1
|
Chowdhury IH, Rahman S, Afroze YJ, Shovah ST. IUPHAR ECR review: Cancer-related anorexia-cachexia in cancer patients: Pathophysiology and treatment. Pharmacol Res 2024; 203:107129. [PMID: 38461961 DOI: 10.1016/j.phrs.2024.107129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 02/20/2024] [Accepted: 03/01/2024] [Indexed: 03/12/2024]
Abstract
Cancer-related anorexia-cachexia (CRAC) comprises one of the most common syndromes of advanced cancer patients. The prevalence of CRAC increases from 50% to 80% before death. CRAC is associated not only with impaired quality of life in patients and family members but also with shorter survival. The management of CRAC is a great challenge in clinical practice. There are no definite practice guidelines yet for the prevention and treatment of CRAC. A multimodal strategy is the most effective way to treat anorexia-cachexia. Numerous medications have been suggested and used in clinical trials, while others are still being studied on experimental animals. These medications include branched-chain amino acids, eicosapentaenoic acid, thalidomide, cytokine inhibitors, steroids, antiserotoninergic medications, and appetite stimulants. The benefits of supportive care interventions and the advancement of exciting new pharmacological medicines for anorexia-cachexia are becoming more widely recognized. Health care professionals need to be aware of the psychosocial and biological effects of anorexia-cachexia, even though knowledge of the underlying molecular causes of the disorder has advanced significantly.
Collapse
Affiliation(s)
| | | | - Yeasmin Jahan Afroze
- Department of Facio-Maxillary Surgical Oncology, National Institute of Cancer Research & Hospital, Dhaka 1212, Bangladesh
| | | |
Collapse
|
2
|
Mata-Pacheco V, Hernandez J, Varma N, Xu J, Sayers S, Le N, Wagner EJ. Dynamic, sex- and diet-specific pleiotropism in the PAC1 receptor-mediated regulation of arcuate proopiomelanocortin and Neuropeptide Y/Agouti related peptide neuronal excitability by anorexigenic ventromedial nucleus PACAP neurons. J Neuroendocrinol 2024; 36:e13357. [PMID: 38056947 DOI: 10.1111/jne.13357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 12/08/2023]
Abstract
This study furthers the investigation of how pituitary adenylate cyclase activating polypeptide (PACAP) and the PAC1 receptor (PAC1R) regulate the homeostatic energy balance circuitry. We hypothesized that apoptotic ablation of PACAP neurones in the hypothalamic ventromedial nucleus (VMN) would affect both energy intake and energy expenditure. We also hypothesized that selective PAC1R knockdown would impair the PACAP-induced excitation in anorexigenic proopiomelanocortin (POMC) neurones and inhibition of orexigenic neuropeptide Y (NPY)/agouti-related peptide (AgRP) neurones in the hypothalamic arcuate nucleus (ARC). The results show CASPASE-3-induced ablation of VMN PACAP neurones leads to increased energy intake and meal frequency as well as decreased energy expenditure in lean animals. The effects were more robust in obese males, whereas we saw the opposite effects in obese females. We then utilized visualized whole-cell patch clamp recordings in hypothalamic slices. PAC1R knockdown in POMC neurones diminishes the PACAP-induced depolarization, increase in firing, decreases in energy intake and meal size, as well as increases in CO2 production and O2 consumption. Similarly, the lack of expression of the PAC1R in NPY/AgRP neurones greatly attenuates the PACAP-induced hyperpolarization, suppression of firing, decreases in energy intake and meal frequency, as well as increases in energy expenditure. The PACAP response in NPY/AgRP neurones switched from predominantly inhibitory to excitatory in fasted animals. Finally, the anorexigenic effect of PACAP was potentiated when oestradiol was injected into the ARC in ovariectomized females. This study demonstrates the critical role of anorexigenic VMN PACAP neurones and the PAC1R in exciting POMC and inhibiting NPY/AgRP neurons to control homeostatic feeding.
Collapse
Affiliation(s)
- Veronica Mata-Pacheco
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, California, USA
| | - Jennifer Hernandez
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Nandini Varma
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Jenny Xu
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Sarah Sayers
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Nikki Le
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Edward J Wagner
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, California, USA
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| |
Collapse
|
3
|
Sayers S, Le N, Hernandez J, Mata‐Pacheco V, Wagner EJ. The vital role of arcuate nociceptin/orphanin FQ neurones in mounting an oestradiol-dependent adaptive response to negative energy balance via inhibition of nearby proopiomelanocortin neurones. J Physiol 2022; 600:4939-4961. [PMID: 36217719 PMCID: PMC9828807 DOI: 10.1113/jp283378] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 09/28/2022] [Indexed: 01/12/2023] Open
Abstract
We tested the hypothesis that N/OFQ neurones in the arcuate nucleus (N/OFQARC ) inhibit proopiomelanocortin (POMCARC ) neurones in a diet- and hormone-dependent manner to promote a more extensive rebound hyperphagia upon re-feeding following an 18 h fast. We utilized intact male or ovariectomized (OVX) female mice subjected to ad libitum-feeding or fasting conditions. N/OFQARC neurones under negative energy balance conditions displayed heightened sensitivity as evidenced by a decreased rheobase threshold, increased firing frequency, and increased burst duration and frequency compared to ad libitum-feeding conditions. Stimulation of N/OFQARC neurones more robustly inhibited POMCARC neurones under fasting conditions compared to ad libitum-feeding conditions. N/OFQARC inhibition of POMCARC neurones is hormone dependent as chemostimulation of N/OFQARC neurones from fasted males and OVX females produced a sizable outward current in POMCARC neurones. Oestradiol (E2 ) markedly attenuated the N/OFQ-induced POMCARC outward current. Additionally, N/OFQ tonically inhibits POMCARC neurones to a greater degree under fasting conditions than in ad libitum-feeding conditions as evidenced by the abrogation of N/OFQ-nociceptin opioid peptide (NOP) receptor signalling and inhibition of N/OFQ release via chemoinhibition of N/OFQARC neurones. Intra-arcuate nucleus application of N/OFQ further elevated the hyperphagic response and increased meal size during the 6 h re-feed period, and these effects were mimicked by chemostimulation of N/OFQARC neurones in vivo. E2 attenuated the robust N/OFQ-induced rebound hyperphagia seen in vehicle-treated OVX females. These data demonstrate that N/OFQARC neurones play a vital role in mitigating the impact of negative energy balance by inhibiting the excitability of anorexigenic neural substrates, an effect that is diminished by E2 in females. KEY POINTS: Nociceptin/orphanin FQ (N/OFQ) promotes increased energy intake and decreased energy expenditure under conditions of positive energy balance in a sex- and hormone-dependent manner. Here it is shown that under conditions of negative energy balance, i.e. fasting, N/OFQ inhibits anorexigenic proopiomelanocortin (POMC) neurones to a greater degree compared to homeostatic conditions due to fasting-induced hyperexcitability of N/OFQ neurones. Additionally, N/OFQ promotes a sustained increase in rebound hyperphagia and increase in meal size during the re-feed period following a fast. These results promote greater understanding of how energy balance influences the anorexigenic circuitry of the hypothalamus, and aid in understanding the neurophysiological pathways implicated in eating disorders promoting cachexia.
Collapse
Affiliation(s)
- Sarah Sayers
- Graduate College of Biomedical SciencesWestern University of Health SciencesPomonaCAUSA
| | - Nikki Le
- Graduate College of Biomedical SciencesWestern University of Health SciencesPomonaCAUSA
| | - Jennifer Hernandez
- Graduate College of Biomedical SciencesWestern University of Health SciencesPomonaCAUSA
| | - Veronica Mata‐Pacheco
- Graduate College of Biomedical SciencesWestern University of Health SciencesPomonaCAUSA
| | - Edward J. Wagner
- College of Osteopathic Medicine of the PacificWestern University of Health SciencesPomonaCAUSA
| |
Collapse
|
4
|
Le N, Sayers S, Mata-Pacheco V, Wagner EJ. The PACAP Paradox: Dynamic and Surprisingly Pleiotropic Actions in the Central Regulation of Energy Homeostasis. Front Endocrinol (Lausanne) 2022; 13:877647. [PMID: 35721722 PMCID: PMC9198406 DOI: 10.3389/fendo.2022.877647] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/30/2022] [Indexed: 12/11/2022] Open
Abstract
Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP), a pleiotropic neuropeptide, is widely distributed throughout the body. The abundance of PACAP expression in the central and peripheral nervous systems, and years of accompanying experimental evidence, indicates that PACAP plays crucial roles in diverse biological processes ranging from autonomic regulation to neuroprotection. In addition, PACAP is also abundantly expressed in the hypothalamic areas like the ventromedial and arcuate nuclei (VMN and ARC, respectively), as well as other brain regions such as the nucleus accumbens (NAc), bed nucleus of stria terminalis (BNST), and ventral tegmental area (VTA) - suggesting that PACAP is capable of regulating energy homeostasis via both the homeostatic and hedonic energy balance circuitries. The evidence gathered over the years has increased our appreciation for its function in controlling energy balance. Therefore, this review aims to further probe how the pleiotropic actions of PACAP in regulating energy homeostasis is influenced by sex and dynamic changes in energy status. We start with a general overview of energy homeostasis, and then introduce the integral components of the homeostatic and hedonic energy balance circuitries. Next, we discuss sex differences inherent to the regulation of energy homeostasis via these two circuitries, as well as the activational effects of sex steroid hormones that bring about these intrinsic disparities between males and females. Finally, we explore the multifaceted role of PACAP in regulating homeostatic and hedonic feeding through its actions in regions like the NAc, BNST, and in particular the ARC, VMN and VTA that occur in sex- and energy status-dependent ways.
Collapse
Affiliation(s)
- Nikki Le
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Sarah Sayers
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Veronica Mata-Pacheco
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Edward J. Wagner
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| |
Collapse
|
5
|
Austin D, May J, Andrade J, Nichols A. Exploring barriers, motivators and solutions to achieve a healthy lifestyle among undergraduate student nurses. BRITISH JOURNAL OF NURSING (MARK ALLEN PUBLISHING) 2022; 31:240-246. [PMID: 35220744 DOI: 10.12968/bjon.2022.31.4.240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Many preregistration student nurses tend to be overweight or obese and have unhealthy lifestyles. AIMS This study aimed to quantify the prevalence of these issues, to identify barriers to adopting a healthy lifestyle as well as potential solutions, and to explore the use of smartphone health apps. METHODS An online questionnaire examined diet and physical activity habits, general health and attitudes to eHealth. An in-class questionnaire with a new sample assessed current lifestyle, barriers to healthier living, support needed to achieve goals and the use of health apps. FINDINGS Half of student nurses were overweight or obese and only 41% met the recommended levels of physical activity. An in-class questionnaire revealed that more than half of them wanted to have a better diet and to exercise more. CONCLUSION Generally, student nurses were not satisfied with their current lifestyles. A lack of motivation and time were the most frequently reported barriers; an intervention focusing on motivational support could be an acceptable and effective means of achieving and sustaining positive behaviour change.
Collapse
Affiliation(s)
- Daniela Austin
- Research Associate, School of Psychology, Faculty of Health, University of Plymouth, Plymouth, daniela
| | - Jon May
- Professor, School of Psychology, Faculty of Health, University of Plymouth, Plymouth
| | - Jackie Andrade
- Professor, School of Psychology, Faculty of Health, University of Plymouth, Plymouth
| | - Andy Nichols
- Lecturer in Nursing Studies, School of Nursing and Midwifery, Faculty of Health, University of Plymouth, Plymouth
| |
Collapse
|
6
|
Masule MV, Rathod S, Agrawal Y, Patil CR, Nakhate KT, Ojha S, Goyal SN, Mahajan UB. Ghrelin mediated regulation of neurosynaptic transmitters in depressive disorders. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2022; 3:100113. [PMID: 35782191 PMCID: PMC9240712 DOI: 10.1016/j.crphar.2022.100113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 06/07/2022] [Accepted: 06/07/2022] [Indexed: 11/16/2022] Open
Abstract
Ghrelin is a peptide released by the endocrine cells of the stomach and the neurons in the arcuate nucleus of the hypothalamus. It modulates both peripheral and central functions. Although ghrelin has emerged as a potent stimulator of growth hormone release and as an orexigenic neuropeptide, the wealth of literature suggests its involvement in the pathophysiology of affective disorders including depression. Ghrelin exhibits a dual role through the advancement and reduction of depressive behavior with nervousness in the experimental animals. It modulates depression-related signals by forming neuronal networks with various neuropeptides and classical neurotransmitter systems. The present review emphasizes the integration and signaling of ghrelin with other neuromodulatory systems concerning depressive disorders. The role of ghrelin in the regulation of neurosynaptic transmission and depressive illnesses implies that the ghrelin system modulation can yield promising antidepressive therapies. Ghrelin is the orexigenic type of neuropeptide. It binds with the growth hormone secretagogue receptor (GHSR). GHSR is ubiquitously present in the various brain regions. Ghrelin is involved in the regulation of depression-related behavior. The review focuses on the neurotransmission and signaling of ghrelin in neuropsychiatric and depressive disorders.
Collapse
Affiliation(s)
- Milind V. Masule
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, 425405, Maharashtra, India
| | - Sumit Rathod
- Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Yogeeta Agrawal
- Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Chandragouda R. Patil
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, 425405, Maharashtra, India
| | - Kartik T. Nakhate
- Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Sameer N. Goyal
- Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
- Corresponding author.
| | - Umesh B. Mahajan
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, 425405, Maharashtra, India
- Corresponding author.
| |
Collapse
|
7
|
Differential Influence of Pueraria lobata Root Extract and Its Main Isoflavones on Ghrelin Levels in Alcohol-Treated Rats. Pharmaceuticals (Basel) 2021; 15:ph15010025. [PMID: 35056082 PMCID: PMC8777655 DOI: 10.3390/ph15010025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/22/2021] [Accepted: 12/22/2021] [Indexed: 12/19/2022] Open
Abstract
The study was carried out on alcohol-preferring male Wistar rats. The following drugs were repeatedly (28×) administered: acamprosate (500 mg/kg, p.o.), naltrexone (0.1 mg/kg, i.p), and Pueraria lobata (kudzu) root extract (KU) (500 mg/kg, p.o.) and its isoflavones: daidzin (40 mg/kg, p.o.) and puerarin (150 mg/kg, p.o.). Their effects on a voluntary alcohol intake were assessed. KU and alcohol were also given for 9 days in an experiment on alcohol tolerance development. Finally, total and active ghrelin levels in peripheral blood serum were measured by ELISA method. Acamprosate, naltrexone, daidzin, and puerarin, reducing the alcohol intake, caused an increase in both forms of ghrelin levels. On the contrary, though KU inhibited the alcohol intake and alcohol tolerance development, it reduced ghrelin levels in alcohol-preferring rats. The changes of ghrelin concentration could play a role as an indicator of the currently used drugs. The other effect on the KU-induced shift in ghrelin levels in the presence of alcohol requires further detailed study.
