collapse
Affiliation(s)
- Yajuan Li
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yaohua Zhang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Qingsong Hu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sergey D Egranov
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Zhen Xing
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Current address: Sanofi U.S., Boston, MA, 02139, USA
| | - Zhao Zhang
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, 77030, USA
| | - Ke Liang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Youqiong Ye
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, 77030, USA
| | - Yinghong Pan
- Department of Biochemistry and Biology, University of Houston, Houston, TX, 77204, USA
- Current address: UPMC Genome Center, Pittsburgh, PA, 15232, USA
| | - Sujash S Chatterjee
- Department of Biochemistry and Biology, University of Houston, Houston, TX, 77204, USA
| | - Brandon Mistretta
- Department of Biochemistry and Biology, University of Houston, Houston, TX, 77204, USA
| | - Tina K Nguyen
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - David H Hawke
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Preethi H Gunaratne
- Department of Biochemistry and Biology, University of Houston, Houston, TX, 77204, USA
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology, and Center for Molecular Medicine, China Medical University, Taichung, 404, Taiwan
- Department of Biotechnology, Asia University, Taichung, 413, Taiwan
| | - Leng Han
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, 77030, USA
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA
| | - Liuqing Yang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- The Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Chunru Lin
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
7
|
Farhid H, Khodkari V, Nazeri MT, Javanbakht S, Shaabani A. Multicomponent reactions as a potent tool for the synthesis of benzodiazepines. Org Biomol Chem 2021; 19:3318-3358. [PMID: 33899847 DOI: 10.1039/d0ob02600j] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Benzodiazepines (BZDs), a diverse class of benzofused seven-membered N-heterocycles, display essential pharmacological properties and play vital roles in some biochemical processes. They have mainly been prescribed as potential therapeutic agents, which interestingly represent various biological activities such as anticancer, anxiolytic, antipsychotic, anticonvulsant, antituberculosis, muscle relaxant, and antimicrobial activities. The extensive biological activities of BZDs in various fields have encouraged medicinal chemists to discover and design novel BZD-based scaffolds as potential therapeutic candidates with the favorite biological activity through an efficient protocol. Although certainly valuable and important, conventional synthetic routes to these bicyclic benzene compounds contain methodologies often requiring multistep procedures, which suffer from waste materials generation and lack of sustainability. By contrast, multicomponent reactions (MCRs) have recently advanced as a green synthetic strategy for synthesizing BZDs with the desired scope. In this regard, MCRs, especially Ugi and Ugi-type reactions, efficiently and conveniently supply various complex synthons, which can easily be converted to the BZDs via suitable post-transformations. Also, MCRs, especially Mannich-type reactions, provide speedy and economic approaches for the one-pot and one-step synthesis of BZDs. As a result, various functionalized-BZDs have been achieved by developing mild, efficient, and high-yielding MCR protocols. This review covers all aspects of the synthesis of BZDs with a particular focus on the MCRs as well as the mechanism chemistry of synthetic protocols. The present manuscript opens a new avenue for organic, medicinal, and industrial chemists to design safe, environmentally benign, and economical methods for the synthesis of new and known BZDs.
Collapse
Affiliation(s)
- Hassan Farhid
- Faculty of Chemistry, Shahid Beheshti University, G. C., P. O. Box 19396-4716, Tehran, Iran.
| | - Vida Khodkari
- Faculty of Chemistry, Shahid Beheshti University, G. C., P. O. Box 19396-4716, Tehran, Iran.
| | - Mohammad Taghi Nazeri
- Faculty of Chemistry, Shahid Beheshti University, G. C., P. O. Box 19396-4716, Tehran, Iran.
| | - Siamak Javanbakht
- Faculty of Chemistry, Shahid Beheshti University, G. C., P. O. Box 19396-4716, Tehran, Iran.
| | - Ahmad Shaabani
- Faculty of Chemistry, Shahid Beheshti University, G. C., P. O. Box 19396-4716, Tehran, Iran. and Peoples' Friendship University of Russia (RUDN University), 6, Miklukho-Maklaya Street, Moscow, 117198, Russian Federation
| |
Collapse
|
8
|
Mehta M, Istfan NW, Apovian CM. Obesity: Overview of Weight Management. Endocr Pract 2021; 27:626-635. [PMID: 33901648 DOI: 10.1016/j.eprac.2021.04.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/02/2021] [Accepted: 04/05/2021] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Obesity is a chronic illness that requires a multifaceted personalized treatment approach. METHODS & FINDINGS Using current guidelines and recent studies in weight management, this article reviews the multiple components of weight management: lifestyle intervention (dietary intervention, physical activity, and behavioral interventions), pharmacotherapy, endoscopic procedures, and surgical procedures. This review briefly discusses specific diets and dietary strategies, compensatory mechanisms acting against weight loss, recent changes to Food and Drug Administration approved antiobesity medications, and technological advances in weight management delivery. CONCLUSION Current literature is lacking large studies on the safety and efficacy of combination therapies involving pharmacotherapy plus 1 or more procedures.
Collapse
Affiliation(s)
- Meetal Mehta
- Brigham and Women's Hospital, Center for Weight Management and Wellness, Section of Endocrinology, Diabetes and Hypertension, Harvard Medical School, Boston, Massachusetts.
