1
|
Gao T, Li R, Hu L, Hu Q, Wen H, Zhou R, Yuan P, Zhang X, Huang L, Zhuo Y, Xu S, Lin Y, Feng B, Che L, Wu D, Fang Z. Probiotic Lactobacillus rhamnosus GG improves insulin sensitivity and offspring survival via modulation of gut microbiota and serum metabolite in a sow model. J Anim Sci Biotechnol 2024; 15:89. [PMID: 38951898 PMCID: PMC11218078 DOI: 10.1186/s40104-024-01046-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/07/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND Sows commonly experience insulin resistance in late gestation and lactation, causing lower feed intake and milk production, which can lead to higher mortality rates in newborn piglets. The probiotic Lactobacillus rhamnosus GG (LGG) is known to improve insulin resistance. However, whether supplementing LGG can improve insulin sensitivity in sows and enhance lactation performance, particularly the early survival of offspring remains unclear. Hence, we explored the effects and mechanisms of supplementing LGG during late gestation and lactation on sow insulin sensitivity, lactation performance, and offspring survival. In total, 20 sows were randomly allocated to an LGG (n = 10) and control group (n = 10). RESULTS In sows, LGG supplementation significantly improved insulin sensitivity during late gestation and lactation, increased feed intake, milk production and colostrum lactose levels in early lactation, and enhanced newborn piglet survival. Moreover, LGG treatment significantly reshaped the gut microbiota in sows, notably increasing microbiota diversity and enriching the relative abundance of insulin sensitivity-associated probiotics such as Lactobacillus, Bifidobacterium, and Bacteroides. Serum metabolite and amino acid profiling in late-gestation sows also revealed decreased branched-chain amino acid and kynurenine serum levels following LGG supplementation. Further analyses highlighted a correlation between mitigated insulin resistance in late pregnancy and lactation by LGG and gut microbiota reshaping and changes in serum amino acid metabolism. Furthermore, maternal LGG enhanced immunity in newborn piglets, reduced inflammation, and facilitated the establishment of a gut microbiota. CONCLUSIONS We provide the first evidence that LGG mitigates insulin resistance in sows and enhances offspring survival by modulating the gut microbiota and amino acid metabolism.
Collapse
Affiliation(s)
- Tianle Gao
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Ran Li
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
- Key Laboratory of Agricultural Product Processing and Nutrition Health (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Food Science, Sichuan Agricultural University, Ya' an, 625014, China
| | - Liang Hu
- Key Laboratory of Agricultural Product Processing and Nutrition Health (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Food Science, Sichuan Agricultural University, Ya' an, 625014, China
| | - Quanfang Hu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Hongmei Wen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Rui Zhou
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Peiqiang Yuan
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Xiaoling Zhang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Lingjie Huang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Yong Zhuo
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Shengyu Xu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Yan Lin
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Bin Feng
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Lianqiang Che
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - De Wu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Zhengfeng Fang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China.
| |
Collapse
|
2
|
Carbone F, Colamatteo A, La Rocca C, Lepore MT, Russo C, De Rosa G, Matarese A, Procaccini C, Matarese G. Metabolic Plasticity of Regulatory T Cells in Health and Autoimmunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1859-1866. [PMID: 38830147 DOI: 10.4049/jimmunol.2400079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/05/2024] [Indexed: 06/05/2024]
Abstract
Immunometabolism has been demonstrated to control immune tolerance and the pathogenic events leading to autoimmunity. Compelling experimental evidence also suggests that intracellular metabolic programs influence differentiation, phenotype, proliferation, and effector functions of anti-inflammatory CD4+CD25+Foxp3+ regulatory T (Treg) cells. Indeed, alterations in intracellular metabolism associate with quantitative and qualitative impairments of Treg cells in several pathological conditions. In this review, we summarize the most recent advances linking how metabolic pathways control Treg cell homeostasis and their alterations occurring in autoimmunity. Also, we analyze how metabolic manipulations could be employed to restore Treg cell frequency and function with the aim to create novel therapeutic opportunities to halt immune-mediated disorders.
Collapse
Grants
- 2022LNHZAP Ministero dell''''Istruzione, dell''''Università e della Ricerca (MIUR)
- PE00000007 Ministero dell''''Istruzione, dell''''Università e della Ricerca (MIUR)
- PE00000006 Ministero dell''''Istruzione, dell''''Università e della Ricerca (MIUR)
- RF-2019-12371111 Italy Ministry of Health | Agenzia Italiana del Farmaco, Ministero della Salute (AIFA)
- PNRR-MAD-2022-12375634 Italy Ministry of Health | Agenzia Italiana del Farmaco, Ministero della Salute (AIFA)
- GR-2018-12366154 Italy Ministry of Health | Agenzia Italiana del Farmaco, Ministero della Salute (AIFA)
- 2022-PRsingle/013 Fondazione Italiana Sclerosi Multipla (FISM)
- P2022T4PKT Ministero dell''''Istruzione, dell''''Università e della Ricerca (MIUR)
- PNRR-MAD-2022-12376126 Italy Ministry of Health | Agenzia Italiana del Farmaco, Ministero della Salute (AIFA)
- GR-2021-12373337 Italy Ministry of Health | Agenzia Italiana del Farmaco, Ministero della Salute (AIFA)
- 2022YMJXYT Ministero dell''''Istruzione, dell''''Università e della Ricerca (MIUR)
- P2022CMK43 Ministero dell''''Istruzione, dell''''Università e della Ricerca (MIUR)
- 20225KH7BZ Ministero dell''''Istruzione, dell''''Università e della Ricerca (MIUR)
Collapse
Affiliation(s)
- Fortunata Carbone
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore," Consiglio Nazionale delle Ricerche, Napoli, Italy
- Unità di Neuroimmunologia, IRCCS-Fondazione Santa Lucia, Roma, Italy
| | - Alessandra Colamatteo
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II," Napoli, Italy
| | - Claudia La Rocca
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore," Consiglio Nazionale delle Ricerche, Napoli, Italy
| | - Maria Teresa Lepore
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore," Consiglio Nazionale delle Ricerche, Napoli, Italy
| | - Claudia Russo
- D.A.I. Medicina di Laboratorio e Trasfusionale, Azienda Ospedaliera Universitaria "Federico II," Napoli, Italy
| | - Giusy De Rosa
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II," Napoli, Italy
| | - Alessandro Matarese
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli "Federico II," Napoli, Italy
| | - Claudio Procaccini
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore," Consiglio Nazionale delle Ricerche, Napoli, Italy
- Unità di Neuroimmunologia, IRCCS-Fondazione Santa Lucia, Roma, Italy
| | - Giuseppe Matarese
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia Sperimentale "G. Salvatore," Consiglio Nazionale delle Ricerche, Napoli, Italy
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II," Napoli, Italy
| |
Collapse
|
3
|
Ling MTM, Govindaraju K, Lokanathan Y, Abidin AZ, Ibrahim B. Mesenchymal stem cell-derived extracellular vesicles for metabolic syndrome therapy: Assessing efficacy with nuclear magnetic resonance spectroscopy. Cell Biochem Funct 2023; 41:1044-1059. [PMID: 37933415 DOI: 10.1002/cbf.3881] [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: 05/27/2023] [Revised: 09/27/2023] [Accepted: 10/21/2023] [Indexed: 11/08/2023]
Abstract
Metabolic syndrome (MetS) represents a cluster of metabolic abnormalities. The prevalence of MetS has surged, transforming it into a pressing public health concern that could potentially affect around 20%-25% of the global population. As MetS continues its ascent, diverse interventions, pharmacological, nonpharmacological and combined have been deployed. Yet, a comprehensive remedy that fully eradicates MetS symptoms remains elusive, compounded by the risks of polypharmacy's emergence. Acknowledging the imperative to grasp MetS's intricate pathologies, deeper insights for future research and therapy optimisation become paramount. Conventional treatments often target specific syndrome elements. However, a novel approach emerges in mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) therapy, promising a holistic shift. MSC-EVs, tiny membranous vesicles secreted by mesenchymal stem cells, have garnered immense attention for their multifaceted bioactivity and regenerative potential. Their ability to modulate inflammation, enhance tissue repair and regulate metabolic pathways has prompted researchers to explore their therapeutic application in MetS. This review primarily aims to provide an overview of how MSC-EVs therapy can improve metabolic parameters in subjects with MetS disease and also introduce the usefulness of NMR spectroscopy in assessing the efficacy of MSC-EVs therapy for treating MetS.
Collapse
Affiliation(s)
- Magdalene Tan Mei Ling
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Kayatri Govindaraju
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Yogeswaran Lokanathan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Asmaa' Zainal Abidin
- Department of Chemistry and Biology, Centre for Defense Foundation Studies, Universiti Pertahanan Nasional Malaysia, Kuala Lumpur, Malaysia
| | - Baharudin Ibrahim
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Universiti Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
4
|
Webster CM, Mittal N, Dhurandhar EJ, Dhurandhar NV. Potential contributors to variation in weight-loss response to liraglutide. Obes Rev 2023:e13568. [PMID: 37069131 DOI: 10.1111/obr.13568] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 03/02/2023] [Accepted: 03/30/2023] [Indexed: 04/19/2023]
Abstract
Obesity treatment requires a chronic state of negative energy balance. Obesity medications can help with this, increasing long-term dietary compliance by promoting satiety or reducing hunger. However, efficacy and safety of obesity medications vary for individuals. Early identification of non-responders to obesity medications may limit drug exposure while optimizing benefits for responders. This review summarizes factors that impact weight-loss response to liraglutide. Factors linked to greater weight loss on liraglutide include being female, not having diabetes, having relatively high baseline weight, and losing at least 4% of initial weight after 16 weeks of treatment. Other covariates that may predict treatment response but require further confirmation include central effects, nausea, gastric emptying of solids, and genotype. Baseline body mass index, race, and age seem less relevant for predicting weight-loss response to liraglutide. Lesser known and harder-to-measure factors such as cerebral blood flow, food cue reactivity, gut hormone levels, and dietary adherence possibly impact variability of response to liraglutide. This information should assist healthcare providers with establishing realistic weight-loss probability for individual patients. Future research should improve the ability to identify responders to liraglutide. Importantly, this review may provide a framework to identify responders to other obesity medications.
Collapse
Affiliation(s)
- Chelsi M Webster
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA
| | - Neha Mittal
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | | | - Nikhil V Dhurandhar
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA
| |
Collapse
|
5
|
Pawlak P, Burren A, Seitz A, Pietsch C. Effects of different acute stressors on the regulation of appetite genes in the carp ( Cyprinus carpio L.) brain. ROYAL SOCIETY OPEN SCIENCE 2023; 10:230040. [PMID: 36816841 PMCID: PMC9929511 DOI: 10.1098/rsos.230040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 01/25/2023] [Indexed: 06/18/2023]
Abstract
Our understanding of the timing of stress responses and specific roles of different regulatory pathways that drive stress responses is incomplete. In particular, the regulation of appetite genes as a consequence of exposure to different stressors has not been studied in sufficient detail in fish. Therefore, a stress trial was conducted with koi carp, aiming at identifying typical effects of stress on regulation of appetite genes. The stressors tank manipulation, air exposure and feed rewarding were chosen. The responses to these stressors were evaluated 10, 30 and 60 min after the stressors were applied. Orexigenic and anorexigenic genes were investigated in four different brain regions (telencephalon, hypothalamus, optic tectum and rhombencephalon). The results show that, apart from the typical appetite regulation in the hypothalamus, the different brain regions also display pronounced responses of appetite genes to the different stressors. In addition, several genes in the serotonergic, dopaminergic and gaba-related pathways were investigated. These genes revealed that rearing in pairs of two and opening of the tank lid affected anorexigenic genes, such as cart and cck, which were not changed by air exposure or feed rewarding. Moreover, distress and eustress led to limited, but distinguishable gene expression pattern changes in the investigated brain regions.
