1
|
Abbasi H, Jourabchi-Ghadim N, Asgarzade A, Mirshekari M, Ebrahimi-Mameghani M. Unveiling the veil of adipokines: A meta-analysis and systematic review in amyotrophic lateral sclerosis. Neuroscience 2024; 563:1-9. [PMID: 39505137 DOI: 10.1016/j.neuroscience.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 10/20/2024] [Accepted: 11/03/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND Adipokines are proposed to be associated with ALS progression through assorted pathways. Therefore, The present meta-analysis explored the link between various adipokines and ALS progression. METHOD International database like PubMed, Scopus, and Web of Science databases were searched to achieve eligible papers published before December 2023. The following PICO structure was utilized: Population (patients with ALS); Intervention (serum concentrations of ghrelin, leptin, and adiponectin), Comparison (with or without controls), and Outcome (ALS progression). the risk of bias of selected papers was assessed through the Newcastle-Ottawa Scale (NOS) tool. RESULTS 11 out of 240 papers were selected for this study which were published between 2010 and 2024. Lower serum leptin concentrations were detected in the ALS compared to control groups (WMD: -0.91, 95% CI:-1.77, -0.05). Serum concentrations of adiponectin were higher in ALS compared to control groups (WMD: 0.41, 95% CI:-0.7, 0.89). Ultimately, The serum concentrations of ghrelin in the ALS groups were lower than control groups (WMD: -1.21, 95% CI: -2.95, 0.53). CONCLUSION Our findings revealed that serum concentrations of ghrelin and leptin were higher in ALS patients compared to control, unlike adiponectin.
Collapse
Affiliation(s)
- Hamid Abbasi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Neda Jourabchi-Ghadim
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Nutrition Research Center, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ali Asgarzade
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Mobin Mirshekari
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mehrangiz Ebrahimi-Mameghani
- Nutrition Research Center, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
2
|
Qiu Y, Li G, Zheng L, Liu W, Li X, Wang X, Chen L. Relationship Between Cognitive Frailty and Mortality in Older Adults: A Systematic Review and Meta-Analysis. J Am Med Dir Assoc 2023; 24:1637-1644.e8. [PMID: 37660724 DOI: 10.1016/j.jamda.2023.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023]
Abstract
OBJECTIVE To synthesize the pooled mortality risk estimate and determine whether cognitive frailty is a predictor of mortality. DESIGN A systematic review and meta-analysis. SETTING AND PARTICIPANTS The participants were community-dwelling older adults aged ≥60 years. METHODS PubMed, Web of Science, Embase, CINAHL, and the Cochrane Library databases were systematically searched. Two researchers independently screened potentially eligible literature, evaluated the quality of the included studies, and then extracted the data. We used STATA, version 15.0 to perform the all data. RESULTS Nineteen studies were included. The association between cognitive frailty and a higher risk of death was statistically significant [hazard ratio (HR), 2.01; 95% CI, 1.84-2.19; P < .001]. The outcomes indicated that cognitive frailty was a critical risk factor for predicting mortality (OR, 4.82; 95% CI, 1.59-14.57; P < .01). Based on different models of cognitive frailty, the results of subgroup analyses revealed that the risk of mortality was the highest in the Frail + mild cognitive impairment group (HR, 2.35; 95% CI, 2.05-2.70; P < .001). The subgroup analyses by region demonstrated that mortality risk was lowest in the European group (HR, 1.63; 95% CI, 1.4-1.87; P < .001). CONCLUSIONS AND IMPLICATIONS This study quantitatively portrays the pooled mortality risk estimate of cognitive frailty. The results suggest that in older adults, cognitive frailty can be a predictor of mortality. The findings could alert health care providers to pay more attention to cognitive frailty.
Collapse
Affiliation(s)
- Yiming Qiu
- School of Nursing, Jilin University, Changchun, Jilin, China
| | - Guichen Li
- School of Nursing, Jilin University, Changchun, Jilin, China
| | - Lufang Zheng
- School of Nursing, Jilin University, Changchun, Jilin, China
| | - Wei Liu
- School of Nursing, Jilin University, Changchun, Jilin, China
| | - Xin Li
- School of Nursing, Jilin University, Changchun, Jilin, China
| | - Xinxin Wang
- The First Hospital of Jilin University, Changchun, Jilin, China.
| | - Li Chen
- School of Nursing, Jilin University, Changchun, Jilin, China; Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
3
|
Zajonz TS, Kunzemann C, Schreiner AL, Beckert F, Schneck E, Boening A, Markmann M, Sander M, Koch C. Potentials of Acetylcholinesterase and Butyrylcholinesterase Alterations in On-Pump Coronary Artery Bypass Surgery in Postoperative Delirium: An Observational Trial. J Clin Med 2023; 12:5245. [PMID: 37629287 PMCID: PMC10455192 DOI: 10.3390/jcm12165245] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/31/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Cardiac surgery is regularly associated with postoperative delirium (POD), affected by neuro-inflammation and changes in cholinergic activity. Therefore, this prospective observational study aimed to evaluate whether pre- and perioperative changes in blood acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) activity were associated with POD development in patients undergoing isolated elective coronary artery bypass graft (CABG) surgery. It included 93 patients. Pre- and postoperative blood AChE and BChE activities were measured with photometric rapid-point-of-care-testing. The Intensive Care Delirium Screening Checklist and the Confusion Assessment Method for the Intensive Care Unit were used to screen patients for POD. POD developed in 20 patients (21.5%), who were older (p = 0.003), had higher EuroSCOREs (p ≤ 0.001), and had longer intensive care unit stays (p < 0.001). On postoperative day one, BChE activity decreased from preoperative values more in patients with (31.9%) than without (23.7%) POD (group difference p = 0.002). Applying a cutoff of ≥32.0% for BChE activity changes, receiver operating characteristic analysis demonstrated a moderate prediction capability for POD (area under the curve = 0.72, p = 0.002). The risk of developing POD was 4.31 times higher with a BChE activity change of ≥32.0% (p = 0.010). Monitoring the pre- to postoperative reduction in BChE activity might be a clinically practicable biomarker for detecting patients at risk of developing POD after CABG surgery.
Collapse
Affiliation(s)
- Thomas S. Zajonz
- Department of Anesthesiology, Operative Intensive Care Medicine and Pain Therapy, University Hospital of Giessen and Marburg, Justus Liebig University of Giessen, 35392 Giessen, Germany; (C.K.); (A.L.S.); (F.B.); (E.S.); (M.M.); (M.S.); (C.K.)
| | - Christian Kunzemann
- Department of Anesthesiology, Operative Intensive Care Medicine and Pain Therapy, University Hospital of Giessen and Marburg, Justus Liebig University of Giessen, 35392 Giessen, Germany; (C.K.); (A.L.S.); (F.B.); (E.S.); (M.M.); (M.S.); (C.K.)
| | - Anna Lena Schreiner
- Department of Anesthesiology, Operative Intensive Care Medicine and Pain Therapy, University Hospital of Giessen and Marburg, Justus Liebig University of Giessen, 35392 Giessen, Germany; (C.K.); (A.L.S.); (F.B.); (E.S.); (M.M.); (M.S.); (C.K.)
| | - Frauke Beckert
- Department of Anesthesiology, Operative Intensive Care Medicine and Pain Therapy, University Hospital of Giessen and Marburg, Justus Liebig University of Giessen, 35392 Giessen, Germany; (C.K.); (A.L.S.); (F.B.); (E.S.); (M.M.); (M.S.); (C.K.)
| | - Emmanuel Schneck
- Department of Anesthesiology, Operative Intensive Care Medicine and Pain Therapy, University Hospital of Giessen and Marburg, Justus Liebig University of Giessen, 35392 Giessen, Germany; (C.K.); (A.L.S.); (F.B.); (E.S.); (M.M.); (M.S.); (C.K.)
| | - Andreas Boening
- Department of Cardiac and Vascular Surgery, University Hospital of Giessen and Marburg, Justus Liebig University of Giessen, 35392 Giessen, Germany;
| | - Melanie Markmann
- Department of Anesthesiology, Operative Intensive Care Medicine and Pain Therapy, University Hospital of Giessen and Marburg, Justus Liebig University of Giessen, 35392 Giessen, Germany; (C.K.); (A.L.S.); (F.B.); (E.S.); (M.M.); (M.S.); (C.K.)
| | - Michael Sander
- Department of Anesthesiology, Operative Intensive Care Medicine and Pain Therapy, University Hospital of Giessen and Marburg, Justus Liebig University of Giessen, 35392 Giessen, Germany; (C.K.); (A.L.S.); (F.B.); (E.S.); (M.M.); (M.S.); (C.K.)
| | - Christian Koch
- Department of Anesthesiology, Operative Intensive Care Medicine and Pain Therapy, University Hospital of Giessen and Marburg, Justus Liebig University of Giessen, 35392 Giessen, Germany; (C.K.); (A.L.S.); (F.B.); (E.S.); (M.M.); (M.S.); (C.K.)
| |
Collapse
|
4
|
Mayor E. Neurotrophic effects of intermittent fasting, calorie restriction and exercise: a review and annotated bibliography. FRONTIERS IN AGING 2023; 4:1161814. [PMID: 37334045 PMCID: PMC10273285 DOI: 10.3389/fragi.2023.1161814] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/09/2023] [Indexed: 06/20/2023]
Abstract
In the last decades, important progress has been achieved in the understanding of the neurotrophic effects of intermittent fasting (IF), calorie restriction (CR) and exercise. Improved neuroprotection, synaptic plasticity and adult neurogenesis (NSPAN) are essential examples of these neurotrophic effects. The importance in this respect of the metabolic switch from glucose to ketone bodies as cellular fuel has been highlighted. More recently, calorie restriction mimetics (CRMs; resveratrol and other polyphenols in particular) have been investigated thoroughly in relation to NSPAN. In the narrative review sections of this manuscript, recent findings on these essential functions are synthesized and the most important molecules involved are presented. The most researched signaling pathways (PI3K, Akt, mTOR, AMPK, GSK3β, ULK, MAPK, PGC-1α, NF-κB, sirtuins, Notch, Sonic hedgehog and Wnt) and processes (e.g., anti-inflammation, autophagy, apoptosis) that support or thwart neuroprotection, synaptic plasticity and neurogenesis are then briefly presented. This provides an accessible entry point to the literature. In the annotated bibliography section of this contribution, brief summaries are provided of about 30 literature reviews relating to the neurotrophic effects of interest in relation to IF, CR, CRMs and exercise. Most of the selected reviews address these essential functions from the perspective of healthier aging (sometimes discussing epigenetic factors) and the reduction of the risk for neurodegenerative diseases (Alzheimer's disease, Huntington's disease, Parkinson's disease) and depression or the improvement of cognitive function.
Collapse
|
5
|
The effect of ghrelin on antioxidant status in the rat's model of Alzheimer's disease induced by amyloid-beta. Biomedicine (Taipei) 2023; 12:44-54. [PMID: 36816173 PMCID: PMC9910231 DOI: 10.37796/2211-8039.1341] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 05/28/2022] [Accepted: 05/30/2022] [Indexed: 11/27/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder associated with amyloid-beta (Aβ) plaque formation and oxidative stress in the brain. Ghrelin has been proven to exert antioxidant activity and neuroprotection in different neurological diseases. This study is going on to examine the effect of ghrelin on antioxidant status in the rat's model of AD induced by Aβ. Cognitive impairment was induced by intra-hippocampal administration of Aβ (10 μg) in Wistar rats and ghrelin (80 μg/kg) was administrated intraperitoneal for ten consecutive days. Behavior was assessed with Morris water maze and passive avoidance tests. Malondialdehyde (MDA) level as a marker of lipid peroxidation was assessed using the thiobarbituric acid. Catalase activity was assayed by the decomposition of H2O2. Antioxidant capacity was determined using the FRAP method. Treatment with ghrelin decreased the hippocampus and serum MDA levels in wild-type rodents and prevented an increase in hippocampal and serum MDA levels in animals receiving Aβ. There was no significant change in the serum catalase activity between the studied groups. Hippocampus catalase activity was reduced in the Aβ group and treatment with ghrelin increased it. The antioxidant capacity of the hippocampus and serum increased in the ghrelin-receiving control group. The hippocampus antioxidant capacity level decreased in the Aβ group, and treatment with ghrelin increased it, but there were no significant changes in the serum antioxidant capacity of animals receiving Aβ. These results provide evidence that the administration of ghrelin has antioxidant properties and protects against hippocampal lipid peroxidation in a rat model of AD.