| | - Nawfal W Istfan
- Brigham and Women's Hospital, Center for Weight Management and Wellness, Section of Endocrinology, Diabetes and Hypertension, Harvard Medical School, Boston, Massachusetts
| | - Caroline M Apovian
- Brigham and Women's Hospital, Center for Weight Management and Wellness, Section of Endocrinology, Diabetes and Hypertension, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
9
|
Anti-Obesity Effect of Pine Needle Extract on High-Fat Diet-Induced Obese Mice. PLANTS 2021; 10:plants10050837. [PMID: 33919440 PMCID: PMC8143554 DOI: 10.3390/plants10050837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/14/2021] [Accepted: 04/17/2021] [Indexed: 12/12/2022]
Abstract
Background: Obesity due to an excessive intake of nutrient disturbs the hypothalamus-mediated energy metabolism subsequently develops metabolic disorders. In this study, we investigated the effect of pine needle extract (PNE) on the hypothalamic proopiomelanocortin (POMC) neurons involved in the regulation of energy balance via melanocortin system and fat tissue metabolism. Methods: We performed electrophysiological and immunohistochemical analyses to determine the effect of PNE on POMC neurons. Mice were fed a normal or high-fat diet for 12 weeks, then received PNE for the last 2 weeks to measure the following physiological indices: Body weight, food intake, fat/lean mass, glucose metabolism, and plasma leptin levels. In addition, changes of thermogenic, lipolytic, and lipogenetic markers were evaluated in brown adipose tissue (BAT) and white adipose tissue (WAT) by western blotting, respectively. Results: PNE increased hypothalamic POMC neuronal activity, and the effect was abolished by blockade of melanocortin 3/4 receptors (MC3/4Rs). PNE decreased body weight, fat mass, plasma leptin levels, and improved glucose metabolism after high-fat-induced obesity. However, PNE did not change the expression of thermogenic markers of the BAT in HFD fed groups, but decreased only the lipogenetic markers of WAT. This study suggests that PNE has a potent anti-obesity effect, inhibiting lipogenesis in WAT, even though HFD-induced leptin resistance-mediated disruption of POMC neuronal activity.
Collapse
|
10
|
Wild LE, Alderete TL, Naik NC, Patterson WB, Berger PK, Jones RB, Plows JF, Goran MI. Specific amino acids but not total protein attenuate postpartum weight gain among Hispanic women from Southern California. Food Sci Nutr 2021; 9:1842-1850. [PMID: 33841803 PMCID: PMC8020954 DOI: 10.1002/fsn3.2085] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 12/17/2022] Open
Abstract
There is a high prevalence of obesity and type 2 diabetes in the United States, particularly among Hispanic women, which may be partly explained by failure to lose gestational weight during the postpartum period. Previous work indicates that protein and amino acids may protect against weight gain; therefore, this study examined the impact of dietary protein and amino acid intake on changes in postpartum weight and the percent of women meeting the Estimated Average Requirement (EAR) for these dietary variables among Hispanic women from the Southern California Mother's Milk Study (n = 99). Multivariable linear regression analysis was used to examine the associations between protein and amino acid intake with change in weight after adjusting for maternal age, height, and energy intake. Women's weight increased from prepregnancy to 1-month and 6-months postpartum (71.1 ± 14.6 vs. 73.1 ± 13.1 vs. 74.5 ± 14.6 kg, p < .0001). Although dietary protein was not associated with weight change (β = -1.09; p = .13), phenylalanine (β = -1.46; p = .04), tryptophan (β = -1.71; p = .009), valine (β = -1.34; p = .04), isoleucine (β = -1.26; p = .045), and cysteine (β = -1.52; p = .02) intake were inversely associated with weight change. Additionally, fewer women met the EAR values for cysteine (11.1%), phenylalanine (60.6%), and methionine (69.7%), whereas most women met the EAR values for tryptophan (92.9%), valine (96.0%), and isoleucine (94.9%). Study results indicate that several essential and conditionally essential amino acids were associated with postpartum weight loss, with a significant portion of women not meeting recommended intake levels for some of these amino acids. These results highlight the importance of postpartum maternal diet as a potential modifiable risk factor.
Collapse
Affiliation(s)
- Laura E. Wild
- Department of Integrative PhysiologyUniversity of Colorado BoulderBoulderCOUSA
| | - Tanya L. Alderete
- Department of Integrative PhysiologyUniversity of Colorado BoulderBoulderCOUSA
| | - Noopur C. Naik
- Department of Integrative PhysiologyUniversity of Colorado BoulderBoulderCOUSA
| | | | - Paige K. Berger
- Department of PediatricsThe Saban Research InstituteChildren's Hospital Los AngelesUniversity of Southern CaliforniaLos AngelesCAUSA
| | - Roshonda B. Jones
- Department of PediatricsThe Saban Research InstituteChildren's Hospital Los AngelesUniversity of Southern CaliforniaLos AngelesCAUSA
| | - Jasmine F. Plows
- Department of PediatricsThe Saban Research InstituteChildren's Hospital Los AngelesUniversity of Southern CaliforniaLos AngelesCAUSA
| | - Michael I. Goran
- Department of PediatricsThe Saban Research InstituteChildren's Hospital Los AngelesUniversity of Southern CaliforniaLos AngelesCAUSA
| |
Collapse
|
11
|
Li S, Kim Y, Chen JDZ, Madhoun MF. Intestinal Electrical Stimulation Alters Hypothalamic Expression of Oxytocin and Orexin and Ameliorates Diet-Induced Obesity in Rats. Obes Surg 2021; 31:1664-1672. [PMID: 33392995 PMCID: PMC10433780 DOI: 10.1007/s11695-020-05177-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 12/17/2020] [Accepted: 12/21/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND Intestinal electrical stimulation (IES) has been proposed as a potential treatment for obesity. The aim of this study was to explore the central mechanism underlying the reduction of food intake and body weight by IES by studying the expression of anorexigenic- and orexigenic-peptide-containing neurons in the hypothalamus. MATERIALS AND METHODS Diet-induced obese (DIO) rats were divided into three groups to receive sham, IES, and pair-feeding for 4 weeks. Food intake was measured automatically and presented as daily and body weight measured weekly. The expressions of oxytocin, an anorexigenic neuropeptide, in the paraventricular nucleus of the hypothalamus (PVN) and the supraoptic nuclei of the hypothalamus (SON) and orexin-A, an orexigenic neuropeptide, in the lateral hypothalamic area (LHA) were studied using immunohistochemistry. RESULTS Compared with sham, IES reduced daily food intake by 28.3% at week 1, 35.6% at week 2, 15.6% at week 3, and 27.1% at week 4. Consistently, IES reduced body weight by 6.3%, compared with a weight gain of 7.2% in sham, and a slight weight loss of 0.5% in pair-feeding. Compared with sham, IES increased the expression of oxytocin-immunoreactive neurons in PVN and SON. Compared with sham, IES decreased the expression of orexin-immunoreactive neurons in LHA. Rats with pair-feeding also showed a relative decease in weight without any changes in the central hormones. CONCLUSION IES reduces food intake and body weight and improves glucose tolerance and insulin sensitivity in DIO rats. Its central mechanisms involve enhancement of anorexigenic peptides and suppression of orexigenic peptides in the hypothalamus.