Collapse
Affiliation(s)
- Paulina Pawlak
- Agronomy, Bern University of Applied Sciences, Zollikofen, Bern CH-2052, Switzerland
- Division of Behavioural Ecology, Institute of Ecology and Evolution, University of Bern, Wohlenstrasse 50a, CH-3032, Hinterkappelen, Bern, Switzerland
| | - Alexander Burren
- Agronomy, Bern University of Applied Sciences, Zollikofen, Bern CH-2052, Switzerland
| | - Andreas Seitz
- Institute of Natural Resource Sciences, Zurich University of Applied Sciences, Wädenswil, Zürich CH-8820, Switzerland
| | - Constanze Pietsch
- Agronomy, Bern University of Applied Sciences, Zollikofen, Bern CH-2052, Switzerland
| |
Collapse
|
6
|
Abstract
BACKGROUND Obesity develops due to an imbalance in energy homeostasis, wherein energy intake exceeds energy expenditure. Accumulating evidence shows that manipulations of dietary protein and their component amino acids affect the energy balance, resulting in changes in fat mass and body weight. Amino acids are not only the building blocks of proteins but also serve as signals regulating multiple biological pathways. SCOPE OF REVIEW We present the currently available evidence regarding the effects of dietary alterations of a single essential amino acid (EAA) on energy balance and relevant signaling mechanisms at both central and peripheral levels. We summarize the association between EAAs and obesity in humans and the clinical use of modifying the dietary EAA composition for therapeutic intervention in obesity. Finally, similar mechanisms underlying diets varying in protein levels and diets altered of a single EAA are described. The current review would expand our understanding of the contribution of protein and amino acids to energy balance control, thus helping discover novel therapeutic approaches for obesity and related diseases. MAJOR CONCLUSIONS Changes in circulating EAA levels, particularly increased branched-chain amino acids (BCAAs), have been reported in obese human and animal models. Alterations in dietary EAA intake result in improvements in fat and weight loss in rodents, and each has its distinct mechanism. For example, leucine deprivation increases energy expenditure, reduces food intake and fat mass, primarily through regulation of the general control nonderepressible 2 (GCN2) and mammalian target of rapamycin (mTOR) signaling. Methionine restriction by 80% decreases fat mass and body weight while developing hyperphagia, primarily through fibroblast growth factor 21 (FGF-21) signaling. Some effects of diets with different protein levels on energy homeostasis are mediated by similar mechanisms. However, reports on the effects and underlying mechanisms of dietary EAA imbalances on human body weight are few, and more investigations are needed in future.
Collapse
Affiliation(s)
- Fei Xiao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China
| | - Feifan Guo
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; Shanghai Jiao Tong University Affiliated Sixth People's Hospital, China.
| |
Collapse
|
7
|
Wu L, Li D, Qin L, Wang Q, Saito Y, Sara R, Fan J. Growth hormone secretagogue receptor deficiency promotes lung cancer growth by affecting the Th17/Treg balance. ANNALS OF TRANSLATIONAL MEDICINE 2022; 9:1696. [PMID: 34988205 PMCID: PMC8667136 DOI: 10.21037/atm-21-5727] [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: 12/22/2020] [Accepted: 11/19/2021] [Indexed: 11/29/2022]
Abstract
Background Cluster of differentiation 4 (CD4+) T cells plays a prominent role in eliminating cancer cells. The balance between T helper (Th)17 and regulatory T (Treg) cells is crucial for optimal immune response and protection against cancer. Growth hormone secretagogue receptor 1a (GHSR1a), a member of the G protein-coupled protein receptor superfamily, plays a critical role in immune cell function. The aim of our study is to investigate the role of GHSR1a in CD4+ T cell differentiation and lung cancer progression. Methods A subcutaneous lung cancer model was used to examine the role of GHSR1a in controlling tumor growth. Lewis lung carcinoma (LLC) cells were subcutaneously implanted into Ghsr1a−/− mice and wild-type (WT) mice. The ratio of Th17 and Treg in the draining lymph node of Ghsr1a−/− mice and WT tumor-bearing mice was detected by fluorescence-activated cell sorting (FACS). The effect of GHSR1a deficiency on Th17 and Treg cell differentiation was examined using an in vitro differentiation assay. The phosphorylation of mammalian target of rapamycin (mTOR), signal transducer, and activator of transcription (STAT)3 and STAT5 signaling was detected with Western blot. Results We found that the ablation of GHSR1a resulted in impaired anti-tumor immunity to control lung cancer growth in vivo. We also demonstrated that the deficiency of GHSR1a promoted a shift in the Th17/Treg balance toward enhanced Treg differentiation and inhibited Th17 differentiation both in vivo and in vitro, which suggests that GHSR1a regulates T cell lineage choices between Th17 and Treg cell commitment in the tumor microenvironment. Mechanistically, the deficiency of GHSR1a resulted in reduced phosphorylation in mTOR and STAT3, and increased phosphorylation in STAT5. Conclusions Our findings showed the important role of GHSR1a in CD4+ T cell differentiation in the context of the lung cancer microenvironment. This research provides a novel molecular target and insights into interventions for the prevention and treatment of lung cancer.
Collapse
Affiliation(s)
- Liang Wu
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongliang Li
- Department of Thoracic Surgery, Shanxi Provincial Tumor Hospital, Taiyuan, China
| | - Linlin Qin
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qingliang Wang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuichi Saito
- Department of Surgery, Teikyo University School of Medicine, Tokyo, Japan
| | - Ricciardi Sara
- Division of Thoracic Surgery, IRCCS University Hospital of Bologna, Bologna, Italy
| | - Jiang Fan
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Li H, Lou R, Xu X, Xu C, Yu Y, Xu Y, Hu L, Xiang Y, Lin X, Tang S. The variations in human orphan G protein-coupled receptor QRFPR affect PI3K-AKT-mTOR signaling. J Clin Lab Anal 2021; 35:e23822. [PMID: 34018631 DOI: 10.1002/jcla.23822] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND QRFPR is a recently identified member of the G protein-coupled receptor and is an orphan receptor for 26Rfa, which plays important role in the regulation of many physiological functions. METHODS Here, we employed whole exome sequencing (WES) to examine the patients with intellectual disability (ID) and difficulty in feeding. We performed SIFT and PolyPhen2 predictions for the variants. The structure model was built from scratch by I-TASSER. Here, results derived from a number of cell-based functional assays, including shRNA experiment, intracellular Ca2+ measurement, the expression of PI3 K-AKT-mTOR, and phosphorylation. The functional effect of QRFPR variants on PI3K-AKT-mTOR signaling was evaluated in vitro transfection experiments. RESULT Here, we identified two QRFPR variants at c.202 T>C (p.Y68H) and c.1111C>T (p.R371W) in 2 unrelated individuals. Structural analysis revealed that p.Y68H and p.R371W variants may affect the side chain structure of adjacent amino acids causing reduced binding of QRFPR to 26Rfa. The results show that QRFPR stimulated by 26Rfa leading to the transient rise of intracellular Ca2+ . The QRFPR variations p.Y68H and p.R371 W can reduce the mobilization of intracellular Ca2+ . The phosphorylation levels of the PI3K, Akt, and mTOR were significantly up- or downregulated by QRFPR overexpression or silencing, respectively. The QRFPR variations inhibited PI3K-AKT-mTOR signaling, resulting in downregulation of p-mTOR. CONCLUSIONS Our findings suggest that QRFPR acts as important role in neurodevelopment, and the effects of QRFPR are likely to be mediated by the Ca2+ -dependent PI3K-AKT-mTOR pathways. Importantly, these findings provide a foundation for future elucidation of GPCR-mediated signaling and the physiological implications.
Collapse
Affiliation(s)
- Huanzheng Li
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China.,Human Aging Research Institute, Nanchang University, Nanchang, China
| | - Ran Lou
- Department of Acupuncture, Wenzhou Central Hospital, Wenzhou, China
| | - Xueqin Xu
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Chenyang Xu
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Yuan Yu
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Yunzhi Xu
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Lin Hu
- Department of Blood Transfusion, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yanbao Xiang
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Xuan Lin
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Shaohua Tang
- Wenzhou Key Laboratory of Birth Defects, Wenzhou Central Hospital, Dingli Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
9
|
Rozjan A, Shan W, Yao Q. Metabolic Consequences of Neuronal HIF1α-Deficiency in Mediobasal Hypothalamus in Mice. Front Endocrinol (Lausanne) 2021; 12:668193. [PMID: 34733235 PMCID: PMC8558296 DOI: 10.3389/fendo.2021.668193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 09/16/2021] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE This study aims to investigate whether hypoxia-inducible factor 1α (HIF1α) in the neurons of the mediobasal hypothalamus is involved in the regulation of body weight, glucose, and lipid metabolism in mice and to explore the underlying molecular mechanisms. METHODS HIF1α flox/flox mice were used. The adeno-associated virus that contained either cre, GFP and syn, or GFP and syn (controls) was injected into the mediobasal hypothalamus to selectively knock out HIF1α in the neurons of the mediobasal hypothalamus. The body weight and food intake were weighed daily. The levels of blood glucose, insulin, total cholesterol (TC), triglyceride (TG), free fatty acid (FFA), high-density lipoprotein (HDL), and low-density lipoprotein (LDL)were tested. Intraperitoneal glucose tolerance test (IPGTT) was performed. The insulin-stimulated Akt phosphorylation in the liver, epididymal fat, and skeletal muscle were examined. Also, the mRNA expression levels of HIF1α, proopiomelanocortin (POMC), neuropeptide Y (NPY), and glucose transporter protein 4 (Glut4) in the hypothalamus were checked. RESULTS After selectively knocking out HIF1α in the neurons of the mediobasal hypothalamus (HIF1αKOMBH), the body weights and food intake of mice increased significantly compared with the control mice (p < 0.001 at 4 weeks). Compared with that of the control group, the insulin level of HIF1αKOMBH mice was 3.5 times higher (p < 0.01). The results of the IPGTT showed that the blood glucose level of the HIF1αKOMBH group at 20-120 min was significantly higher than that of the control group (p < 0.05). The serum TC, FFA, HDL, and LDL content of the HIF1αKOMBH group was significantly higher than those of the control group (p < 0.05). Western blot results showed that compared with those in the control group, insulin-induced AKT phosphorylation levels in liver, epididymal fat, and skeletal muscle in the HIF1αKOMBH group were not as significantly elevated as in the control group. Reverse transcription-polymerase chain reaction (RT-PCR) results in the whole hypothalamus showed a significant decrease in Glut4 mRNA expression. And the mRNA expression levels of HIF1α, POMC, and NPY of the HIF1αKOMBH group decreased significantly in ventral hypothalamus. CONCLUSIONS The hypothalamic neuronal HIF1α plays an important role in the regulation of body weight balance in mice under normoxic condition. In the absence of hypothalamic neuronal HIF1α, the mice gained weight with increased appetite, accompanied with abnormal glucose and lipid metabolism. POMC and Glut4 may be responsible for this effect of HIF1α.
Collapse
Affiliation(s)
- Azmat Rozjan
- Department of Physiology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - Weibi Shan
- Department of Dermatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Qiaoling Yao
- Department of Physiology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
- *Correspondence: Qiaoling Yao,
| |
Collapse
|
10
|
Gao J, Li X, Wang Y, Cao Y, Yao D, Sun L, Qin L, Qiu H, Zhan X. Adipocyte-derived extracellular vesicles modulate appetite and weight through mTOR signalling in the hypothalamus. Acta Physiol (Oxf) 2020; 228:e13339. [PMID: 31278836 DOI: 10.1111/apha.13339] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 06/29/2019] [Accepted: 07/01/2019] [Indexed: 12/13/2022]
Abstract
AIM Type 2 diabetes and obesity are diseases related to surplus energy in the body. Abnormal interaction between the hypothalamus and adipose tissues is a key trigger of energy metabolism dysfunction. Extracellular vesicles (EVs) regulate intercellular communication by transporting intracellular cargo to recipient cells thereby altering the function of recipient cells. This study aimed to evaluate whether adipocyte-derived EVs can act on hypothalamic neurons to modulate energy intake and to identify the EV-associated non-coding RNAs. METHODS Confocal imaging was used to trace the uptake of labelled adipocyte-derived exosomes by hypothalamic anorexigenic POMC neurons. The effects of adipocyte-derived EVs on the mammalian target of rapamycin (mTOR) signalling pathway in POMC neurons were evaluated based on mRNA and protein expression in vitro using quantitative real-time PCR and western blotting. In addition, adipocyte-derived EVs were injected into recipient mice, and changes in mice body weight and daily food intake were monitored. The biological effects of the EV-associated MALAT1 on POMC neurons were explored. RESULTS Adipocyte-derived EVs were successfully transferred into POMC neurons in vitro. Results showed that adipocytes of obese mice secreted MALAT1-containing EVs, which increased appetite and weight when administered to lean mice. Conversely, adipocyte-derived EVs from lean mice decreased food intake and weight when administered to obese mice. CONCLUSION Adipocyte-derived EVs play important roles in mediating the interaction between adipocytes and hypothalamic neurons. Adipocyte-derived EVs can regulate POMC expression through the hypothalamic mTOR signalling in vivo and in vitro, thereby affecting body energy intake.