Collapse
|
8
|
Vintró-Alcaraz C, Baenas I, Lozano-Madrid M, Granero R, Ruiz-Canela M, Babio N, Corella D, Fitó M, Martínez JA, Alonso-Gómez ÁM, Wärnberg J, Vioque J, Romaguera D, López-Miranda J, Estruch R, Bernal-López MR, Lapetra J, Sánchez-Villegas A, Bueno-Cavanillas A, Tur JA, Martin-Sánchez V, Pintó X, Delgado-Rodríguez M, Matía-Martín P, Vidal J, Cárdenas JJ, Daimiel L, Ros E, Razquin C, Díaz-López A, González JI, Forcano L, Zulet MDLÁ, Bello-Mora MC, Valenzuela-Guerrero S, García de la Hera M, Konieczna J, García-Ríos A, Casas R, Gómez-Pérez AM, García-Arellano A, Bulló M, Sorli JV, Cuenca-Royo A, Abete I, Salaverria-Lete I, Basterra-Gortari FJ, de la Torre R, Salas-Salvadó J, Fernández-Aranda F. Psychological and metabolic risk factors in older adults with a previous history of eating disorder: A cross-sectional study from the Predimed-Plus study. EUROPEAN EATING DISORDERS REVIEW 2021; 29:575-587. [PMID: 33908163 DOI: 10.1002/erv.2833] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 01/05/2023]
Abstract
GOALS To explore affective and cognitive status, later in life, in individuals with and without previous history of eating disorder (ED), and also its association with higher risk for metabolic syndrome (MetS) symptomatology. METHODS A cross-sectional analysis of 6756 adults, aged 55-75 years with overweight/obesity and MetS participating in the Predimed-Plus study was conducted. Participants completed self-reported questionnaires to examine lifetime history of ED, according to DSM-5 criteria, and other psychopathological and neurocognitive factors. Anthropometric and metabolic measurements were also collected. RESULTS Of the whole sample, 24 individuals (0.35%) reported a previous history of ED. In this subsample, there were more women and singles compared to their counterparts, but they also presented higher levels of depressive symptoms and higher cognitive impairment, but also higher body mass index (BMI) and severe obesity, than those without lifetime ED. CONCLUSIONS This is one of the first studies to analyse the cognitive and metabolic impact of a previous history of ED. The results showed that previous ED was associated with greater affective and cognitive impairment, but also with higher BMI, later in life. No other MetS risk factors were found, after controlling for relevant variables.
Collapse
Affiliation(s)
- Cristina Vintró-Alcaraz
- Department of Psychiatry, Bellvitge University Hospital-IDIBELL, Barcelona, Spain.,Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Isabel Baenas
- Department of Psychiatry, Bellvitge University Hospital-IDIBELL, Barcelona, Spain.,Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - María Lozano-Madrid
- Department of Psychiatry, Bellvitge University Hospital-IDIBELL, Barcelona, Spain.,Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Roser Granero
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Department de Psicobiologia i Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Miguel Ruiz-Canela
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Department of Preventive Medicine and Public Health, Navarra Health Research Institute (IDISNA), University of Navarra, Pamplona, Spain
| | - Nancy Babio
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Unitat de Nutrició Humana, Reus, Spain.,Nutrition Unit, University Hospital of Sant Joan de Reus, Reus, Spain.,Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Dolores Corella
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Department of Preventive Medicine, University of Valencia, Valencia, Spain
| | - Montserrat Fitó
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Cardiovascular risk and Nutrition group, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - José Alfredo Martínez
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain.,Precision Nutrition Program IMDEA Food, CEI UAM +CSIC, Madrid, Spain
| | - Ángel M Alonso-Gómez
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Bioaraba Health Research Institute, Osakidetza Basque Health Service, Araba University Hospital, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Julia Wärnberg
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,School of Health Sciences, Instituto de Investigación Biomédica de Málaga, IBIMA, University of Málaga, Málaga, Spain
| | - Jesús Vioque
- Instituto de Investigación Sanitaria y Biomédica de Alicante, ISABIAL-UMH, Alicante, Spain.,CIBER de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
| | - Dora Romaguera
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Research Group on Nutritional Epidemiology & Cardiovascular Physiopathology (NUTRECOR), Health Research Institute of the Balearic Islands (IdISBa), University Hospital Son Espases (HUSE), Palma de Mallorca, Spain
| | - José López-Miranda
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Lipids and Atherosclerosis Unit, Department of Internal Medicine, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Córdoba, Spain
| | - Ramon Estruch
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Department of Internal Medicine, Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - María Rosa Bernal-López
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Internal Medicine Department, Regional University Hospital of Malaga, Instituto de Investigación Biomédica de Málaga (IBIMA), University of Málaga, Málaga, Spain
| | - José Lapetra
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Department of Family Medicine, Research Unit, Distrito Sanitario Atención Primaria Sevilla, Sevilla, Spain
| | - Almudena Sánchez-Villegas
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Institute for Biomedical Research, University of Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Aurora Bueno-Cavanillas
- Department of Preventive Medicine, University of Granada, Granada, Spain.,CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.,Instituto de Investigación Biosanitaria de Granada (ibs.Granada), Servicio Andaluz de Salud/Universidad de Granada, Granada, Spain
| | - Josep A Tur
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Research Group on Nutritional Epidemiology & Cardiovascular Physiopathology (NUTRECOR), Health Research Institute of the Balearic Islands (IdISBa), University Hospital Son Espases (HUSE), Palma de Mallorca, Spain.,Research Group on Community Nutrition & Oxidative Stress, University of Balearic Islands, Palma, Spain
| | - Vicente Martin-Sánchez
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,CIBER de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain.,Institute of Biomedicine (IBIOMED), University of León, León, Spain
| | - Xavier Pintó
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Lipids and Vascular Risk Unit, Internal Medicine, Hospital Universitario de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | - Pilar Matía-Martín
- Department of Endocrinology and Nutrition, Instituto de Investigación Sanitaria Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Josep Vidal
- Department of Endocrinology, IDIBAPS, Hospital Clinic, University of Barcelona, Barcelona, Spain.,CIBER Diabetes y Enfermedades Metabólicas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Jersy J Cárdenas
- Department of Endocrinology and Nutrition, University Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - Lidia Daimiel
- Nutritional Control of the Epigenome Group, Precision Nutrition and Obesity Program, IMDEA Food, CEI UAM + CSIC, Madrid, Spain
| | - Emilio Ros
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Lipid Clinic, Endocrinology and Nutrition Service, Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Cristina Razquin
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Department of Preventive Medicine and Public Health, Navarra Health Research Institute (IDISNA), University of Navarra, Pamplona, Spain
| | - Andrés Díaz-López
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Unitat de Nutrició Humana, Reus, Spain.,Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain.,Serra Hunter Fellow, Universitat Rovira i Virgili, Reus, Spain
| | - José I González
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Department of Preventive Medicine, University of Valencia, Valencia, Spain
| | - Laura Forcano
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Integrative Pharmacology and Systems Neurosciences, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - María de Los Ángeles Zulet
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain
| | - Maria C Bello-Mora
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Bioaraba Health Research Institute, Osakidetza Basque Health Service, Araba University Hospital, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Silvia Valenzuela-Guerrero
- School of Health Sciences, Instituto de Investigación Biomédica de Málaga, IBIMA, University of Málaga, Málaga, Spain
| | - Manoli García de la Hera
- Instituto de Investigación Sanitaria y Biomédica de Alicante, ISABIAL-UMH, Alicante, Spain.,CIBER de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
| | - Jadwiga Konieczna
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Research Group on Nutritional Epidemiology & Cardiovascular Physiopathology (NUTRECOR), Health Research Institute of the Balearic Islands (IdISBa), University Hospital Son Espases (HUSE), Palma de Mallorca, Spain
| | - Antonio García-Ríos
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Lipids and Atherosclerosis Unit, Department of Internal Medicine, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Córdoba, Spain
| | - Rosa Casas
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Department of Internal Medicine, Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Ana M Gómez-Pérez
- Department of Endocrinology, Virgen de la Victoria Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain
| | - Ana García-Arellano
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Department of Preventive Medicine and Public Health, Navarra Health Research Institute (IDISNA), University of Navarra, Pamplona, Spain.,Osasunbidea, Servicio Navarro de Salud, Pamplona, Spain
| | - Mònica Bulló
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Unitat de Nutrició Humana, Reus, Spain.,Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - José V Sorli
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Department of Preventive Medicine, University of Valencia, Valencia, Spain
| | - Aida Cuenca-Royo
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Integrative Pharmacology and Systems Neurosciences, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Itziar Abete
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain
| | - Itziar Salaverria-Lete
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Bioaraba Health Research Institute, Osakidetza Basque Health Service, Araba University Hospital, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain
| | - Francisco Javier Basterra-Gortari
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Department of Preventive Medicine and Public Health, Navarra Health Research Institute (IDISNA), University of Navarra, Pamplona, Spain.,Osasunbidea, Servicio Navarro de Salud, Pamplona, Spain
| | - Rafael de la Torre
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Integrative Pharmacology and Systems Neurosciences, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Jordi Salas-Salvadó
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Unitat de Nutrició Humana, Reus, Spain.,Nutrition Unit, University Hospital of Sant Joan de Reus, Reus, Spain.,Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Fernando Fernández-Aranda
- Department of Psychiatry, Bellvitge University Hospital-IDIBELL, Barcelona, Spain.,Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.,Department of Clinical Sciences, School of Medicine, University of Barcelona, Barcelona, Spain
| |
Collapse
|
9
|
Mosavat M, Mirsanjari M, Arabiat D, Smyth A, Whitehead L. The Role of Sleep Curtailment on Leptin Levels in Obesity and Diabetes Mellitus. Obes Facts 2021; 14:214-221. [PMID: 33756469 PMCID: PMC8138234 DOI: 10.1159/000514095] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 12/23/2020] [Indexed: 12/15/2022] Open
Abstract
Emerging evidence has identified sleep as a significant, but modifiable, risk factor for metabolic syndrome, diabetes, and obesity. Leptin, an adipocyte-derived peptide and a regulator of food intake and energy expenditure, has been shown to be associated with a short sleep duration in the pathophysiology of obesity and consequently type 2 diabetes. This review focuses on the current evidence indicating the effects of a short sleep duration on the regulation of leptin concentration in association with obesity and diabetes mellitus. In summary, the evidence suggests that sleep deprivation, by affecting leptin regulation, may lead to obesity and consequently development of type 2 diabetes through increased appetite and food intake. However, findings on the role of leptin in diabetes due to sleep deprivation are contradictory, and further studies with larger sample sizes are needed to confirm previous findings.
Collapse
Affiliation(s)
- Maryam Mosavat
- School of Nursing and Midwifery, Edith Cowan University, Joondalup, Washington, Australia,
| | - Mitra Mirsanjari
- Mazandaran University of Medical Sciences, Emam Khomeini Hospital, Mazandaran, Iran
| | - Diana Arabiat
- School of Nursing and Midwifery, Edith Cowan University, Joondalup, Washington, Australia
- Maternal and Child Nursing Department, The University of Jordan, Amman, Jordan
| | - Aisling Smyth
- School of Nursing and Midwifery, Edith Cowan University, Joondalup, Washington, Australia
| | - Lisa Whitehead
- School of Nursing and Midwifery, Edith Cowan University, Joondalup, Washington, Australia
| |
Collapse
|
10
|
Gastelum C, Perez L, Hernandez J, Le N, Vahrson I, Sayers S, Wagner EJ. Adaptive Changes in the Central Control of Energy Homeostasis Occur in Response to Variations in Energy Status. Int J Mol Sci 2021; 22:2728. [PMID: 33800452 PMCID: PMC7962960 DOI: 10.3390/ijms22052728] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/15/2022] Open
Abstract
Energy homeostasis is regulated in coordinate fashion by the brain-gut axis, the homeostatic energy balance circuitry in the hypothalamus and the hedonic energy balance circuitry comprising the mesolimbcortical A10 dopamine pathway. Collectively, these systems convey and integrate information regarding nutrient status and the rewarding properties of ingested food, and formulate it into a behavioral response that attempts to balance fluctuations in consumption and food-seeking behavior. In this review we start with a functional overview of the homeostatic and hedonic energy balance circuitries; identifying the salient neural, hormonal and humoral components involved. We then delve into how the function of these circuits differs in males and females. Finally, we turn our attention to the ever-emerging roles of nociceptin/orphanin FQ (N/OFQ) and pituitary adenylate cyclase-activating polypeptide (PACAP)-two neuropeptides that have garnered increased recognition for their regulatory impact in energy homeostasis-to further probe how the imposed regulation of energy balance circuitry by these peptides is affected by sex and altered under positive (e.g., obesity) and negative (e.g., fasting) energy balance states. It is hoped that this work will impart a newfound appreciation for the intricate regulatory processes that govern energy homeostasis, as well as how recent insights into the N/OFQ and PACAP systems can be leveraged in the treatment of conditions ranging from obesity to anorexia.
Collapse
Affiliation(s)
- Cassandra Gastelum
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (C.G.); (L.P.); (J.H.); (N.L.); (I.V.); (S.S.)
| | - Lynnea Perez
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (C.G.); (L.P.); (J.H.); (N.L.); (I.V.); (S.S.)
| | - Jennifer Hernandez
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (C.G.); (L.P.); (J.H.); (N.L.); (I.V.); (S.S.)
| | - Nikki Le
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (C.G.); (L.P.); (J.H.); (N.L.); (I.V.); (S.S.)
| | - Isabella Vahrson
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (C.G.); (L.P.); (J.H.); (N.L.); (I.V.); (S.S.)
| | - Sarah Sayers
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (C.G.); (L.P.); (J.H.); (N.L.); (I.V.); (S.S.)
| | - Edward J. Wagner
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (C.G.); (L.P.); (J.H.); (N.L.); (I.V.); (S.S.)