Collapse
Affiliation(s)
- Shiying Li
- Veterans Research Education Foundation, Oklahoma City Veterans Health Care System, Oklahoma City, OK, USA
- Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI, USA
| | - Yeram Kim
- Veterans Research Education Foundation, Oklahoma City Veterans Health Care System, Oklahoma City, OK, USA
| | - Jiande D Z Chen
- Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI, USA
| | - Mohammad F Madhoun
- Veterans Research Education Foundation, Oklahoma City Veterans Health Care System, Oklahoma City, OK, USA.
- Division of Digestive Diseases and Nutrition, Department of Internal Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK, USA.
| |
Collapse
|
12
|
De la Fuente-Reynoso AL, Barrios De Tomasi E, Juárez J. Differential effects of citalopram on the intake of high fat or high carbohydrates diets in female and male rats. Nutr Neurosci 2021; 25:1477-1487. [PMID: 33427121 DOI: 10.1080/1028415x.2020.1870198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Chronic administration of selective serotonin reuptake inhibitors (SSRI), usually prescribed as antidepressants, decreases total energy intake; however, at present the differential effect on the intake of distinct macronutrients and on female vs. male organisms is not clear. On this basis, female and male adult Wistar rats were exposed to two types of diets: (1) a standard balanced diet (BD); or (2) two types of diets simultaneously, (a) one high in carbohydrates (HC); the other (b) high in fat (HF). Both study groups were given a dose of 10 mg/kg/day i.p. of citalopram or a vehicle for 21 days. Food and water consumption and body weight were recorded daily at baseline (BL), during treatment (TX), and post-treatment (PTx1-PTx2). The male rats exposed to BD reduced total energy consumption during treatment with citalopram, but body weight gain decreased both females and males compared to BL. During exposure to the two types of diets, citalopram treatment reduced fat consumption with respect to BL and PTx1 only in the male group. This group also decreased its total energy consumption during TX compared to PTx1. Finally, the females gained less body weight in TX than PTx1, while weight gain in the males during TX decreased with respect to BL and PTx1. Results show a differential effect of citalopram on females vs. males that was dependent on the type of macronutrient administered.
Collapse
Affiliation(s)
- Amparo L De la Fuente-Reynoso
- Laboratorio de Farmacología y Conducta, Instituto de Neurociencias, CUCBA, Universidad de Guadalajara, Guadalajara, México
| | - Eliana Barrios De Tomasi
- Laboratorio de Farmacología y Conducta, Instituto de Neurociencias, CUCBA, Universidad de Guadalajara, Guadalajara, México
| | - Jorge Juárez
- Laboratorio de Farmacología y Conducta, Instituto de Neurociencias, CUCBA, Universidad de Guadalajara, Guadalajara, México
| |
Collapse
|
13
|
Wang L, Yu CC, Li J, Tian Q, Du YJ. Mechanism of Action of Acupuncture in Obesity: A Perspective From the Hypothalamus. Front Endocrinol (Lausanne) 2021; 12:632324. [PMID: 33868169 PMCID: PMC8050351 DOI: 10.3389/fendo.2021.632324] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 03/16/2021] [Indexed: 01/22/2023] Open
Abstract
Obesity is a prevalent metabolic disease caused by an imbalance in food intake and energy expenditure. Although acupuncture is widely used in the treatment of obesity in a clinical setting, its mechanism has not been adequately elucidated. As the key pivot of appetite signals, the hypothalamus receives afferent and efferent signals from the brainstem and peripheral tissue, leading to the formation of a complex appetite regulation circuit, thereby effectively regulating food intake and energy homeostasis. This review mainly discusses the relationship between the hypothalamic nuclei, related neuropeptides, brainstem, peripheral signals, and obesity, as well as mechanisms of acupuncture on obesity from the perspective of the hypothalamus, exploring the current evidence and therapeutic targets for mechanism of action of acupuncture in obesity.
Collapse
Affiliation(s)
- Li Wang
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Chao-Chao Yu
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- The Fourth Clinical College, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Jia Li
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Qing Tian
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan-Jun Du
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- *Correspondence: Yan-Jun Du,
| |
Collapse
|
14
|
Espinosa-Salinas I, San-Cristobal R, Colmenarejo G, Loria-Kohen V, Molina S, Reglero G, Ramirez de Molina A, Martinez JA. Polymorphic Appetite Effects on Waist Circumference Depend on rs3749474 CLOCK Gene Variant. Nutrients 2020; 12:E1846. [PMID: 32575803 PMCID: PMC7353411 DOI: 10.3390/nu12061846] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/16/2020] [Accepted: 06/18/2020] [Indexed: 11/29/2022] Open
Abstract
Chronobiological aspects controlled by CLOCK genes may influence obesity incidence. Although there are studies that show an association between the expression of these genes and energy intake, waist circumference or abdominal obesity phenotypes, interactions with appetite have been insufficiently investigated in relation to chrononutrition. The objective was to identify interactions between CLOCK genetic variants involved in appetite status. A total of 442 subjects (329 women, 113 men; aged 18 to 65 years) were recruited. Anthropometric, dietary and lifestyle data were collected by trained nutritionists. Participants were classified according to their appetite feelings with a Likert scale. Multiple linear regression models were used to examine associations of the type genotype x appetite status on adiposity-related variables. p values were corrected by the Bonferroni method. A significant influence was found concerning the effects of appetite on waist circumference with respect to rs3749474 CLOCK polymorphism (p < 0.001). An additive model analysis (adjusted by age, gender, exercise and energy intake) showed that risk allele carriers, increased the waist circumference around 14 cm (β = 14.1, CI = 6.3-22.0) by each increment in the level of appetite. The effects of appetite on waist circumference may be partly modulated by the rs3749474 CLOCK polymorphism.