Collapse
Affiliation(s)
- Jie Gao
- Department of Endocrinology First Affiliated Hospital of Harbin Medical University Harbin China
| | - Xinyu Li
- Department of Endocrinology First Affiliated Hospital of Harbin Medical University Harbin China
| | - You Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics Chinese Academy of Sciences Beijing China
| | - Yan Cao
- Department of Endocrinology First Affiliated Hospital of Harbin Medical University Harbin China
| | - Dengju Yao
- Software and Microelectronics School Harbin University of Science and Technology Harbin China
| | - Lijie Sun
- Department of Endocrinology First Affiliated Hospital of Harbin Medical University Harbin China
| | - Lv Qin
- Department of Endocrinology First Affiliated Hospital of Harbin Medical University Harbin China
| | - Hui Qiu
- Department of Endocrinology First Affiliated Hospital of Harbin Medical University Harbin China
| | - Xiaorong Zhan
- Department of Endocrinology First Affiliated Hospital of Harbin Medical University Harbin China
| |
Collapse
|
11
|
Wang WJ, Zhang H, Chen ZQ, Zhang W, Liu XM, Fang JY, Liu FJ, Kwak-Kim J. Endometrial TGF-β, IL-10, IL-17 and autophagy are dysregulated in women with recurrent implantation failure with chronic endometritis. Reprod Biol Endocrinol 2019; 17:2. [PMID: 30606202 PMCID: PMC6317248 DOI: 10.1186/s12958-018-0444-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 12/12/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Chronic endometritis (CE) is a condition which results in reduced receptivity of embryos by dysregulated lymphocyte subsets, abnormal expression of cytokines, chemokines and other regulatory molecules in the endometrium (EM). Macroautophagy (autophagy), the highly conserved cellular homeostasis pathway, plays an essential role in the development and function of T lymphocytes, and supports T cell lineage stability and survival fitness. The possible relationships between autophagy and local cytokine milieus in repeated implantation failure (RIF) with CE have not been elucidated yet. METHODS This case-control study was performed at a large reproductive medicine center between February 2015 and July 2016. Seventy-five recurrent implantation falliure women with CE who had "strawberry aspect" and 75 women with male factor infertility were included. In this study, endometrial expressions of IL-17, IL-10, TGF-β and autophagy related molecules, including LC3-II and mTORC1 were investigated by qRT-PCR, Western blot, immunofluorescence and immunohistochemistry assays. RESULTS The expression of IL-17 was significantly higher in patients with CE compared to women with male factor infertility, while the expressions of IL-10 and TGF-β were significantly lower. Moreover, the expression of autophagy (LC3-II) is increased, while the expression of mTORC1 was impaired. CONCLUSIONS CE is associated with shifted cytokine milieu towards Th17 over Treg immunity in endometrium through impaired autophagy by decreased mTORC1.
Collapse
Affiliation(s)
- Wen-Juan Wang
- Reproduction Medical Center, Yantai Yuhuangding Hospital of Qingdao University, 20 Yuhuangding East Road, Yantai, 264000, People's Republic of China.
| | - Hong Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, 1055th Sanxiang road, Suzhou, 215004, People's Republic of China
| | - Zhong-Quan Chen
- Department of anatomy, Binzhou Medical University, Binhai Road, Yantai, 26400, People's Republic of China
| | - Wei Zhang
- Reproduction Medical Center, Yantai Yuhuangding Hospital of Qingdao University, 20 Yuhuangding East Road, Yantai, 264000, People's Republic of China
| | - Xue-Mei Liu
- Reproduction Medical Center, Yantai Yuhuangding Hospital of Qingdao University, 20 Yuhuangding East Road, Yantai, 264000, People's Republic of China
| | - Jiang-Ye Fang
- Reproduction Medical Center, Yantai Yuhuangding Hospital of Qingdao University, 20 Yuhuangding East Road, Yantai, 264000, People's Republic of China
| | - Fu-Jun Liu
- Central Laboratory, Yantai Yuhuangding Hospital of Qingdao University, 20 Yuhuangding East Road, Yantai, 264000, People's Republic of China
| | - Joanne Kwak-Kim
- Reproductive Medicine and Immunology, Department of Obstetrics and Gynecology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, 830 West Court, Suite 400, Vernon Hills, IL, 60061, USA.
| |
Collapse
|
12
|
Chen Z, Herting MM, Chatzi L, Belcher BR, Alderete TL, McConnell R, Gilliland F. Regional and traffic-related air pollutants are associated with higher consumption of fast food and trans fat among adolescents. Am J Clin Nutr 2019; 109:99-108. [PMID: 30596809 PMCID: PMC6358030 DOI: 10.1093/ajcn/nqy232] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 06/26/2018] [Accepted: 08/09/2018] [Indexed: 12/21/2022] Open
Abstract
Background Air pollution exposures are novel contributors to the growing childhood obesity epidemic. One possible mechanism linking air pollution exposures and obesity is through changes in food consumption patterns. Objective The aim of this study was to examine the longitudinal association between childhood exposure to air pollutants and changes in diet among adolescents. Design School-age children were enrolled in the Southern California Children's Health Study during 1993-1994 (n = 3100) and were followed for 4-8 y. Community-level regional air pollutants [e.g., nitrogen dioxide (NO2), elemental carbon (EC), and fine particles with aerodynamic diameter <2.5 µm (PM2.5)] were measured at central monitoring stations. Line dispersion modeling was used to estimate concentrations of traffic-related air pollutants based on nitrogen oxides (NOx) at participants' residential addresses. In addition, self-reported diet information was collected annually using a structured youth/adolescent food-frequency questionnaire during 1997-2001. Generalized linear mixed-effects models were used in the association analyses. Results Higher exposures to regional and traffic-related air pollutants were associated with intake of a high-trans-fat diet, after adjusting for confounders including socioeconomic status and access to fast food in the community. A 2-SD (12.2 parts per billion) increase in regional NO2 exposure was associated with a 34% increased risk of consuming a high-trans-fat diet compared with a low-trans-fat diet (OR: 1.34; 95% CI: 1.05, 1.72). In addition, higher exposures to acid vapor, EC, PM2.5, and non-freeway NOx were all associated with higher consumption of dietary trans fat (all P < 0.04). Notably, higher exposures to regional NO2, acid vapor, and EC were also associated with a higher consumption of fast food (all P < 0.05). Conclusions Childhood exposures to regional and traffic-related air pollutants were associated with increased consumption by adolescents of trans fat and fast foods. Our results indicate that air pollution exposures may contribute to obesogenic behaviors. This study was registered at clinicaltrials.gov as NCT03379298.
Collapse
Affiliation(s)
- Zhanghua Chen
- Division of Environmental Health and Preventive Medicine
| | | | - Leda Chatzi
- Division of Environmental Health and Preventive Medicine
| | - Britni R Belcher
- Institute for Health Promotion and Disease Prevention Research, Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA
| | - Tanya L Alderete
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO
| | - Rob McConnell
- Division of Environmental Health and Preventive Medicine
| | | |
Collapse
|
13
|
Liang H, Nie J, Van Skike CE, Valentine JM, Orr ME. Mammalian Target of Rapamycin at the Crossroad Between Alzheimer's Disease and Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1128:185-225. [PMID: 31062331 DOI: 10.1007/978-981-13-3540-2_10] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Accumulating evidence suggests that Alzheimer's disease may manifest as a metabolic disorder with pathology and/or dysfunction in numerous tissues. Adults with Alzheimer's disease suffer with significantly more comorbidities than demographically matched Medicare beneficiaries (Zhao et al, BMC Health Serv Res 8:108, 2008b). Reciprocally, comorbid health conditions increase the risk of developing Alzheimer's disease (Haaksma et al, PLoS One 12(5):e0177044, 2017). Type 2 diabetes mellitus is especially notable as the disease shares many overlapping pathologies observed in patients with Alzheimer's disease, including hyperglycemia, hyperinsulinemia, insulin resistance, glucose intolerance, dyslipidemia, inflammation, and cognitive dysfunction, as described in Chap. 8 of this book (Yoshitake et al, Neurology 45(6):1161-1168, 1995; Leibson et al, Am J Epidemiol 145(4):301-308, 1997; Ott et al, Neurology 53(9):1937-1942, 1999; Voisin et al, Rev Med Interne 24(Suppl 3):288s-291s, 2003; Janson et al. Diabetes 53(2):474-481, 2004; Ristow M, J Mol Med (Berl) 82(8):510-529, 2004; Whitmer et al, BMJ 330(7504):1360, 2005, Curr Alzheimer Res 4(2):103-109, 2007; Ohara et al, Neurology 77(12):1126-1134, 2011). Although nondiabetic older adults also experience age-related cognitive decline, diabetes is uniquely associated with a twofold increased risk of Alzheimer's disease, as described in Chap. 2 of this book (Yoshitake et al, Neurology 45(6):1161-1168, 1995; Leibson et al, Am J Epidemiol 145(4):301-308, 1997; Ott et al. Neurology 53(9):1937-1942, 1999; Ohara et al, Neurology 77(12):1126-1134, 2011). Good glycemic control has been shown to improve cognitive status (Cukierman-et al, Diabetes Care 32(2):221-226, 2009), and the use of insulin sensitizers is correlated with a lower rate of cognitive decline in older adults (Morris JK, Burns JM, Curr Neurol Neurosci Rep 12(5):520-527, 2012). At the molecular level, the mechanistic/mammalian target of rapamycin (mTOR) plays a key role in maintaining energy homeostasis. Nutrient availability and cellular stress information, both extracellular and intracellular, are integrated and transduced through mTOR signaling pathways. Aberrant regulation of mTOR occurs in the brains of patients with Alzheimer's disease and in numerous tissues of individuals with type 2 diabetes (Mannaa et al, J Mol Med (Berl) 91(10):1167-1175, 2013). Moreover, modulating mTOR activity with a pharmacological inhibitor, rapamycin, provides wide-ranging health benefits, including healthy life span extension in numerous model organisms (Vellai et al, Nature 426(6967):620, 2003; Jia et al, Development 131(16):3897-3906, 2004; Kapahi et al, Curr Biol 14(10):885-890, 2004; Kaeberlein et al, Science 310(5751):1193-1196, 2005; Powers et al, Genes Dev 20(2):174-184, 2006; Harrison et al, Nature 460(7253):392-395, 2009; Selman et al, Science 326(5949):140-144, 2009; Sharp ZD, Strong R, J Gerontol A Biol Sci Med Sci 65(6):580-589, 2010), which underscores its importance to overall organismal health and longevity. In this chapter, we discuss the physiological role of mTOR signaling and the consequences of mTOR dysregulation in the brain and peripheral tissues, with emphasis on its relevance to the development of Alzheimer's disease and link to type 2 diabetes.
Collapse
Affiliation(s)
- Hanyu Liang
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jia Nie
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Candice E Van Skike
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Joseph M Valentine
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Miranda E Orr
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
- San Antonio Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, TX, USA.
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, San Antonio, TX, USA.
| |
Collapse
|
14
|
Sahu M, Anamthathmakula P, Sahu A. Hypothalamic PDE3B deficiency alters body weight and glucose homeostasis in mouse. J Endocrinol 2018; 239:93-105. [PMID: 30307157 PMCID: PMC6190684 DOI: 10.1530/joe-18-0304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Pharmacological studies have suggested hypothalamic phosphodiesterase-3B to mediate leptin and insulin action in regulation of energy homeostasis. Whereas Pde3b-null mice show altered energy homeostasis, it is unknown whether this is due to ablation of Pde3b in the hypothalamus. Thus, to address the functional significance of hypothalamic phosphodiesterase-3B, we used Pde3bflox/flox and Nkx2.1-Cre mice to generate Pde3b Nkx2.1KD mice that showed 50% reduction of phosphodiesterase-3B in the hypothalamus. To determine the effect of partial ablation of phosphodiesterase-3B in the hypothalamus on energy and glucose homeostasis, males and females were subjected to either a low- or high-fat diet for 19–21 weeks. Only female but not male Pde3b Nkx2.1KD mice on the low-fat diet showed increased body weight from 13 weeks onward with increased food intake, decreased fat pad weights and hypoleptinemia. Glucose tolerance was improved in high-fat diet-fed male Pde3b Nkx2.1KD mice in association with decreased phosphoenolpyruvate carboxykinase-1 and glucose-6-phosphatase mRNA levels in the liver. Also, insulin sensitivity was increased in male Pde3b Nkx2.1KD mice on the low-fat diet. Changes in body weight or in glucose homeostasis were not associated with any alteration in hypothalamic proopiomelanocortin, neuropepide Y and agouti-related peptide mRNA levels. These results suggest that partial loss of phosphodiesterase-3B in the hypothalamus produces a sex-specific response in body weight and glucose homeostasis, and support a role, at least in part, for hypothalamic phosphodiesterase-3B in regulation of energy and glucose homeostasis in mice.