Collapse
|
6
|
Amin AM, Mostafa H, Khojah HMJ. Insulin resistance in Alzheimer's disease: The genetics and metabolomics links. Clin Chim Acta 2023; 539:215-236. [PMID: 36566957 DOI: 10.1016/j.cca.2022.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/16/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with significant socioeconomic burden worldwide. Although genetics and environmental factors play a role, AD is highly associated with insulin resistance (IR) disorders such as metabolic syndrome (MS), obesity, and type two diabetes mellitus (T2DM). These findings highlight a shared pathogenesis. The use of metabolomics as a downstream systems' biology (omics) approach can help to identify these shared metabolic traits and assist in the early identification of at-risk groups and potentially guide therapy. Targeting the shared AD-IR metabolic trait with lifestyle interventions and pharmacological treatments may offer promising AD therapeutic approach. In this narrative review, we reviewed the literature on the AD-IR pathogenic link, the shared genetics and metabolomics biomarkers between AD and IR disorders, as well as the lifestyle interventions and pharmacological treatments which target this pathogenic link.
Collapse
Affiliation(s)
- Arwa M Amin
- Department of Clinical and Hospital Pharmacy, College of Pharmacy, Taibah University, Madinah, Saudi Arabia.
| | - Hamza Mostafa
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Food Innovation Network (XIA), Nutrition and Food Safety Research Institute (INSA), Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona (UB), 08028 Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Hani M J Khojah
- Department of Clinical and Hospital Pharmacy, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| |
Collapse
|
7
|
Harris BN, Roberts BR, DiMarco GM, Maldonado KA, Okwunwanne Z, Savonenko AV, Soto PL. Hypothalamic-pituitary-adrenal (HPA) axis activity and anxiety-like behavior during aging: A test of the glucocorticoid cascade hypothesis in amyloidogenic APPswe/PS1dE9 mice. Gen Comp Endocrinol 2023; 330:114126. [PMID: 36122793 PMCID: PMC10250074 DOI: 10.1016/j.ygcen.2022.114126] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/06/2022] [Accepted: 09/13/2022] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a progressive, dementing, whole-body disorder that presents with decline in cognitive, behavioral, and emotional functions, as well as endocrine dysregulation. The etiology of AD is not fully understood but stress- and anxiety-related hormones may play a role in its development and trajectory. The glucocorticoid cascade hypothesis posits that levels of glucocorticoids increase with age, leading to dysregulated negative feedback, further elevated glucocorticoids, and resulting neuropathology. We examined the impact of age (from 2 to 10 months) and stressor exposure (predator odor) on hormone levels (corticosterone and ghrelin), anxiety-like behavior (open field and light dark tests), and memory-related behavior (novel object recognition; NOR), and whether these various measures correlated with neuropathology (hippocampus and cortex amyloid beta, Aβ) in male and female APPswe/PS1dE9 transgenic and non-transgenic mice. Additionally, we performed exploratory analyses to probe if the open field and light dark test as commonly used tasks to assess anxiety levels were correlated. Consistent with the glucocorticoid cascade hypothesis, baseline corticosterone increased with age. Predator odor exposure elevated corticosterone at each age, but in contrast to the glucocorticoid cascade hypothesis, the magnitude of stressor-induced elevations in corticosterone levels did not increase with age. Overall, transgenic mice had higher post-stressor, but not baseline, corticosterone than non-transgenic mice, and across both genotypes, females consistently had higher (baseline and post-stressor) corticosterone than males. Behavior in the open field test primarily showed decreased locomotion with age, and this was pronounced in transgenic females. Anxiety-like behaviors in the light dark test were exacerbated following predator odor, and female transgenic mice were the most impacted. Compared to transgenic males, transgenic females had higher Aβ concentrations and showed more anxiety-like behavior. Performance on the NOR did not differ significantly between genotypes. Lastly, we did not find robust, statistically significant correlations among corticosterone, ghrelin, recognition memory, anxiety-like behaviors, or Aβ, suggesting outcomes are not strongly related on the individual level. Our data suggest that despite Aβ accumulation in the hippocampus and cortex, male and female APPswePS1dE9 transgenic mice do not differ robustly from their non-transgenic littermates in physiological, endocrine, and behavioral measures at the range of ages studied here.
Collapse
Affiliation(s)
- Breanna N Harris
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, United States.
| | - Breanna R Roberts
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, United States
| | - Giuliana M DiMarco
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, United States; Center for Molecular and Behavioral Neuroscience, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | | | - Zenobia Okwunwanne
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, United States
| | - Alena V Savonenko
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Paul L Soto
- Department of Psychology, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
8
|
Meanti R, Bresciani E, Rizzi L, Coco S, Zambelli V, Dimitroulas A, Molteni L, Omeljaniuk RJ, Locatelli V, Torsello A. Potential Applications for Growth Hormone Secretagogues Treatment of Amyotrophic Lateral Sclerosis. Curr Neuropharmacol 2023; 21:2376-2394. [PMID: 36111771 PMCID: PMC10616926 DOI: 10.2174/1570159x20666220915103613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/18/2022] [Accepted: 08/01/2022] [Indexed: 11/22/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) arises from neuronal death due to complex interactions of genetic, molecular, and environmental factors. Currently, only two drugs, riluzole and edaravone, have been approved to slow the progression of this disease. However, ghrelin and other ligands of the GHS-R1a receptor have demonstrated interesting neuroprotective activities that could be exploited in this pathology. Ghrelin, a 28-amino acid hormone, primarily synthesized and secreted by oxyntic cells in the stomach wall, binds to the pituitary GHS-R1a and stimulates GH secretion; in addition, ghrelin is endowed with multiple extra endocrine bioactivities. Native ghrelin requires esterification with octanoic acid for binding to the GHS-R1a receptor; however, this esterified form is very labile and represents less than 10% of circulating ghrelin. A large number of synthetic compounds, the growth hormone secretagogues (GHS) encompassing short peptides, peptoids, and non-peptidic moieties, are capable of mimicking several biological activities of ghrelin, including stimulation of GH release, appetite, and elevation of blood IGF-I levels. GHS have demonstrated neuroprotective and anticonvulsant effects in experimental models of pathologies both in vitro and in vivo. To illustrate, some GHS, currently under evaluation by regulatory agencies for the treatment of human cachexia, have a good safety profile and are safe for human use. Collectively, evidence suggests that ghrelin and cognate GHS may constitute potential therapies for ALS.
Collapse
Affiliation(s)
- Ramona Meanti
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza, 20900, Italy
| | - Elena Bresciani
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza, 20900, Italy
| | - Laura Rizzi
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza, 20900, Italy
| | - Silvia Coco
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza, 20900, Italy
| | - Vanessa Zambelli
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza, 20900, Italy
| | - Anna Dimitroulas
- Faculty of Health and Medical Sciences, University of Surrey, Stag Hill, Guildford, GU2 7XH, United Kingdom
| | - Laura Molteni
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza, 20900, Italy
| | - Robert J. Omeljaniuk
- Department of Biology, Lakehead University, 955 Oliver Rd, Thunder Bay, Ontario, P7B 5E1, Canada
| | - Vittorio Locatelli
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza, 20900, Italy
| | - Antonio Torsello
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza, 20900, Italy
| |
Collapse
|
9
|
Gupta S, Mukhopadhyay S, Mitra A. Therapeutic potential of GHSR-1A antagonism in alcohol dependence, a review. Life Sci 2022; 291:120316. [PMID: 35016882 DOI: 10.1016/j.lfs.2022.120316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 11/28/2022]
Abstract
Growth hormone secretagogue receptor type 1A (GHSR-1A) is a functional receptor of orexigenic peptide ghrelin and is highly expressed in mesolimbic dopaminergic systems that regulate incentive value of artificial reward in substance abuse. Interestingly, GHSR-1A has also shown ligand-independent constitutive activity. Alcohol use disorder (AUD) is one of the growing concerns worldwide as it involves complex neuro-psycho-endocrinological interactions. Positive correlation of acylated ghrelin and alcohol-induced human brain response in the right and left ventral striatum are evident. In the last decade, the beneficial effects of ghrelin receptor (GHSR-1A) antagonism to suppress artificial reward circuitries and induce self-control for alcohol consumption have drawn significant attention from researchers. In this updated review, we summarize the available recent preclinical, clinical, and experimental data to discuss functional, molecular actions of central ghrelin-GHSR-1A signaling in different craving levels for alcohol as well as to promote "GHSR-1A antagonism" as one of the potential therapies in early abstinence.
Collapse
Affiliation(s)
- Shreyasi Gupta
- Department of Zoology, Triveni Devi Bhalotia College, Raniganj, Paschim Bardhaman 713 347, West Bengal, India
| | - Sanchari Mukhopadhyay
- Department of Psychiatry, National Institute of Mental Health and Neurosciences, Hombegowda Nagar, Bengaluru 560029, India
| | - Arkadeep Mitra
- Department of Zoology, City College, 102/1, Raja Rammohan Sarani, Kolkata 700 009, West Bengal, India.
| |
Collapse
|
10
|
Lin DT, Kao NJ, Cross TWL, Lee WJ, Lin SH. nEffects of ketogenic diet on cognitive functions of mice fed high-fat-high-cholesterol diet. J Nutr Biochem 2022; 104:108974. [DOI: 10.1016/j.jnutbio.2022.108974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 12/07/2021] [Accepted: 01/31/2022] [Indexed: 12/28/2022]
|
11
|
Herrera-Martínez Y, Alzas Teomiro C, León Idougourram S, Molina Puertas MJ, Calañas Continente A, Serrano Blanch R, Castaño JP, Gálvez Moreno MÁ, Gahete MD, Luque RM, Herrera-Martínez AD. Sarcopenia and Ghrelin System in the Clinical Outcome and Prognosis of Gastroenteropancreatic Neuroendocrine Neoplasms. Cancers (Basel) 2021; 14:cancers14010111. [PMID: 35008278 PMCID: PMC8750458 DOI: 10.3390/cancers14010111] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/11/2021] [Accepted: 12/13/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Malnutrition and sarcopenia affect clinical outcomes in cancer patients. Nutritional evaluation in patients with neuroendocrine neoplasms (NENs) is not routinely performed. Currently, the evaluation of sarcopenia using CT scans is the gold standard in cancer patients, additionally, anthropometric, biochemical and molecular analysis of patients with gastroenteropancreatic NENs at diagnosis was perfomed. The expression levels of key ghrelin system components were assessed in 63 tumor samples. Results: Nutritional parameters were similar in GEP-NEN tumors of different origin. Relapsed disease was associated with decreased BMI. Patients who presented with weight loss at diagnosis had significantly lower overall survival (108 (25–302) vs. 263 (79–136) months). Ghrelin O-acyltransferase (GOAT) enzyme expression was higher in these patients. The prevalence of sarcopenia using CT images reached 87.2%. Mortality was observed only in patients with sarcopenia. Muscle evaluation was correlated with biochemical parameters but not with the expression of ghrelin system components. Conclusion: Survival is related to the nutritional status of patients with GEP-NENs and also to the molecular expression of some relevant ghrelin system components. Routine nutritional evaluation should be performed in these patients, in order to prescribe appropriate nutritional support, when necessary, for increasing quality of life and improving clinical outcomes. Abstract Background: Malnutrition and sarcopenia affect clinical outcomes and treatment response in cancer patients. Patients with neuroendocrine neoplasms (NENs) may present with additional symptoms related to tumor localization in the gastrointestinal tract and hormone secretion, increasing the risk and effects of sarcopenia. Aim: To explore the presence of malnutrition and sarcopenia in gastroenteropancreatic (GEP)-NEN patients, their relation to tumor characteristics, patient outcomes, survival and the molecular expression of ghrelin system components in the tumor. Patients and methods: One-hundred-and-four patients were included. Anthropometric, biochemical and CT-scans at diagnosis were evaluated. The expression levels of key ghrelin system components were assessed in 63 tumor samples. Results: Nutritional parameters were similar in GEP-NEN tumors of different origin. Relapsed disease was associated with decreased BMI. Patients who presented with weight loss at diagnosis had significantly lower overall survival (108 (25–302) vs. 263 (79–136) months). Ghrelin O-acyltransferase (GOAT) enzyme expression was higher in these patients. The prevalence of sarcopenia using CT images reached 87.2%. Mortality was observed only in patients with sarcopenia. Muscle evaluation was correlated with biochemical parameters but not with the expression of ghrelin system components. Conclusion: Survival is related to the nutritional status of patients with GEP-NENs and also to the molecular expression of some relevant ghrelin system components. Routine nutritional evaluation should be performed in these patients, in order to prescribe appropriate nutritional support, when necessary, for increasing quality of life and improving clinical outcomes.