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
11
|
Homeostasis Research Model Based on Yin-Yang Theory: Five Examples. Chin J Integr Med 2021; 27:403-407. [PMID: 33433846 DOI: 10.1007/s11655-021-2861-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2020] [Indexed: 10/22/2022]
Abstract
In recent years, the ancient Yin-Yang theory has been gradually adopted by modern researchers, especially European and American scholars, and it has also been applied to modern scientific research on sleep, viruses, metabolism, cancer, genes, autoimmune diseases, and so on. It is very promising and fruitful results have been reported. However, the understanding of the connotations of Yin-Yang theory is not sufficient and thorough enough in these studies. If we understand and apply Yin-Yang theory more comprehensively, it may provide us with additional potential mechanisms and research directions worthy of study. On the basis of promoting a comprehensive understanding of all three connotations of Yin-Yang theory, this review attempts to illustrate this theory, summarize its applications in modern scientific research, and reveal the potential research direction of modern medicine.
Collapse
|
12
|
Gopalakrishnan S, Kannan NN. Only time will tell: the interplay between circadian clock and metabolism. Chronobiol Int 2020; 38:149-167. [PMID: 33345624 DOI: 10.1080/07420528.2020.1842436] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In most organisms ranging from cyanobacteria to humans, the endogenous timekeeping system temporally coordinates the behavioral, physiological, and metabolic processes with a periodicity close to 24 h. The timing of these daily rhythms is orchestrated by the synchronized oscillations of both the central pacemaker in the brain and the peripheral clocks located across multiple organs and tissues. A growing body of evidence suggests that the central circadian clock and peripheral clocks residing in the metabolically active tissues are incredibly well coordinated to confer coherent metabolic homeostasis. The interplay between nutrient metabolism and circadian rhythms can occur at various levels supported by the molecular clock network, multiple systemic mechanisms, and the neuroendocrine signaling pathways. While studies suggest the reciprocal regulation between circadian clock and metabolism, it is important to understand the precise mechanisms and the underlying pathways involved in the cross-talk among circadian oscillators and diverse metabolic networks. In addition to the internal synchronization of the metabolic rhythms, feeding time is considered as a potential external synchronization cue that fine tunes the timing of the circadian rhythms in metabolic peripheral clocks. A deeper understanding of how the timing of food intake and the diet composition drive the tissue-specific metabolic rhythms across the body is concomitantly important to develop novel therapeutic strategies for the metabolic disorders arising from circadian misalignment. This review summarizes the recent advancements in the circadian clock regulation of nutrient metabolism and discusses the current understanding of the metabolic feedback signals that link energy metabolism with the circadian clock.
Collapse
Affiliation(s)
- Swetha Gopalakrishnan
- Chronobiology Laboratory, School of Biology, Indian Institute of Science Education and Research (IISER) , Thiruvananthapuram, India
| | - Nisha N Kannan
- Chronobiology Laboratory, School of Biology, Indian Institute of Science Education and Research (IISER) , Thiruvananthapuram, India
| |
Collapse
|
13
|
Laar RA, Shi S, Ashraf MA, Khan MN, Bibi J, Liu Y. Impact of Physical Activity on Challenging Obesity in Pakistan: A Knowledge, Attitude, and Practice (KAP) Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17217802. [PMID: 33113780 PMCID: PMC7662990 DOI: 10.3390/ijerph17217802] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 01/08/2023]
Abstract
Physical activity (PA) refers to any action produced by skeletal muscle that consumes energy. According to the World Health Organization (WHO), PA is the primary element that can improve health at the community level. Obviously, PA plays an important role in the social, physical, and mental development of men and women, as well as in balancing weight. However, the large-scale negative impacts of physical inactivity on health-related issues are also recognized globally, such as obesity, which is the source of many non-communication diseases (NCDs). In Pakistan alone, 46% of deaths occur due to NCD. The majority of NCD deaths are linked to obesity, and Pakistan is the ninth most obese country in the world. Research on obesity caused by sedentary work in Pakistan is lacking, especially among university employees. To fill this gap, the current study mainly focuses on the rising non-communicable disease (NCD) rates among university employees in Pakistan due to a lack of exercise (obesity, in this case), with the help of a self-designed knowledge, attitude, and practice (KAP) questionnaire. Five universities in the Sindh province of Pakistan were surveyed (n = 276), following the concept of Yin-Yang as a theoretical lens. The results of the current study show that the knowledge, behaviors, and attitudes of university employees have a great influence on their body mass index (BMI). The study shows that Pakistani residents' (especially teaching staff) perceptions and attitudes towards obesity and PA have been instructive, but their practices need to be improved.
Collapse
Affiliation(s)
- Rizwan Ahmed Laar
- School of Sports Science and Physical Education, Nanjing Normal University, Nanjing 210023, China; (R.A.L.); (Y.L.)
| | - Shusheng Shi
- School of Sports Science and Physical Education, Nanjing Normal University, Nanjing 210023, China; (R.A.L.); (Y.L.)
- Correspondence:
| | - Muhammad Azeem Ashraf
- Research Institute of Educational Science, Hunan University, Changsha 410082, China;
| | - Muhammad Naeem Khan
- Department of Sociology, School of social and Behavioral Science, Nanjing University, Nanjing 210023, China;
| | - Jannat Bibi
- School of Physical Education, Shaanxi Normal University, Xi’an 710119, China;
| | - Yibing Liu
- School of Sports Science and Physical Education, Nanjing Normal University, Nanjing 210023, China; (R.A.L.); (Y.L.)
| |
Collapse
|
14
|
Horiuchi K, Kogiso T, Sagawa T, Ito T, Taniai M, Miura K, Hattori M, Morisada N, Hashimoto E, Tokushige K. Bardet-Biedl Syndrome Caused by Skipping of SCLT1 Complicated by Microvesicular Steatohepatitis. Intern Med 2020; 59:2719-2724. [PMID: 33132306 PMCID: PMC7691027 DOI: 10.2169/internalmedicine.5045-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
We treated the case of a 22-year-old male patient with liver dysfunction. At 1 year of age, hepatic fibrosis was suspected. In addition, due to the presence of retinitis pigmentosa, renal failure, obesity, mental retardation, and hypogonadism, he was diagnosed with Bardet-Biedl syndrome (BBS). Skipping of exons 14 and 17 in the sodium channel and clathrin linker 1 (SCLT1) gene was observed. At 22 years of age, the liver enzyme levels were further elevated and a diagnosis of microvesicular steatohepatitis was made. Insulin resistance, a reduction of muscle mass, an impairment of the fatty acid metabolism, and hyperleptinemia in this syndrome may cause steatohepatitis.
Collapse
Affiliation(s)
- Kentaro Horiuchi
- Institute of Gastroenterology, Department of Internal Medicine, Tokyo Women's Medical University, Japan
| | - Tomomi Kogiso
- Institute of Gastroenterology, Department of Internal Medicine, Tokyo Women's Medical University, Japan
| | - Takaomi Sagawa
- Institute of Gastroenterology, Department of Internal Medicine, Tokyo Women's Medical University, Japan
| | - Taito Ito
- Institute of Gastroenterology, Department of Internal Medicine, Tokyo Women's Medical University, Japan
| | - Makiko Taniai
- Institute of Gastroenterology, Department of Internal Medicine, Tokyo Women's Medical University, Japan
| | - Kenichiro Miura
- Department of Pediatric Nephrology, Tokyo Women's Medical University, Japan
| | - Motoshi Hattori
- Department of Pediatric Nephrology, Tokyo Women's Medical University, Japan
| | - Naoya Morisada
- Department of Pediatrics, Kobe University Graduate School of Medicine, Japan
| | | | - Katsutoshi Tokushige
- Institute of Gastroenterology, Department of Internal Medicine, Tokyo Women's Medical University, Japan
| |
Collapse
|
15
|
The interrelationship of body mass index with gray matter volume and resting-state functional connectivity of the hypothalamus. Int J Obes (Lond) 2019; 44:1097-1107. [PMID: 31796869 PMCID: PMC8643195 DOI: 10.1038/s41366-019-0496-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 10/26/2019] [Accepted: 11/06/2019] [Indexed: 11/08/2022]
Abstract
Background The hypothalamus plays an important role in regulating body weight through its interactions with multiple brain circuits involved in distinct aspects of feeding behavior. Yet, how hypothalamic gray matter volume (GMV) and connectivity may be related to individual differences in body weight remains unclear. We tested the hypothesis that the hypothalamus shows enhanced resting-state functional connectivity (rsFC) with regions of the reward, motivation, and motor circuits in positive correlation with body mass index (BMI) and the opposite with those associated with inhibitory control. We further examined the interdependent relationships between hypothalamic GMV, connectivity, and body weight. Methods Using seed-based rsFC and voxel-based morphometry analyses, we examined the relationship between the rsFC and GMV of the hypothalamus and BMI in 105 healthy humans. Additionally, we employed mediation analyses to characterize the inter-relationships between hypothalamic connectivity, GMV, and BMI. Results A whole-brain multiple regression showed that BMI was positively correlated with hypothalamic rsFC with the insula, thalamus, globus pallidus, and cerebellum, and negatively correlated with hypothalamic rsFC with the superior parietal lobule. Thus, higher BMI was associated with enhanced hypothalamic connectivity with regions involved in motivated feeding and reduced connectivity with those in support of cognitive control of food intake. A second whole-brain multiple regression revealed a positive correlation between hypothalamic GMV and the hypothalamus-posterior insula connectivity. Finally, the relationship between hypothalamic GMV and BMI was significantly and bidirectionally mediated by the hypothalamus-posterior insula connectivity. Conclusions The current findings suggest that the hypothalamus differentially interacts with the motivation, motor, and control circuits to regulate BMI. We further found evidence for the interdependence of hypothalamic structure, function, and body weight, which provides potential insights into the brain mechanisms of obesity.
Collapse
|
16
|
Mills JG, Larkin TA, Deng C, Thomas SJ. Weight gain in Major Depressive Disorder: Linking appetite and disordered eating to leptin and ghrelin. Psychiatry Res 2019; 279:244-251. [PMID: 30878306 DOI: 10.1016/j.psychres.2019.03.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/08/2019] [Accepted: 03/09/2019] [Indexed: 01/12/2023]
Abstract
Major Depressive Disorder (MDD) involves changes in appetite and weight, with a subset of individuals at an increased risk of weight gain. Pathways to weight gain may include appetite disturbances, excess eating, and dysregulation of appetite hormones. However, little research has simultaneously examined relationships between hormones, eating behaviours and MDD symptoms. Plasma ghrelin and leptin, biometrics, eating behaviours and psychopathology were compared between depressed (n = 60) and control (n = 60) participants. Depressed participants were subcategorised into those with increased or decreased appetite/weight for comparison by subtype. The Dutch Eating Behaviours Questionnaire and Yale Food Addiction Scale measured eating behaviours. Disordered eating was higher in MDD than controls, in females than males, and in depressed individuals with increased, compared to decreased, appetite/weight. Leptin levels were higher in females only. Leptin levels correlated positively, and ghrelin negatively, with disordered eating. The results provide further evidence for high levels of disordered eating in MDD, particularly in females. The correlations suggest that excessive eating in MDD is significantly linked to appetite hormones, indicating that it involves physiological, rather than purely psychological, factors. Further, longitudinal, research is needed to better understand whether hormonal factors play a causal role in excessive eating in MDD.
Collapse
Affiliation(s)
- Jessica G Mills
- Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, Australia.
| | - Theresa A Larkin
- Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, Australia
| | - Chao Deng
- Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, Australia; Antipsychotic Research Laboratory, University of Wollongong, Australia
| | - Susan J Thomas
- Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, Australia
| |
Collapse
|
17
|
Tonhajzerova I, Mestanikova A, Jurko A, Grendar M, Langer P, Ondrejka I, Jurko T, Hrtanek I, Cesnekova D, Mestanik M. Arterial stiffness and haemodynamic regulation in adolescent anorexia nervosa versus obesity. Appl Physiol Nutr Metab 2019; 45:81-90. [PMID: 31163115 DOI: 10.1139/apnm-2018-0867] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cardiovascular complications contribute to higher morbidity and mortality in patients with anorexia nervosa. We aimed to study biomarkers of cardiovascular risk in anorexic, normal-weight, and obese adolescents with focus on complex cardiovascular autonomic regulation and early arteriosclerotic damage. We examined 20 adolescent girls with anorexia nervosa, 20 obese girls, and 20 healthy normal-weight controls. Collected data: body composition analysis, 5 min recordings of R-R intervals and beat-to-beat blood pressure (BP), and arterial stiffness evaluated using cardio-ankle vascular index (CAVI). Evaluated parameters: beat-to-beat heart rate and BP variability, haemodynamic parameters (total peripheral resistance (TPR) cardiac output), CAVI, and anthropometric indices, including novel body roundness index (BRI). Adolescents with anorexia nervosa had increased CAVI associated with lower arterial constriction indexed by low-frequency band of BP variability compared with normal-weight peers (p = 0.03, p = 0.04, respectively) and obese adolescents (p < 0.01, p = 0.01, respectively). After normalization of CAVI and TPR by BRI, the relationship between CAVI and TPR was significant for all groups with the highest slope in the anorexia nervosa group (R2 = 0.724, p < 0.01). This is the first study revealing early arteriosclerotic damage in anorexic girls with increased CAVI. Complex analysis of cardiovascular autonomic regulation, and early arteriosclerotic, hemodynamic, and anthropometric changes in spectrum anorexia nervosa, normal weight, and obesity could help to understand the mechanisms of increased cardiovascular risk in malnutrition. Novelty Girls with anorexia nervosa showed signs of early arteriosclerotic damage indexed by CAVI. Insufficient sympathetic cardiovascular control was found already in adolescents with anorexia nervosa. The effect of body composition on CAVI was best predicted by novel body roundness index.