Collapse
Affiliation(s)
- Maitrayee Sahu
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Prashanth Anamthathmakula
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Abhiram Sahu
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
15
|
de Oliveira JC, de Moura EG, Miranda RA, de Moraes AMP, Barella LF, da Conceição EPS, Gomes RM, Ribeiro TA, Malta A, Martins IP, Franco CCDS, Lisboa PC, Mathias PCDF. Low-protein diet in puberty impairs testosterone output and energy metabolism in male rats. J Endocrinol 2018; 237:243-254. [PMID: 29599416 DOI: 10.1530/joe-17-0606] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 03/28/2018] [Indexed: 11/08/2022]
Abstract
We examined the long-term effects of protein restriction during puberty on the function of hypothalamic-pituitary-adrenal (HPA) and hypothalamic-pituitary-gonadal (HPG) axes in male rats. Male Wistar rats from the age of 30 to 60 days were fed a low-protein diet (4%, LP). A normal-protein diet (20.5%) was reintroduced to rats from the age of 60 to 120 days. Control rats were fed a normal-protein diet throughout life (NP). Rats of 60 or 120 days old were killed. Food consumption, body weight, visceral fat deposits, lipid profile, glycemia, insulinemia, corticosteronemia, adrenocorticotropic hormone (ACTH), testosteronemia and leptinemia were evaluated. Glucose-insulin homeostasis, pancreatic-islet insulinotropic response, testosterone production and hypothalamic protein expression of the androgen receptor (AR), glucocorticoid receptor (GR) and leptin signaling pathway were also determined. LP rats were hypophagic, leaner, hypoglycemic, hypoinsulinemic and hypoleptinemic at the age of 60 days (P < 0.05). These rats exhibited hyperactivity of the HPA axis, hypoactivity of the HPG axis and a weak insulinotropic response (P < 0.01). LP rats at the age of 120 days were hyperphagic and exhibited higher visceral fat accumulation, hyperleptinemia and dyslipidemia; lower blood ACTH, testosterone and testosterone release; and reduced hypothalamic expression of AR, GR and SOCS3, with a higher pSTAT3/STAT3 ratio (P < 0.05). Glucose-insulin homeostasis was disrupted and associated with hyperglycemia, hyperinsulinemia and increased insulinotropic response of the pancreatic islets. The cholinergic and glucose pancreatic-islet responses were small in 60-day-old LP rats but increased in 120-day-old LP rats. The hyperactivity of the HPA axis and the suppression of the HPG axis caused by protein restriction at puberty contributed to energy and metabolic disorders as long-term consequences.
Collapse
Affiliation(s)
- Júlio Cezar de Oliveira
- Laboratório de Biologia Celular da Secreção, Departamento de Biotecnologia, Genética e Biologia Celular, Universidade Estadual de Maringá, Maringá, Brazil
- Departamento de Ciências Fisiológicas, Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Ciências da Saúde, Universidade Federal de Mato Grosso, Sinop, Brazil
| | - Egberto Gaspar de Moura
- Departamento de Ciências Fisiológicas, Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rosiane Aparecida Miranda
- Laboratório de Biologia Celular da Secreção, Departamento de Biotecnologia, Genética e Biologia Celular, Universidade Estadual de Maringá, Maringá, Brazil
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Maria Praxedes de Moraes
- Laboratório de Biologia Celular da Secreção, Departamento de Biotecnologia, Genética e Biologia Celular, Universidade Estadual de Maringá, Maringá, Brazil
| | - Luiz Felipe Barella
- Laboratório de Biologia Celular da Secreção, Departamento de Biotecnologia, Genética e Biologia Celular, Universidade Estadual de Maringá, Maringá, Brazil
| | - Ellen Paula Santos da Conceição
- Departamento de Ciências Fisiológicas, Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rodrigo Mello Gomes
- Departamento de Ciências Fisiológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Tatiane Aparecida Ribeiro
- Laboratório de Biologia Celular da Secreção, Departamento de Biotecnologia, Genética e Biologia Celular, Universidade Estadual de Maringá, Maringá, Brazil
| | - Ananda Malta
- Laboratório de Biologia Celular da Secreção, Departamento de Biotecnologia, Genética e Biologia Celular, Universidade Estadual de Maringá, Maringá, Brazil
| | - Isabela Peixoto Martins
- Laboratório de Biologia Celular da Secreção, Departamento de Biotecnologia, Genética e Biologia Celular, Universidade Estadual de Maringá, Maringá, Brazil
| | - Claudinéia Conationi da Silva Franco
- Laboratório de Biologia Celular da Secreção, Departamento de Biotecnologia, Genética e Biologia Celular, Universidade Estadual de Maringá, Maringá, Brazil
| | - Patrícia Cristina Lisboa
- Departamento de Ciências Fisiológicas, Laboratório de Fisiologia Endócrina, Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paulo Cezar de Freitas Mathias
- Laboratório de Biologia Celular da Secreção, Departamento de Biotecnologia, Genética e Biologia Celular, Universidade Estadual de Maringá, Maringá, Brazil
| |
Collapse
|
16
|
Bolster DR, Rahn M, Kamil AG, Bristol LT, Goltz SR, Leidy HJ, Blaze Mt M, Nunez MA, Guo E, Wang J, Harkness LS. Consuming Lower-Protein Nutrition Bars with Added Leucine Elicits Postprandial Changes in Appetite Sensations in Healthy Women. J Nutr 2018; 148:693-701. [PMID: 29897544 DOI: 10.1093/jn/nxy023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 05/18/2017] [Accepted: 01/30/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Higher-protein meals (>25 g protein/meal) have been associated with enhanced satiety but the role of amino acids is unclear. Leucine has been proposed to stimulate satiety in rodents but has not been assessed in humans. OBJECTIVE We assessed the acute effects of lower-protein nutrition bars, enhanced with a leucine peptide (LP), on postprandial appetite sensations in combination with plasma leucine and peptide YY (PYY) in healthy women. METHODS Utilizing a double-blind randomized crossover design, 40 healthy women [28 ± 7.5 y; body mass index (BMI, in kg/m2): 23.5 ± 2.4] consumed the following isocaloric (180 kcal) pre-loads on 3 separate visits: control bar [9 g protein with 0 g added LP (0-g LP)] or treatment bars [11 g protein with 2 g added LP (2-g LP) or 13 g protein with 3 g added LP (3-g LP)]. Pre- and postprandial hunger, desire to eat, prospective food consumption (PFC), fullness, and plasma leucine were assessed every 30 min for 240 min. Plasma PYY was assessed hourly for 240 min (n = 24). RESULTS Main effects of time (P < 0.0001) and treatment (P < 0.03) were detected for postprandial hunger, desire to eat, PFC, and fullness. Post hoc analyses revealed that the 2-g and 3-g LP bars elicited greater increases in fullness and greater decreases in PFC compared with 0-g LP (all, P < 0.05) with no differences between the 2-g and 3-g LP bars. The 2-g bar elicited greater decreases in hunger and desire to eat compared with the 0-g LP bar (both, P ≤ 0.01), whereas 3-g LP did not. Appetite incremental areas under the curves (iAUCs) and PYY outcomes were not different between bars. A treatment × time interaction was detected for plasma leucine with increases occurring in a leucine-dose-dependent manner (P < 0.0001). CONCLUSION Despite the dose-dependent increases in plasma leucine following the consumption of lower-protein bars enhanced with LP, only the 2-g LP bar elicited consistent postprandial changes in select appetite sensations compared with the 0-g LP bar. This study was registered on clinicaltrials.gov as NCT02091570.
Collapse
Affiliation(s)
| | - Maike Rahn
- PepsiCo R&D Nutrition Sciences, Purchase, NY
| | | | | | | | - Heather J Leidy
- Department of Nutrition Science, Purdue University, West Lafayette, IN
| | | | | | | | | | | |
Collapse
|
17
|
Perez-Alvarez MJ, Villa Gonzalez M, Benito-Cuesta I, Wandosell FG. Role of mTORC1 Controlling Proteostasis after Brain Ischemia. Front Neurosci 2018; 12:60. [PMID: 29497356 PMCID: PMC5818460 DOI: 10.3389/fnins.2018.00060] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/24/2018] [Indexed: 01/24/2023] Open
Abstract
Intense efforts are being undertaken to understand the pathophysiological mechanisms triggered after brain ischemia and to develop effective pharmacological treatments. However, the underlying molecular mechanisms are complex and not completely understood. One of the main problems is the fact that the ischemic damage is time-dependent and ranges from negligible to massive, involving different cell types such as neurons, astrocytes, microglia, endothelial cells, and some blood-derived cells (neutrophils, lymphocytes, etc.). Thus, approaching such a complicated cellular response generates a more complex combination of molecular mechanisms, in which cell death, cellular damage, stress and repair are intermixed. For this reason, animal and cellular model systems are needed in order to dissect and clarify which molecular mechanisms have to be promoted and/or blocked. Brain ischemia may be analyzed from two different perspectives: that of oxygen deprivation (hypoxic damage per se) and that of deprivation of glucose/serum factors. For investigations of ischemic stroke, middle cerebral artery occlusion (MCAO) is the preferred in vivo model, and uses two different approaches: transient (tMCAO), where reperfusion is permitted; or permanent (pMCAO). As a complement to this model, many laboratories expose different primary cortical neuron or neuronal cell lines to oxygen-glucose deprivation (OGD). This ex vivo model permits the analysis of the impact of hypoxic damage and the specific response of different cell types implicated in vivo, such as neurons, glia or endothelial cells. Using in vivo and neuronal OGD models, it was recently established that mTORC1 (mammalian Target of Rapamycin Complex-1), a protein complex downstream of PI3K-Akt pathway, is one of the players deregulated after ischemia and OGD. In addition, neuroprotective intervention either by estradiol or by specific AT2R agonists shows an important regulatory role for the mTORC1 activity, for instance regulating vascular endothelial growth factor (VEGF) levels. This evidence highlights the importance of understanding the role of mTORC1 in neuronal death/survival processes, as it could be a potential therapeutic target. This review summarizes the state-of-the-art of the complex kinase mTORC1 focusing in upstream and downstream pathways, their role in central nervous system and their relationship with autophagy, apoptosis and neuroprotection/neurodegeneration after ischemia/hypoxia.
Collapse
Affiliation(s)
- Maria J Perez-Alvarez
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain.,Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain.,Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | - Mario Villa Gonzalez
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain.,Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
| | - Irene Benito-Cuesta
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain.,Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | - Francisco G Wandosell
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain.,Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, Madrid, Spain
| |
Collapse
|
18
|
Ardestani A, Lupse B, Kido Y, Leibowitz G, Maedler K. mTORC1 Signaling: A Double-Edged Sword in Diabetic β Cells. Cell Metab 2018; 27:314-331. [PMID: 29275961 DOI: 10.1016/j.cmet.2017.11.004] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 10/11/2017] [Accepted: 11/15/2017] [Indexed: 12/21/2022]
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) is a central regulator of metabolic and nutrient cues that integrates environmental inputs into downstream signaling pathways to control cellular metabolism, growth, and survival. While numerous in vitro and in vivo studies reported the positive functions of mTORC1 in the regulation of β cell survival and proliferation under physiological conditions, more recent work demonstrates the opposite in the long term; this is exemplified by the constitutive inappropriate hyper-activation of mTORC1 in diabetic islets or β cells under conditions of increased β cell stress and metabolic demands. These recent findings uncover mTORC1's importance as an emerging significant player in the development and progression of β cell failure in type 2 diabetes and suggest that mTORC1 may act as a "double edge sword" in the regulation of β cell mass and function in response to metabolic stress such as nutrient overload and insulin resistance.
Collapse
Affiliation(s)
- Amin Ardestani
- University of Bremen, Centre for Biomolecular Interactions Bremen, Bremen 28359, Germany.
| | - Blaz Lupse
- University of Bremen, Centre for Biomolecular Interactions Bremen, Bremen 28359, Germany
| | - Yoshiaki Kido
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; Division of Metabolism and Disease, Department of Biophysics, Kobe University Graduate School of Health Sciences, Kobe 654-0142, Japan
| | - Gil Leibowitz
- Endocrinology and Metabolism Service and the Hadassah Diabetes Unit, Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Kathrin Maedler
- University of Bremen, Centre for Biomolecular Interactions Bremen, Bremen 28359, Germany.
| |
Collapse
|
19
|
Burmeister MA, Brown JD, Ayala JE, Stoffers DA, Sandoval DA, Seeley RJ, Ayala JE. The glucagon-like peptide-1 receptor in the ventromedial hypothalamus reduces short-term food intake in male mice by regulating nutrient sensor activity. Am J Physiol Endocrinol Metab 2017; 313:E651-E662. [PMID: 28811293 PMCID: PMC6109646 DOI: 10.1152/ajpendo.00113.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/27/2017] [Accepted: 08/10/2017] [Indexed: 12/25/2022]
Abstract
Pharmacological activation of the glucagon-like peptide-1 receptor (GLP-1R) in the ventromedial hypothalamus (VMH) reduces food intake. Here, we assessed whether suppression of food intake by GLP-1R agonists (GLP-1RA) in this region is dependent on AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR). We found that pharmacological inhibition of glycolysis, and thus activation of AMPK, in the VMH attenuates the anorectic effect of the GLP-1R agonist exendin-4 (Ex4), indicating that glucose metabolism and inhibition of AMPK are both required for this effect. Furthermore, we found that Ex4-mediated anorexia in the VMH involved mTOR but not acetyl-CoA carboxylase, two downstream targets of AMPK. We support this by showing that Ex4 activates mTOR signaling in the VMH and Chinese hamster ovary (CHO)-K1 cells. In contrast to the clear acute pharmacological impact of the these receptors on food intake, knockdown of the VMH Glp1r conferred no changes in energy balance in either chow- or high-fat-diet-fed mice, and the acute anorectic and glucose tolerance effects of peripherally dosed GLP-1RA were preserved. These results show that the VMH GLP-1R regulates food intake by engaging key nutrient sensors but is dispensable for the effects of GLP-1RA on nutrient homeostasis.