Collapse
Affiliation(s)
| | - Carlos Alzas Teomiro
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, 14004 Cordova, Spain; (C.A.T.); (S.L.I.); (M.J.M.P.); (A.C.C.); (R.S.B.); (J.P.C.); (M.Á.G.M.); (M.D.G.); (R.M.L.)
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, 14004 Cordova, Spain
| | - Soraya León Idougourram
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, 14004 Cordova, Spain; (C.A.T.); (S.L.I.); (M.J.M.P.); (A.C.C.); (R.S.B.); (J.P.C.); (M.Á.G.M.); (M.D.G.); (R.M.L.)
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, 14004 Cordova, Spain
| | - María José Molina Puertas
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, 14004 Cordova, Spain; (C.A.T.); (S.L.I.); (M.J.M.P.); (A.C.C.); (R.S.B.); (J.P.C.); (M.Á.G.M.); (M.D.G.); (R.M.L.)
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, 14004 Cordova, Spain
| | - Alfonso Calañas Continente
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, 14004 Cordova, Spain; (C.A.T.); (S.L.I.); (M.J.M.P.); (A.C.C.); (R.S.B.); (J.P.C.); (M.Á.G.M.); (M.D.G.); (R.M.L.)
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, 14004 Cordova, Spain
| | - Raquel Serrano Blanch
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, 14004 Cordova, Spain; (C.A.T.); (S.L.I.); (M.J.M.P.); (A.C.C.); (R.S.B.); (J.P.C.); (M.Á.G.M.); (M.D.G.); (R.M.L.)
- Medical Oncology Service, Reina Sofia University Hospital, 14004 Cordova, Spain
| | - Justo P. Castaño
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, 14004 Cordova, Spain; (C.A.T.); (S.L.I.); (M.J.M.P.); (A.C.C.); (R.S.B.); (J.P.C.); (M.Á.G.M.); (M.D.G.); (R.M.L.)
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, 14014 Cordova, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 14004 Cordova, Spain
| | - María Ángeles Gálvez Moreno
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, 14004 Cordova, Spain; (C.A.T.); (S.L.I.); (M.J.M.P.); (A.C.C.); (R.S.B.); (J.P.C.); (M.Á.G.M.); (M.D.G.); (R.M.L.)
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, 14004 Cordova, Spain
| | - Manuel D. Gahete
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, 14004 Cordova, Spain; (C.A.T.); (S.L.I.); (M.J.M.P.); (A.C.C.); (R.S.B.); (J.P.C.); (M.Á.G.M.); (M.D.G.); (R.M.L.)
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, 14014 Cordova, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 14004 Cordova, Spain
| | - Raúl M. Luque
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, 14004 Cordova, Spain; (C.A.T.); (S.L.I.); (M.J.M.P.); (A.C.C.); (R.S.B.); (J.P.C.); (M.Á.G.M.); (M.D.G.); (R.M.L.)
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, 14014 Cordova, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 14004 Cordova, Spain
| | - Aura D. Herrera-Martínez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, 14004 Cordova, Spain; (C.A.T.); (S.L.I.); (M.J.M.P.); (A.C.C.); (R.S.B.); (J.P.C.); (M.Á.G.M.); (M.D.G.); (R.M.L.)
- Endocrinology and Nutrition Service, Reina Sofia University Hospital, 14004 Cordova, Spain
- Correspondence:
| |
Collapse
|
12
|
Sun P, Su L, Zhu H, Li X, Guo Y, Du X, Zhang L, Qin C. Gut Microbiota Regulation and Their Implication in the Development of Neurodegenerative Disease. Microorganisms 2021; 9:microorganisms9112281. [PMID: 34835406 PMCID: PMC8621510 DOI: 10.3390/microorganisms9112281] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/19/2021] [Accepted: 10/27/2021] [Indexed: 12/12/2022] Open
Abstract
In recent years, human gut microbiota have become one of the most promising areas of microorganism research; meanwhile, the inter-relation between the gut microbiota and various human diseases is a primary focus. As is demonstrated by the accumulating evidence, the gastrointestinal tract and central nervous system interact through the gut–brain axis, which includes neuronal, immune-mediated and metabolite-mediated pathways. Additionally, recent progress from both preclinical and clinical studies indicated that gut microbiota play a pivotal role in gut–brain interactions, whereas the imbalance of the gut microbiota composition may be associated with the pathogenesis of neurological diseases (particularly neurodegenerative diseases), the underlying mechanism of which is insufficiently studied. This review aims to highlight the relationship between gut microbiota and neurodegenerative diseases, and to contribute to our understanding of the function of gut microbiota in neurodegeneration, as well as their relevant mechanisms. Furthermore, we also discuss the current application and future prospects of microbiota-associated therapy, including probiotics and fecal microbiota transplantation (FMT), potentially shedding new light on the research of neurodegeneration.
Collapse
Affiliation(s)
- Peilin Sun
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS), Beijing 100021, China; (P.S.); (L.S.); (H.Z.); (X.L.); (Y.G.); (X.D.); (L.Z.)
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Chinese Academy of Medical Sciences (CAMS), Beijing 100021, China
| | - Lei Su
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS), Beijing 100021, China; (P.S.); (L.S.); (H.Z.); (X.L.); (Y.G.); (X.D.); (L.Z.)
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Chinese Academy of Medical Sciences (CAMS), Beijing 100021, China
| | - Hua Zhu
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS), Beijing 100021, China; (P.S.); (L.S.); (H.Z.); (X.L.); (Y.G.); (X.D.); (L.Z.)
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Chinese Academy of Medical Sciences (CAMS), Beijing 100021, China
| | - Xue Li
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS), Beijing 100021, China; (P.S.); (L.S.); (H.Z.); (X.L.); (Y.G.); (X.D.); (L.Z.)
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Chinese Academy of Medical Sciences (CAMS), Beijing 100021, China
| | - Yaxi Guo
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS), Beijing 100021, China; (P.S.); (L.S.); (H.Z.); (X.L.); (Y.G.); (X.D.); (L.Z.)
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Chinese Academy of Medical Sciences (CAMS), Beijing 100021, China
| | - Xiaopeng Du
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS), Beijing 100021, China; (P.S.); (L.S.); (H.Z.); (X.L.); (Y.G.); (X.D.); (L.Z.)
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Chinese Academy of Medical Sciences (CAMS), Beijing 100021, China
| | - Ling Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS), Beijing 100021, China; (P.S.); (L.S.); (H.Z.); (X.L.); (Y.G.); (X.D.); (L.Z.)
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Chinese Academy of Medical Sciences (CAMS), Beijing 100021, China
| | - Chuan Qin
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS), Beijing 100021, China; (P.S.); (L.S.); (H.Z.); (X.L.); (Y.G.); (X.D.); (L.Z.)
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Chinese Academy of Medical Sciences (CAMS), Beijing 100021, China
- Correspondence: ; Tel.: +86-10-8777-8141
| |
Collapse
|
13
|
Ferguson SA, Panos JJ, Sloper D, Varma V, Sarkar S. Alzheimer's disease: a step closer to understanding type 3 diabetes in African Americans. Metab Brain Dis 2021; 36:1803-1816. [PMID: 34021875 DOI: 10.1007/s11011-021-00754-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 05/10/2021] [Indexed: 10/21/2022]
Abstract
Alzheimer's disease (AD) is the fourth leading cause of death in the United States and the most common cause of adult-onset dementia. Recent results suggest an increased prevalence and severity in African Americans compared to Caucasians. Our understanding of the potential mechanism(s) underlying this ethnicity difference is limited. We previously described ethnicity-related differences in levels of neurodegenerative proteins and cytokines/chemokines in the BA21 region of African Americans and Caucasians with AD. Here, similar multiplex assays were used to examine those endpoints in patient postmortem cerebrospinal fluid (CSF). Additionally, we measured levels of C-peptide, ghrelin, gastric inhibitory polypeptide (GIP), glucagon-like peptide-1 (GLP-1), glucagon, insulin, leptin, PAI-1, resistin, and visfatin using a human diabetes 10-plex assay. The cytokine and chemokine assays revealed that levels of 26 chemokines or cytokines differed significantly with ethnicity, and three of those were significantly associated with gender. The neurodegenerative disease panel indicated that levels of soluble RAGE were significantly elevated in African Americans compared to Caucasians. All measures in the diabetes disease panel assay were significantly elevated in African Americans: ghrelin, GIP, GLP-1, glucagon, insulin, and visfatin. Through peripheral sample analysis, these results provide further evidence that ethnicity is critically involved in the manifestation of AD.
Collapse
Affiliation(s)
- Sherry A Ferguson
- Division of Neurotoxicology, National Center for Toxicological Research/Food and Drug Administration, Jefferson, AR, 72079, USA
| | - John J Panos
- Division of Neurotoxicology, National Center for Toxicological Research/Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Daniel Sloper
- Division of Systems Biology, National Center for Toxicological Research/Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Vijayalakshmi Varma
- Division of Systems Biology, National Center for Toxicological Research/Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Sumit Sarkar
- Division of Neurotoxicology, National Center for Toxicological Research/Food and Drug Administration, Jefferson, AR, 72079, USA.
| |
Collapse
|
14
|
Protective Features of Calorie Restriction on Cuprizone-induced Demyelination via Modulating Microglial Phenotype. J Chem Neuroanat 2021; 116:102013. [PMID: 34391881 DOI: 10.1016/j.jchemneu.2021.102013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/29/2021] [Accepted: 08/07/2021] [Indexed: 01/21/2023]
Abstract
Multiple sclerosis (MS) is an immune-mediated demyelinating disorder in the central nervous system (CNS) with no definitive treatment, but it can be alleviated by changing life habits. Calorie restriction (CR) is effective in preventing or treating metabolic and autoimmune disorders. CR is one of the helpful approaches to control the progression of MS. In the present study, we investigated the preventive effect of caloric restriction on cuprizone induced-demyelination, a model of multiple sclerosis. To induce acute demyelination in C57/BL6 mice, we added 0.2% Cuprizone (CPZ) to their diet for 6 weeks. To induce calorie restriction, 10% Carboxymethyl cellulose (CMC) was added to the diet as a dietary cellulose fiber for 6 weeks. Remyelination was studied by luxol fast blue (LFB) staining. Microglia activity, M1 and M2 microglial/macrophage phenotypes were assessed by immunohistochemistry of Iba-1, iNOS and Arg-1, respectively. The expression of targeted genes was assessed by the real-time polymerase chain reaction. Luxol fast blue (LFB) staining showed that the CR regimen could decrease the cuprizone-induced demyelination process (p < 0.01). Moreover, the CR application could improve balance and motor performance in cuprizone-intoxicated mice by significantly enhancing protein and gene expression of Sirt1, M2 microglial phenotype marker (Arg-1) and Akt1 gene expression, also decreased M1 microglial phenotype marker (iNOS), Akt2 and P53 gene expressions (p < 0.05). Cumulatively, it can be concluded that caloric restriction was able to counteract MS symptoms through alleviating inflammatory responses.