Collapse
Affiliation(s)
- Ingrid Tonhajzerova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4C, 036 01 Martin, Slovak Republic.,Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4D, 036 01 Martin, Slovak Republic
| | - Andrea Mestanikova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4C, 036 01 Martin, Slovak Republic.,Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4D, 036 01 Martin, Slovak Republic
| | - Alexander Jurko
- Pediatric Cardiology, Kollarova 13, 036 01 Martin, Slovak Republic
| | - Marian Grendar
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4D, 036 01 Martin, Slovak Republic
| | - Peter Langer
- Research Center, St. Anne's University Hospital Brno, 656 91 Brno, Czech Republic; Institute of Scientific Instruments of the ASCR, v.v.i., Kralovopolska 147, 612 64 Brno, Czech Republic
| | - Igor Ondrejka
- Psychiatric Clinic, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Hospital Martin, Kollarova 2, 036 59 Martin, Slovak Republic
| | - Tomas Jurko
- Department of Neonatology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Hospital Martin, Kollarova 2, 036 59 Martin, Slovak Republic
| | - Igor Hrtanek
- Psychiatric Clinic, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Hospital Martin, Kollarova 2, 036 59 Martin, Slovak Republic
| | - Dana Cesnekova
- Psychiatric Clinic, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Hospital Martin, Kollarova 2, 036 59 Martin, Slovak Republic
| | - Michal Mestanik
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4C, 036 01 Martin, Slovak Republic.,Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Mala Hora 4D, 036 01 Martin, Slovak Republic
| |
Collapse
|
18
|
Nour HA, El Sawaf AL, Elewa SM, El Sayed Y. Strength and independence of associations between ghrelin, leptin, adiponectin and insulin in stimulating basic functions to energy metabolism. ALEXANDRIA JOURNAL OF MEDICINE 2019. [DOI: 10.1016/j.ajme.2013.05.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Hoda A. Nour
- Physiology Department, Medical Research Institute, Alexandria University, Egypt
| | - Amel L. El Sawaf
- Physiology Department, Medical Research Institute, Alexandria University, Egypt
| | - Samia M. Elewa
- Physiology Department, Medical Research Institute, Alexandria University, Egypt
| | - Yosria El Sayed
- Physiology Department, Medical Research Institute, Alexandria University, Egypt
| |
Collapse
|
19
|
Sergi D, Campbell FM, Grant C, Morris AC, Bachmair EM, Koch C, McLean FH, Muller A, Hoggard N, de Roos B, Porteiro B, Boekschoten MV, McGillicuddy FC, Kahn D, Nicol P, Benzler J, Mayer CD, Drew JE, Roche HM, Muller M, Nogueiras R, Dieguez C, Tups A, Williams LM. SerpinA3N is a novel hypothalamic gene upregulated by a high-fat diet and leptin in mice. GENES AND NUTRITION 2018; 13:28. [PMID: 30519364 PMCID: PMC6263559 DOI: 10.1186/s12263-018-0619-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 11/06/2018] [Indexed: 02/08/2023]
Abstract
Background Energy homeostasis is regulated by the hypothalamus but fails when animals are fed a high-fat diet (HFD), and leptin insensitivity and obesity develops. To elucidate the possible mechanisms underlying these effects, a microarray-based transcriptomics approach was used to identify novel genes regulated by HFD and leptin in the mouse hypothalamus. Results Mouse global array data identified serpinA3N as a novel gene highly upregulated by both a HFD and leptin challenge. In situ hybridisation showed serpinA3N expression upregulation by HFD and leptin in all major hypothalamic nuclei in agreement with transcriptomic gene expression data. Immunohistochemistry and studies in the hypothalamic clonal neuronal cell line, mHypoE-N42 (N42), confirmed that alpha 1-antichymotrypsin (α1AC), the protein encoded by serpinA3, is localised to neurons and revealed that it is secreted into the media. SerpinA3N expression in N42 neurons is upregulated by palmitic acid and by leptin, together with IL-6 and TNFα, and all three genes are downregulated by the anti-inflammatory monounsaturated fat, oleic acid. Additionally, palmitate upregulation of serpinA3 in N42 neurons is blocked by the NFκB inhibitor, BAY11, and the upregulation of serpinA3N expression in the hypothalamus by HFD is blunted in IL-1 receptor 1 knockout (IL-1R1−/−) mice. Conclusions These data demonstrate that serpinA3 expression is implicated in nutritionally mediated hypothalamic inflammation.
Collapse
Affiliation(s)
- Domenico Sergi
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | - Fiona M Campbell
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | - Christine Grant
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | - Amanda C Morris
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | | | - Christiane Koch
- 2Department of Animal Physiology, Faculty of Biology, Philipps University Marburg, Karl-von-Frisch Str. 8, 35043 Marburg, Germany.,3Centre for Neuroendocrinology and Brain Health Research Centre, Department of Physiology, School of Medical Sciences, University of Otago, Dunedin, 9054 New Zealand
| | - Fiona H McLean
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | - Aifric Muller
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | - Nigel Hoggard
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | - Baukje de Roos
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | - Begona Porteiro
- 4Department of Physiology, University of Santiago de Compostela, 15705 Santiago de Compostela, Spain.,5CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Mark V Boekschoten
- 6Nutrition, Metabolism and Genomics Group, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | - Fiona C McGillicuddy
- 7Nutrigenomics Research Group, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Darcy Kahn
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | - Phyllis Nicol
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | - Jonas Benzler
- 2Department of Animal Physiology, Faculty of Biology, Philipps University Marburg, Karl-von-Frisch Str. 8, 35043 Marburg, Germany.,3Centre for Neuroendocrinology and Brain Health Research Centre, Department of Physiology, School of Medical Sciences, University of Otago, Dunedin, 9054 New Zealand
| | - Claus-Dieter Mayer
- 8Biomathematics & Statistics Scotland (BioSS), Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | - Janice E Drew
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | - Helen M Roche
- 7Nutrigenomics Research Group, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Michael Muller
- 9Nutrigenomics and Systems Nutrition Group, Norwich Medical School, University of East Anglia, Norwich, NR4 7UQ UK
| | - Ruben Nogueiras
- 4Department of Physiology, University of Santiago de Compostela, 15705 Santiago de Compostela, Spain.,5CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Carlos Dieguez
- 4Department of Physiology, University of Santiago de Compostela, 15705 Santiago de Compostela, Spain.,5CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Alexander Tups
- 2Department of Animal Physiology, Faculty of Biology, Philipps University Marburg, Karl-von-Frisch Str. 8, 35043 Marburg, Germany.,3Centre for Neuroendocrinology and Brain Health Research Centre, Department of Physiology, School of Medical Sciences, University of Otago, Dunedin, 9054 New Zealand
| | - Lynda M Williams
- 1Rowett Institute, University of Aberdeen, Aberdeen, AB25 2ZD UK
| |
Collapse
|
20
|
Mills JG, Thomas SJ, Larkin TA, Pai NB, Deng C. Problematic eating behaviours, changes in appetite, and weight gain in Major Depressive Disorder: The role of leptin. J Affect Disord 2018; 240:137-145. [PMID: 30071417 DOI: 10.1016/j.jad.2018.07.069] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 06/22/2018] [Accepted: 07/22/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Appetite and weight changes are core symptoms of Major Depressive Disorder (MDD), and those with MDD are at increased risk of obesity, cardiovascular disease and metabolic disorders. Leptin promotes satiety, with leptin dysregulation and resistance noted in obesity. However, the role of leptin in weight changes in MDD is not established. This study investigates leptin levels in relation to appetite and weight changes and problematic eating behaviours in MDD. METHODS Plasma leptin levels, psychopathology and biometrics were compared between participants meeting DSM-5 diagnostic criteria for MDD (n = 63) and healthy controls (n = 60). Depressed participants were also sub-categorised according to increased, decreased or unchanged appetite and weight. The Dutch Eating Behaviour Questionnaire and Yale Food Addiction Scale were examined in a subset of participants with MDD. RESULTS Females with increased appetite/weight had higher leptin levels than those with stable or reduced appetite/weight, however males showed the opposite effect. Leptin levels were positively correlated with problematic eating behaviours. One quarter of the depressed subset, all females, met the Yale criteria for food addiction, approximately double the rates reported in general community samples. LIMITATIONS The study is limited by a cross sectional design and a small sample size in the subset analysis of eating behaviours. CONCLUSIONS The results provide new information about associations between leptin, sex-specific weight and appetite changes and problematic eating behaviours, which may be risk factors for cardiovascular and metabolic diseases in MDD, particularly in females. Future longitudinal research investigating leptin as a risk factor for weight gain in MDD is warranted, and may lead to early interventions aimed at preventing weight gain in at-risk individuals.
Collapse
Affiliation(s)
- Jessica G Mills
- School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, Australia.
| | - Susan J Thomas
- School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, Australia
| | - Theresa A Larkin
- School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, Australia
| | - Nagesh B Pai
- School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, Australia
| | - Chao Deng
- School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, Australia; Illawarra Health and Medical Research Institute, University of Wollongong, Australia; Antipsychotic Research Laboratory, University of Wollongong, Australia
| |
Collapse
|
21
|
Bederman IR, Pora G, O'Reilly M, Poleman J, Spoonhower K, Puchowicz M, Perez A, Erokwu BO, Rodriguez-Palacios A, Flask CA, Drumm ML. Absence of leptin signaling allows fat accretion in cystic fibrosis mice. Am J Physiol Gastrointest Liver Physiol 2018; 315:G685-G698. [PMID: 30118352 PMCID: PMC6293256 DOI: 10.1152/ajpgi.00344.2017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 07/09/2018] [Accepted: 07/16/2018] [Indexed: 01/31/2023]
Abstract
Negative energy balance is a prevalent feature of cystic fibrosis (CF). Pancreatic insufficiency, elevated energy expenditure, lung disease, and malnutrition, all characteristic of CF, contribute to the negative energy balance causing low body-growth phenotype. As low body weight and body mass index strongly correlate with poor lung health and survival of patients with CF, improving energy balance is an important clinical goal (e.g., high-fat diet). CF mouse models also exhibit negative energy balance (growth retardation and high energy expenditure), independent from exocrine pancreatic insufficiency, lung disease, and malnutrition. To improve energy balance through increased caloric intake and reduced energy expenditure, we disrupted leptin signaling by crossing the db/db leptin receptor allele with mice carrying the R117H Cftr mutation. Compared with db/db mice, absence of leptin signaling in CF mice (CF db/db) resulted in delayed and moderate hyperphagia with lower de novo lipogenesis and lipid deposition, producing only moderately obese CF mice. Greater body length was found in db/db mice but not in CF db/db, suggesting CF-dependent effect on bone growth. The db/db genotype resulted in lower energy expenditure regardless of Cftr genotype leading to obesity. Despite the db/db genotype, the CF genotype exhibited high respiratory quotient indicating elevated carbohydrate oxidation, thus limiting carbohydrates for lipogenesis. In summary, db/db-linked hyperphagia, elevated lipogenesis, and morbid obesity were partially suppressed by reduced CFTR activity. CF mice still accrued large amounts of adipose tissue in contrast to mice fed a high-fat diet, thus highlighting the importance of dietary carbohydrates and not simply fat for energy balance in CF. NEW & NOTEWORTHY We show that cystic fibrosis (CF) mice are able to accrue fat under conditions of carbohydrate overfeeding, increased lipogenesis, and decreased energy expenditure, although length was unaffected. High-fat diet feeding failed to improve growth in CF mice. Morbid db/db-like obesity was reduced in CF double-mutant mice by reduced CFTR activity.
Collapse
Affiliation(s)
- Ilya R Bederman
- Department of Pediatrics, Case Western Reserve University , Cleveland, Ohio
| | - Gavriella Pora
- Department of Pediatrics, Case Western Reserve University , Cleveland, Ohio
| | - Maureen O'Reilly
- Department of Pediatrics, Case Western Reserve University , Cleveland, Ohio
| | - James Poleman
- Department of Pediatrics, Case Western Reserve University , Cleveland, Ohio
| | | | - Michelle Puchowicz
- Department of Nutrition, Case Western Reserve University , Cleveland, Ohio
| | - Aura Perez
- Department of Pediatrics, Case Western Reserve University , Cleveland, Ohio
| | | | - Alex Rodriguez-Palacios
- Department of Medicine, School of Medicine, Case Western Reserve University , Cleveland, Ohio
| | - Chris A Flask
- Department of Pediatrics, Case Western Reserve University , Cleveland, Ohio
- Department of Radiology, Case Western Reserve University , Cleveland, Ohio
- Department of Biomedical Engineering, School of Engineering, Case Western Reserve University , Cleveland, Ohio
| | - Mitchell L Drumm
- Department of Pediatrics, Case Western Reserve University , Cleveland, Ohio
| |
Collapse
|
22
|
Chouzouris TM, Dovolou E, Rekkas CA, Georgoulias P, Athanasiou LV, Amiridis GS. A study on ghrelin and LH secretion after short fasting and on ghrelin levels at perioestrual period in dairy cattle. Reprod Domest Anim 2018; 54:91-99. [PMID: 30171634 DOI: 10.1111/rda.13321] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 08/13/2018] [Indexed: 12/19/2022]
Abstract
In two experiments, we studied (a) the changes of LH secretion in heifers under different feeding schedules and (b) total ghrelin concentration at oestrus in cows and heifers. In experiment one, synchronized heifers were allocated in three groups (R, regularly fed controls; F, fasted; and F-F fasted-fed). One day after the completion of the oestrous induction protocol, group F and F-F animals stayed without feed for 24 hr; thereafter, feed was provided to R and F-F cattle; 2 hr later, GnRH was administered to all animals. Blood samples were collected for ghrelin, progesterone, LH and cortisol concentrations. Fasting caused increased ghrelin concentrations in groups F and F-F, while in response to GnRH, LH surge was significantly attenuated in groups F and F-F compared to R. In experiment 2, lactating cows and heifers were used. On day 9 of a synchronized cycle, PGF2α was administered, and blood samples were collected twice daily until the third day after oestrus and analysed for progesterone, estradiol, ghrelin, glucose and BHBA concentrations. No difference was recorded between groups in steroids and BHBA concentrations. In comparison to mid-luteal values, ghrelin concentrations significantly increased at perioestrual period in cows, but not in heifers. This study provides evidence that starving-induced elevated ghrelin concentrations can have suppressing effect on LH secretion, even after ghrelin's restoration to basal values and that during oestrus, ghrelin secretion is differently regulated in cows and heifers, likely being independent from oestradiol concentrations. Further research is required to identify the determining factors that drive the different regulation of ghrelin secretion in cows and heifers.