Collapse
Affiliation(s)
- Melissa A Burmeister
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida;
| | - Jacob D Brown
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida
| | - Jennifer E Ayala
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida
| | - Doris A Stoffers
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Darleen A Sandoval
- Department of Surgery, University of Michigan Health System, Ann Arbor, Michigan
| | - Randy J Seeley
- Department of Surgery, University of Michigan Health System, Ann Arbor, Michigan
| | - Julio E Ayala
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida
| |
Collapse
|
20
|
Yu M, Chen Y, Zeng H, Zheng Y, Fu G, Zhu W, Broeckel U, Aggarwal P, Turner A, Neale G, Guy C, Zhu N, Chi H, Wen R, Wang D. PLCγ-dependent mTOR signalling controls IL-7-mediated early B cell development. Nat Commun 2017; 8:1457. [PMID: 29133930 PMCID: PMC5684131 DOI: 10.1038/s41467-017-01388-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 09/14/2017] [Indexed: 01/01/2023] Open
Abstract
The precise molecular mechanism underlying the regulation of early B cell lymphopoiesis is unclear. The PLCγ signaling pathway is critical for antigen receptor-mediated lymphocyte activation, but its function in cytokine signaling is unknown. Here we show that PLCγ1/PLCγ2 double deficiency in mice blocks early B cell development at the pre-pro-B cell stage and renders B cell progenitors unresponsive to IL-7. PLCγ pathway inhibition blocks IL-7-induced activation of mTOR, but not Stat5. The PLCγ pathway activates mTOR through the DAG/PKC signaling branch, independent of the conventional Akt/TSC/Rheb signaling axis. Inhibition of PLCγ/PKC-induced mTOR activation impairs IL-7-mediated B cell development. PLCγ1/PLCγ2 double-deficient B cell progenitors have reduced expression of genes related to B cell lineage, IL-7 signaling, and cell cycle. Thus, IL-7 receptor controls early B lymphopoiesis through activation of mTOR via PLCγ/DAG/PKC signaling, not via Akt/Rheb signaling.
Collapse
Affiliation(s)
- Mei Yu
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI, 53226, USA
| | - Yuhong Chen
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI, 53226, USA
| | - Hu Zeng
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Yongwei Zheng
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI, 53226, USA
| | - Guoping Fu
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI, 53226, USA
| | - Wen Zhu
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI, 53226, USA
- Interdisciplinary Program in Biomedical Sciences, Milwaukee, WI, 53226, USA
| | - Ulrich Broeckel
- Section of Genomic Pediatrics, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Praful Aggarwal
- Section of Genomic Pediatrics, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Amy Turner
- Section of Genomic Pediatrics, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Geoffrey Neale
- Hartwell Center for Bioinformatics and Biotechnology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Cliff Guy
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Nan Zhu
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI, 53226, USA
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Renren Wen
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI, 53226, USA
| | - Demin Wang
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI, 53226, USA.
- Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian, 350117, China.
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| |
Collapse
|
21
|
Bird JK, Raederstorff D, Weber P, Steinert RE. Cardiovascular and Antiobesity Effects of Resveratrol Mediated through the Gut Microbiota. Adv Nutr 2017; 8:839-849. [PMID: 29141969 PMCID: PMC5682996 DOI: 10.3945/an.117.016568] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Encouraging scientific research into the health effects of dietary bioactive resveratrol has been confounded by its rapid first-pass metabolism, which leads to low in vivo bioavailability. Preliminary studies have shown that resveratrol can modulate gut microbiota composition, undergo biotransformation to active metabolites via the intestinal microbiota, or affect gut barrier function. In rodents, resveratrol can modify the relative Bacteroidetes:Firmicutes ratio and reverse the gut microbial dysbiosis caused by a high-fat diet. By upregulating the expression of genes involved in maintaining tight junctions between intestinal cells, resveratrol contributes to gut barrier integrity. The composition of the gut microbiome and rapid metabolism of resveratrol determines the production of resveratrol metabolites, which are found at greater concentrations in humans after ingestion than their parent molecule and can have similar biological effects. Resveratrol may affect cardiovascular risk factors such as elevated blood cholesterol or trimethylamine N-oxide concentrations. Modulating the composition of the gut microbiota by resveratrol may affect central energy metabolism and modify concentrations of satiety hormones to produce antiobesity effects. Encouraging research from animal models could be tested in humans.
Collapse
Affiliation(s)
- Julia K Bird
- Human Nutrition and Health, DSM Nutritional Products, Basel, Switzerland, and
| | - Daniel Raederstorff
- Human Nutrition and Health, DSM Nutritional Products, Basel, Switzerland, and
| | - Peter Weber
- Human Nutrition and Health, DSM Nutritional Products, Basel, Switzerland, and
| | - Robert E Steinert
- Human Nutrition and Health, DSM Nutritional Products, Basel, Switzerland, and,Department of Surgery, Division of Visceral and Transplantation Surgery, University Hospital Zürich, Zürich, Switzerland
| |
Collapse
|
22
|
Kuz’mina VV. Role of functional blocks in the evolution of exotrophy in vertebrates (as exemplified by fish and mammals). J EVOL BIOCHEM PHYS+ 2017. [DOI: 10.1134/s0022093017030012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
23
|
Role of DNA methylation in the dietary restriction mediated cellular memory. GeroScience 2017; 39:331-345. [PMID: 28477138 PMCID: PMC5505897 DOI: 10.1007/s11357-017-9976-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 04/11/2017] [Indexed: 01/13/2023] Open
Abstract
An important facet of dietary restriction (DR) that has been largely overlooked is that DR can have early effects that create a cellular memory, which persists even when DR is discontinued. The goal of this study was to determine if DNA methylation played a role in the cellular memory of DR by examining the effect of short-term DR on gene expression and DNA methylation and determining if the changes in expression and DNA methylation persist when DR is discontinued and mice returned to ad libitum (AL) feeding. We show that DR can induce substantial changes in gene expression within 1 month of its implementation in various tissues, and more interestingly, ~19–50% of these changes in gene expression persist across the tissues even when DR is discontinued. We then determined whether DR induced changes in DNA methylation in the promoter of three candidate genes identified from our gene expression analysis (Pomc, Hsph1, and Nts1) that correlated with the changes in the expression of these genes. Decreased methylation at three specific CG sites in the promoter of the Nts1 gene encompassing the distal consensus AP-1 site was correlated with increased Nts1 expression. Both the promoter hypomethylation and increased Nts1 expression persisted even after DR was discontinued and mice fed AL, supporting our hypothesis that DNA methylation could play a role in the memory effect of DR. The changes in DNA methylation in the Nts1 gene are likely to occur in intestinal stem cells and could play a role in preserving the intestinal stem cell pool in DR mice.
Collapse
|
24
|
Rhinehart EM. Mechanisms linking energy balance and reproduction: impact of prenatal environment. Horm Mol Biol Clin Investig 2016; 25:29-43. [PMID: 26943613 DOI: 10.1515/hmbci-2016-0004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 02/09/2016] [Indexed: 01/21/2023]
Abstract
The burgeoning field of metabolic reproduction regulation has been gaining momentum due to highly frequent discoveries of new neuroendocrine factors regulating both energy balance and reproduction. Universally throughout the animal kingdom, energy deficits inhibit the reproductive axis, which demonstrates that reproduction is acutely sensitive to fuel availability. Entrainment of reproductive efforts with energy availability is especially critical for females because they expend large amounts of energy on gestation and lactation. Research has identified an assortment of both central and peripheral factors involved in the metabolic regulation of reproduction. From an evolutionary perspective, these mechanisms likely evolved to optimize reproductive fitness in an environment with an unpredictable food supply and regular bouts of famine. To be effective, however, the mechanisms responsible for the metabolic regulation of reproduction must also retain developmental plasticity to allow organisms to adapt their reproductive strategies to their particular niche. In particular, the prenatal environment has emerged as a critical developmental window for programming the mechanisms responsible for the metabolic control of reproduction. This review will discuss the current knowledge about hormonal and molecular mechanisms that entrain reproduction with prevailing energy availability. In addition, it will provide an evolutionary, human life-history framework to assist in the interpretation of findings on gestational programming of the female reproductive function, with a focus on pubertal timing as an example. Future research should aim to shed light on mechanisms underlying the prenatal modulation of the adaptation to an environment with unstable resources in a way that optimizes reproductive fitness.
Collapse
|
25
|
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
|
26
|
Imbalanced insulin action in chronic over nutrition: Clinical harm, molecular mechanisms, and a way forward. Atherosclerosis 2016; 247:225-82. [PMID: 26967715 DOI: 10.1016/j.atherosclerosis.2016.02.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 12/31/2015] [Accepted: 02/02/2016] [Indexed: 02/08/2023]
Abstract
The growing worldwide prevalence of overnutrition and underexertion threatens the gains that we have made against atherosclerotic cardiovascular disease and other maladies. Chronic overnutrition causes the atherometabolic syndrome, which is a cluster of seemingly unrelated health problems characterized by increased abdominal girth and body-mass index, high fasting and postprandial concentrations of cholesterol- and triglyceride-rich apoB-lipoproteins (C-TRLs), low plasma HDL levels, impaired regulation of plasma glucose concentrations, hypertension, and a significant risk of developing overt type 2 diabetes mellitus (T2DM). In addition, individuals with this syndrome exhibit fatty liver, hypercoagulability, sympathetic overactivity, a gradually rising set-point for body adiposity, a substantially increased risk of atherosclerotic cardiovascular morbidity and mortality, and--crucially--hyperinsulinemia. Many lines of evidence indicate that each component of the atherometabolic syndrome arises, or is worsened by, pathway-selective insulin resistance and responsiveness (SEIRR). Individuals with SEIRR require compensatory hyperinsulinemia to control plasma glucose levels. The result is overdrive of those pathways that remain insulin-responsive, particularly ERK activation and hepatic de-novo lipogenesis (DNL), while carbohydrate regulation deteriorates. The effects are easily summarized: if hyperinsulinemia does something bad in a tissue or organ, that effect remains responsive in the atherometabolic syndrome and T2DM; and if hyperinsulinemia might do something good, that effect becomes resistant. It is a deadly imbalance in insulin action. From the standpoint of human health, it is the worst possible combination of effects. In this review, we discuss the origins of the atherometabolic syndrome in our historically unprecedented environment that only recently has become full of poorly satiating calories and incessant enticements to sit. Data are examined that indicate the magnitude of daily caloric imbalance that causes obesity. We also cover key aspects of healthy, balanced insulin action in liver, endothelium, brain, and elsewhere. Recent insights into the molecular basis and pathophysiologic harm from SEIRR in these organs are discussed. Importantly, a newly discovered oxide transport chain functions as the master regulator of the balance amongst different limbs of the insulin signaling cascade. This oxide transport chain--abbreviated 'NSAPP' after its five major proteins--fails to function properly during chronic overnutrition, resulting in this harmful pattern of SEIRR. We also review the origins of widespread, chronic overnutrition. Despite its apparent complexity, one factor stands out. A sophisticated junk food industry, aided by subsidies from willing governments, has devoted years of careful effort to promote overeating through the creation of a new class of food and drink that is low- or no-cost to the consumer, convenient, savory, calorically dense, yet weakly satiating. It is past time for the rest of us to overcome these foes of good health and solve this man-made epidemic.