Collapse
|
15
|
Noda M, Liu J, Long J. Neuroprotective and Preventative Effects of Molecular Hydrogen. Curr Pharm Des 2021; 27:585-591. [PMID: 33076798 DOI: 10.2174/1381612826666201019103020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 08/12/2020] [Indexed: 11/22/2022]
Abstract
One of the beneficial effects of molecular hydrogen (H2, hydrogen gas) is neuroprotection and prevention of neurological disorders. It is important and useful if taking H2 every day can prevent or ameliorate the progression of neurodegenerative disorders, such as Parkinson's disease or Alzheimer's disease, both lacking specific therapeutic drugs. There are several mechanisms of how H2 protects neuronal damage. Anti-oxidative, anti-inflammatory, and the regulation of the endocrine system via stomach-brain connection seem to play an important role. At the cellular and tissue level, H2 appears to prevent the production of reactive oxygen species (ROS), and not only hydroxy radical (•OH) but also superoxide. In Parkinson's disease model mice, chronic intake of H2 causes the release of ghrelin from the stomach. In Alzheimer's disease model mice, sex-different neuroprotection is observed by chronic intake of H2. In female mice, declines of estrogen and estrogen receptor-β (ERβ) are prevented by H2, upregulating brain-derived neurotrophic factor (BDNF) and its receptor, tyrosine kinase receptor B (TrkB). The question of how drinking H2 upregulates the release of ghrelin or attenuates the decline of estrogen remains to be investigated and the mechanism of how H2 modulates endocrine systems and the fundamental question of what or where is the target of H2 needs to be elucidated for a better understanding of the effects of H2.
Collapse
Affiliation(s)
- Mami Noda
- Laboratory of Pathophysiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Jiankang Liu
- Center for Mitochondrial Biology and Medicine and Center for Translational Medicine, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Jiangang Long
- Center for Mitochondrial Biology and Medicine and Center for Translational Medicine, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
16
|
Wang H, Dou S, Zhu J, Shao Z, Wang C, Cheng B. Regulatory effects of ghrelin on endoplasmic reticulum stress, oxidative stress, and autophagy: Therapeutic potential. Neuropeptides 2021; 85:102112. [PMID: 33333485 DOI: 10.1016/j.npep.2020.102112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 10/29/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023]
Abstract
Ghrelin is a regulatory peptide that is the endogenous ligand of the growth hormone secretagogue 1a (GHS-R1a) which belongs to the G protein-coupled receptor family. Ghrelin and GHS-R1a are widely expressed in the central and peripheral tissues and play therapeutic potential roles in the cytoprotection of many internal organs. Endoplasmic reticulum stress (ERS), oxidative stress, and autophagy dysfunction, which are involved in various diseases. In recent years, accumulating evidence has suggested that ghrelin exerts protective effects by regulating ERS, oxidative stress, and autophagy in diverse diseases. This review article summarizes information about the roles of the ghrelin system on ERS, oxidative stress, and autophagy in multiple diseases. It is suggested that ghrelin positively affects the treatment of diseases and may be considered as a therapeutic drug in many illnesses.
Collapse
Affiliation(s)
- Huiqing Wang
- Cheeloo College of Medicine, Shandong University, 250014 Jinan, China
| | - Shanshan Dou
- Neurobiology Institute, Jining Medical University, 272067 Jining, China
| | - Junge Zhu
- Cheeloo College of Medicine, Shandong University, 250014 Jinan, China
| | - Ziqi Shao
- Cheeloo College of Medicine, Shandong University, 250014 Jinan, China
| | - Chunmei Wang
- Neurobiology Institute, Jining Medical University, 272067 Jining, China
| | - Baohua Cheng
- Neurobiology Institute, Jining Medical University, 272067 Jining, China.
| |
Collapse
|
17
|
Kim S, Nam Y, Shin SJ, Park YH, Jeon SG, Kim JI, Kim MJ, Moon M. The Potential Roles of Ghrelin in Metabolic Syndrome and Secondary Symptoms of Alzheimer's Disease. Front Neurosci 2020; 14:583097. [PMID: 33071750 PMCID: PMC7543232 DOI: 10.3389/fnins.2020.583097] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
Abstract
Although the major causative factors of Alzheimer's disease (AD) are the accumulation of amyloid β and hyperphosphorylated tau, AD can also be caused by metabolic dysfunction. The major clinical symptom of AD is cognitive dysfunction. However, AD is also accompanied by various secondary symptoms such as depression, sleep-wake disturbances, and abnormal eating behaviors. Interestingly, the orexigenic hormone ghrelin has been suggested to have beneficial effects on AD-related metabolic syndrome and secondary symptoms. Ghrelin improves lipid distribution and alters insulin sensitivity, effects that are hypothesized to delay the progression of AD. Furthermore, ghrelin can relieve depression by enhancing the secretion of hormones such as serotonin, noradrenaline, and orexin. Moreover, ghrelin can upregulate the expression of neurotrophic factors such as brain-derived neurotrophic factor and modulate the release of proinflammatory cytokines such as tumor necrosis factor α and interleukin 1β. Ghrelin alleviates sleep-wake disturbances by increasing the levels of melatonin, melanin-concentrating hormone. Ghrelin reduces the risk of abnormal eating behaviors by increasing neuropeptide Y and γ-aminobutyric acid. In addition, ghrelin increases food intake by inhibiting fatty acid biosynthesis. However, despite the numerous studies on the role of ghrelin in the AD-related pathology and metabolic disorders, there are only a few studies that investigate the effects of ghrelin on secondary symptoms associated with AD. In this mini review, our purpose is to provide the insights of future study by organizing the previous studies for the role of ghrelin in AD-related pathology and metabolic disorders.
Collapse
Affiliation(s)
- Sujin Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, South Korea
| | - Yunkwon Nam
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, South Korea
| | - Soo Jung Shin
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, South Korea
| | - Yong Ho Park
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, South Korea
| | - Seong Gak Jeon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, South Korea.,Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu, South Korea
| | - Jin-Il Kim
- Department of Nursing, College of Nursing, Jeju National University, Jeju-si, South Korea
| | - Min-Jeong Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, South Korea
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, South Korea
| |
Collapse
|
18
|
Breithaupt L, Chunga-Iturry N, Lyall AE, Cetin-Karayumak S, Becker KR, Thomas JJ, Slattery M, Makris N, Plessow F, Pasternak O, Holsen LM, Kubicki M, Misra M, Lawson EA, Eddy KT. Developmental stage-dependent relationships between ghrelin levels and hippocampal white matter connections in low-weight anorexia nervosa and atypical anorexia nervosa. Psychoneuroendocrinology 2020; 119:104722. [PMID: 32512249 PMCID: PMC8629489 DOI: 10.1016/j.psyneuen.2020.104722] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/29/2020] [Accepted: 05/18/2020] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Disruptions in homeostatic and hedonic food motivation are proposed to underlie anorexia nervosa (AN) and atypical AN, restrictive eating disorders which commonly onset in puberty. Ghrelin, a neuroprotective hormone that drives hedonic eating is increased in AN and is expressed in the hippocampus. White matter (WM) undergoes significant change during puberty in regions involved in food motivation, particularly WM tracts connected with the hippocampus. The association between ghrelin and WM region of interest (ROI) with hippocampal connections in restrictive eating disorders, particularly in adolescence during key neurodevelopmental growth, is unknown. METHODS We evaluated fasting plasma ghrelin and WM microstructure (measured by free-water corrected fractional anisotropy (FA-t)) in WM ROIs with hippocampal connections - the fornix and the hippocampal portion of the cingulum - in 56 adolescent females (age range: 11.9 - 22.1 y; mean: 19.0 y) with low-weight eating disorders including AN and atypical AN (N = 36) and healthy controls (N = 20). RESULTS FA-t in the fornix or hippocampal portion of the fornix did not differ between groups. Ghrelin was higher in AN/atypical AN vs. HC and was positively correlated with puberty stage in the AN/atypical AN group, but not the HC group. The correlation between ghrelin and FA-t in the fornix was significantly different in females with AN/atypical AN compared to controls. In AN/atypical AN, pubertal stage moderated the relation between fasting plasma ghrelin and FA-t in the fornix: higher fasting ghrelin was associated with lower FA-t in the fornix in late-post-puberty, but was not associated with FA-t in the early to mid stages of puberty. CONCLUSIONS In post-pubertal females with low-weight AN/atypical AN, higher levels of ghrelin are associated with lower FA-t in the fornix. This relationship is not evident in the early to mid stages of puberty in AN/atypical AN or in HC, and may reflect a lack of possible neuroprotective effects of ghrelin in late-post puberty only. Understanding the effects of ghrelin on WM microstructure longitudinally and following recovery from AN/Atypical AN and how this differs across pubertal stages will be an important next step. These findings could ultimately inform treatment staging and aid in diagnosis and detection of AN/atypical AN.
Collapse
Affiliation(s)
- Lauren Breithaupt
- Eating Disorders Clinical and Research Program, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Natalia Chunga-Iturry
- Harvard Medical School, Boston, MA, USA; Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Boston, USA
| | - Amanda E Lyall
- Harvard Medical School, Boston, MA, USA; Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Boston, USA
| | - Suheyla Cetin-Karayumak
- Harvard Medical School, Boston, MA, USA; Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Boston, USA
| | - Kendra R Becker
- Eating Disorders Clinical and Research Program, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Jennifer J Thomas
- Eating Disorders Clinical and Research Program, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Meghan Slattery
- Neuroendocrine Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Nikos Makris
- Harvard Medical School, Boston, MA, USA; Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Boston, USA; Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States of America; Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Franziska Plessow
- Harvard Medical School, Boston, MA, USA; Neuroendocrine Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Ofer Pasternak
- Harvard Medical School, Boston, MA, USA; Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Boston, USA; Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Laura M Holsen
- Harvard Medical School, Boston, MA, USA; Division of Women's Health, Brigham and Women's Hospital, Boston, MA, USA
| | - Marek Kubicki
- Harvard Medical School, Boston, MA, USA; Psychiatry Neuroimaging Laboratory, Brigham and Women's Hospital, Boston, USA; Department of Neurology, Massachusetts General Hospital, Charlestown, MA, United States of America; Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Madhusmita Misra
- Harvard Medical School, Boston, MA, USA; Neuroendocrine Unit, Massachusetts General Hospital, Boston, MA, USA; Division of Pediatric Endocrinology, Massachusetts General Hospital, Boston, MA, USA
| | - Elizabeth A Lawson
- Harvard Medical School, Boston, MA, USA; Neuroendocrine Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Kamryn T Eddy
- Eating Disorders Clinical and Research Program, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| |
Collapse
|
19
|
Yahfoufi N, Matar C, Ismail N. Adolescence and Aging: Impact of Adolescence Inflammatory Stress and Microbiota Alterations on Brain Development, Aging, and Neurodegeneration. J Gerontol A Biol Sci Med Sci 2020; 75:1251-1257. [PMID: 31917834 PMCID: PMC7302172 DOI: 10.1093/gerona/glaa006] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Indexed: 12/16/2022] Open
Abstract
Puberty/adolescence is a critical phase during neurodevelopment with numerous structural, neurochemical, and molecular changes occurring in response to genetic and environmental signals. A consequence of this major neuronal reorganizing and remodeling is a heightened level of vulnerability to stressors and immune challenges. The gut microbiota is a fundamental modulator of stress and immune responses and has been found to play a role in mental health conditions and neurodegenerative disorders. Environmental insults (stress, infection, neuroinflammation, and use of antibiotics) during adolescence can result in dysbiosis subsidizing the development of brain disorders later in life. Also, pubertal neuroinflammatory insults can alter neurodevelopment, impact brain functioning in an enduring manner, and contribute to neurological disorders related to brain aging, such as Alzheimer's disease, Parkinson's disease, and depression. Exposure to probiotics during puberty can mitigate inflammation, reverse dysbiosis, and decrease vulnerabilities to brain disorders later in life. The goal of this review is to reveal the consequences of pubertal exposure to stress and immune challenges on the gut microbiota, immune reactivity within the brain, and the risk or resilience to stress-induced mental illnesses and neurodegenerative disorders. We propose that the consumption of probiotics during adolescence contribute to the prevention of brain pathologies in adulthood.