Collapse
Affiliation(s)
- Isabel Espinosa-Salinas
- IMDEA-Food Institute, CEI UAM+CSIC, 28049 Madrid, Spain; (R.S.-C.); (G.C.); (V.L.-K.); (S.M.); (G.R.); (J.A.M.)
| | - Rodrigo San-Cristobal
- IMDEA-Food Institute, CEI UAM+CSIC, 28049 Madrid, Spain; (R.S.-C.); (G.C.); (V.L.-K.); (S.M.); (G.R.); (J.A.M.)
| | - Gonzalo Colmenarejo
- IMDEA-Food Institute, CEI UAM+CSIC, 28049 Madrid, Spain; (R.S.-C.); (G.C.); (V.L.-K.); (S.M.); (G.R.); (J.A.M.)
| | - Viviana Loria-Kohen
- IMDEA-Food Institute, CEI UAM+CSIC, 28049 Madrid, Spain; (R.S.-C.); (G.C.); (V.L.-K.); (S.M.); (G.R.); (J.A.M.)
| | - Susana Molina
- IMDEA-Food Institute, CEI UAM+CSIC, 28049 Madrid, Spain; (R.S.-C.); (G.C.); (V.L.-K.); (S.M.); (G.R.); (J.A.M.)
| | - Guillermo Reglero
- IMDEA-Food Institute, CEI UAM+CSIC, 28049 Madrid, Spain; (R.S.-C.); (G.C.); (V.L.-K.); (S.M.); (G.R.); (J.A.M.)
- Institute of Food Science Research (CIAL) CEI UAM+CSIC, 28049 Madrid, Spain
| | - Ana Ramirez de Molina
- IMDEA-Food Institute, CEI UAM+CSIC, 28049 Madrid, Spain; (R.S.-C.); (G.C.); (V.L.-K.); (S.M.); (G.R.); (J.A.M.)
| | - J. Alfredo Martinez
- IMDEA-Food Institute, CEI UAM+CSIC, 28049 Madrid, Spain; (R.S.-C.); (G.C.); (V.L.-K.); (S.M.); (G.R.); (J.A.M.)
- Center for Nutrition Research (CIN), Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
- Center of Biomedical Research in Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| |
Collapse
|
15
|
Duque AP, Rodrigues Junior LF, Mediano MFF, Tibiriça E, De Lorenzo A. Emerging concepts in metabolically healthy obesity. AMERICAN JOURNAL OF CARDIOVASCULAR DISEASE 2020; 10:48-61. [PMID: 32685263 PMCID: PMC7364278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 05/15/2020] [Indexed: 06/11/2023]
Abstract
Obesity is a major risk factor for noncommunicable diseases that is responsible for more than 70% of early deaths in the world. In the 1980's decade, some studies started to describe a "benign" obesity phenotype, named "metabolically healthy obesity" (MHO), which represents obesity without comorbidities such as hypertension, cardiovascular diseases, insulin resistance, diabetes, dyslipidemia or metabolic syndrome. However, it is still unclear if this "benign" obesity phenotype is really favorable or just a transition status to unhealthy obesity and if these subjects presented subclinical levels of cardiovascular risk that are not commonly detected. To further elucidate these issues, the investigation of pathophysiological mechanisms that can increase cardiovascular risk in MHO individuals, such as hormones and cytokines, may offer some responses. In parallel, the evaluation of subclinical cardiovascular derangement, using the systemic microcirculation as a proxy, may be an alternative to anticipate overt cardiovascular disease. Overall, further studies are needed to better understand the pathophysiology of MHO as well as to identify high-risk individuals who deserve more intensive management.
Collapse
Affiliation(s)
- Alice P Duque
- Department of Research and Education, National Institute of CardiologyRio de Janeiro, RJ, Brazil
| | - Luiz F Rodrigues Junior
- Department of Research and Education, National Institute of CardiologyRio de Janeiro, RJ, Brazil
- Department of Physiological Sciences, Biomedical Institute, Federal University of The State of Rio de JaneiroRio de Janeiro, RJ, Brazil
| | - Mauro F F Mediano
- Department of Research and Education, National Institute of CardiologyRio de Janeiro, RJ, Brazil
- Evandro Chagas National Institute of Infectious Disease, Oswaldo Cruz FoundationRio de Janeiro, RJ, Brazil
| | - Eduardo Tibiriça
- Department of Research and Education, National Institute of CardiologyRio de Janeiro, RJ, Brazil
| | - Andrea De Lorenzo
- Department of Research and Education, National Institute of CardiologyRio de Janeiro, RJ, Brazil
| |
Collapse
|
16
|
Falls BA, Zhang Y. Insights into the Allosteric Mechanism of Setmelanotide (RM-493) as a Potent and First-in-Class Melanocortin-4 Receptor (MC4R) Agonist To Treat Rare Genetic Disorders of Obesity through an in Silico Approach. ACS Chem Neurosci 2019; 10:1055-1065. [PMID: 30048591 DOI: 10.1021/acschemneuro.8b00346] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Human melanocortin-4 receptor (hMC4R) mutations have been implicated as the cause for about 6-8% of all severe obesity cases. Drug-like molecules that are able to rescue the functional activity of mutated receptors are highly desirable to combat genetic obesity among this population of patients. One such molecule is the selective MC4R agonist RM-493 (setmelanotide). While this molecule has been shown to activate mutated receptors with 20-fold higher potency over the endogenous agonist, little is known about its binding mode and how it effectively interacts with hMC4R despite the presence of mutations. In this study, a MC4R homology model was constructed based on the X-ray crystal structure of the adenosine A2A receptor in the active state. Four MC4R mutations commonly found in genetically obese patients and known to effect ligand binding in vitro were introduced into the constructed model. RM-493 was then docked into the wild-type and mutated models in order to better elucidate the possible binding modes for this promising drug candidate and assess how it may be interacting with MC4R to effectively activate receptor polymorphisms. The results reflected the orthosteric interactions of both the endogenous and synthetic ligands with the MC4R, which is supported by the site-directed mutagenesis studies. Meanwhile it helped explain the decremental affinity and potency of these ligands with the receptor polymorphisms. More significantly, our findings indicated that the structural characteristics of RM-493 may allow for enhanced receptor-ligand interactions, particularly through those with the putative allosteric binding sites, which facilitated the ligand to stabilize the active state of native and mutant MC4Rs to maintain reasonably high affinity and potency.