Collapse
Affiliation(s)
- Thomas Markos Chouzouris
- Department of Obstetrics and Reproduction, Veterinary Faculty, University of Thessaly, Karditsa, Greece
| | - Eleni Dovolou
- Department of Obstetrics and Reproduction, Veterinary Faculty, University of Thessaly, Karditsa, Greece
| | | | - Panagiotis Georgoulias
- Department of Nuclear Medicine, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Lambrini V Athanasiou
- Department of Medicine, Veterinary Faculty, University of Thessaly, Karditsa, Greece
| | - Georgios S Amiridis
- Department of Obstetrics and Reproduction, Veterinary Faculty, University of Thessaly, Karditsa, Greece
| |
Collapse
|
23
|
Kirsch M, Mertens W. On the Drive Specificity of Freudian Drives for the Generation of SEEKING Activities: The Importance of the Underestimated Imperative Motor Factor. Front Psychol 2018; 9:616. [PMID: 29774002 PMCID: PMC5943553 DOI: 10.3389/fpsyg.2018.00616] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 04/12/2018] [Indexed: 12/19/2022] Open
Abstract
Doubters of Freud's theory of drives frequently mentioned that his approach is outdated and therefore cannot be useful for solving current problems in patients with mental disorders. At present, many scientists believe that affects rather than drives are of utmost importance for the emotional life and the theoretical framework of affective neuroscience, developed by Panksepp, strongly underpinned this view. Panksepp evaluated seven so-called command systems and the SEEKING system is therein of central importance. Panksepp used Pankseppian drives as inputs for the SEEKING system but noted the missing explanation of drive-specific generation of SEEKING activities in his description. Drive specificity requires dual action of the drive: the activation of a drive-specific brain area and the release of the neurotransmitter dopamine. Noticeably, as Freud claimed drive specificity too, it was here analyzed whether a Freudian drive can evoke the generation of drive-specific SEEKING activities. Special importance was addressed to the imperative motor factor in Freud's drive theory because Panksepp's formulations focused on neural pathways without specifying underlying neurotransmitter/endocrine factors impelling motor activity. As Panksepp claimed sleep as a Pankseppian drive, we firstly had to classified sleep as a Freudian drive by using three evaluated criteria for a Freudian drive. After that it was possible to identify the imperative motor factors of hunger, thirst, sex, and sleep. Most importantly, all of these imperative motor factors can both activate a drive-specific brain area and release dopamine from dopaminergic neurons, i.e., they can achieve the so-called drive specificity. Surprisingly, an impaired Freudian drive can alter via endocrinological pathways the concentration of the imperative motor factor of a second Freudian drive, obviously in some independence to the level of the metabolic deficit, thereby offering the possibility to modulate the generation of SEEKING activities of this second Freudian drive. This novel possibility might help to refine the general understanding of the action of Freudian drives. As only imperative motor factors of Freudian drives can guarantee drive specificity for the generation of SEEKING activities, the impact of Freud's construct Eros (with its constituents hunger, thirst, sex, and sleep) should be revisited.
Collapse
Affiliation(s)
- Michael Kirsch
- Institute of Physiological Chemistry, University Hospital Essen, Essen, Germany
| | - Wolfgang Mertens
- Division of Clinical Psychology and Psychotherapy, Department of Psychology, Faculty of Psychology and Educational Sciences, Ludwig Maximilian University of Munich, Munich, Germany
| |
Collapse
|
24
|
Saga of ghrelin: its structure, actions, and therapeutic uses. ASIAN BIOMED 2018. [DOI: 10.2478/abm-2010-0112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Abstract
The fat-regulating hormones, adiponectin, ghrelin, and leptin have been studied extensively because therapeutic modality might be gleaned from their augmentation or blockade. Since a link between the hormone and control of hunger was found, ghrelin levels in the human body have received a great deal of attention over the past several years. This mini-review summarizes the role of ghrelin, describing its actions and therapeutic uses.
Collapse
|
25
|
Ding C, Lim LL, Xu L, Kong APS. Sleep and Obesity. J Obes Metab Syndr 2018; 27:4-24. [PMID: 31089536 PMCID: PMC6489488 DOI: 10.7570/jomes.2018.27.1.4] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/08/2018] [Accepted: 03/09/2018] [Indexed: 12/30/2022] Open
Abstract
Rising global prevalence and incidence of obesity lead to increased cardiovascular-renal complications and cancers. Epidemiological studies reported a worldwide trend towards suboptimal sleep duration and poor sleep quality in parallel with this obesity epidemic. From rodents and human models, it is highly plausible that abnormalities in sleep, both quantity and quality, impact negatively on energy metabolism. While excess dietary intake and physical inactivity are the known drivers of the obesity epidemic, promotion of healthy sleep habits has emerged as a new target to combat obesity. In this light, present review focuses on the existing literature examining the relationship between sleep physiology and energy homeostasis. Notably, sleep dysregulation perturbs the metabolic milieu via alterations in hormones such as leptin and ghrelin, eating behavior, neuroendocrine and autonomic nervous systems. In addition, shift work and trans-meridian air travel may exert a negative influence on the hypothalamic-pituitary-adrenal axis and trigger circadian misalignment, leading to impaired glucose tolerance and increased fat accumulation. Amassing evidence has also suggested that uncoupling of the circadian clock can increase the risk of adverse metabolic health. Given the importance of sleep in maintaining energy homeostasis and that it is potentially modifiable, promoting good sleep hygiene may create new avenues for obesity prevention and treatment.
Collapse
Affiliation(s)
- Chenzhao Ding
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong,
China
| | - Lee Ling Lim
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong,
China
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur,
Malaysia
| | - Li Xu
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong,
China
| | - Alice Pik Shan Kong
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong,
China
| |
Collapse
|
26
|
Avendaño MS, Vazquez MJ, Tena-Sempere M. Disentangling puberty: novel neuroendocrine pathways and mechanisms for the control of mammalian puberty. Hum Reprod Update 2018; 23:737-763. [PMID: 28961976 DOI: 10.1093/humupd/dmx025] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 08/01/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Puberty is a complex developmental event, controlled by sophisticated regulatory networks that integrate peripheral and internal cues and impinge at the brain centers driving the reproductive axis. The tempo of puberty is genetically determined but is also sensitive to numerous modifiers, from metabolic and sex steroid signals to environmental factors. Recent epidemiological evidence suggests that the onset of puberty is advancing in humans, through as yet unknown mechanisms. In fact, while much knowledge has been gleaned recently on the mechanisms responsible for the control of mammalian puberty, fundamental questions regarding the intimate molecular and neuroendocrine pathways responsible for the precise timing of puberty and its deviations remain unsolved. OBJECTIVE AND RATIONALE By combining data from suitable model species and humans, we aim to provide a comprehensive summary of our current understanding of the neuroendocrine mechanisms governing puberty, with particular focus on its central regulatory pathways, underlying molecular basis and mechanisms for metabolic control. SEARCH METHODS A comprehensive MEDLINE search of articles published mostly from 2003 to 2017 has been carried out. Data from cellular and animal models (including our own results) as well as clinical studies focusing on the pathophysiology of puberty in mammals were considered and cross-referenced with terms related with central neuroendocrine mechanisms, metabolic control and epigenetic/miRNA regulation. OUTCOMES Studies conducted during the last decade have revealed the essential role of novel central neuroendocrine pathways in the control of puberty, with a prominent role of kisspeptins in the precise regulation of the pubertal activation of GnRH neurosecretory activity. In addition, different transmitters, including neurokinin-B (NKB) and, possibly, melanocortins, have been shown to interplay with kisspeptins in tuning puberty onset. Alike, recent studies have documented the role of epigenetic mechanisms, involving mainly modulation of repressors that target kisspeptins and NKB pathways, as well as microRNAs and the related binding protein, Lin28B, in the central control of puberty. These novel pathways provide the molecular and neuroendocrine basis for the modulation of puberty by different endogenous and environmental cues, including nutritional and metabolic factors, such as leptin, ghrelin and insulin, which are known to play an important role in pubertal timing. WIDER IMPLICATIONS Despite recent advancements, our understanding of the basis of mammalian puberty remains incomplete. Complete elucidation of the novel neuropeptidergic and molecular mechanisms summarized in this review will not only expand our knowledge of the intimate mechanisms responsible for puberty onset in humans, but might also provide new tools and targets for better prevention and management of pubertal deviations in the clinical setting.
Collapse
Affiliation(s)
- M S Avendaño
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Avda. Menéndez Pidal, s/n, 14004 Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, Faculty of Medicine, University of Córdoba, Avda. Menéndez Pidal s/n. 14004 Córdoba, Spain.,Hospital Universitario Reina Sofia, Avda. Menéndez Pidal, s/n, 14004 Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Avda. Menéndez Pidal, s/n, 14004 Córdoba, Spain
| | - M J Vazquez
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Avda. Menéndez Pidal, s/n, 14004 Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, Faculty of Medicine, University of Córdoba, Avda. Menéndez Pidal s/n. 14004 Córdoba, Spain.,Hospital Universitario Reina Sofia, Avda. Menéndez Pidal, s/n, 14004 Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Avda. Menéndez Pidal, s/n, 14004 Córdoba, Spain
| | - M Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Avda. Menéndez Pidal, s/n, 14004 Córdoba, Spain.,Department of Cell Biology, Physiology and Immunology, Faculty of Medicine, University of Córdoba, Avda. Menéndez Pidal s/n. 14004 Córdoba, Spain.,Hospital Universitario Reina Sofia, Avda. Menéndez Pidal, s/n, 14004 Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Avda. Menéndez Pidal, s/n, 14004 Córdoba, Spain.,FiDiPro Program, Department of Physiology, University of Turku, Kiinamyllynkatu 10, FIN-20520 Turku, Finland
| |
Collapse
|
27
|
Yang SJ, Do HJ, Jung Y, Hwang GS, Shin MJ. Voglibose-mediated alterations in neurometabolomic profiles in the hypothalamus of high-fat diet-fed mice. Nutr Neurosci 2018; 22:760-767. [PMID: 29495953 DOI: 10.1080/1028415x.2018.1443995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The alpha-glucosidase inhibitor voglibose (VO) was recently reported to have a protective effect against weight gain as well as affect various metabolic changes related to food intake and gut-brain signaling. We hypothesized that VO prevents weight gain by altering neurometabolome profiles in the hypothalamus to reduce food intake. To test this hypothesis, we assessed metabolite profiles in the hypothalamus of standard diet- or high-fat (HF) diet-fed mice in the absence or presence of VO. In total, 29 male C57BL/6 mice were divided into 3 groups: (1) lean control, (2) HF, and (3) HF + VO. Vehicle or VO was administered for 12 weeks. The results showed that there were alterations in levels of metabolites across several metabolic pathways in the hypothalamus. VO treatment increased levels of many amino acids including arginine, glutamine, histidine, isoleucine, leucine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine in the hypothalamus. In addition, levels of 2-hydroxy-2-methyl-butyric acid in the hypothalamus were significantly increased after VO administration in HF diet-fed mice. Among lipid metabolites, levels of fatty acids were higher in the hypothalamus of VO-treated mice than in that of HF diet-fed mice. In terms of the energy status, the ATP/ADP ratio was higher in the hypothalamus of VO-treated mice (P < 0.001), thereby indicating an energy surplus. In conclusion, VO supplementation altered metabolite profiles in the hypothalamus to enhance catabolism, which is possibly responsible for the hypophagic effect of VO in HF diet-fed mice.
Collapse
Affiliation(s)
- Soo Jin Yang
- Department of Food and Nutrition, Seoul Women's University , Seoul , Republic of Korea
| | - Hyun Ju Do
- Korean Medicine Application Center, Korea Institute of Oriental Medicine , Daegu , Republic of Korea
| | - Youngae Jung
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute , Seoul , Republic of Korea
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute , Seoul , Republic of Korea
| | - Min-Jeong Shin
- Department of Public Health Sciences, BK21PLUS Program in Embodiment: Health-Society Interaction, Graduate School, Korea University , Seoul , Republic of Korea
| |
Collapse
|
28
|
Effects of pregnancy and short-lasting acute feed restriction on total ghrelin concentration and metabolic parameters in dairy cattle. Theriogenology 2018; 106:141-148. [DOI: 10.1016/j.theriogenology.2017.10.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 10/02/2017] [Accepted: 10/07/2017] [Indexed: 02/01/2023]
|
29
|
Functional Interrogation of the AgRP Neural Circuits in Control of Appetite, Body Weight, and Behaviors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1090:1-16. [PMID: 30390282 DOI: 10.1007/978-981-13-1286-1_1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurons expressing agouti-related protein (AgRP), the so-called hunger neurons, protect mammals from starvation by promoting food-seeking behaviors (Trends Neurosci 36:504-512, 2013). Now an increasing amount of evidence show that these hunger-sensing neurons not only motivate animals to forage and ingest food but also help conserve energy by inhibiting innate processes that demand large amounts of energy such as growth, reproduction, and stress response. It has further been perceived that AgRP neurons transmit signals with negative valence to reward and cognitive centers so as to engage the motivational behavior toward seeking and obtaining foods (Physiol Behav 190:34-42, 2017). Recent advancement in genome editing and neurotechniques unleashed an escalated research of uniquely defined neuronal populations and neural circuits underlying the behavioral regulation of body weight and food responses (Nat Biotechnol 32:347-355, 2014; Proc Natl Acad Sci 113, 2016). In this chapter we will review literatures describing the functional organization of the AgRP circuit and its correlative signaling components that influence ingestive, foraging, motivational, and cognitive responses, a framework that reshaped our thinking toward the new hope and challenges in treatment of obesity and eating disorders.
Collapse
|
30
|
Gangadharan A, Choi SE, Hassan A, Ayoub NM, Durante G, Balwani S, Kim YH, Pecora A, Goy A, Suh KS. Protein calorie malnutrition, nutritional intervention and personalized cancer care. Oncotarget 2017; 8:24009-24030. [PMID: 28177923 PMCID: PMC5410360 DOI: 10.18632/oncotarget.15103] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 01/23/2017] [Indexed: 12/27/2022] Open
Abstract
Cancer patients often experience weight loss caused by protein calorie malnutrition (PCM) during the course of the disease or treatment. PCM is expressed as severe if the patient has two or more of the following characteristics: obvious significant muscle wasting, loss of subcutaneous fat; nutritional intake of <50% of recommended intake for 2 weeks or more; bedridden or otherwise significantly reduced functional capacity; weight loss of >2% in 1 week, 5% in 1 month, or 7.5% in 3 months. Cancer anorexia-cachexia syndrome (CACS) is a multifactorial condition of advanced PCM associated with underlying illness (in this case cancer) and is characterized by loss of muscle with or without loss of fat mass. Cachexia is defined as weight loss of more than 5% of body weight in 12 months or less in the presence of chronic disease. Hence with a chronic illness on board even a small amount of weight loss can open the door to cachexia. These nutritional challenges can lead to severe morbidity and mortality in cancer patients. In the clinic, the application of personalized medicine and the ability to withstand the toxic effects of anti-cancer therapies can be optimized when the patient is in nutritional homeostasis and is free of anorexia and cachexia. Routine assessment of nutritional status and appropriate intervention are essential components of the effort to alleviate effects of malnutrition on quality of life and survival of patients.