Collapse
|
27
|
Pimentel GD, Contreras C, López M. Fatty Acids and Hypothalamic Dysfunction in Obesity. HANDBOOK OF LIPIDS IN HUMAN FUNCTION 2016:557-582. [DOI: 10.1016/b978-1-63067-036-8.00021-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
28
|
Abstract
Obesity ensues from an imbalance between energy intake and expenditure that results from gene-environment interactions, which favour a positive energy balance. A society that promotes unhealthy food and encourages sedentary lifestyle (that is, an obesogenic environment) has become a major contributory factor in excess fat deposition in individuals predisposed to obesity. Energy homeostasis relies upon control of energy intake as well as expenditure, which is in part determined by the themogenesis of brown adipose tissue and mediated by the sympathetic nervous system. Several areas of the brain that constitute cognitive and autonomic brain systems, which in turn form networks involved in the control of appetite and thermogenesis, also contribute to energy homeostasis. These networks include the dopamine mesolimbic circuit, as well as the opioid, endocannabinoid and melanocortin systems. The activity of these networks is modulated by peripheral factors such as hormones derived from adipose tissue and the gut, which access the brain via the circulation and neuronal signalling pathways to inform the central nervous system about energy balance and nutritional status. In this Review, I focus on the determinants of energy homeostasis that have emerged as prominent factors relevant to obesity.
Collapse
Affiliation(s)
- Denis Richard
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725 Chemin Sainte-Foy, Québec, QC G1V 4G5, Canada
| |
Collapse
|
29
|
Abstract
Leptin is an adipose-derived cytokine that has an important role in bodyweight homeostasis and energy balance. There are a number of studies which have suggested that leptin and its receptors dysregulation play a critical role in the development of malignancies including hematological malignancies, mainly via activation of the JAK/STAT pathway which regulates downstream signaling pathways such as PI3K/AKT signaling and ERK1/2. In this review, current understandings of leptin/leptin receptors mediated pathogenesis in various lymphoid malignancies are described. Blocking of the leptin receptor might be a unique therapeutic approach for many hematological malignancies.
Collapse
Affiliation(s)
- Shahab Uddin
- a Translational Research Institute, Academic Health System, Hamad Medical Corporation , Doha , Qatar
| | - Ramzi M Mohammad
- a Translational Research Institute, Academic Health System, Hamad Medical Corporation , Doha , Qatar
| |
Collapse
|
30
|
Contreras C, Gonzalez F, Fernø J, Diéguez C, Rahmouni K, Nogueiras R, López M. The brain and brown fat. Ann Med 2015; 47:150-68. [PMID: 24915455 PMCID: PMC4438385 DOI: 10.3109/07853890.2014.919727] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Accepted: 04/25/2014] [Indexed: 02/06/2023] Open
Abstract
Brown adipose tissue (BAT) is a specialized organ responsible for thermogenesis, a process required for maintaining body temperature. BAT is regulated by the sympathetic nervous system (SNS), which activates lipolysis and mitochondrial uncoupling in brown adipocytes. For many years, BAT was considered to be important only in small mammals and newborn humans, but recent data have shown that BAT is also functional in adult humans. On the basis of this evidence, extensive research has been focused on BAT function, where new molecules, such as irisin and bone morphogenetic proteins, particularly BMP7 and BMP8B, as well as novel central factors and new regulatory mechanisms, such as orexins and the canonical ventomedial nucleus of the hypothalamus (VMH) AMP- activated protein kinase (AMPK)-SNS-BAT axis, have been discovered and emerged as potential drug targets to combat obesity. In this review we provide an overview of the complex central regulation of BAT and how different neuronal cell populations co-ordinately work to maintain energy homeostasis.
Collapse
Affiliation(s)
- Cristina Contreras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria , Santiago de Compostela, 15782 , Spain
| | | | | | | | | | | | | |
Collapse
|
31
|
Chen S, Liu D, Wu J, Xu B, Lu K, Zhu W, Chen M. Effect of inhibiting the signal of mammalian target of rapamycin on memory T cells. Transplant Proc 2015; 46:1642-8. [PMID: 24935341 DOI: 10.1016/j.transproceed.2013.10.063] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 10/28/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND Mammalian target of rapamycin (mTOR) signaling is crucial to the activation and proliferation of T cells. Memory T cells can significantly hinder the induction of transplant tolerance. Resent research demonstrates that mTOR signaling regulates the survival and function of memory T cells. MATERIALS AND METHODS Naïve T cells were adoptively transferred to Rag(-/-) mice to generate similar memory T cells that undergo homeostatic proliferation. These memory T cells were then used to examine the effect of mTOR inhibition on the function of memory T cells. The effect of inhibiting mTOR signaling on the apoptosis of memory T cells was also examined. RESULTS Quantitative reverse-transcription polymerase chain reaction analysis showed that the expression of mTOR signaling was substantially lower in memory T cells. The levels of interferon-γ, interleukin (IL)-2, IL-4, and IL-10 decreased after mTOR inhibition; the expression of Bcl-2 increased in memory CD8(+) T cells and decreased in memory CD4(+) T cells; and Bax increased in memory CD4(+) T cells and decreased in memory CD8(+) T cells. Memory CD4(+) T cells were more sensitive to apoptotic cell death in this model after mTOR inhibition. Memory CD8(+) T cells were not affected by mTOR inhibition. CONCLUSIONS mTOR was crucial to homeostatic proliferation-induced memory T cells. The critical mechanisms of mTOR signaling inhibition are suppressed the functions of memory T cells and promoted the apoptosis of memory CD4(+) T cells.
Collapse
Affiliation(s)
- S Chen
- Medical School of Southeast University, Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nan Jing, China
| | - D Liu
- Medical School of Southeast University, Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nan Jing, China
| | - J Wu
- Department of Urology, Affiliated Nanjing First Hospital, Nanjing Medical University, Nan Jing, China
| | - B Xu
- Medical School of Southeast University, Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nan Jing, China
| | - K Lu
- Medical School of Southeast University, Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nan Jing, China
| | - W Zhu
- Medical School of Southeast University, Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nan Jing, China
| | - M Chen
- Medical School of Southeast University, Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nan Jing, China.
| |
Collapse
|
32
|
Leptin induced GRP78 expression through the PI3K-mTOR pathway in neuronal cells. Sci Rep 2014; 4:7096. [PMID: 25403445 PMCID: PMC4235288 DOI: 10.1038/srep07096] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 10/30/2014] [Indexed: 12/15/2022] Open
Abstract
Leptin is a circulating hormone that plays a critical role in regulating energy expenditure and food intake. Evidence to suggest the involvement of endoplasmic reticulum (ER) stress in the development of obesity is increasing. To adapt against ER stress, cells trigger the unfolded protein response (UPR). The 78 kDa glucose-regulated protein (GRP78) is an ER chaperone that protects cells against ER stress by inducing protein folding. In the present study, we hypothesized that leptin may activate UPR and protect against ER stress associated with obesity. SH-SY5Y, a human neuroblastoma cell line stably transfected with the Ob-Rb leptin receptor (SH-SY5Y-ObRb), was treated with leptin. We demonstrated that leptin induced GRP78 expression. We then validated the mechanism responsible for the leptin-induced expression of GRP78. Interestingly, leptin-induced GRP78 expression was not dependent on IRE1-XBP1 pathway. On the other hand, the PI3K inhibitor, LY294002, and mTOR inhibitor, rapamycin, inhibited the leptin-induced expression of GRP78. These results suggested that the leptin-induced expression of GRP78 may be dependent on the PI3K-mTOR pathway. Leptin specifically induced GRP78 because the induction of the ER-apoptotic marker, CHOP, was not detected in leptin-treated cells. Therefore, leptin may upregulate the expression of GRP78, thereby protecting against ER stress associated with obesity.
Collapse
|
33
|
Abstract
The mechanistic target of rapamycin (mTOR) signaling pathway is a crucial cellular signaling hub that, like the nervous system itself, integrates internal and external cues to elicit critical outputs including growth control, protein synthesis, gene expression, and metabolic balance. The importance of mTOR signaling to brain function is underscored by the myriad disorders in which mTOR pathway dysfunction is implicated, such as autism, epilepsy, and neurodegenerative disorders. Pharmacological manipulation of mTOR signaling holds therapeutic promise and has entered clinical trials for several disorders. Here, we review the functions of mTOR signaling in the normal and pathological brain, highlighting ongoing efforts to translate our understanding of cellular physiology into direct medical benefit for neurological disorders.
Collapse
|
34
|
Cassano S, Pucino V, La Rocca C, Procaccini C, De Rosa V, Marone G, Matarese G. Leptin modulates autophagy in human CD4+CD25- conventional T cells. Metabolism 2014; 63:1272-9. [PMID: 25060689 PMCID: PMC4180014 DOI: 10.1016/j.metabol.2014.06.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 06/13/2014] [Accepted: 06/14/2014] [Indexed: 12/29/2022]
Abstract
OBJECTIVE In this report we show that the adipocytokine leptin directly modulates autophagy in human CD4(+)CD25(-) conventional (Tconv) T cells. RESULTS In vitro treatment with recombinant human leptin determined an inhibition of autophagy during T cell receptor (TCR) stimulation, and this phenomenon was dose- and time-dependent. The events were secondary to the activation of the mammalian-target of rapamycin (mTOR)-pathway induced by leptin, as testified by its reversion induced by mTOR inhibition with rapamycin. At molecular level these phenomena associated with Bcl-2 up-regulation and its interaction with Beclin-1, whose complex exerts a negative effect on autophagy. MATERIALS/METHODS The impact of leptin on autophagy of Tconv cells was determined at biochemical level by western blotting and by flow cytometry; the interaction between BCL-2 and Beclin-1 by co-immunoprecipitation assays. CONCLUSIONS Our results, suggest that in unconditioned, freshly-isolated human Tconv cells, autophagy and proliferation are controlled by leptin during TCR-engagement, and that both phenomena occur alternatively indicating a balance between these processes during immune activation.
Collapse
Affiliation(s)
- Silvana Cassano
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), 80131 Napoli, Italy
| | - Valentina Pucino
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli "Federico II", 80131 Napoli, Italy
| | - Claudia La Rocca
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), 80131 Napoli, Italy; Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131 Napoli, Italy
| | - Claudio Procaccini
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), 80131 Napoli, Italy
| | - Veronica De Rosa
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), 80131 Napoli, Italy; Unità di NeuroImmunologia, IRCCS-Fondazione Santa Lucia, 00143 Roma, Italy
| | - Gianni Marone
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli "Federico II", 80131 Napoli, Italy; Centro Interdipartimentale di Ricerca in Scienze Immunologiche di Base e Cliniche (CISI), Università di Napoli "Federico II", 80131 Napoli, Italy
| | - Giuseppe Matarese
- Dipartimento di Medicina e Chirurgia, Facoltà di Medicina e Chirurgia, Università di Salerno, Baronissi Campus, 84081 Baronissi, Salerno, Italy; IRCCS-MultiMedica, 20138 Milano, Italy.
| |
Collapse
|
35
|
Wei T, Lu L, Shen Q, Fang C. [Advances of the relationship between leptin system and non-small cell lung cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2014; 17:347-50. [PMID: 24758911 PMCID: PMC6000024 DOI: 10.3779/j.issn.1009-3419.2014.04.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
瘦素系统在肺部炎症反应、癌症发生发展等过程中发挥重要作用,但是其在肿瘤微环境中的作用机理、对肺癌的诊断价值仍不明晰。本文就瘦素系统与非小细胞肺癌之间的关系,从瘦素及其受体在循环和肿瘤组织中的表达变化、瘦素信号转导通路、瘦素与调节性T细胞的相互作用和瘦素及其受体的基因多态性等方面进行叙述,以期为非小细胞肺癌的诊治提供新方法。
Collapse
Affiliation(s)
- Tengfei Wei
- Department of Laboratory Diagnosis, Changhai Hospital, the Second Military Medical University, Shanghai 200433, China
| | - Longkun Lu
- Department of Laboratory Diagnosis, Changhai Hospital, the Second Military Medical University, Shanghai 200433, China
| | - Qian Shen
- Department of Laboratory Diagnosis, Changhai Hospital, the Second Military Medical University, Shanghai 200433, China
| | - Chaoping Fang
- Department of Laboratory Diagnosis, Changhai Hospital, the Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
36
|
Genetic deletion of Rheb1 in the brain reduces food intake and causes hypoglycemia with altered peripheral metabolism. Int J Mol Sci 2014; 15:1499-510. [PMID: 24451134 PMCID: PMC3907882 DOI: 10.3390/ijms15011499] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Revised: 12/12/2013] [Accepted: 01/07/2014] [Indexed: 02/05/2023] Open
Abstract
Excessive food/energy intake is linked to obesity and metabolic disorders, such as diabetes. The hypothalamus in the brain plays a critical role in the control of food intake and peripheral metabolism. The signaling pathways in hypothalamic neurons that regulate food intake and peripheral metabolism need to be better understood for developing pharmacological interventions to manage eating behavior and obesity. Mammalian target of rapamycin (mTOR), a serine/threonine kinase, is a master regulator of cellular metabolism in different cell types. Pharmacological manipulations of mTOR complex 1 (mTORC1) activity in hypothalamic neurons alter food intake and body weight. Our previous study identified Rheb1 (Ras homolog enriched in brain 1) as an essential activator of mTORC1 activity in the brain. Here we examine whether central Rheb1 regulates food intake and peripheral metabolism through mTORC1 signaling. We find that genetic deletion of Rheb1 in the brain causes a reduction in mTORC1 activity and impairs normal food intake. As a result, Rheb1 knockout mice exhibit hypoglycemia and increased lipid mobilization in adipose tissue and ketogenesis in the liver. Our work highlights the importance of central Rheb1 signaling in euglycemia and energy homeostasis in animals.