Collapse
Affiliation(s)
- Bethany A. Falls
- Department of Medicinal Chemistry, Virginia Commonwealth University, 800 East Leigh Street, Richmond, Virginia 23298, United States
| | - Yan Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University, 800 East Leigh Street, Richmond, Virginia 23298, United States
| |
Collapse
|
17
|
Minakova E, Lang J, Medel-Matus JS, Gould GG, Reynolds A, Shin D, Mazarati A, Sankar R. Melanotan-II reverses autistic features in a maternal immune activation mouse model of autism. PLoS One 2019; 14:e0210389. [PMID: 30629642 PMCID: PMC6328175 DOI: 10.1371/journal.pone.0210389] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 12/21/2018] [Indexed: 12/19/2022] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder characterized by impaired social interactions, difficulty with communication, and repetitive behavior patterns. In humans affected by ASD, there is a male pre-disposition towards the condition with a male to female ratio of 4:1. In part due to the complex etiology of ASD including genetic and environmental interplay, there are currently no available medical therapies to improve the social deficits of ASD. Studies in rodent models and humans have shown promising therapeutic effects of oxytocin in modulating social adaptation. One pharmacological approach to stimulating oxytocinergic activity is the melanocortin receptor 4 agonist Melanotan-II (MT-II). Notably the effects of oxytocin on environmental rodent autism models has not been investigated to date. We used a maternal immune activation (MIA) mouse model of autism to assess the therapeutic potential of MT-II on autism-like features in adult male mice. The male MIA mice exhibited autism-like features including impaired social behavioral metrics, diminished vocal communication, and increased repetitive behaviors. Continuous administration of MT-II to male MIA mice over a seven-day course resulted in rescue of social behavioral metrics. Normal background C57 male mice treated with MT-II showed no significant alteration in social behavioral metrics. Additionally, there was no change in anxiety-like or repetitive behaviors following MT-II treatment of normal C57 mice, though there was significant weight loss following subacute treatment. These data demonstrate MT-II as an effective agent for improving autism-like behavioral deficits in the adult male MIA mouse model of autism.
Collapse
Affiliation(s)
- Elena Minakova
- Department of Pediatrics, Division of Neonatology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Jordan Lang
- Department of Internal Medicine, Huntington Memorial Hospital, Pasadena, California, United States of America
| | - Jesus-Servando Medel-Matus
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Georgianna G. Gould
- University of Texas Health Science Center at San Antonio, Department of Cellular and Integrative Physiology, San Antonio, Texas, United States of America
| | - Ashley Reynolds
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Don Shin
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Andrey Mazarati
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Children's Discovery and Innovation Institute at UCLA, Los Angeles, California, United States of America
| | - Raman Sankar
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Children's Discovery and Innovation Institute at UCLA, Los Angeles, California, United States of America
| |
Collapse
|
18
|
Yu Y, Park SJ, Beyak MJ. Inducible nitric oxide synthase-derived nitric oxide reduces vagal satiety signalling in obese mice. J Physiol 2018; 597:1487-1502. [PMID: 30565225 DOI: 10.1113/jp276894] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 11/19/2018] [Indexed: 12/22/2022] Open
Abstract
KEY POINTS Obesity is associated with disrupted satiety regulation. Mice with diet-induced obesity have reduced vagal afferent neuronal excitability and a decreased afferent response to satiety signals. A low grade inflammation occurs in obesity with increased expression of inducible nitric oxide synthase (iNOS). Inhibition of iNOS in diet-induced obese mice restored vagal afferent neuronal excitability, increased the afferent response to satiety mediators and distention of the gut, and reduced short-term energy intake. A prolonged inhibition of iNOS reduced energy intake and body weight gain during the first week, and reduced amounts of epididymal fat after 3 weeks. We identified a novel pathway underlying disrupted satiety regulation in obesity. Blocking of this pathway might be clinically useful for the management of obesity. ABSTRACT Vagal afferents regulate feeding by transmitting satiety signals to the brain. Mice with diet-induced obesity have reduced vagal afferent sensitivity to satiety signals. We investigated whether inducible nitric oxide synthase (iNOS)-derived NO contributed to this reduction. C57BL/6J mice were fed a high- or low-fat diet for 6-8 weeks. Nodose ganglia and jejunum were analysed by immunoblotting for iNOS expression; NO production was measured using a fluorometric assay. Nodose neuron excitability and intestinal afferent sensitivity were evaluated by whole-cell patch clamp and in vitro afferent recording, respectively. Expression of iNOS and production of NO were increased in nodose ganglia and the small intestine in obese mice. Inhibition of iNOS in obese mice by pre-treatment with an iNOS inhibitor increased nodose neuron excitability via 2-pore-domain K+ channel leak currents, restored afferent sensitivity to satiety signals and reduced short-term energy intake. Obese mice given the iNOS inhibitor daily for 3 weeks had reduced energy intake and decreased body weight gain during the first week, compared to mice given saline, and lower amounts of epididymal fat at the end of 3 weeks. Inhibition of iNOS or blocking the action of iNOS-derived NO on vagal afferent pathways might comprise therapeutic strategies for hyperphagia and obesity.