Collapse
Affiliation(s)
- Nour Yahfoufi
- Cellular and Molecular Medicine Department, Faculty of Medicine, University of Ottawa, Ontario, Canada
| | - Chantal Matar
- Cellular and Molecular Medicine Department, Faculty of Medicine, University of Ottawa, Ontario, Canada
- School of Nutrition, Faculty of Health Sciences, University of Ottawa, Ontario, Canada
| | - Nafissa Ismail
- School of Psychology, Faculty of Social Sciences, University of Ottawa, Ontario, Canada
| |
Collapse
|
20
|
Yadegary A, Nazari-Serenjeh F, Darbandi N. Synergistic improvement effect of nicotine-ghrelin co-injection into the anterior ventral tegmental area on morphine-induced amnesia. Neuropeptides 2020; 80:102025. [PMID: 32029269 DOI: 10.1016/j.npep.2020.102025] [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: 06/06/2019] [Revised: 01/15/2020] [Accepted: 01/29/2020] [Indexed: 11/15/2022]
Abstract
In the present study the effect of ghrelin or ghrelin/nicotine injection into the anterior ventral tegmental area (aVTA) on morphine-induced amnesia in passive avoidance learning have been evaluated. Also, the role of the aVTA nicotinic receptors in possible ghrelin-induced effects has been investigated. All animals were bilaterally implanted with chronic cannulas in the aVTA. A step-through type passive avoidance task was used for measurement of memory. We found that post-training subcutaneous (s.c.) injection of morphine (0.5-7.5 mg/kg) dose-dependently reduced the step-through latency, indicating morphine-induced amnesia. Post-training bilateral infusion of ghrelin (0.3, 1.5 and 3 nmol/μl) in a dose-dependent manner reversed amnesia induced by morphine (7.5 mg/kg, s.c.). Furthermore, reversal effect of ghrelin (3 nmol/μl) was blocked by pre-treatment of intra-aVTA administration of mecamylamine (1-3 μg/rat), a nicotinic acetylcholine receptor antagonist. Intra-aVTA administration of the higher dose of mecamylamine (3 μg/rat) into the aVTA by itself decreased the step-through latency and induced amnesia. In addition, post-training intra-aVTA administration of nicotine (0.25, 0.5, 1 μg/rat) which alone cannot affect memory consolidation, decreased significantly the amnesia induced by morphine (7.5 mg/kg, s.c.). Co-treatment of an ineffective dose of ghrelin (0.3 nmol/μl) with an ineffective dose of nicotine (0.25 μg/rat) significantly increased step-through latency of morphine (7.5 mg/kg, s.c.) treated animals, indicating the synergistic effect of the drugs. Taken together, our results suggest that intra-aVTA administration of ghrelin reversed morphine-induced amnesia and that ghrelin interacts synergistically with nicotine to mitigate morphine-induced amnesia.
Collapse
Affiliation(s)
- Atena Yadegary
- Department of Biology, Faculty of Science, Arak University, Arak, Iran
| | | | - Niloufar Darbandi
- Department of Biology, Faculty of Science, Arak University, Arak, Iran
| |
Collapse
|
21
|
Barra R, Morgan C, Sáez-Briones P, Reyes-Parada M, Burgos H, Morales B, Hernández A. Facts and hypotheses about the programming of neuroplastic deficits by prenatal malnutrition. Nutr Rev 2020; 77:65-80. [PMID: 30445479 DOI: 10.1093/nutrit/nuy047] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Studies in rats have shown that a decrease in either protein content or total dietary calories results in molecular, structural, and functional changes in the cerebral cortex and hippocampus, among other brain regions, which lead to behavioral disturbances, including learning and memory deficits. The neurobiological bases underlying those effects depend at least in part on fetal programming of the developing brain, which in turn relies on epigenetic regulation of specific genes via stable and heritable modifications of chromatin. Prenatal malnutrition also leads to epigenetic programming of obesity, and obesity on its own can lead to poor cognitive performance in humans and experimental animals, complicating understanding of the factors involved in the fetal programming of neuroplasticity deficits. This review focuses on the role of epigenetic mechanisms involved in prenatal malnutrition-induced brain disturbances, which are apparent at a later postnatal age, through either a direct effect of fetal programming on brain plasticity or an indirect effect on the brain mediated by the postnatal development of obesity.
Collapse
Affiliation(s)
- Rafael Barra
- School of Medicine, Faculty of Medical Sciences, University of Santiago de Chile, Santiago, Chile
| | - Carlos Morgan
- Laboratory of Nutrition and Metabolic Regulation, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - Patricio Sáez-Briones
- School of Medicine, Faculty of Medical Sciences, University of Santiago de Chile, Santiago, Chile
| | - Miguel Reyes-Parada
- School of Medicine, Faculty of Medical Sciences, University of Santiago de Chile, Santiago, Chile.,Facultad de Ciencias de la Salud Universidad Autónoma de Chile, Talca, Chile
| | - Héctor Burgos
- Núcleo Disciplinar Psicología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.,Center of Innovation on Information Technologies for Social Applications (CITIAPS), University of Santiago de Chile, Santiago, Chile
| | - Bernardo Morales
- Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - Alejandro Hernández
- Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| |
Collapse
|
22
|
Ma L, Chan P. Understanding the Physiological Links Between Physical Frailty and Cognitive Decline. Aging Dis 2020; 11:405-418. [PMID: 32257550 PMCID: PMC7069469 DOI: 10.14336/ad.2019.0521] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 05/21/2019] [Indexed: 12/14/2022] Open
Abstract
Declines in both physical and cognitive function are associated with increasing age. Understanding the physiological link between physical frailty and cognitive decline may allow us to develop interventions that prevent and treat both conditions. Although there is significant epidemiological evidence linking physical frailty to cognitive decline, a complete understanding of the underpinning biological basis of the two disorders remains fragmented. This narrative review discusses insights into the potential roles of chronic inflammation, impaired hypothalamic-pituitary axis stress response, imbalanced energy metabolism, mitochondrial dysfunction, oxidative stress, and neuroendocrine dysfunction linking physical frailty with cognitive decline. We highlight the importance of easier identification of strategic approaches delaying the progression and onset of physical frailty and cognitive decline as well as preventing disability in the older population.
Collapse
Affiliation(s)
- Lina Ma
- 1Department of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing Institute of Geriatrics, Beijing, China.,2China National Clinical Research Center for Geriatric Medicine, Beijing, China
| | - Piu Chan
- 1Department of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing Institute of Geriatrics, Beijing, China.,2China National Clinical Research Center for Geriatric Medicine, Beijing, China.,3Department of Neurology and Neurobiology, Xuanwu Hospital, Capital Medical University, Beijing, China.,4Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Key Laboratory for Parkinson's Disease, Parkinson Disease Center of Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
23
|
Elfil M, Kamel S, Kandil M, Koo BB, Schaefer SM. Implications of the Gut Microbiome in Parkinson's Disease. Mov Disord 2020; 35:921-933. [DOI: 10.1002/mds.28004] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/06/2020] [Accepted: 02/04/2020] [Indexed: 12/14/2022] Open
Affiliation(s)
- Mohamed Elfil
- Department of NeurologyYale University New Haven Connecticut USA
| | - Serageldin Kamel
- Department of NeurologyYale University New Haven Connecticut USA
| | - Mohamed Kandil
- Department of NeurologyYale University New Haven Connecticut USA
| | - Brian B. Koo
- Department of NeurologyYale University New Haven Connecticut USA
- Center for Neuroepidemiology and Clinical Neurologic Research Yale New Haven Connecticut USA
- Department of NeurologyConnecticut Veterans Affairs Healthcare System West Haven Connecticut USA
| | - Sara M. Schaefer
- Department of NeurologyYale University New Haven Connecticut USA
| |
Collapse
|
24
|
Panza F, Lozupone M, Solfrizzi V, Sardone R, Dibello V, Di Lena L, D'Urso F, Stallone R, Petruzzi M, Giannelli G, Quaranta N, Bellomo A, Greco A, Daniele A, Seripa D, Logroscino G. Different Cognitive Frailty Models and Health- and Cognitive-related Outcomes in Older Age: From Epidemiology to Prevention. J Alzheimers Dis 2019; 62:993-1012. [PMID: 29562543 PMCID: PMC5870024 DOI: 10.3233/jad-170963] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Frailty, a critical intermediate status of the aging process that is at increased risk for negative health-related events, includes physical, cognitive, and psychosocial domains or phenotypes. Cognitive frailty is a condition recently defined by operationalized criteria describing coexisting physical frailty and mild cognitive impairment (MCI), with two proposed subtypes: potentially reversible cognitive frailty (physical frailty/MCI) and reversible cognitive frailty (physical frailty/pre-MCI subjective cognitive decline). In the present article, we reviewed the framework for the definition, different models, and the current epidemiology of cognitive frailty, also describing neurobiological mechanisms, and exploring the possible prevention of the cognitive frailty progression. Several studies suggested a relevant heterogeneity with prevalence estimates ranging 1.0–22.0% (10.7–22.0% in clinical-based settings and 1.0–4.4% in population-based settings). Cross-sectional and longitudinal population-based studies showed that different cognitive frailty models may be associated with increased risk of functional disability, worsened quality of life, hospitalization, mortality, incidence of dementia, vascular dementia, and neurocognitive disorders. The operationalization of clinical constructs based on cognitive impairment related to physical causes (physical frailty, motor function decline, or other physical factors) appears to be interesting for dementia secondary prevention given the increased risk for progression to dementia of these clinical entities. Multidomain interventions have the potential to be effective in preventing cognitive frailty. In the near future, we need to establish more reliable clinical and research criteria, using different operational definitions for frailty and cognitive impairment, and useful clinical, biological, and imaging markers to implement intervention programs targeted to improve frailty, so preventing also late-life cognitive disorders.
Collapse
Affiliation(s)
- Francesco Panza
- Department of Medical Sciences, Geriatric Unit and Laboratory of Gerontology and Geriatrics, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy.,Department of Basic Medicine, Neurodegenerative Disease Unit, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy.,Department of Clinical Research in Neurology, University of Bari Aldo Moro, "Pia Fondazione Cardinale G. Panico", Tricase, Lecce, Italy
| | - Madia Lozupone
- Department of Basic Medicine, Neurodegenerative Disease Unit, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Vincenzo Solfrizzi
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari "Aldo Moro", Bari, Italy
| | - Rodolfo Sardone
- National Institute of Gastroenterology "Saverio de Bellis", Research Hospital, Castellana Grotte Bari, Italy
| | - Vittorio Dibello
- Interdisciplinary Department of Medicine (DIM), Section of Dentistry, University of Bari Aldo Moro, Bari, Italy
| | - Luca Di Lena
- National Institute of Gastroenterology "Saverio de Bellis", Research Hospital, Castellana Grotte Bari, Italy
| | - Francesca D'Urso
- Department of Clinical and Experimental Medicine, Psychiatric Unit, University of Foggia, Foggia, Italy
| | - Roberta Stallone
- Department of Basic Medicine, Neurodegenerative Disease Unit, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Massimo Petruzzi
- Interdisciplinary Department of Medicine (DIM), Section of Dentistry, University of Bari Aldo Moro, Bari, Italy
| | - Gianluigi Giannelli
- National Institute of Gastroenterology "Saverio de Bellis", Research Hospital, Castellana Grotte Bari, Italy
| | | | - Antonello Bellomo
- Department of Clinical and Experimental Medicine, Psychiatric Unit, University of Foggia, Foggia, Italy
| | - Antonio Greco
- Department of Medical Sciences, Geriatric Unit and Laboratory of Gerontology and Geriatrics, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy
| | - Antonio Daniele
- Institute of Neurology, Catholic University of Sacred Heart, Rome, Italy
| | - Davide Seripa
- Department of Medical Sciences, Geriatric Unit and Laboratory of Gerontology and Geriatrics, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy
| | - Giancarlo Logroscino
- Department of Basic Medicine, Neurodegenerative Disease Unit, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy.,Department of Clinical Research in Neurology, University of Bari Aldo Moro, "Pia Fondazione Cardinale G. Panico", Tricase, Lecce, Italy
| |
Collapse
|
25
|
Farokhnia M, Faulkner ML, Piacentino D, Lee MR, Leggio L. Ghrelin: From a gut hormone to a potential therapeutic target for alcohol use disorder. Physiol Behav 2019; 204:49-57. [DOI: 10.1016/j.physbeh.2019.02.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 01/24/2019] [Accepted: 02/06/2019] [Indexed: 12/22/2022]
|
26
|
Gan ZS, Stein SC, Swanson R, Guan S, Garcia L, Mehta D, Smith DH. Blood Biomarkers for Traumatic Brain Injury: A Quantitative Assessment of Diagnostic and Prognostic Accuracy. Front Neurol 2019; 10:446. [PMID: 31105646 PMCID: PMC6498532 DOI: 10.3389/fneur.2019.00446] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 04/12/2019] [Indexed: 12/18/2022] Open
Abstract
Blood biomarkers have been explored for their potential to provide objective measures in the assessment of traumatic brain injury (TBI). However, it is not clear which biomarkers are best for diagnosis and prognosis in different severities of TBI. Here, we compare existing studies on the discriminative abilities of serum biomarkers for four commonly studied clinical situations: detecting concussion, predicting intracranial damage after mild TBI (mTBI), predicting delayed recovery after mTBI, and predicting adverse outcome after severe TBI (sTBI). We conducted a literature search of publications on biomarkers in TBI published up until July 2018. Operating characteristics were pooled for each biomarker for comparison. For detecting concussion, 4 biomarker panels and creatine kinase B type had excellent discriminative ability. For detecting intracranial injury and the need for a head CT scan after mTBI, 2 biomarker panels, and hyperphosphorylated tau had excellent operating characteristics. For predicting delayed recovery after mTBI, top candidates included calpain-derived αII-spectrin N-terminal fragment, tau A, neurofilament light, and ghrelin. For predicting adverse outcome following sTBI, no biomarker had excellent performance, but several had good performance, including markers of coagulation and inflammation, structural proteins in the brain, and proteins involved in homeostasis. The highest-performing biomarkers in each of these categories may provide insight into the pathophysiologies underlying mild and severe TBI. With further study, these biomarkers have the potential to be used alongside clinical and radiological data to improve TBI diagnostics, prognostics, and evidence-based medical management.