Collapse
|
37
|
Penney CC, Volkoff H. Peripheral injections of cholecystokinin, apelin, ghrelin and orexin in cavefish (Astyanax fasciatus mexicanus): effects on feeding and on the brain expression levels of tyrosine hydroxylase, mechanistic target of rapamycin and appetite-related hormones. Gen Comp Endocrinol 2014; 196:34-40. [PMID: 24287340 DOI: 10.1016/j.ygcen.2013.11.015] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 11/15/2013] [Accepted: 11/18/2013] [Indexed: 12/27/2022]
Abstract
The effects of intraperitoneal injections of cholecystokinin (CCK), apelin, ghrelin, and orexin on food intake were examined in the blind cavefish Astyanax fasciatus mexicanus. CCK (50ng/g) induced a decrease in food intake whereas apelin (100ng/g), orexin (100ng/g), and ghrelin (100ng/g) induced an increase in food intake as compared to saline-injected control fish. In order to better understand the central mechanism by which these hormones act, we examined the effects of injections on the brain mRNA expression of two metabolic enzymes, tyrosine hydroxylase (TH), and mechanistic target of rapamycin (mTOR), and of appetite-regulating peptides, CCK, orexin, apelin and cocaine and amphetamine regulated transcript (CART). CCK injections induced a decrease in brain apelin injections, apelin injections induced an increase in TH, mTOR, and orexin brain expressions, orexin treatment increased brain TH expression and ghrelin injections induced an increase in mTOR and orexin brain expressions. CART expression was not affected by any of the injection treatments. Our results suggest that the enzymes TH and mTOR and the hormones CCK, apelin, orexin, and ghrelin all regulate food intake in cavefish through a complex network of interactions.
Collapse
Affiliation(s)
- Carla C Penney
- Departments of Biology and Biochemistry, Memorial University of Newfoundland, St. John's, NL A1B 3X9, Canada
| | - Hélène Volkoff
- Departments of Biology and Biochemistry, Memorial University of Newfoundland, St. John's, NL A1B 3X9, Canada.
| |
Collapse
|
38
|
Delporte C. Structure and physiological actions of ghrelin. SCIENTIFICA 2013; 2013:518909. [PMID: 24381790 PMCID: PMC3863518 DOI: 10.1155/2013/518909] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 11/10/2013] [Indexed: 05/30/2023]
Abstract
Ghrelin is a gastric peptide hormone, discovered as being the endogenous ligand of growth hormone secretagogue receptor. Ghrelin is a 28 amino acid peptide presenting a unique n-octanoylation modification on its serine in position 3, catalyzed by ghrelin O-acyl transferase. Ghrelin is mainly produced by a subset of stomach cells and also by the hypothalamus, the pituitary, and other tissues. Transcriptional, translational, and posttranslational processes generate ghrelin and ghrelin-related peptides. Homo- and heterodimers of growth hormone secretagogue receptor, and as yet unidentified receptors, are assumed to mediate the biological effects of acyl ghrelin and desacyl ghrelin, respectively. Ghrelin exerts wide physiological actions throughout the body, including growth hormone secretion, appetite and food intake, gastric secretion and gastrointestinal motility, glucose homeostasis, cardiovascular functions, anti-inflammatory functions, reproductive functions, and bone formation. This review focuses on presenting the current understanding of ghrelin and growth hormone secretagogue receptor biology, as well as the main physiological effects of ghrelin.
Collapse
Affiliation(s)
- Christine Delporte
- Laboratory of Pathophysiological and Nutritional Biochemistry, Université Libre de Bruxelles, 808 Route de Lennik, Bat G/E-CP611, 1070 Brussels, Belgium
| |
Collapse
|
39
|
Guzmán-Quevedo O, Da Silva Aragão R, Pérez García G, Matos RJB, de Sa Braga Oliveira A, de Castro RM, Bolaños-Jiménez F. Impaired hypothalamic mTOR activation in the adult rat offspring born to mothers fed a low-protein diet. PLoS One 2013; 8:e74990. [PMID: 24040371 PMCID: PMC3767644 DOI: 10.1371/journal.pone.0074990] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 08/08/2013] [Indexed: 12/23/2022] Open
Abstract
Several epidemiological and experimental studies have clearly established that maternal malnutrition induces a high risk of developing obesity and related metabolic diseases in the offspring. To determine if altered nutrient sensing might underlie this enhanced disease susceptibility, here we examined the effects of perinatal protein restriction on the activation of the nutrient sensor mTOR in response to acute variations in the nutritional status of the organism. Female Wistar rats were fed isocaloric diets containing either 17% protein (control) or 8% protein (PR) throughout pregnancy and lactation. At weaning offspring received standard chow and at 4 months of age the effects of fasting or fasting plus re-feeding on the phosphorylation levels of mTOR and its downstream target S6 ribosomal protein (rpS6) in the hypothalamus were assessed by immuno-fluorescence and western blot. Under ad libitum feeding conditions, PR rats exhibited decreased mTOR and rpS6 phosphorylation in the arcuate (ARC) and ventromedial (VMH) hypothalamic nuclei. Moreover, the phosphorylation of mTOR and rpS6 in these hypothalamic nuclei decreased with fasting in control but not in PR animals. Conversely, PR animals exhibited enhanced number of pmTOR imunostained cells in the paraventricular nucleus (PVN) and fasting decreased the activation of mTOR in the PVN of malnourished but not of control rats. These alterations occurred at a developmental stage at which perinatally-undernourished animals do not show yet obesity or glucose intolerance. Collectively, our observations suggest that altered hypothalamic nutrient sensing in response to an inadequate foetal and neonatal energetic environment is one of the basic mechanisms of the developmental programming of metabolic disorders and might play a causing role in the development of the metabolic syndrome induced by malnutrition during early life.
Collapse
Affiliation(s)
- Omar Guzmán-Quevedo
- Unité Mixte de recherche 1280 Physiologie des Adaptations Nutritionnelles, Institut National de la Recherche Agronomique, Nantes, France
- Université de Nantes, Nantes Atlantique Université, Nantes, France
| | - Raquel Da Silva Aragão
- Unité Mixte de recherche 1280 Physiologie des Adaptations Nutritionnelles, Institut National de la Recherche Agronomique, Nantes, France
- Université de Nantes, Nantes Atlantique Université, Nantes, France
- Departamento de Nutrição, Centro de Ciências da Saúde, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - Georgina Pérez García
- Unité Mixte de recherche 1280 Physiologie des Adaptations Nutritionnelles, Institut National de la Recherche Agronomique, Nantes, France
- Université de Nantes, Nantes Atlantique Université, Nantes, France
| | - Rhowena J. B. Matos
- Núcleo de Educação Física e Ciências do Esporte, Universidade Federal de Pernambuco, Vitória de Santo Antão, Pernambuco, Brazil
| | - André de Sa Braga Oliveira
- Unité Mixte de recherche 1280 Physiologie des Adaptations Nutritionnelles, Institut National de la Recherche Agronomique, Nantes, France
- Université de Nantes, Nantes Atlantique Université, Nantes, France
- Departamento de Nutrição, Centro de Ciências da Saúde, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - Raul Manhães de Castro
- Departamento de Nutrição, Centro de Ciências da Saúde, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - Francisco Bolaños-Jiménez
- Unité Mixte de recherche 1280 Physiologie des Adaptations Nutritionnelles, Institut National de la Recherche Agronomique, Nantes, France
- Université de Nantes, Nantes Atlantique Université, Nantes, France
- * E-mail:
| |
Collapse
|
40
|
Pierre JF, Heneghan AF, Lawson CM, Wischmeyer PE, Kozar RA, Kudsk KA. Pharmaconutrition Review. JPEN J Parenter Enteral Nutr 2013; 37:51S-65S. [DOI: 10.1177/0148607113493326] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Joseph F. Pierre
- Veterans Administration Surgical Services, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison
| | - Aaron F. Heneghan
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison
| | - Christy M. Lawson
- Department of Surgery, University of Tennessee Medical Center, Knoxville
| | | | - Rosemary A. Kozar
- Department of Surgery, University of Texas–Houston Health Science Center, Houston
| | - Kenneth A. Kudsk
- Veterans Administration Surgical Services, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison
| |
Collapse
|
41
|
Schneider JE, Wise JD, Benton NA, Brozek JM, Keen-Rhinehart E. When do we eat? Ingestive behavior, survival, and reproductive success. Horm Behav 2013; 64:702-28. [PMID: 23911282 DOI: 10.1016/j.yhbeh.2013.07.005] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 07/21/2013] [Accepted: 07/22/2013] [Indexed: 12/13/2022]
Abstract
The neuroendocrinology of ingestive behavior is a topic central to human health, particularly in light of the prevalence of obesity, eating disorders, and diabetes. The study of food intake in laboratory rats and mice has yielded some useful hypotheses, but there are still many gaps in our knowledge. Ingestive behavior is more complex than the consummatory act of eating, and decisions about when and how much to eat usually take place in the context of potential mating partners, competitors, predators, and environmental fluctuations that are not present in the laboratory. We emphasize appetitive behaviors, actions that bring animals in contact with a goal object, precede consummatory behaviors, and provide a window into motivation. Appetitive ingestive behaviors are under the control of neural circuits and neuropeptide systems that control appetitive sex behaviors and differ from those that control consummatory ingestive behaviors. Decreases in the availability of oxidizable metabolic fuels enhance the stimulatory effects of peripheral hormones on appetitive ingestive behavior and the inhibitory effects on appetitive sex behavior, putting a new twist on the notion of leptin, insulin, and ghrelin "resistance." The ratio of hormone concentrations to the availability of oxidizable metabolic fuels may generate a critical signal that schedules conflicting behaviors, e.g., mate searching vs. foraging, food hoarding vs. courtship, and fat accumulation vs. parental care. In species representing every vertebrate taxa and even in some invertebrates, many putative "satiety" or "hunger" hormones function to schedule ingestive behavior in order to optimize reproductive success in environments where energy availability fluctuates.
Collapse
Affiliation(s)
- Jill E Schneider
- Department of Biological Sciences, Lehigh University, 111 Research Drive, Bethlehem, PA 18015, USA
| | | | | | | | | |
Collapse
|
42
|
Kim HJ, Park EY, Oh MJ, Park SS, Shin KH, Choi SH, Chun BG, Kim DH. Central administration of metformin into the third ventricle of C57BL/6 mice decreases meal size and number and activates hypothalamic S6 kinase. Am J Physiol Regul Integr Comp Physiol 2013; 305:R499-505. [DOI: 10.1152/ajpregu.00099.2013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Administration of metformin is known to reduce both body weight and food intake. Although the hypothalamus is recognized as a critical regulator of energy balance and body weight, there is currently no evidence for an effect of metformin in the hypothalamus. Therefore, we sought to determine the central action of metformin on energy balance and body weight, as well as its potential involvement with key hypothalamic energy sensors, including adenosine monophosphate-activated protein kinase (AMPK) and S6 kinase (S6K). We used meal pattern analysis and a conditioned taste aversion (CTA) test and measured energy expenditure in C56BL/6 mice administered metformin (0, 7.5, 15, or 30 μg) into the third ventricle (I3V). Furthermore, we I3V-administered either control or metformin (30 μg) and compared the phosphorylation of AMPK and S6K in the mouse mediobasal hypothalamus. Compared with the control, I3V administration of metformin decreased body weight and food intake in a dose-dependent manner and did not result in CTA. Furthermore, the reduction in food intake induced by I3V administration of metformin was accomplished by decreases in both nocturnal meal size and number. Compared with the control, I3V administration of metformin significantly increased phosphorylation of S6K at Thr389 and AMPK at Ser485/491 in the mediobasal hypothalamus, while AMPK phosphorylation at Thr172 was not significantly altered. Moreover, I3V rapamycin pretreatment restored the metformin-induced anorexia and weight loss. These results suggest that the reduction in food intake induced by the central administration of metformin in the mice may be mediated by activation of S6K pathway.