Collapse
Affiliation(s)
- Yang Yu
- Gastrointestinal Disease Research Unit, Kingston General Hospital, Queen's University, Kingston, ON, Canada
| | - Sung Jin Park
- Gastrointestinal Disease Research Unit, Kingston General Hospital, Queen's University, Kingston, ON, Canada
| | - Michael J Beyak
- Gastrointestinal Disease Research Unit, Kingston General Hospital, Queen's University, Kingston, ON, Canada
| |
Collapse
|
19
|
Bliss ES, Whiteside E. The Gut-Brain Axis, the Human Gut Microbiota and Their Integration in the Development of Obesity. Front Physiol 2018; 9:900. [PMID: 30050464 PMCID: PMC6052131 DOI: 10.3389/fphys.2018.00900] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 06/21/2018] [Indexed: 12/17/2022] Open
Abstract
Obesity is a global epidemic, placing socioeconomic strain on public healthcare systems, especially within the so-called Western countries, such as Australia, United States, United Kingdom, and Canada. Obesity results from an imbalance between energy intake and energy expenditure, where energy intake exceeds expenditure. Current non-invasive treatments lack efficacy in combating obesity, suggesting that obesity is a multi-faceted and more complex disease than previously thought. This has led to an increase in research exploring energy homeostasis and the discovery of a complex bidirectional communication axis referred to as the gut-brain axis. The gut-brain axis is comprised of various neurohumoral components that allow the gut and brain to communicate with each other. Communication occurs within the axis via local, paracrine and/or endocrine mechanisms involving a variety of gut-derived peptides produced from enteroendocrine cells (EECs), including glucagon-like peptide 1 (GLP1), cholecystokinin (CCK), peptide YY3-36 (PYY), pancreatic polypeptide (PP), and oxyntomodulin. Neural networks, such as the enteric nervous system (ENS) and vagus nerve also convey information within the gut-brain axis. Emerging evidence suggests the human gut microbiota, a complex ecosystem residing in the gastrointestinal tract (GIT), may influence weight-gain through several inter-dependent pathways including energy harvesting, short-chain fatty-acids (SCFA) signalling, behaviour modifications, controlling satiety and modulating inflammatory responses within the host. Hence, the gut-brain axis, the microbiota and the link between these elements and the role each plays in either promoting or regulating energy and thereby contributing to obesity will be explored in this review.
Collapse
Affiliation(s)
- Edward S. Bliss
- School of Health and Wellbeing, University of Southern Queensland, Toowoomba, QLD, Australia
| | | |
Collapse
|
20
|
Cardiovascular Outcome Trials of Diabetes and Obesity Drugs: Implications for Conditional Approval and Early Phase Clinical Development. Pharmaceut Med 2017. [DOI: 10.1007/s40290-017-0209-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
21
|
Krentz AJ, Fujioka K, Hompesch M. Evolution of pharmacological obesity treatments: focus on adverse side-effect profiles. Diabetes Obes Metab 2016; 18:558-70. [PMID: 26936802 DOI: 10.1111/dom.12657] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 12/14/2015] [Accepted: 02/27/2016] [Indexed: 12/19/2022]
Abstract
Pharmacotherapy directed toward reducing body weight may provide benefits for both curbing obesity and lowering the risk of obesity-associated comorbidities; however, many weight loss medications have been withdrawn from the market because of serious adverse effects. Examples include pulmonary hypertension (aminorex), cardiovascular toxicity, e.g. flenfluramine-induced valvopathy, stroke [phenylpropanolamine (PPA)], excess non-fatal cardiovascular events (sibutramine), and neuro-psychiatric issues (rimonabant; approved in Europe, but not in the USA). This negative experience has helped mould the current drug development and approval process for new anti-obesity drugs. Differences between the US Food and Drug Administration (FDA) and the European Medicines Agency, however, in perceptions of risk-benefit considerations for individual drugs have resulted in discrepancies in approval and/or withdrawal of weight-reducing medications. Thus, two drugs recently approved by the FDA, i.e. lorcaserin and phentermine + topiramate extended release, are not available in Europe. In contrast, naltrexone sustained release (SR)/bupropion SR received FDA approval, and liraglutide 3.0 mg was recently approved in both the USA and Europe. Regulatory strategies adopted by the FDA to manage the potential for uncommon but potentially serious post-marketing toxicity include: (i) risk evaluation and mitigation strategy programmes; (ii) stipulating post-marketing safety trials; (iii) considering responder rates and limiting cumulative exposure by discontinuation if weight loss is not attained within a reasonable timeframe; and (iv) requiring large cardiovascular outcome trials before or after approval. We chronicle the adverse effects of anti-obesity pharmacotherapy and consider how the history of high-profile toxicity issues has shaped the current regulatory landscape for new and future weight-reducing drugs.
Collapse
Affiliation(s)
- A J Krentz
- Profil Institute for Clinical Research, Chula Vista, CA, USA
| | - K Fujioka
- Center for Weight Management, Department of Diabetes and Endocrinology, Scripps Clinic Del Mar, San Diego, CA, USA
| | - M Hompesch
- Profil Institute for Clinical Research, Chula Vista, CA, USA
| |
Collapse
|
22
|
Wang S, Xiang N, Yang L, Zhu C, Zhu X, Wang L, Gao P, Xi Q, Zhang Y, Shu G, Jiang Q. Linoleic acid and stearic acid elicit opposite effects on AgRP expression and secretion via TLR4-dependent signaling pathways in immortalized hypothalamic N38 cells. Biochem Biophys Res Commun 2016; 471:566-71. [PMID: 26879142 DOI: 10.1016/j.bbrc.2016.02.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 02/09/2016] [Indexed: 12/28/2022]
|
23
|
Buhmann H, le Roux CW, Bueter M. The gut-brain axis in obesity. Best Pract Res Clin Gastroenterol 2014; 28:559-71. [PMID: 25194175 DOI: 10.1016/j.bpg.2014.07.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Revised: 06/11/2014] [Accepted: 07/05/2014] [Indexed: 01/31/2023]
Abstract
Currently the only effective treatment for morbid obesity with a proven mortality benefit is surgical intervention. The underlying mechanisms of these surgical techniques are unclear, but alterations in circulating gut hormone levels have been demonstrated to be at least one contributing factor. Gut hormones seem to communicate information from the gastrointestinal tract to the regulatory appetite centres within the central nervous system (CNS) via the so-called 'Gut-Brain-Axis'. Such information may be transferred to the CNS either via vagal or non-vagal afferent nerve signalling or directly via blood circulation. Complex neural networks, distributed throughout the forebrain and brainstem, are in control of feeding and energy homoeostasis. This article aims to review how appetite is potentially regulated by these gastrointestinal hormones. Identification of the underlying mechanisms of appetite and weight control may pave the way to develop better surgical techniques and new therapies in the future.