Collapse
Affiliation(s)
- Zoe S Gan
- University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Sherman C Stein
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Randel Swanson
- Department of Physical Medicine and Rehabilitation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Rehabilitation Medicine Service, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States.,Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States.,Department of Neurosurgery, Perelman School of Medicine, Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, PA, United States
| | - Shaobo Guan
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Lizette Garcia
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Devanshi Mehta
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Douglas H Smith
- Department of Neurosurgery, Perelman School of Medicine, Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
27
|
Stutz B, Nasrallah C, Nigro M, Curry D, Liu ZW, Gao XB, Elsworth JD, Mintz L, Horvath TL. Dopamine neuronal protection in the mouse Substantia nigra by GHSR is independent of electric activity. Mol Metab 2019; 24:120-138. [PMID: 30833218 PMCID: PMC6531791 DOI: 10.1016/j.molmet.2019.02.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 02/15/2019] [Accepted: 02/16/2019] [Indexed: 12/25/2022] Open
Abstract
Objective Dopamine neurons in the Substantia nigra (SN) play crucial roles in control of voluntary movement. Extensive degeneration of this neuronal population is the cause of Parkinson's disease (PD). Many factors have been linked to SN DA neuronal survival, including neuronal pacemaker activity (responsible for maintaining basal firing and DA tone) and mitochondrial function. Dln-101, a naturally occurring splice variant of the human ghrelin gene, targets the ghrelin receptor (GHSR) present in the SN DA cells. Ghrelin activation of GHSR has been shown to protect SN DA neurons against 1-methyl-4-phenyl-1,2,5,6 tetrahydropyridine (MPTP) treatment. We decided to compare the actions of Dln-101 with ghrelin and identify the mechanisms associated with neuronal survival. Methods Histologial, biochemical, and behavioral parameters were used to evaluate neuroprotection. Inflammation and redox balance of SN DA cells were evaluated using histologial and real-time PCR analysis. Designer Receptors Exclusively Activated by Designer Drugs (DREADD) technology was used to modulate SN DA neuron electrical activity and associated survival. Mitochondrial dynamics in SN DA cells was evaluated using electron microscopy data. Results Here, we report that the human isoform displays an equivalent neuroprotective factor. However, while exogenous administration of mouse ghrelin electrically activates SN DA neurons increasing dopamine output, as well as locomotion, the human isoform significantly suppressed dopamine output, with an associated decrease in animal motor behavior. Investigating the mechanisms by which GHSR mediates neuroprotection, we found that dopamine cell-selective control of electrical activity is neither sufficient nor necessary to promote SN DA neuron survival, including that associated with GHSR activation. We found that Dln101 pre-treatment diminished MPTP-induced mitochondrial aberrations in SN DA neurons and that the effect of Dln101 to protect dopamine cells was dependent on mitofusin 2, a protein involved in the process of mitochondrial fusion and tethering of the mitochondria to the endoplasmic reticulum. Conclusions Taken together, these observations unmasked a complex role of GHSR in dopamine neuronal protection independent on electric activity of these cells and revealed a crucial role for mitochondrial dynamics in some aspects of this process. Dln101 is a human splice-variant of the ghrelin gene with different expression pattern. Ghrelin and Dln101 display equivalent levels of neuroprotection of SN DA cells. Modulation of electrical activity of SN DA cells is not relevant for neuroprotection. Mitochondrial fusion protein 2 (MFN 2) blocks DLN101-induced mitochondrial fusion in SN DA neurons and prevents DLN101-induced neuroprotection.
Collapse
Affiliation(s)
- Bernardo Stutz
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, USA.
| | - Carole Nasrallah
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, USA; Interdepartmental Neuroscience Program, USA
| | - Mariana Nigro
- Department of Obstetrics, Gynecology and Reproductive Sciences, USA
| | | | - Zhong-Wu Liu
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, USA
| | - Xiao-Bing Gao
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, USA
| | | | | | - Tamas L Horvath
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Department of Comparative Medicine, USA; Interdepartmental Neuroscience Program, USA; Department of Obstetrics, Gynecology and Reproductive Sciences, USA; Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Anatomy and Histology, University of Veterinary Medicine, Budapest, 1078, Hungary.
| |
Collapse
|
28
|
Chen XY, Du YF, Chen L. Neuropeptides Exert Neuroprotective Effects in Alzheimer's Disease. Front Mol Neurosci 2019; 11:493. [PMID: 30687008 PMCID: PMC6336706 DOI: 10.3389/fnmol.2018.00493] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 12/21/2018] [Indexed: 01/03/2023] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder characterized by cognitive deficits and neuronal loss. Deposition of beta-amyloid peptide (Aβ) causes neurotoxicity through the formation of plaques in brains of Alzheimer's disease. Numerous studies have indicated that the neuropeptides including ghrelin, neurotensin, pituitary adenylate cyclase-activating polypeptide (PACAP), neuropeptide Y, substance P and orexin are closely related to the pathophysiology of Alzheimer's disease. The levels of neuropeptides and their receptors change in Alzheimer's disease. These neuropeptides exert neuroprotective roles mainly through preventing Aβ accumulation, increasing neuronal glucose transport, increasing the production of neurotrophins, inhibiting endoplasmic reticulum stress and autophagy, modulating potassium channel activity and hippocampal long-term potentiation. Therefore, the neuropeptides may function as potential drug targets in the prevention and cure of Alzheimer's disease.
Collapse
Affiliation(s)
- Xin-Yi Chen
- Department of Physiology and Pathophysiology, Qingdao University, Qingdao, China.,Department of Neurology, Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Yi-Feng Du
- Department of Neurology, Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Lei Chen
- Department of Physiology and Pathophysiology, Qingdao University, Qingdao, China
| |
Collapse
|
29
|
Ghrelin octanoylation by ghrelin O-acyltransferase: Unique protein biochemistry underlying metabolic signaling. Biochem Soc Trans 2019; 47:169-178. [PMID: 30626708 DOI: 10.1042/bst20180436] [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] [Received: 10/18/2018] [Revised: 11/26/2018] [Accepted: 11/28/2018] [Indexed: 02/08/2023]
Abstract
Ghrelin is a small peptide hormone that requires a unique post-translational modification, serine octanoylation, to bind and activate the GHS-R1a receptor. Ghrelin signaling is implicated in a variety of neurological and physiological processes, but is most well known for its roles in controlling hunger and metabolic regulation. Ghrelin octanoylation is catalyzed by ghrelin O-acyltransferase (GOAT), a member of the membrane-bound O-acyltransferase (MBOAT) enzyme family. From the status of ghrelin as the only substrate for GOAT in the human genome to the source and requirement for the octanoyl acyl donor, the ghrelin-GOAT system is defined by multiple unique aspects within both protein biochemistry and endocrinology. In this review, we examine recent advances in our understanding of the interactions and mechanisms leading to ghrelin modification by GOAT, discuss the potential sources for the octanoyl acyl donor required for ghrelin's activation, and summarize the current landscape of molecules targeting ghrelin octanoylation through GOAT inhibition.
Collapse
|
30
|
Yoshino Y, Funahashi Y, Nakata S, Ozaki Y, Yamazaki K, Yoshida T, Mori T, Mori Y, Ochi S, Iga JI, Ueno SI. Ghrelin cascade changes in the peripheral blood of Japanese patients with Alzheimer's disease. J Psychiatr Res 2018; 107:79-85. [PMID: 30366284 DOI: 10.1016/j.jpsychires.2018.10.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/17/2018] [Accepted: 10/18/2018] [Indexed: 11/26/2022]
Abstract
The neuroprotective effect of ghrelin has recently been reported in Alzheimer's disease (AD). Ghrelin is converted from des-acyl ghrelin to the activated form, acyl ghrelin, by membrane bound o-acyltransferase 4 (MBOAT4), and then binds to growth hormone secretagogue receptor (GHS-R). We examined the levels of plasma acyl/des-acyl ghrelin in 75 AD subjects and age- and sex-matched controls, as well as the DNA methylation and mRNA expression of MBOAT4 and GHS-R in peripheral leukocytes. The acyl ghrelin concentration was significantly higher in AD subjects than in controls (2.18 ± 1.25 vs. 1.49 ± 2.3, p = 0.001). The methylation rate of MBOAT4 CpG 2 was significantly lower in AD subjects than in controls (4.0 ± 0.9 vs. 4.7 ± 1.2, p < 0.001). The mRNA expression levels of MBOAT4 and GHS-R1b were significantly higher in AD subjects than in controls (MBOAT4: 1.10 ± 0.48 vs. 1.0 ± 0.55, p = 0.049; GHS-R1b: 1.76 ± 3.18 vs. 1.0 ± 1.56, p = 0.030). These changes in the ghrelin cascade in peripheral blood may reflect those in the brain, and may be a neuroprotective biomarker in AD.
Collapse
Affiliation(s)
- Yuta Yoshino
- Department of Neuropsychiatry, Molecules and Function, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Yu Funahashi
- Department of Neuropsychiatry, Molecules and Function, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Shunsuke Nakata
- Department of Neuropsychiatry, Molecules and Function, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Yuki Ozaki
- Department of Neuropsychiatry, Molecules and Function, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Kiyohiro Yamazaki
- Department of Neuropsychiatry, Molecules and Function, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Taku Yoshida
- Department of Neuropsychiatry, Molecules and Function, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Takaaki Mori
- Department of Neuropsychiatry, Molecules and Function, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Yoko Mori
- Department of Neuropsychiatry, Molecules and Function, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Shinichiro Ochi
- Department of Neuropsychiatry, Molecules and Function, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Jun-Ichi Iga
- Department of Neuropsychiatry, Molecules and Function, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan.
| | - Shu-Ichi Ueno
- Department of Neuropsychiatry, Molecules and Function, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan
| |
Collapse
|
31
|
Goshadrou F, Arefi Oskouie A, Eslami M, Nobakht Mothlagh Ghoochani BF. Effect of ghrelin on serum metabolites in Alzheimer's disease model rats; a metabolomics studies based on 1H-NMR technique. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2018; 21:1245-1254. [PMID: 30627368 PMCID: PMC6312673 DOI: 10.22038/ijbms.2018.30596.7373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/21/2018] [Indexed: 11/06/2022]
Abstract
OBJECTIVES Alzheimer's disease (AD) is dysfunction of the central nervous system and as a neurodegenerative disease. The objective of this work is to investigate metabolic profiling in the serum of animal models of AD compared to healthy controls and then to peruse the role of ghrelin as a therapeutic approach for the AD. MATERIALS AND METHODS Nuclear magnetic resonance (NMR) technique was used for identification of metabolites that are differentially expressed in the serum of a rat model of the AD with or without ghrelin treatment. Using multivariate statistical analysis, models were built and indicated. RESULTS There were significant differences and high predictive power between AD and ghrelin-treated groups. The area under curve (AUC) of receiver operating characteristic (ROC) curve and Q2 were 0.870 and 0.759, respectively. A biomarker panel consisting of 14 metabolites was identified to discriminate the AD from the control group. Another panel of 12 serum metabolites was used to differentiate AD models from treated models. CONCLUSION Both panels had good agreements with clinical diagnosis. Analysis of the results displayed that ghrelin improved memory and cognitive abilities. Affected pathways by ghrelin included oxidative stress, and osteoporosis pathways and vascular risk factors.