Collapse
Affiliation(s)
- Hyun-Ju Kim
- Department of Pharmacology, Korea University College of Medicine, Seoul, Republic of Korea; and
| | - Eun-Young Park
- Department of Pharmacology, Korea University College of Medicine, Seoul, Republic of Korea; and
| | - Mi-Jeong Oh
- Department of Pharmacology, Korea University College of Medicine, Seoul, Republic of Korea; and
| | - Sung-Soo Park
- Department of Surgery, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kyung-Ho Shin
- Department of Pharmacology, Korea University College of Medicine, Seoul, Republic of Korea; and
| | - Sang-Hyun Choi
- Department of Pharmacology, Korea University College of Medicine, Seoul, Republic of Korea; and
| | - Boe-Gwun Chun
- Department of Pharmacology, Korea University College of Medicine, Seoul, Republic of Korea; and
| | - Dong-Hoon Kim
- Department of Pharmacology, Korea University College of Medicine, Seoul, Republic of Korea; and
| |
Collapse
|
43
|
Singhal SS, Figarola J, Singhal J, Reddy MA, Liu X, Berz D, Natarajan R, Awasthi S. RLIP76 protein knockdown attenuates obesity due to a high-fat diet. J Biol Chem 2013; 288:23394-406. [PMID: 23821548 DOI: 10.1074/jbc.m113.480194] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Feeding a Western high-fat diet (HFD) to C57BL/6 mice induces obesity, associated with a chronic inflammatory state, lipid transport, and metabolic derangements, and organ system effects that particularly prominent in the kidneys. Here, we report that RLIP76 homozygous knock-out (RLIP76(-/-)) mice are highly resistant to obesity as well as these other features of metabolic syndrome caused by HFD. The normal increase in pro-inflammatory and fibrotic markers associated with HFD induced obesity in wild-type C57B mice was broadly and nearly completely abrogated in RLIP76(-/-) mice. This is a particularly striking finding because chemical markers of oxidative stress including lipid hydroperoxides and alkenals were significantly higher in RLIP76(-/-) mice. Whereas HFD caused marked suppression of AMPK in wild-type C57B mice, RLIP76(-/-) mice had baseline activation of AMP-activated protein kinase, which was not further affected by HFD. The baseline renal function was reduced in RLIP76(-/-) mice as compared with wild-type, but was unaffected by HFD, in marked contrast to severe renal impairment and glomerulopathy in the wild-type mice given HFD. Our findings confirm a fundamental role of RLIP76 in regulating the function of obesity-promoting pro-inflammatory cytokines, and provide a novel mechanism for targeted therapy of obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Sharad S Singhal
- Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope, Comprehensive Cancer Center, Duarte, California 91010, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Zhang Q, Yu J, Liu B, Lv Z, Xia T, Xiao F, Chen S, Guo F. Central activating transcription factor 4 (ATF4) regulates hepatic insulin resistance in mice via S6K1 signaling and the vagus nerve. Diabetes 2013; 62:2230-9. [PMID: 23454693 PMCID: PMC3712031 DOI: 10.2337/db12-1050] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent studies have revealed that the central nervous system, particularly the hypothalamus, is critical for regulating insulin sensitivity in peripheral tissues. The aim of our current study is to investigate the possible involvement of hypothalamic activating transcription factor 4 (ATF4) in the regulation of insulin sensitivity in the liver. Here, we show that overexpression of ATF4 in the hypothalamus resulting from intracerebroventricular injection of adenovirus expressing ATF4 induces hepatic insulin resistance in mice and that inhibition of hypothalamic ATF4 by intracerebroventricular adenovirus expressing a dominant-negative ATF4 variant has the opposite effect. We also show that hypothalamic ATF4-induced insulin resistance is significantly blocked by selective hepatic vagotomy or by inhibiting activity of the mammalian target of rapamycin (mTOR) downstream target S6K1. Finally, we show that inhibition of hypothalamic ATF4 reverses hepatic insulin resistance induced by acute brain endoplasmic reticulum (ER) stress. Taken together, our study describes a novel central pathway regulating hepatic insulin sensitivity that is mediated by hypothalamic ATF4/mTOR/S6K1 signaling and the vagus nerve and demonstrates an important role for hypothalamic ATF4 in brain ER stress-induced hepatic insulin resistance. These results may lead to the identification of novel therapeutic targets for treating insulin resistance and associated metabolic diseases.
Collapse
|
45
|
Nazarians-Armavil A, Menchella JA, Belsham DD. Cellular insulin resistance disrupts leptin-mediated control of neuronal signaling and transcription. Mol Endocrinol 2013; 27:990-1003. [PMID: 23579487 DOI: 10.1210/me.2012-1338] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Central resistance to the actions of insulin and leptin is associated with the onset of obesity and type 2 diabetes mellitus, whereas leptin and insulin signaling is essential for both glucose and energy homeostasis. Although it is known that leptin resistance can lead to attenuated insulin signaling, whether insulin resistance can lead to or exacerbate leptin resistance is unknown. To investigate the molecular events underlying crosstalk between these signaling pathways, immortalized hypothalamic neuronal models, rHypoE-19 and mHypoA-2/10, were used. Prolonged insulin exposure was used to induce cellular insulin resistance, and thereafter leptin-mediated regulation of signal transduction and gene expression was assessed. Leptin directly repressed agouti-related peptide mRNA levels but induced urocortin-2, insulin receptor substrate (IRS)-1, IRS2, and IR transcription, through leptin-mediated phosphatidylinositol 3-kinase/Akt activation. Neuronal insulin resistance, as assessed by attenuated Akt phosphorylation, blocked leptin-mediated signal transduction and agouti-related peptide, urocortin-2, IRS1, IRS2, and insulin receptor synthesis. Insulin resistance caused a substantial decrease in insulin receptor protein levels, forkhead box protein 1 phosphorylation, and an increase in suppressor of cytokine signaling 3 protein levels. Cellular insulin resistance may cause or exacerbate neuronal leptin resistance and, by extension, obesity. It is essential to unravel the effects of neuronal insulin resistance given that both peripheral, as well as the less widely studied central insulin resistance, may contribute to the development of metabolic, reproductive, and cardiovascular disorders. This study provides improved understanding of the complex cellular crosstalk between insulin-leptin signal transduction that is disrupted during neuronal insulin resistance.
Collapse
Affiliation(s)
- Anaies Nazarians-Armavil
- Department of Physiology, University of Toronto, Medical Sciences Building 3247A, 1 Kings College Circle, Toronto, Ontario, Canada M5S 1A8
| | | | | |
Collapse
|
46
|
Woods SC. From conditioned hypoglycemia to obesity: following the data. Physiol Behav 2013; 121:19-24. [PMID: 23352822 DOI: 10.1016/j.physbeh.2013.01.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 01/03/2013] [Indexed: 01/24/2023]
Abstract
While a graduate student in the late 1960s I trained rats to lower their blood glucose in response to an arbitrary cue, a phenomenon called conditioned hypoglycemia. Over many years as my colleagues and I attempted to understand the underlying physiology of conditioned insulin secretion and conditioned hypoglycemia, it became clear that there were many implications that were highly important, including the entry of insulin into the brain, the existence of insulin receptors in certain brain areas, neural reflexes that project to insulin-secreting B-cells in the pancreas, the entrainment of those reflexes to improve the efficiency of meal-taking, and the possibility of adiposity signals from the body to the brain that influence behavior and metabolism. This article summarizes how we tackled each of these areas of research.
Collapse
Affiliation(s)
- Stephen C Woods
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, 2170 East Galbraith Road, Cincinnati, OH 45237, USA.
| |
Collapse
|
47
|
Castellano JM, Tena-Sempere M. Metabolic Regulation of Kisspeptin. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 784:363-83. [DOI: 10.1007/978-1-4614-6199-9_17] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
48
|
Herzer S, Silahtaroglu A, Meister B. Locked nucleic acid-based in situ hybridisation reveals miR-7a as a hypothalamus-enriched microRNA with a distinct expression pattern. J Neuroendocrinol 2012; 24:1492-504. [PMID: 22775435 DOI: 10.1111/j.1365-2826.2012.02358.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 06/26/2012] [Accepted: 07/03/2012] [Indexed: 12/11/2022]
Abstract
MicroRNAs (miRNAs) are short (∼22 nucleotides) noncoding RNA molecules that post-transcriptionally repress the expression of protein-coding genes by binding to 3'-untranslated regions of the target mRNAs. To identify miRNAs selectively expressed within the hypothalamus, a part of the brain that controls vital bodily functions, we employed locked nucleic acid (LNA)-fluorescent in situ hybridisation (FISH). The expression pattern of the mature miRNAs miR-7a, miR-7b, miR-137 and miR-153 in mouse brain tissue sections was investigated. Although all studied miRNAs were present in the hypothalamus, miR-7a, was the only miRNA found to be enriched in the hypothalamus, with low or no expression in other parts of the central nervous system (CNS). Within the hypothalamus, strong miR-7a expression was distinct and restricted to some hypothalamic nuclei and adjacent areas. miR-7a expression was particularly prominent in the subfornical organ, as well as the suprachiasmatic, paraventricular, periventricular, supraoptic, dorsomedial and arcuate nuclei. Identical expression patterns for miR-7a were seen in mouse and rat hypothalamus. By combining LNA-FISH with immunohistochemistry, it was shown that miR-7a was preferentially present in small orexigenic neuropeptide Y/agouti-related protein-containing-neurones located in the ventromedial aspect of the arcuate nucleus but not in large pro-opiomelanocortin/cocaine- and amphetamine-regulated transcript-containing anorexigenic neurones of the ventrolateral part of the arcuate nucleus. The limited and distinct expression of miR-7a in the CNS suggests that miR-7a has a role in post-transcriptional regulation in hypothalamic neurones. Particularly strong expression of miR-7a in neurones located in the ventromedial division of the arcuate nucleus, a subregion with a weak blood-brain barrier, raises the possibility that miR-7a is influenced by circulating hormones and is a regulator of the genes involved in body weight control.
Collapse
Affiliation(s)
- S Herzer
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | | |
Collapse
|
49
|
Blouet C, Schwartz GJ. Brainstem nutrient sensing in the nucleus of the solitary tract inhibits feeding. Cell Metab 2012; 16:579-87. [PMID: 23123165 PMCID: PMC3537851 DOI: 10.1016/j.cmet.2012.10.003] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2012] [Revised: 08/23/2012] [Accepted: 10/09/2012] [Indexed: 01/16/2023]
Abstract
Direct detection of circulating nutrients by the central nervous system has been implicated in the regulation of energy balance, and the mediobasal hypothalamus is considered as the primary sensing site mediating these effects. Neurons sensitive to energyrelated signals have also been identified outside the hypothalamus, particularly within the caudomedial nucleus of the solitary tract (cmNTS) in brainstem, but the consequences of direct cmNTS nutrient detection on energy balance remain poorly characterized. Here we determined the behavioral and metabolic consequences of direct L-leucine detection by the cmNTS and investigated the intracellular signaling and neurochemical pathways implicated in cmNTS L-leucine sensing in rats. Our results support the distributed nature of central nutrient detection, evidence a role for the cmNTS S6K1 pathway in the regulation of meal size and body weight, and suggest that the cmNTS integrates direct cmNTS nutrient detection with gut-derived, descending forebrain, and adiposity signals of energy availability to regulate food intake.
Collapse
Affiliation(s)
- Clemence Blouet
- Department of Medicine, Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | |
Collapse
|
50
|
Procaccini C, De Rosa V, Galgani M, Carbone F, Cassano S, Greco D, Qian K, Auvinen P, Calì G, Stallone G, Formisano L, La Cava A, Matarese G. Leptin-induced mTOR activation defines a specific molecular and transcriptional signature controlling CD4+ effector T cell responses. THE JOURNAL OF IMMUNOLOGY 2012; 189:2941-53. [PMID: 22904304 DOI: 10.4049/jimmunol.1200935] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The sensing by T cells of metabolic and energetic changes in the microenvironment can determine the differentiation, maturation, and activation of these cells. Although it is known that mammalian target of rapamycin (mTOR) gauges nutritonal and energetic signals in the extracellular milieu, it is not known how mTOR and metabolism influence CD4+CD25-FOXP3- effector T cell (Teff) responses. In this article, we show that leptin-induced activation of mTOR, which, in turn, controls leptin production and signaling, causes a defined cellular, biochemical, and transcriptional signature that determine the outcome of Teff responses, both in vitro and in vivo. The blockade of leptin/leptin receptor signaling, induced by genetic means or by starvation, leads to impaired mTOR activity that inhibits the proliferation of Teffs in vivo. Notably, the transcriptional signature of Teffs in the presence of leptin blockade appears similar to that observed in rapamycin-treated Teffs. These results identify a novel link between nutritional status and Teff responses through the leptin-mTOR axis and define a potential target for Teff modulation in normal and pathologic conditions.
Collapse
Affiliation(s)
- Claudio Procaccini
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche, Napoli 80131, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|