Collapse
Affiliation(s)
- Helena Buhmann
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Switzerland
| | - Carel W le Roux
- Diabetes Complications Research Centre, Conway Institute, School of Medicine and Medical Sciences, University College Dublin, Ireland; Gastrosurgical Laboratory, University of Gothenburg, Sweden
| | - Marco Bueter
- Department of Surgery, Division of Visceral and Transplantation Surgery, University Hospital Zurich, Zurich, Switzerland; Center of Integrative Human Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
24
|
Valsamakis G, Lois K, Kumar S, Mastorakos G. New molecular targets in the pathophysiology of obesity and available treatment options under investigation. Clin Obes 2014; 4:209-19. [PMID: 25826792 DOI: 10.1111/cob.12064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 05/12/2014] [Accepted: 05/14/2014] [Indexed: 12/22/2022]
Abstract
The pharmacotherapy of obesity has historically recorded an overall poor safety and efficacy profile largely because of the complex mechanisms involved in the pathophysiology of obesity. It is hoped that a better understanding of the regulation of body weight will lead us to the development of effective and safer drugs. Recent advances in our understanding of the regulation of energy homeostasis has allowed the design of novel anti-obesity drugs targeting specific molecules crucial for the modulation of energy balance, including drugs that induce satiety, modulate nutrient absorption or influence metabolism or lipogenesis. Almost a decade after the Food and Drug Administration approved the first weight loss medication, it recently approved two novel anti-obesity drugs Belviq (lorcaserin) and Qsymia (topiramate and phentermine), thus signalling the beginning of a new era in the pharmacotherapy of obesity. It is believed that the next generation of weight-loss drugs will be based on combination treatments with gut hormones in a manner that mimics the changes underlying surgically induced weight loss thus introducing the so called 'bariatric pharmacotherapy'. An in-depth understanding of the interrelated physiological and behavioural effects of these new molecules together with the development of new treatment paradigms is needed so that future disappointments in the field of obesity pharmacotherapy may be avoided.
Collapse
Affiliation(s)
- G Valsamakis
- Endocrine Unit, 2nd Department of Obs and Gynae, Areteeion University Hospital, Athens Medical School National and Kapodistrian University of Athens, Athens, Greece; WISDEM Centre for Diabetes, Endocrinology and Metabolism, Warwick Medical School, University of Warwick, Coventry, UK
| | | | | | | |
Collapse
|
25
|
Bossowski A, Czarnocka B, Harasymczuk J, Moniuszko A, Bardadin K, Lyczkowska A, Hanusek K, Bossowska A. Identification of GPR39 receptor and ghrelin receptor in thyroid tissues in paediatric patients with immune and non-immune thyroid diseases. Horm Res Paediatr 2013; 79:130-6. [PMID: 23485550 DOI: 10.1159/000347218] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 01/17/2013] [Indexed: 11/19/2022] Open
Abstract
The preproghrelin gene is responsible for generating ghrelin and obestatin, two gastric peptides with opposite effects on food intake. Obestatin suppresses food intake and digestive motility through interaction with GPR39 (GPCR). Ghrelin is supposed to be a link connecting metabolism and energy homeostasis with growth as the result of activation of the growth hormone secretagogue receptor (GHSR).The aim of the current study was to assess the expression of preproghrelin, GPR39 and GHSR in thyroid tissues from patients with Graves' disease (GD; n = 15), non-toxic nodular goiter (NTNG; n = 10) and toxic nodular goiter (TNG; n = 10). GPR39 and GHSR in thyroid tissues were detected by immunohistochemistry and Western blot, revealing higher expression of both proteins in GD patients (+++; ++) in comparison with NTNG (+; +) and TNG (++; +) patients. GPR39 was present in thyroid autoimmune disease, NTNG and TNG at band p51 (kDa). The ghrelin receptor was identified in all study groups at p70. mRNA expression for preproghrelin was found in thyroid tissues from patients with immune and non-immune thyroid diseases. We conclude that the expression of the ghrelin receptor family in thyroid tissues may suggest a role of gastric peptides in thyroid functions. mRNA of preproghrelin expression is a proof of ghrelin gene-derived peptide presence in thyroid tissues.
Collapse
Affiliation(s)
- Artur Bossowski
- Department of Paediatric Endocrinology, Diabetology with Cardiology Division, Medical University of Białystok, Białystok, Poland.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
NESS06SM reduces body weight with an improved profile relative to SR141716A. Pharmacol Res 2013; 74:94-108. [PMID: 23756200 DOI: 10.1016/j.phrs.2013.06.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 06/01/2013] [Accepted: 06/02/2013] [Indexed: 01/21/2023]
Abstract
We have recently synthesized a new series of 4,5-dihydrobenzo-oxa-cycloheptapyrazole derivatives with the aim to discover novel CB1 antagonist agents characterized by anti-obesity activity comparable to that of SR141716A but with reduced adverse effects such as anxiety and depression. Within the novel class, the CB1 antagonist 8-chloro-1-(2,4-dichlorophenyl)-N-piperidin-1-yl-4,5-dihydrobenzo-1H-6-oxa-cyclohepta(1,2-c)pyrazole-3-carboxamide (NESS06SM) has been selected as lead compound. We found that NESS06SM is a CB1 neutral antagonist, characterized by poor blood-brain barrier permeability. Moreover, NESS06SM chronic treatment determined both anti-obesity effect and cardiovascular risk factor improvement in C57BL/6N Diet Induced Obesity (DIO) mice fed with fat diet (FD mice). In fact, the mRNA gene expression in Central Nervous System (CNS) and peripheral tissues by real time PCR, showed a significant increase of orexigenic peptides and a decrease of anorexigenic peptides elicited by NESS06SM treatment, compared to control mice fed with the same diet. Moreover, in contrast to SR141716A treatment, the chronic administration of NESS06SM did not change mRNA expression of both monoaminergic transporters and neurotrophins highly related with anxiety and mood disorders. Our results suggest that NESS06SM reduces body weight and it can restore the disrupted expression profile of genes linked to the hunger-satiety circuit without altering monoaminergic transmission probably avoiding SR141716A side effects. Therefore the novel CB1 neutral antagonist could represent a useful candidate agent for the treatment of obesity and its metabolic complications.