Collapse
Affiliation(s)
- Anju Gangadharan
- The Genomics and Biomarkers Program, JT Cancer Center, Hackensack University Medical Center, Hackensack Meridian Health, Hackensack, NJ, USA
| | - Sung Eun Choi
- Department of Family, Nutrition, and Exercise Sciences, Queens College, The City University of New York, Flushing, NY, USA
| | - Ahmed Hassan
- The Genomics and Biomarkers Program, JT Cancer Center, Hackensack University Medical Center, Hackensack Meridian Health, Hackensack, NJ, USA
| | - Nehad M Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Gina Durante
- Department of Clinical Nutrition, Baystate Medical Center, Springfield, MA, USA
| | - Sakshi Balwani
- The Genomics and Biomarkers Program, JT Cancer Center, Hackensack University Medical Center, Hackensack Meridian Health, Hackensack, NJ, USA
| | - Young Hee Kim
- Department of Clinical Nutrition, Baystate Medical Center, Springfield, MA, USA
| | - Andrew Pecora
- Clinical Divisions, JT Cancer Center, Hackensack University Medical Center, Hackensack Meridian Health, Hackensack, NJ, USA
| | - Andre Goy
- Clinical Divisions, JT Cancer Center, Hackensack University Medical Center, Hackensack Meridian Health, Hackensack, NJ, USA
| | - K Stephen Suh
- The Genomics and Biomarkers Program, JT Cancer Center, Hackensack University Medical Center, Hackensack Meridian Health, Hackensack, NJ, USA
| |
Collapse
|
31
|
Hypothalamic and inflammatory basis of hypertension. Clin Sci (Lond) 2017; 131:211-223. [PMID: 28057892 DOI: 10.1042/cs20160001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 11/07/2016] [Accepted: 11/21/2016] [Indexed: 02/07/2023]
Abstract
Hypertension is a major health problem with great consequences for public health. Despite its role as the primary cause of significant morbidity and mortality associated with cardiovascular disease, the pathogenesis of essential hypertension remains largely unknown. The central nervous system (CNS) in general, and the hypothalamus in particular, are intricately involved in the development and maintenance of hypertension. Over the last several decades, the understanding of the brain's role in the development of hypertension has dramatically increased. This brief review is to summarize the neural mechanisms of hypertension with a focus on neuroendocrine and neurotransmitter involvement, highlighting recent findings that suggest that hypothalamic inflammation disrupts key signalling pathways to affect the central control of blood pressure, and therefore suggesting future development of interventional strategies that exploit recent findings pertaining to the hypothalamic control of blood pressure as well as the inflammatory-sympathetic mechanisms involved in hypertension.
Collapse
|
32
|
Miller GD. Appetite Regulation: Hormones, Peptides, and Neurotransmitters and Their Role in Obesity. Am J Lifestyle Med 2017; 13:586-601. [PMID: 31662725 DOI: 10.1177/1559827617716376] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 05/18/2017] [Accepted: 05/31/2017] [Indexed: 12/29/2022] Open
Abstract
Understanding body weight regulation will aid in the development of new strategies to combat obesity. This review examines energy homeostasis and food intake behaviors, specifically with regards to hormones, peptides, and neurotransmitters in the periphery and central nervous system, and their potential role in obesity. Dysfunction in feeding signals by the brain is a factor in obesity. The hypothalamic (arcuate nucleus) and brainstem (nucleus tractus solitaris) areas integrate behavioral, endocrine, and autonomic responses via afferent and efferent pathways from and to the brainstem and peripheral organs. Neurons present in the arcuate nucleus express pro-opiomelanocortin, Neuropeptide Y, and Agouti Related Peptide, with the former involved in lowering food intake, and the latter two acutely increasing feeding behaviors. Action of peripheral hormones from the gut, pancreas, adipose, and liver are also involved in energy homeostasis. Vagal afferent neurons are also important in regulating energy homeostasis. Peripheral signals respond to the level of stored and currently available fuel. By studying their actions, new agents maybe developed that disable orexigenic responses and enhance anorexigenic signals. Although there are relatively few medications currently available for obesity treatment, a number of agents are in development that work through these pathways.
Collapse
Affiliation(s)
- Gary D Miller
- Department of Health and Exercise Science, Wake Forest University, Winston-Salem, North Carolina
| |
Collapse
|
33
|
Mason BL. Feeding Systems and the Gut Microbiome: Gut-Brain Interactions With Relevance to Psychiatric Conditions. PSYCHOSOMATICS 2017; 58:574-580. [PMID: 28716445 DOI: 10.1016/j.psym.2017.06.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/05/2017] [Accepted: 06/06/2017] [Indexed: 12/26/2022]
Abstract
BACKGROUND Physical and mental health is dependent on the environment, and feeding is a prime example of this environmental exchange. While the hypothalamus controls both feeding behavior and the stress response, the integration of the neural control centers and the peripheral gut allows for disruption in the gastrointestinal systems and dysfunctional communication to the brain. OBJECTIVE The purpose of this review is to familiarize clinicians with the physiology controlling feeding behavior and its implications for psychiatric conditions, such as anorexia nervosa and depression. Growing understanding of how integrated bacterial life is in the body has shown that gut bacteria regulate basic physiologic processes and are implicated in various disease states and contribute to regulation of mood. Responses to stress have effects on feeding behavior and mood and the regulation of the stress response by the gut microbiota could contribute to the dysfunction seen in patients with psychiatric illnesses. CONCLUSIONS Gut microbiota may contribute to dysfunction in psychiatric illnesses. New opportunities to modulate existing gut microbiota using probiotics could be novel targets for clinical interventions.
Collapse
Affiliation(s)
- Brittany L Mason
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX.
| |
Collapse
|
34
|
Afshar SF, Zawaski JA, Inoue T, Rendon DA, Zieske AW, Punia JN, Sabek OM, Gaber MW. Investigating the Abscopal Effects of Radioablation on Shielded Bone Marrow in Rodent Models Using Multimodality Imaging. Radiat Res 2017; 188:56-65. [PMID: 28475423 DOI: 10.1667/rr14692.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The abscopal effect is the response to radiation at sites that are distant from the irradiated site of an organism, and it is thought to play a role in bone marrow (BM) recovery by initiating responses in the unirradiated bone marrow. Understanding the mechanism of this effect has applications in treating BM failure (BMF) and BM transplantation (BMT), and improving survival of nuclear disaster victims. Here, we investigated the use of multimodality imaging as a translational tool to longitudinally assess bone marrow recovery. We used positron emission tomography/computed tomography (PET/CT), magnetic resonance imaging (MRI) and optical imaging to quantify bone marrow activity, vascular response and marrow repopulation in fully and partially irradiated rodent models. We further measured the effects of radiation on serum cytokine levels, hematopoietic cell counts and histology. PET/CT imaging revealed a radiation-induced increase in proliferation in the shielded bone marrow (SBM) compared to exposed bone marrow (EBM) and sham controls. T2-weighted MRI showed radiation-induced hemorrhaging in the EBM and unirradiated SBM. In the EBM and SBM groups, we found alterations in serum cytokine and hormone levels and in hematopoietic cell population proportions, and histological evidence of osteoblast activation at the bone marrow interface. Importantly, we generated a BMT mouse model using fluorescent-labeled bone marrow donor cells and performed fluorescent imaging to reveal the migration of bone marrow cells from shielded to radioablated sites. Our study validates the use of multimodality imaging to monitor bone marrow recovery and provides evidence for the abscopal response in promoting bone marrow recovery after irradiation.
Collapse
Affiliation(s)
- Solmaz F Afshar
- a Department of Surgery, Houston Methodist Hospital Research Institute, Houston, Texas
| | - Janice A Zawaski
- b Hematology-Oncology Section, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Taeko Inoue
- b Hematology-Oncology Section, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - David A Rendon
- b Hematology-Oncology Section, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Arthur W Zieske
- d Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas
| | - Jyotinder N Punia
- c Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - Omaima M Sabek
- a Department of Surgery, Houston Methodist Hospital Research Institute, Houston, Texas
| | - M Waleed Gaber
- b Hematology-Oncology Section, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
35
|
Alé A, Zhang Y, Han C, Cai D. Obesity-associated extracellular mtDNA activates central TGFβ pathway to cause blood pressure increase. Am J Physiol Endocrinol Metab 2017; 312:E161-E174. [PMID: 27894066 PMCID: PMC5374298 DOI: 10.1152/ajpendo.00337.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 10/25/2016] [Accepted: 11/09/2016] [Indexed: 02/07/2023]
Abstract
Hypothalamic inflammation was recently found to mediate obesity-related hypertension, but the responsible upstream mediators remain unexplored. In this study, we show that dietary obesity is associated with extracellular release of mitochondrial DNA (mtDNA) into the cerebrospinal fluid and that central delivery of mtDNA mimics transforming growth factor-β (TGFβ) excess to activate downstream signaling pathways. Physiological study reveals that central administration of mtDNA or TGFβ is sufficient to cause hypertension in mice. Knockout of the TGFβ receptor in proopiomelanocortin neurons counteracts the hypertensive effect of not only TGFβ but also mtDNA excess, while the hypertensive action of central mtDNA can be blocked pharmacologically by a TGFβ receptor antagonist or genetically by TGFβ receptor knockout. Finally, we confirm that obesity-induced hypertension can be reversed through central treatment with TGFβ receptor antagonist. In conclusion, circulating mtDNA in the brain employs neural TGFβ pathway to mediate a central inflammatory mechanism of obesity-related hypertension.
Collapse
MESH Headings
- Animals
- Benzamides/pharmacology
- Blood Pressure/immunology
- Blotting, Western
- DNA, Mitochondrial/cerebrospinal fluid
- DNA, Mitochondrial/immunology
- DNA, Mitochondrial/metabolism
- DNA, Mitochondrial/pharmacology
- Diet, High-Fat
- Dioxoles/pharmacology
- Hypertension/etiology
- Hypertension/immunology
- Hypothalamus/immunology
- Hypothalamus/metabolism
- Male
- Mice
- Mice, Knockout
- Neurons/immunology
- Neurons/metabolism
- Obesity/complications
- Obesity/immunology
- Pro-Opiomelanocortin/metabolism
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/immunology
- Receptor, Transforming Growth Factor-beta Type II
- Receptors, Transforming Growth Factor beta/antagonists & inhibitors
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/immunology
- Third Ventricle
- Transforming Growth Factor beta/immunology
- Transforming Growth Factor beta1/pharmacology
Collapse
Affiliation(s)
- Albert Alé
- Department of Molecular Pharmacology, Diabetes Research Center, and Institute for Aging Research, Albert Einstein College of Medicine, New York, New York
| | - Yalin Zhang
- Department of Molecular Pharmacology, Diabetes Research Center, and Institute for Aging Research, Albert Einstein College of Medicine, New York, New York
| | - Cheng Han
- Department of Molecular Pharmacology, Diabetes Research Center, and Institute for Aging Research, Albert Einstein College of Medicine, New York, New York
| | - Dongsheng Cai
- Department of Molecular Pharmacology, Diabetes Research Center, and Institute for Aging Research, Albert Einstein College of Medicine, New York, New York
| |
Collapse
|
36
|
Prokosch ML, Hill SE. A Method for Manipulating Blood Glucose and Measuring Resulting Changes in Cognitive Accessibility of Target Stimuli. J Vis Exp 2016. [PMID: 27585282 DOI: 10.3791/54211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Much research in social psychology has investigated the impact of bodily energy need on cognition and decision-making. As such, blood glucose, the body's primary energy source, has been of special interest to researchers for years. Fluctuations in blood glucose have been linked to a variety of changes in cognitive and behavioral processes, such as self-control, political attitudes, and eating behavior. To help meet growing interest in the links between bodily energy need and these processes, this manuscript offers a simple methodology to experimentally manipulate blood glucose using a fasting procedure followed by administration of a sugar-sweetened, unsweetened, or artificially-sweetened beverage. This is followed by presentation of a method for measuring resulting changes in implicit cognition using a lexical decision-task. In this task, participants are asked to identify whether strings of letters are words or non-words and response latencies are recorded. Sample results from a recent publication are presented as an example of the applications for the experimental manipulation of blood glucose and the lexical decision task measures.
Collapse
Affiliation(s)
| | - Sarah E Hill
- Department of Psychology, Texas Christian University;
| |
Collapse
|
37
|
Castellano JM, Tena-Sempere M. Metabolic control of female puberty: potential therapeutic targets. Expert Opin Ther Targets 2016; 20:1181-93. [PMID: 27409160 DOI: 10.1080/14728222.2016.1212015] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION The onset of puberty in females is highly sensitive to the nutritional status and the amount of energy reserves of the organism. This metabolic information is sensed and transmitted to hypothalamic GnRH neurons, considered to be ultimately responsible for triggering puberty through the coordinated action of different peripheral hormones, central neurotransmitters, and molecular mediators. AREAS COVERED This article will review and discuss (i) the relevant actions of the adipose hormone leptin, as a stimulatory/permissive signal, and the gut hormone ghrelin, as an inhibitory factor, in the metabolic control of female puberty; (ii) the crucial role of the hypothalamic kisspeptin neurons, recently emerged as essential gatekeepers of puberty, in transmitting this metabolic information to GnRH neurons; and (iii) the potential involvement of key cellular energy sensors, such as mTOR, as molecular mediators in this setting. EXPERT OPINION The thorough characterization of the physiological roles of the above elements in the metabolic control of female puberty, along with the discovery of novel factors, pathways, and mechanisms involved, will promote our understanding of the complex networks connecting metabolism and puberty and, ultimately, will aid in the design of target-specific treatments for female pubertal disorders linked to conditions of metabolic stress.