Collapse
Affiliation(s)
- Fatemeh Goshadrou
- Faculty of Paramedical Sciences, Department of Basic Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afsaneh Arefi Oskouie
- Faculty of Paramedical Sciences, Department of Basic Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Eslami
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
32
|
Herrera-Martínez AD, Gahete MD, Sánchez-Sánchez R, Alors-Perez E, Pedraza-Arevalo S, Serrano-Blanch R, Martínez-Fuentes AJ, Gálvez-Moreno MA, Castaño JP, Luque RM. Ghrelin-O-Acyltransferase (GOAT) Enzyme as a Novel Potential Biomarker in Gastroenteropancreatic Neuroendocrine Tumors. Clin Transl Gastroenterol 2018; 9:196. [PMID: 30297816 PMCID: PMC6175927 DOI: 10.1038/s41424-018-0058-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 08/16/2018] [Accepted: 08/17/2018] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES The association between the presence and alterations of the components of the ghrelin system and the development and progression of neuroendocrine tumors (NETs) is still controversial and remains unclear. METHODS Here, we systematically evaluated the expression levels (by quantitative-PCR) of key ghrelin system components of in gastroenteropancreatic (GEP)-NETs, as compared to non-tumor adjacent (NTA; n = 42) and normal tissues (NT; n = 14). Then, we analyzed their putative associations with clinical-histological characteristics. RESULTS The results indicate that ghrelin and its receptor GHSR1a are present in a high proportion of normal tissues, while the enzyme ghrelin-O-acyltransferase (GOAT) and the splicing variants In1-ghrelin and GHSR1b were present in a lower proportion of normal tissues. In contrast, all ghrelin system components were present in a high proportion of tumor and NTA tissues. GOAT was significantly overexpressed (by quantitative-PCR (qPCR)) in tumor samples compared to NTA, while a trend was found for ghrelin, In1-ghrelin and GHSR1a. In addition, expression of these components displayed significant correlations with key clinical parameters. The marked overexpression of GOAT in tumor samples compared to NTA regions was confirmed by IHC, revealing that this enzyme is particularly overexpressed in gastrointestinal NETs, where it is directly correlated with tumor diameter. CONCLUSIONS These results provide novel information on the presence and potential pathophysiological implications of the ghrelin system components in GEP-NETs, wherein GOAT might represent a novel diagnostic biomarker.
Collapse
Affiliation(s)
- Aura D Herrera-Martínez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Endocrinology and Nutrition Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Manuel D Gahete
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain
| | - Rafael Sánchez-Sánchez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Pathology Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Emilia Alors-Perez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain
| | - Sergio Pedraza-Arevalo
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain
| | - Raquel Serrano-Blanch
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Medical Oncology Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Antonio J Martínez-Fuentes
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain
| | - Maria A Gálvez-Moreno
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain. .,Endocrinology and Nutrition Service, Reina Sofia University Hospital, Córdoba, Spain.
| | - Justo P Castaño
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain. .,Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain. .,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain. .,Reina Sofia University Hospital, Córdoba, Spain.
| | - Raúl M Luque
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain. .,Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain. .,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain. .,Reina Sofia University Hospital, Córdoba, Spain.
| |
Collapse
|
33
|
Ghrelin agonist HM01 attenuates chemotherapy-induced neurotoxicity in rodent models. Eur J Pharmacol 2018; 840:89-103. [PMID: 30268665 DOI: 10.1016/j.ejphar.2018.09.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 09/25/2018] [Accepted: 09/26/2018] [Indexed: 12/22/2022]
Abstract
Chemotherapy-Induced Peripheral Neurotoxicity (CIPN) is often dose-limiting and impacts life quality and survival of cancer patients. Ghrelin agonists have neuroprotectant effects and may have a role in treating or preventing CIPN. We evaluated the CNS-penetrant ghrelin agonist HM01 in three experimental models of CIPN at doses of 3-30 mg/kg p.o. daily monitoring orexigenic properties, nerve conduction, mechanical allodynia, and intra-epidermal nerve fiber density (IENFD). In a cisplatin-based study, rats were dosed daily for 3 days (0.5 mg/kg i.p.) + HM01. Cisplatin treatment induced mechanical hypersensitivity which was significantly reduced by HM01. In a second study, oxaliplatin was administered to mice (6 mg/kg i.p. 3 times/week for 4 weeks) resulting in significant digital nerve conduction velocity (NCV) deficits and reduction of IENFD. Concurrent HM01 dose dependently prevented the decline in NCV and attenuated the reduction in IENFD. Pharmacokinetic studies showed HM01 accumulation in the dorsal root ganglia and sciatic nerves which reached concentrations > 10 fold that of plasma. In a third model, HM01 was tested in preventive and therapeutic paradigms in a bortezomib-based rat model (0.2 mg/kg i.v., 3 times/week for 8 weeks). In the preventive setting, HM01 blocked bortezomib-induced hyperalgesia and IENFD reduction at all doses tested. In the therapeutic setting, significant effect was observed, but only at the highest dose. Altogether, the robust peripheral nervous system penetration of HM01 and its ability to improve multiple oxaliplatin-, cisplatin-, and bortezomib-induced neurotoxicities suggest that HM01 may be a useful neuroprotective adjuvant for CIPN.
Collapse
|
34
|
Eslami M, Sadeghi B, Goshadrou F. Chronic ghrelin administration restores hippocampal long-term potentiation and ameliorates memory impairment in rat model of Alzheimer's disease. Hippocampus 2018; 28:724-734. [PMID: 30009391 DOI: 10.1002/hipo.23002] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 06/09/2018] [Accepted: 06/16/2018] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease (AD), as a common age-related dementia, is a progressive manifestation of cognitive decline following synaptic failure resulted majorly by senile plaques composed of deposits of amyloid beta (Aβ). Ghrelin is a multifunctional peptide hormone with receptors present in various brain tissues including hippocampus and has been associated with neuroprotection, neuromodulation, and memory processing. Here, we investigated the neuroprotective and therapeutic effects of intracerebroventricular (icv) ghrelin infusion for 2 weeks on passive avoidance learning (PAL), memory retention, and synaptic plasticity in the hippocampal dentate gyrus (DG) and CA1 of both normal rats and Aβ1-42-induced neurotoxicity in AD model. Male Wistar rats were evaluated for their passive memory performance using a shuttle box while some groups had already received Aβ1-42 and/or chronic ghrelin. Using field potential recording, the induction of short- and long-term potentiation (STP and LTP) was studied in DG granule cells along with the LTP changes in CA1 pyramidal neurons through stimulation of the medial perforant path (mPP) and Schaffer collaterals (SCs), respectively. Our results demonstrated that chronic ghrelin treatment not only improved memory processing and retrieval in normal rats during the PAL task, but also promoted memory retention and alleviated memory loss by amelioration of Aβ1-42-induced synaptic plasticity impairment in AD subjects through augmentation of field excitatory postsynaptic potential (fEPSP) slope that led to LTP restitution in both the mPP-DG and the CA3-CA1 synapses. Meanwhile, STP was not significantly changed, meaning that although ghrelin enhanced postsynaptic excitability in DG, it did not change presynaptic transmitter release significantly. This suggests the involvement of postsynaptic mechanisms in long-term ghrelin-enhanced memory. In conclusion, it can be inferred that chronic ghrelin administration has an auspicious therapeutic value for impaired cognitive performance and memory deficits in AD-like neuropathology.
Collapse
Affiliation(s)
- Maryam Eslami
- Department of Physiology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahman Sadeghi
- Department of Physiology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Biochemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Fatemeh Goshadrou
- Department of Physiology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
35
|
Mir JF, Zagmutt S, Lichtenstein MP, García-Villoria J, Weber M, Gracia A, Fabriàs G, Casas J, López M, Casals N, Ribes A, Suñol C, Herrero L, Serra D. Ghrelin Causes a Decline in GABA Release by Reducing Fatty Acid Oxidation in Cortex. Mol Neurobiol 2018; 55:7216-7228. [PMID: 29396649 PMCID: PMC6096967 DOI: 10.1007/s12035-018-0921-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 01/21/2018] [Indexed: 10/26/2022]
Abstract
Lipid metabolism, specifically fatty acid oxidation (FAO) mediated by carnitine palmitoyltransferase (CPT) 1A, has been described to be an important actor of ghrelin action in hypothalamus. However, it is not known whether CPT1A and FAO mediate the effect of ghrelin on the cortex. Here, we show that ghrelin produces a differential effect on CPT1 activity and γ-aminobutyric acid (GABA) metabolism in the hypothalamus and cortex of mice. In the hypothalamus, ghrelin enhances CPT1A activity while GABA transaminase (GABAT) activity, a key enzyme in GABA shunt metabolism, is unaltered. However, in cortex CPT1A activity and GABAT activity are reduced after ghrelin treatment. Furthermore, in primary cortical neurons, ghrelin reduces GABA release through a CPT1A reduction. By using CPT1A floxed mice, we have observed that genetic ablation of CPT1A recapitulates the effect of ghrelin on GABA release in cortical neurons, inducing reductions in mitochondrial oxygen consumption, cell content of citrate and α-ketoglutarate, and GABA shunt enzyme activity. Taken together, these observations indicate that ghrelin-induced changes in CPT1A activity modulate mitochondrial function, yielding changes in GABA metabolism. This evidence suggests that the action of ghrelin on GABA release is region specific within the brain, providing a basis for differential effects of ghrelin in the central nervous system.