Collapse
|
27
|
Kim GW, Lin JE, Waldman SA. GUCY2C: at the intersection of obesity and cancer. Trends Endocrinol Metab 2013; 24:165-73. [PMID: 23375388 PMCID: PMC3617062 DOI: 10.1016/j.tem.2013.01.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 12/26/2012] [Accepted: 01/02/2013] [Indexed: 12/13/2022]
Abstract
Guanylyl cyclase C (GUCY2C) has canonical centrality in defense of key intestinal homeostatic mechanisms, encompassing fluid and electrolyte balance, epithelial dynamics, antitumorigenesis, and intestinal barrier function. Recent discoveries expand the homeostatic role of GUCY2C to reveal a novel gut-brain endocrine axis regulating appetite, anchored by hypothalamic GUCY2C which is responsive to intestine-derived uroguanylin. Thus, GUCY2C may represent a new target for anti-obesity pharmacotherapy. Moreover, the coincident regulation of energy balance and tumor suppression by a single hormone receptor system suggests that the GUCY2C axis might contribute to the established relationship between obesity and colorectal cancer. This confluence suggests that hormone supplementation to reconstitute GUCY2C signaling may be an elegant strategy to reverse both pathophysiologic processes.
Collapse
Affiliation(s)
- Gilbert W Kim
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | |
Collapse
|
28
|
Ceccarelli E, Guarino EG, Merlotti D, Patti A, Gennari L, Nuti R, Dotta F. Beyond glycemic control in diabetes mellitus: effects of incretin-based therapies on bone metabolism. Front Endocrinol (Lausanne) 2013; 4:73. [PMID: 23785355 PMCID: PMC3684850 DOI: 10.3389/fendo.2013.00073] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 06/03/2013] [Indexed: 12/31/2022] Open
Abstract
Diabetes mellitus (DM) and osteoporosis (OP) are common disorders with a significant health burden, and an increase in fracture risk has been described both in type 1 (T1DM) and in type 2 (T2DM) diabetes. The pathogenic mechanisms of impaired skeletal strength in diabetes remain to be clarified in details and they are only in part reflected by a variation in bone mineral density. In T2DM, the occurrence of low bone turnover together with a decreased osteoblast activity and compromised bone quality has been shown. Of note, some antidiabetic drugs (e.g., thiazolidinediones, insulin) may deeply affect bone metabolism. In addition, the recently introduced class of incretin-based drugs (i.e., GLP-1 receptor agonists and DPP-4 inhibitors) is expected to exert potentially beneficial effects on bone health, possibly due to a bone anabolic activity of GLP-1, that can be either direct or indirect through the involvement of thyroid C cells. Here we will review the established as well as the putative effects of incretin hormones and of incretin-based drugs on bone metabolism, both in preclinical models and in man, taking into account that such therapeutic strategy may be effective not only to achieve a good glycemic control, but also to improve bone health in diabetic patients.
Collapse
Affiliation(s)
- Elena Ceccarelli
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Elisa G. Guarino
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Daniela Merlotti
- Internal Medicine Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Aurora Patti
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
- Internal Medicine Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Luigi Gennari
- Internal Medicine Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Ranuccio Nuti
- Internal Medicine Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Francesco Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
- Fondazione Umberto Di Mario ONLUS, Siena, Italy
- *Correspondence: Francesco Dotta, U.O.C. Diabetologia, University of Siena, Policlinico Le Scotte, Viale Bracci 18, Siena 53100, Italy e-mail:
| |
Collapse
|
29
|
Abstract
Drugs that augment the incretin system [glucagon like peptide (GLP) agonists and dipeptidyl peptidase-4 (DPP-4) inhibitors] represent a novel class of anti-hyperglycemic agents that have shown to improve the health and survival of beta-cells (improvement in postprandial hyperglycemia) and suppress glucagon (improvement in fasting hyperglycemia). The incretins represent a large family of molecules referred to as the "glucagon superfamily of peptide hormones" of which more than 90% of the physiological effects of incretins are accomplished by GLP-1(7-37) and GLP1(7-36) amide and gastric insulinotropic peptide (GIP). GLP-1 mediates its effects via the GLP-1 receptor, which has a wide tissue distribution [pancreas, lung, heart, vascular smooth muscle cells, endothelial cells, macrophages and monocytes, kidney, gastrointestinal tract (stomach and intestine), central nervous system (neoortex, cerebellum, hypothalamus, hippocampus, brainstem nucleus tractus solitarius) and peripheral nervous system]. This would imply that the incretin system has effects outside the pancreas. Over time data has accumulated to suggest that therapies that augment the incretin system has beneficial pleiotrophic effects. The incretins have shown to possess a cardiac-friendly profile, preserve neuronal cells and safeguard from neuronal degeneration, improve hepatic inflammation and hepatosteatosis, improve insulin resistance, promote weight loss and induce satiety. There is growing evidence that they may also be renoprotective promoting wound healing and bone health.
Collapse
Affiliation(s)
- Vishal Gupta
- Department of Endocrinology, Jaslok Hospital and Research Centre, Mumbai, India
| |
Collapse
|
30
|
Tratamiento farmacológico de la obesidad. REVISTA MÉDICA CLÍNICA LAS CONDES 2012. [DOI: 10.1016/s0716-8640(12)70295-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|