Collapse
Affiliation(s)
- Juan M Castellano
- a Department of Cell Biology, Physiology and Immunology , University of Córdoba , Córdoba , Spain.,b CIBER Fisiopatología de la Obesidad y Nutrición , Instituto de Salud Carlos III , Córdoba , Spain.,c Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC)/Hospital Universitario Reina Sofia , Córdoba , Spain
| | - Manuel Tena-Sempere
- a Department of Cell Biology, Physiology and Immunology , University of Córdoba , Córdoba , Spain.,b CIBER Fisiopatología de la Obesidad y Nutrición , Instituto de Salud Carlos III , Córdoba , Spain.,c Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC)/Hospital Universitario Reina Sofia , Córdoba , Spain.,d FiDiPro Program, Department of Physiology , University of Turku , Turku , Finland
| |
Collapse
|
38
|
Clarke IJ, Arbabi L. New concepts of the central control of reproduction, integrating influence of stress, metabolic state, and season. Domest Anim Endocrinol 2016; 56 Suppl:S165-79. [PMID: 27345314 DOI: 10.1016/j.domaniend.2016.03.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 03/02/2016] [Accepted: 03/02/2016] [Indexed: 10/21/2022]
Abstract
Gonadotropin releasing hormone is the primary driver of reproductive function and pulsatile GnRH secretion from the brain causes the synthesis and secretion of LH and FSH from the pituitary gland. Recent work has revealed that the secretion of GnRH is controlled at the level of the GnRH secretory terminals in the median eminence. At this level, projections of kisspeptin cells from the arcuate nucleus of the hypothalamus are seen to be closely associated with fibers and terminals of GnRH cells. Direct application of kisspeptin into the median eminence causes release of GnRH. The kisspeptin cells are activated at the time of a natural "pulse" secretion of GnRH, as reflected in the secretion of LH. This appears to be due to input to the kisspeptin cells from glutamatergic cells in the basal hypothalamus, indicating that more than 1 neural element is involved in the secretion of GnRH. Because the GnRH secretory terminals are outside the blood-brain barrier, factors such as kisspeptin may be administered systemically to cause GnRH secretion; this offers opportunities for manipulation of the reproductive axis using factors that do not cross the blood-brain barrier. In particular, kisspeptin or analogs of the same may be used to activate reproduction in the nonbreeding season of domestic animals. Another brain peptide that influences reproductive function is gonadotropin inhibitory hormone (GnIH). Work in sheep shows that this peptide acts on GnRH neuronal perikarya, but projections to the median eminence also allow secretion into the hypophysial portal blood and action of GnIH on pituitary gonadotropes. GnIH cells are upregulated in anestrus, and infusion of GnIH can block the ovulatory surge in GnRH and/or LH secretion. Metabolic status may also affect the secretion of reproduction, and this could involve action of gut peptides and leptin. Neuropeptide Y and Y-receptor ligands have a negative impact on reproduction, and Neuropeptide Y production is markedly increased in negative energy balance; this may be the cause of lowered GnRH and gonadotropin secretion in this state. There is a complex interaction between appetite-regulating peptide neurons and kisspeptin neurons that enables the former to regulate the latter both positively and negatively. In terms of how GnRH secretion is reduced during stress, recent data indicate that GnIH cells are integrally involved, with increased input to the GnRH cells. The secretion of GnIH into the portal blood is not increased during stress, so the negative effect is most likely effected at the level of GnRH neuronal cell bodies.
Collapse
Affiliation(s)
- I J Clarke
- Department of Physiology, Monash University, Clayton, VIC 3800, Australia.
| | - L Arbabi
- Department of Physiology, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
39
|
Wang SP, Gao YL, Liu G, Deng D, Chen RJ, Zhang YZ, Li LL, Wen QQ, Hou YQ, Feng ZM, Guo ZH. Molecular cloning, characterization and expression of the energy homeostasis-associated gene in piglet. J Zhejiang Univ Sci B 2016; 16:524-32. [PMID: 26055914 DOI: 10.1631/jzus.b1400260] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The energy homeostasis-associated (Enho) gene encodes a secreted protein, adropin, which regulates the expression of hepatic lipogenic genes and adipose tissue peroxisome proliferator-activated receptor γ, a major regulator of lipogenesis. In the present study, the porcine (Sus scrofa) homologue of the Enho gene, which was named pEnho, was amplified by reverse transcriptase polymerase chain reaction (RT-PCR) using oligonucleotide primers derived from in silico sequences. The gene sequence was submitted into the GenBank of NCBI, and the access number is GQ414763. The pEnho encodes a protein of 76 amino acids which shows 75% similarity to Homo sapiens adropin. The expression profile of pEnho in tissues (liver, muscle, anterior jejunum, posterior jejunum, and ileum) was determined by quantitative real-time RT-PCR. pEnho was localized on porcine chromosome 10 and no introns were found. In conclusion, pEnho was cloned and analysed with the aim of increasing knowledge about glucose and lipid metabolism in piglets and helping to promote the health and growth of piglets through adropin regulation.
Collapse
Affiliation(s)
- Sheng-ping Wang
- Research Center of Healthy Breeding Livestock & Poultry, Hunan Engineering & Research Center of Animal & Poultry Science, Key Lab Agro-ecology Processing Subtropical Region, Scientific Observational and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; Hunan Institute of Microbiology, Changsha 410009, China; Rice Research Institute of Sichuan Agricultural University, Chengdu 625014, China; Fujian Aonong Biotechnology Corporation, Xiamen 361007, China; Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan Polytechnic University, Wuhan 430023, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abreu AP, Kaiser UB. Pubertal development and regulation. Lancet Diabetes Endocrinol 2016; 4:254-264. [PMID: 26852256 PMCID: PMC5192018 DOI: 10.1016/s2213-8587(15)00418-0] [Citation(s) in RCA: 318] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 10/22/2015] [Accepted: 10/22/2015] [Indexed: 12/12/2022]
Abstract
Puberty marks the end of childhood and is a period when individuals undergo physiological and psychological changes to achieve sexual maturation and fertility. The hypothalamic-pituitary-gonadal axis controls puberty and reproduction and is tightly regulated by a complex network of excitatory and inhibitory factors. This axis is active in the embryonic and early postnatal stages of life and is subsequently restrained during childhood, and its reactivation culminates in puberty initiation. The mechanisms underlying this reactivation are not completely known. The age of puberty onset varies between individuals and the timing of puberty initiation is associated with several health outcomes in adult life. In this Series paper, we discuss pubertal markers, epidemiological trends of puberty initiation over time, and the mechanisms whereby genetic, metabolic, and other factors control secretion of gonadotropin-releasing hormone to determine initiation of puberty.
Collapse
Affiliation(s)
- Ana Paula Abreu
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ursula B Kaiser
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
41
|
Chavan R, Feillet C, Costa SSF, Delorme JE, Okabe T, Ripperger JA, Albrecht U. Liver-derived ketone bodies are necessary for food anticipation. Nat Commun 2016; 7:10580. [PMID: 26838474 PMCID: PMC4742855 DOI: 10.1038/ncomms10580] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Accepted: 12/30/2015] [Indexed: 12/16/2022] Open
Abstract
The circadian system has endowed animals with the ability to anticipate recurring food availability at particular times of day. As daily food anticipation (FA) is independent of the suprachiasmatic nuclei, the central pacemaker of the circadian system, questions arise of where FA signals originate and what role components of the circadian clock might play. Here we show that liver-specific deletion of Per2 in mice abolishes FA, an effect that is rescued by viral overexpression of Per2 in the liver. RNA sequencing indicates that Per2 regulates β-hydroxybutyrate (βOHB) production to induce FA leading to the conclusion that liver Per2 is important for this process. Unexpectedly, we show that FA originates in the liver and not in the brain. However, manifestation of FA involves processing of the liver-derived βOHB signal in the brain, indicating that the food-entrainable oscillator is not located in a single tissue but is of systemic nature.
Collapse
Affiliation(s)
- Rohit Chavan
- Department of Biology, Unit of Biochemistry, University of Fribourg, Fribourg 1700, Switzerland
| | - Céline Feillet
- Department of Biology, Unit of Biochemistry, University of Fribourg, Fribourg 1700, Switzerland
| | - Sara S Fonseca Costa
- Department of Biology, Unit of Biochemistry, University of Fribourg, Fribourg 1700, Switzerland
| | - James E Delorme
- Department of Biology, Unit of Biochemistry, University of Fribourg, Fribourg 1700, Switzerland
| | - Takashi Okabe
- Department of Biology, Unit of Biochemistry, University of Fribourg, Fribourg 1700, Switzerland
| | - Jürgen A Ripperger
- Department of Biology, Unit of Biochemistry, University of Fribourg, Fribourg 1700, Switzerland
| | - Urs Albrecht
- Department of Biology, Unit of Biochemistry, University of Fribourg, Fribourg 1700, Switzerland
| |
Collapse
|
42
|
Ghrelin Actions on Somatotropic and Gonadotropic Function in Humans. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 138:3-25. [PMID: 26940384 DOI: 10.1016/bs.pmbts.2015.11.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ghrelin, a 28 amino-acid octanoylated peptide predominantly produced by the stomach, was discovered to be the natural ligand of the type 1a GH secretagogue receptor (GHS-R1a). It was thus considered as a natural GHS additional to GHRH, although later on ghrelin has mostly been considered a major orexigenic factor. The GH-releasing action of ghrelin takes place both directly on pituitary cells and through modulation of GHRH from the hypothalamus; some functional antisomatostatin action has also been shown. However, ghrelin is much more than a natural GH secretagogue. In fact, it also modulates lactotroph and corticotroph secretion in humans as well as in animals and plays a relevant role in the modulation of the hypothalamic-pituitary-gonadal function. Several studies have indicated that ghrelin plays an inhibitory effect on gonadotropin pulsatility, is involved in the regulation of puberty onset in animals, and may regulate spermatogenesis, follicular development and ovarian cell functions in humans. In this chapter ghrelin actions on the GH/IGF-I and the gonadal axes will be revised. The potential therapeutic role of ghrelin as a treatment of catabolic conditions will also be discussed.
Collapse
|
43
|
Abstract
There exists a continuous spectrum of overeating, where at the extremes there are casual overindulgences and at the other a 'pathological' drive to consume palatable foods. It has been proposed that pathological eating behaviors may be the result of addictive appetitive behavior and loss of ability to regulate the consumption of highly processed foods containing refined carbohydrates, fats, salt, and caffeine. In this review, we highlight the genetic similarities underlying substance addiction phenotypes and overeating compulsions seen in individuals with binge eating disorder. We relate these similarities to findings from neuroimaging studies on reward processing and clinical diagnostic criteria based on addiction phenotypes. The abundance of similarities between compulsive overeating and substance addictions puts forth a case for a 'food addiction' phenotype as a valid, diagnosable disorder.
Collapse
|
44
|
Sofer S, Eliraz A, Madar Z, Froy O. Concentrating carbohydrates before sleep improves feeding regulation and metabolic and inflammatory parameters in mice. Mol Cell Endocrinol 2015. [PMID: 26206716 DOI: 10.1016/j.mce.2015.06.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
New evidance highlights the importance of food timing. Recently, we showed that a low-calorie diet with carbohydrates eaten mostly at dinner changed diurnal hormone secretion and led to greater weight loss and improved metabolic status in obese people. Herein, we set out to test whether concentrated-carbohydrates diet (CCD), in which carbohydrates are fed only before sleep, leads to an improved metabolic status in mouse hypothalamus and peripheral tissues. Diet-induced obese mice were given concentrated or distributed carbohydrate diet for 6 weeks. Obese mice fed CCD ate 8.3% less, were 9.3% leaner and had 39.7% less fat mass. Leptin, ghrelin and adiponectin displayed altered secretion. In addition, these mice exhibited an improved biochemical and inflammatory status. In the hypothalamus, anorexigenic signals were up-regulated and orexigenic signals were down-regulated. In peripheral tissues, CCD promoted adiponectin signaling, repressed gluconeogenesis, enhanced lipid oxidation and lowered inflammation, thus ameliorating the major risk factors of obesity.
Collapse
Affiliation(s)
- Sigal Sofer
- Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel; Meuhedet Medical Services, Diet and Nutrition Department, Tel Aviv, Israel
| | - Abraham Eliraz
- Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Zecharia Madar
- Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel.
| | - Oren Froy
- Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel.
| |
Collapse
|
45
|
Celik O, Aydin S, Celik N, Yilmaz M. Peptides: Basic determinants of reproductive functions. Peptides 2015; 72:34-43. [PMID: 26074346 DOI: 10.1016/j.peptides.2015.05.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/28/2015] [Accepted: 05/29/2015] [Indexed: 12/22/2022]
Abstract
Mammalian reproduction is a costly process in terms of energy consumption. The critical information regarding metabolic status is signaled to the hypothalamus mainly through peripheral peptides from the adipose tissue and gastrointestinal tract. Changes in energy stores produce fluctuations in leptin, insulin, ghrelin and glucose signals that feedback mainly to the hypothalamus to regulate metabolism and fertility. In near future, possible effects of the nutritional status on GnRH regulation can be evaluated by measuring serum or tissue levels of leptin and ghrelin in patiens suffering from infertility. The fact that leptin and ghrelin are antagonistic in their effects on GnRH neurons, their respective agonistic and antagonistic roles make them ideal candidates to use instead of GnRH agonist and antagonist. Similarly, kisspeptin expressing neurons are likely to mediate the well-established link between energy balance and reproductive functions. Exogenous kisspeptin can be used for physiological ovarian hyperstimulation for in-vitro fertilization. Moreover, kisspeptin antagonist therapy can be used for the treatment of postmenapousal women, precocious puberty, PCOS, endometriosis and uterine fibroids. In this review, we will analyze the central mechanisms involved in the integration of metabolic information and their contribution to the control of the reproductive function. Particular attention will be paid to summarize the participation of leptin, kisspeptin, ghrelin, NPY, orexin, urocortin, VIP, insulin, galanin, galanin like peptide, oxytocin, agouti gene-related peptide, and POMC neurons in this process and their possible interactions to contribute to the metabolic control of reproduction.
Collapse
Affiliation(s)
- Onder Celik
- Private Clinic, Department of Obstetrics and Gynecology, İzmir, Turkey
| | - Suleyman Aydin
- Firat University, School of Medicine, Department of Medical Biochemistry (Firat Hormones Research Group), 23119 Elazig, Turkey.
| | - Nilufer Celik
- Behcet Uz Children's Hospital, Department of Biochemistry, İzmir, Turkey
| | - Musa Yilmaz
- Firat University, School of Medicine, Department of Medical Biochemistry (Firat Hormones Research Group), 23119 Elazig, Turkey
| |
Collapse
|
46
|
Abstract
The endocrine hypothalamus constitutes those cells which project to the median eminence and secrete neurohormones into the hypophysial portal blood to act on cells of the anterior pituitary gland. The entire endocrine system is controlled by these peptides. In turn, the hypothalamic neuroendocrine cells are regulated by feedback signals from the endocrine glands and other circulating factors. The neuroendocrine cells are found in specific regions of the hypothalamus and are regulated by afferents from higher brain centers. Integrated function is clearly complex and the networks between and amongst the neuroendocrine cells allows fine control to achieve homeostasis. The entry of hormones and other factors into the brain, either via the cerebrospinal fluid or through fenestrated capillaries (in the basal hypothalamus) is important because it influences the extent to which feedback regulation may be imposed. Recent evidence of the passage of factors from the pars tuberalis and the median eminence casts a new layer in our understanding of neuroendocrine regulation. The function of neuroendocrine cells and the means by which pulsatile secretion is achieved is best understood for the close relationship between gonadotropin releasing hormone and luteinizing hormone, which is reviewed in detail. The secretion of other neurohormones is less rigid, so the relationship between hypothalamic secretion and the relevant pituitary hormones is more complex.
Collapse
Affiliation(s)
- I J Clarke
- Monash University, Department of Physiology, Clayton, Australia
| |
Collapse
|
47
|
Mandelblat-Cerf Y, Ramesh RN, Burgess CR, Patella P, Yang Z, Lowell BB, Andermann ML. Arcuate hypothalamic AgRP and putative POMC neurons show opposite changes in spiking across multiple timescales. eLife 2015; 4. [PMID: 26159614 PMCID: PMC4498165 DOI: 10.7554/elife.07122] [Citation(s) in RCA: 195] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 06/19/2015] [Indexed: 12/18/2022] Open
Abstract
Agouti-related-peptide (AgRP) neurons—interoceptive neurons in the arcuate nucleus of the hypothalamus (ARC)—are both necessary and sufficient for driving feeding behavior. To better understand the functional roles of AgRP neurons, we performed optetrode electrophysiological recordings from AgRP neurons in awake, behaving AgRP-IRES-Cre mice. In free-feeding mice, we observed a fivefold increase in AgRP neuron firing with mounting caloric deficit in afternoon vs morning recordings. In food-restricted mice, as food became available, AgRP neuron firing dropped, yet remained elevated as compared to firing in sated mice. The rapid drop in spiking activity of AgRP neurons at meal onset may reflect a termination of the drive to find food, while residual, persistent spiking may reflect a sustained drive to consume food. Moreover, nearby neurons inhibited by AgRP neuron photostimulation, likely including satiety-promoting pro-opiomelanocortin (POMC) neurons, demonstrated opposite changes in spiking. Finally, firing of ARC neurons was also rapidly modulated within seconds of individual licks for liquid food. These findings suggest novel roles for antagonistic AgRP and POMC neurons in the regulation of feeding behaviors across multiple timescales. DOI:http://dx.doi.org/10.7554/eLife.07122.001 Appetite is controlled in part by the opposing actions of the ‘hunger hormone’ (called ghrelin) and the ‘satiety hormone’ (called leptin). Ghrelin is released by the stomach when empty and stimulates appetite, whereas leptin is released by fat stores and induces feelings of fullness. Both hormones travel via the bloodstream and are detected by a region of the brain called the hypothalamus. Ghrelin and leptin act specifically on a group of cells in the hypothalamus that contains at least two major cell types: AgRP neurons and POMC neurons. Electrode recordings from slices of mouse brain show that AgRP neurons fire more rapidly at night—when mice normally feed—than during the day, whereas POMC neurons do the opposite. This suggests that the activity of AgRP neurons drives food-seeking behavior, whereas POMC firing inhibits it. However, the absence of circulating hormones such as leptin and ghrelin in brain slices makes it difficult to draw firm conclusions about the role of these cells in controlling appetite. Mandelblat-Cerf, Ramesh, Burgess et al. have addressed this issue by performing the first recordings of spiking activity in individual AgRP neurons and other cells that are likely to be POMC neurons in awake mice. Consistent with the results of slice experiments, the firing rate of AgRP neurons increased steadily over the course of the day, suggesting that their activity signals an increasing need for food. Furthermore, as soon as food became available, the firing rate of the AgRP neurons suddenly dropped—even though the animals' energy reserves would still have been low. These results are consistent with the findings of two recent studies reported earlier this year that used different methods to indirectly measure neuronal activity in awake mice. Notably, even after the drop in activity, the firing rates of AgRP neurons remained above those recorded in fully sated mice—which possibly reflects the fact that the animals' energy reserves were still low. The putative POMC neurons generally showed opposite effects to the AgRP neurons. The results of these electrode recordings in awake mice thus suggest that AgRP and POMC neurons together maintain a drive to seek out food sources as energy reserves fall, and to refrain from doing so when energy reserves are plentiful. Moreover, the seemingly paradoxical drop in AgRP firing and increase in POMC firing upon receiving food may act as a signal to temporarily stop searching for food, so that feeding itself can begin. Alternatively, since the release of satiety hormones after eating a meal is slow, these rapid changes in firing may provide more immediate feedback to the neuronal circuits that regulate the drives to seek and consume food. DOI:http://dx.doi.org/10.7554/eLife.07122.002
Collapse
Affiliation(s)
- Yael Mandelblat-Cerf
- Department of Endocrinology, Beth Israel Deaconess Medical Center, Boston, United States
| | - Rohan N Ramesh
- Department of Endocrinology, Beth Israel Deaconess Medical Center, Boston, United States
| | - Christian R Burgess
- Department of Endocrinology, Beth Israel Deaconess Medical Center, Boston, United States
| | - Paola Patella
- Department of Endocrinology, Beth Israel Deaconess Medical Center, Boston, United States
| | - Zongfang Yang
- Department of Endocrinology, Beth Israel Deaconess Medical Center, Boston, United States
| | - Bradford B Lowell
- Department of Endocrinology, Beth Israel Deaconess Medical Center, Boston, United States
| | - Mark L Andermann
- Department of Endocrinology, Beth Israel Deaconess Medical Center, Boston, United States
| |
Collapse
|
48
|
Rojczyk E, Pałasz A, Wiaderkiewicz R. Effect of short and long-term treatment with antipsychotics on orexigenic/anorexigenic neuropeptides expression in the rat hypothalamus. Neuropeptides 2015; 51:31-42. [PMID: 25888224 DOI: 10.1016/j.npep.2015.04.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 03/27/2015] [Accepted: 04/01/2015] [Indexed: 12/31/2022]
Abstract
Among numerous side effects of antipsychotic drugs (neuroleptics), one of the leading problems is a significant weight gain caused by disturbances in energy homeostasis. The hypothalamus is considered an important target for neuroleptics and contains some neuronal circuits responsible for food intake regulation, so we decided to study which hypothalamic signaling pathways connected with energy balance control are modified by antipsychotic drugs of different generations. We created an expression profile of different neuropeptides after single-dose and chronic neuroleptic administration. Experiments were carried out on adult male Sprague-Dawley rats injected intraperitoneally for 1 day or for 28 days by three neuroleptics: olanzapine, chlorpromazine and haloperidol. Hypothalami were isolated in order to perform PCR reactions and also whole brains were sliced for immunohistochemical analysis. We assessed the expression of orexigenic/anorexigenic neuropeptides and their receptors--neuropeptide Y (NPY), NPY receptor type 1 (Y1R), preproorexin (PPOX), orexin A, orexin receptor type 1 (OX1R) and 2 (OX2R), nucleobindin 2 (NUCB2), nesfatin-1, proopiomelanocortin (POMC), alpha-melanotropin (α-MSH) and melanocortin receptor type 4 (MC4R)--both on the mRNA and protein levels. We have shown that antipsychotics of different generations administered chronically have the ability to upregulate PPOX, orexin A and Y1R expression with little or no effect on orexigenic receptors (OX1R, OX2R) and NPY. Interestingly, antipsychotics also increased the level of some anorexigenic factors (POMC, α-MSH and MC4R), but at the same time strongly downregulated NUCB2 and nesfatin-1 signaling--a newly discovered neuropeptide known as a food-intake inhibiting factor. Our results may contribute to a better understanding of mechanisms responsible for antipsychotics' side effects. They also underline the complex nature of interactions between classical monoamine receptors and hypothalamic peptidergic pathways, which has potential clinical applications.
Collapse
Affiliation(s)
- Ewa Rojczyk
- Department of Histology, Faculty of Medicine in Katowice, Medical University of Silesia, 18 Medyków Street, 40-752 Katowice, Poland.
| | - Artur Pałasz
- Department of Histology, Faculty of Medicine in Katowice, Medical University of Silesia, 18 Medyków Street, 40-752 Katowice, Poland
| | - Ryszard Wiaderkiewicz
- Department of Histology, Faculty of Medicine in Katowice, Medical University of Silesia, 18 Medyków Street, 40-752 Katowice, Poland
| |
Collapse
|
49
|
Folch J, Patraca I, Martínez N, Pedrós I, Petrov D, Ettcheto M, Abad S, Marin M, Beas-Zarate C, Camins A. The role of leptin in the sporadic form of Alzheimer's disease. Interactions with the adipokines amylin, ghrelin and the pituitary hormone prolactin. Life Sci 2015; 140:19-28. [PMID: 25998028 DOI: 10.1016/j.lfs.2015.05.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 05/05/2015] [Accepted: 05/11/2015] [Indexed: 12/11/2022]
Abstract
Leptin (Lep) is emerging as a pivotal molecule involved in both the early events and the terminal phases of Alzheimer's disease (AD). In the canonical pathway, Lep acts as an anorexigenic factor via its effects on hypothalamic nucleus. However, additional functions of Lep in the hippocampus and cortex have been unravelled in recent years. Early events in the sporadic form of AD likely involve cellular level alterations which can have an effect on food intake and metabolism. Thus, AD can be conceivably interpreted as a multiorgan pathology that not only results in a dramatic neuronal loss in brain areas such as the hippocampus and the cortex (ultimately leading to a significant cognitive impairment) but as a disease which also affects body-weight homeostasis. According to this view, body-weight control disruptions are to be expected in both the early- and late-stage AD, concomitant with changes in serum Lep content, alterations in Lep transport across the blood-brain barrier (BBB) and Lep receptor-related signalling abnormalities. Lep is a member of the adipokine family of molecules, while the Lep receptor belongs to the class I cytokine receptors. Since cellular response to adipokine signalling can be either potentiated or diminished as a result of specific ligand-receptor interactions, Lep interactions with other members of the adipokine family including amylin, ghrelin and hormones such as prolactin require further investigation. In this review, we provide a general perspective on the functions of Lep in the brain, with a particular focus on the sporadic AD.
Collapse
Affiliation(s)
- Jaume Folch
- Unitats de Bioquímica i Farmacologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, C./ St. Llorenç 21, 43201 Reus, Tarragona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos Tercero, Madrid, Spain
| | - Iván Patraca
- Unitats de Bioquímica i Farmacologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, C./ St. Llorenç 21, 43201 Reus, Tarragona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos Tercero, Madrid, Spain
| | - Nohora Martínez
- Unitats de Bioquímica i Farmacologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, C./ St. Llorenç 21, 43201 Reus, Tarragona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos Tercero, Madrid, Spain
| | - Ignacio Pedrós
- Unitats de Bioquímica i Farmacologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, C./ St. Llorenç 21, 43201 Reus, Tarragona, Spain; Unitat de Farmacologia i Farmacognòsia Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Dmitry Petrov
- Unitat de Farmacologia i Farmacognòsia Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos Tercero, Madrid, Spain
| | - Miren Ettcheto
- Unitat de Farmacologia i Farmacognòsia Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos Tercero, Madrid, Spain
| | - Sonia Abad
- Unitat de Farmacologia i Farmacognòsia Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos Tercero, Madrid, Spain
| | - Miguel Marin
- Centro de Biotecnología, Universidad Nacional de Loja, Av. Pío Jaramillo Alvarado y Reinaldo Espinosa, La Argelia, Loja, Ecuador
| | - Carlos Beas-Zarate
- Departamento de Biología Celular y Molecular, C.U.C.B.A., Universidad de Guadalajara and División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Mexico; Instituto Mexicano del Seguro Social (IMSS), Sierra Mojada 800, Col. Independencia, Guadalajara, Jalisco 44340, Mexico
| | - Antoni Camins
- Unitat de Farmacologia i Farmacognòsia Facultat de Farmàcia, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos Tercero, Madrid, Spain; Centro de Biotecnología, Universidad Nacional de Loja, Av. Pío Jaramillo Alvarado y Reinaldo Espinosa, La Argelia, Loja, Ecuador.
| |
Collapse
|
50
|
Nøhr MK, Egerod KL, Christiansen SH, Gille A, Offermanns S, Schwartz TW, Møller M. Expression of the short chain fatty acid receptor GPR41/FFAR3 in autonomic and somatic sensory ganglia. Neuroscience 2015; 290:126-37. [PMID: 25637492 DOI: 10.1016/j.neuroscience.2015.01.040] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 01/05/2015] [Accepted: 01/06/2015] [Indexed: 01/14/2023]
Abstract
G-protein-coupled receptor 41 (GPR41) also called free fatty acid receptor 3 (FFAR3) is a Gαi-coupled receptor activated by short-chain fatty acids (SCFAs) mainly produced from dietary complex carbohydrate fibers in the large intestine as products of fermentation by microbiota. FFAR3 is expressed in enteroendocrine cells, but has recently also been shown to be present in sympathetic neurons of the superior cervical ganglion. The aim of this study was to investigate whether the FFAR3 is present in other autonomic and sensory ganglia possibly influencing gut physiology. Cryostat sections were cut of autonomic and sensory ganglia of a transgenic reporter mouse expressing the monomeric red fluorescent protein (mRFP) gene under the control of the FFAR3 promoter. Control for specific expression was also done by immunohistochemistry with an antibody against the reporter protein. mRFP expression was as expected found not only in neurons of the superior cervical ganglion, but also in sympathetic ganglia of the thoracic and lumbar sympathetic trunk. Further, neurons in prevertebral ganglia expressed the mRFP reporter. FFAR3-mRFP-expressing neurons were also present in both autonomic and sensory ganglia such as the vagal ganglion, the spinal dorsal root ganglion and the trigeminal ganglion. No expression was observed in the brain or spinal cord. By use of radioactive-labeled antisense DNA probes, mRNA encoding the FFAR3 was found to be present in cells of the same ganglia. Further, the expression of the FFAR3 in the ganglia of the transgenic mice was confirmed by immunohistochemistry using an antibody directed against the receptor protein, and double labeling colocalized mRFP and the FFAR3-protein in the same neurons. Finally, quantitative real-time polymerase chain reaction (qRT-PCR) on extracts from the ganglia supported the presence mRNA encoding the FFAR3 in most of the investigated tissues. These data indicate that FFAR3 is expressed on postganglionic sympathetic and sensory neurons in both the autonomic and somatic peripheral nervous system and that SCFAs act not only through the enteroendocrine system but also directly by modifying physiological reflexes integrating the peripheral nervous system and the gastro-intestinal tract.
Collapse
Affiliation(s)
- M K Nøhr
- Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - K L Egerod
- Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - S H Christiansen
- Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - A Gille
- Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany; Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Maybachstrasse 14, 68169 Mannheim, Germany.
| | - S Offermanns
- Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany.
| | - T W Schwartz
- Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - M Møller
- Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|