Collapse
Affiliation(s)
- Joan Francesc Mir
- Department of Biochemistry and Physiology, Facultat de Farmàcia i Ciències de l'Alimentació and Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Av. Joan XXIII, 27-30, E-08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain
| | - Sebastián Zagmutt
- Department of Biochemistry and Physiology, Facultat de Farmàcia i Ciències de l'Alimentació and Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Av. Joan XXIII, 27-30, E-08028, Barcelona, Spain
| | - Mathieu P Lichtenstein
- Institut d'Investigacions Biomèdiques de Barcelona, Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Judit García-Villoria
- Sección de Errores Congénitos del Metabolismo - IBC, Servicio de Bioquímica y Genética Molecular, Hospital Clínic, IDIBAPS, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Minéia Weber
- Department of Biochemistry and Physiology, Facultat de Farmàcia i Ciències de l'Alimentació and Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Av. Joan XXIII, 27-30, E-08028, Barcelona, Spain
| | - Ana Gracia
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain
- Nutrition and Food Science Area, School of Pharmacy, Universidad del País Vasco/Euskal Herriko Unibersitatea, Leioa, Spain
| | - Gemma Fabriàs
- Research Unit on BioActive Molecules, Department of Biomedicinal Chemistry, Institute of Advanced Chemistry of Catalonia (IQAC)/CSIC, Barcelona, Spain
| | - Josefina Casas
- Research Unit on BioActive Molecules, Department of Biomedicinal Chemistry, Institute of Advanced Chemistry of Catalonia (IQAC)/CSIC, Barcelona, Spain
| | - Miguel López
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782, Santiago de Compostela, Spain
- Departament de Ciències Bàsiques, Facultat de Medicina i Ciències de la Salut, Universitat Internacional de Catalunya (UIC), Barcelona, Spain
| | - Núria Casals
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain
- Departament de Ciències Bàsiques, Facultat de Medicina i Ciències de la Salut, Universitat Internacional de Catalunya (UIC), Barcelona, Spain
| | - Antònia Ribes
- Sección de Errores Congénitos del Metabolismo - IBC, Servicio de Bioquímica y Genética Molecular, Hospital Clínic, IDIBAPS, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Cristina Suñol
- Institut d'Investigacions Biomèdiques de Barcelona, Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, Facultat de Farmàcia i Ciències de l'Alimentació and Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Av. Joan XXIII, 27-30, E-08028, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain
| | - Dolors Serra
- Department of Biochemistry and Physiology, Facultat de Farmàcia i Ciències de l'Alimentació and Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Av. Joan XXIII, 27-30, E-08028, Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain.
| |
Collapse
|
36
|
Farokhnia M, Lee MR, Farinelli LA, Ramchandani VA, Akhlaghi F, Leggio L. Pharmacological manipulation of the ghrelin system and alcohol hangover symptoms in heavy drinking individuals: Is there a link? Pharmacol Biochem Behav 2018; 172:39-49. [PMID: 30030128 DOI: 10.1016/j.pbb.2018.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 06/23/2018] [Accepted: 07/16/2018] [Indexed: 12/14/2022]
Abstract
Ghrelin, an orexigenic peptide synthesized in the stomach, is a key player in the gut-brain axis. In addition to its role in regulating food intake and energy homeostasis, ghrelin has been shown to modulate alcohol-related behaviors. Alcohol consumption frequently results in hangover, an underexplored phenomenon with considerable medical, psychological, and socioeconomic consequences. While the pathophysiology of hangover is not clear, contributions of mechanisms such as alcohol-induced metabolic/endocrine changes, inflammatory/immune response, oxidative stress, and gut dysbiosis have been reported. Interestingly, these mechanisms considerably overlap with ghrelin's physiological functions. Here, we investigated whether pharmacological manipulation of the ghrelin system may affect alcohol hangover symptoms. Data were obtained from two placebo-controlled laboratory studies. The first study tested the effects of intravenous (IV) ghrelin and consisted of two experiments: a progressive-ratio IV alcohol self-administration (IV-ASA) and a fixed-dose IV alcohol clamp. The second study tested the effects of an oral ghrelin receptor inverse agonist (PF-5190457) and included a fixed-dose oral alcohol administration experiment. Alcohol hangover data were collected the morning after each alcohol administration experiment using the Acute Hangover Scale (AHS). IV ghrelin, compared to placebo, significantly reduced alcohol hangover after IV-ASA (p = 0.04) and alcohol clamp (p = 0.04); PF-5190457 had no significant effect on AHS scores. Females reported significantly higher hangover symptoms than males following the IV-ASA experiment (p = 0.04), but no gender × drug condition (ghrelin vs. placebo) effect was found. AHS total scores were positively correlated with peak subjective responses, including 'stimulation' (p = 0.08), 'sedation' (p = 0.009), 'feel high' (p = 0.05), and 'feel intoxicated' (p = 0.03) during the IV-ASA. IV ghrelin blunted the positive association between alcohol sedation and hangover as shown by trend-level drug × sedation effect (p = 0.08). This is the first study showing that exogenous ghrelin administration, but not ghrelin receptor inverse agonism, affects hangover symptoms. Future research should investigate the potential mechanism(s) underlying this effect.
Collapse
Affiliation(s)
- Mehdi Farokhnia
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Mary R Lee
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Lisa A Farinelli
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Vijay A Ramchandani
- Section on Human Psychopharmacology, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Fatemeh Akhlaghi
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Lorenzo Leggio
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA; Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA.
| |
Collapse
|
37
|
MK-0677, a Ghrelin Agonist, Alleviates Amyloid Beta-Related Pathology in 5XFAD Mice, an Animal Model of Alzheimer's Disease. Int J Mol Sci 2018; 19:ijms19061800. [PMID: 29912176 PMCID: PMC6032329 DOI: 10.3390/ijms19061800] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 05/31/2018] [Accepted: 06/06/2018] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder characterized by cognitive deficits, neuroinflammation, and neuronal death. The primary pathogenic cause is believed to be the accumulation of pathogenic amyloid beta (Aβ) assemblies in the brain. Ghrelin, which is a peptide hormone predominantly secreted from the stomach, is an endogenous ligand for the growth hormone secretagogue-receptor type 1a (GHS-R1a). MK-0677 is a ghrelin agonist that potently stimulates the GHS-R1a ghrelin receptor. Interestingly, previous studies have shown that ghrelin improves cognitive impairments and attenuates neuronal death and neuroinflammation in several neurological disorders. However, it is unknown whether MK-0677 can affect Aβ accumulation or Aβ-mediated pathology in the brains of patients with AD. Therefore, we examined the effects of MK-0677 administration on AD-related pathology in 5XFAD mice, an Aβ-overexpressing transgenic mouse model of AD. MK-0677 was intraperitoneally administered to three-month-old 5XFAD mice. To visualize Aβ accumulation, neuroinflammation, and neurodegeneration, thioflavin-S staining and immunostaining with antibodies against Aβ (4G8), ionized calcium-binding adaptor molecule 1 (Iba-1), glial fibrillary acidic protein (GFAP), neuronal nuclear antigen (NeuN), and synaptophysin were conducted in the neocortex of 5XFAD and wild-type mice, and to evaluate changes of phosphorylated cyclic adenosine monophosphate (cAMP) response element binding protein (pCREB) levels, immunostaining with antibody against pCREB was performed in dentate gyrus of the hippocampus of 5XFAD and wild-type mice. The histological analyses indicated that MK-0677-treated 5XFAD mice showed reduced Aβ deposition, gliosis, and neuronal and synaptic loss in the deep cortical layers, and inhibited the decrement of pCREB levels in dentate gyrus of the hippocampus compared to vehicle-treated 5XFAD mice. Our results showed that activation of the ghrelin receptor with MK-0677 inhibited the Aβ burden, neuroinflammation, and neurodegeneration, which suggested that MK-0677 might have potential as a treatment of the early phase of AD.
Collapse
|
38
|
Çeri V, Aykutlu HC, Görker I, Akça ÖF, Tarakçıoğlu MC, Aksoy UM, Kaya H, Sertdemir M, İnce E, Kadak MT, Yalçın GY, Guliyev C, Bilgiç A, Çiftçi E, Tekin K, Tuna ZO, Oğuzdoğan B, Duman NS, Semerci B, Üneri ÖŞ, Karabekiroglu K, Mutluer T, Nebioglu M, Başgül ŞS, Naharcı Mİ, Maden Ö, Hocaoğlu Ç, Durmaz O, Usta H, Boşgelmez Ş, Puşuroğlu M, Eser HY, Kaçar M, Çakır M, Karatepe HT, Işık Ü, Kara H, Yeloğlu ÇH, Yazıcı E, Gündüz A, Karataş KS, Yavlal F, Uzun N, Yazici AB, Bodur Ş, Aslan EA, Batmaz S, Çelik F, Açıkel SB, Topal Z, Altunsoy N, Tulacı ÖD, Demirel ÖF, Çıtak S, Çak HT, Artık AB, Özçetin A, Özdemir I, Çelik FGH, Kültür SEÇ, Çipil A, Ay R, Arman AR, Yazıcı KU, Yuce AE, Yazıcı İP, Kurt E, Kaçar AŞ, Erbil N, Poyraz CA, Altın GE, Şahin B, Kılıç Ö, Turan Ş, Aydın M, Kuru E, Bozkurt A, Güleç H, İnan MY, Şevik AE, Baykal S, Karaer Y, Yanartaş O, Aksu H, Ergün S, Görmez A, Yıldız M, Bag S, Özkanoğlu FK, Caliskan M, Yaşar AB, Konuk E, Altın M, Bulut S, Bulut GÇ, Tulacı RG, Küpeli NY, Enver N, Tasci İ, Kani AS, Bahçeci B, Oğuz G, Şenyuva G, Ünal GT, Yektaş Ç, Örüm MH, Göka E, Gıca Ş, Şahmelikoğlu Ö, Dinç GŞ, Erşan S, Erşan E, Ceylan MF, Hesapçıoğlu ST, Solmaz M, Balcioglu YH, Cetin M, Tosun M, Yurteri N, Ulusoy S, Karadere ME, Kivrak Y, Görmez V. Symposium Oral Presentations. PSYCHIAT CLIN PSYCH 2018. [DOI: 10.1080/24750573.2018.1464274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Affiliation(s)
- Veysi Çeri
- Marmara University Pendik Research and Training Hospital, Child and Adolescent Psychiatry Clinic, Istanbul, Turkey
| | - Hasan Cem Aykutlu
- Department of Child and Adolescent Psychiatry, Trakya University School of Medicine, Edirne, Turkey
| | - Işık Görker
- Department of Child and Adolescent Psychiatry, Trakya University School of Medicine, Edirne, Turkey
| | - Ömer Faruk Akça
- Department of Child and Adolescent Psychiatry, Necmettin Erbakan University Meram School of Medicine, Konya, Turkey
| | - Mahmut Cem Tarakçıoğlu
- Health Sciences University Kanuni Sultan Süleyman Research and Training Hospital, Istanbul, Turkey
| | - Umut Mert Aksoy
- Health Sciences University Kanuni Sultan Süleyman Research and Training Hospital, Istanbul, Turkey
| | - Heysem Kaya
- Department of Computer Engineering, Çorlu Faculty of Engineering, Namık Kemal University, Çorlu, Tekirdağ, Turkey
| | - Merve Sertdemir
- Department of Child and Adolescent Psychiatry, Necmettin Erbakan University Meram School of Medicine, Konya, Turkey
| | - Ezgi İnce
- Department of Psychiatry, Istanbul University Istanbul School of Medicine, Istanbul, Turkey
| | - Muhammed Tayyib Kadak
- Department of Child and Adolescent Psychiatry, Istanbul University Cerrahpaşa School of Medicine, Istanbul, Turkey
| | | | | | - Ayhan Bilgiç
- Department of Child and Adolescent Psychiatry, Necmettin Erbakan University Meram School of Medicine, Konya, Turkey
| | - Elvan Çiftçi
- Department of Psychiatry, Erenkoy Research and Training Hospital, Istanbul, Turkey
| | | | | | | | | | - Bengi Semerci
- Department of Psychology, Hasan Kalyoncu University, Gaziantep, Turkey
| | - Özden Şükran Üneri
- Department of Child and Adolescent Psychiatry, Yıldırım Beyazıt University School of Medicine, Ankara, Turkey
| | | | - Tuba Mutluer
- Koç University Hospital, Department of Child and Adolescent Psychiatry, Istanbul, Turkey
| | - Melike Nebioglu
- Health Sciences University, Haydarpaşa Numune Research and Training Hospital, Istanbul, Turkey
| | | | - Mehmet İlkin Naharcı
- Division of Geriatrics, Department of Internal Medicine, Health Sciences University, Ankara, Turkey
| | - Özgür Maden
- SBÜ Sultan Abdülhamid Han Education and Training Hospital, Department of Psychiatry, Istanbul, Turkey
| | - Çiçek Hocaoğlu
- Department of Psychiatry, Recep Tayyip Erdogan University School of Medicine, Rize, Turkey
| | - Onur Durmaz
- Erenköy Mental Health and Neurology Research and Training Hospital, Department of Psychiatry, Istanbul, Turkey
| | - Haluk Usta
- Erenköy Mental Health and Neurology Research and Training Hospital, Department of Psychiatry, Istanbul, Turkey
| | - Şükriye Boşgelmez
- Kocaeli Derince Research and Training Hospital, Psychiatry Clinic, Kocaeli, Turkey
| | | | - Hale Yapıcı Eser
- KOÇ University School of Medicine, Istanbul, Turkey
- KOÇ University Research Center FOR Translational Medicine (Kuttam), Istanbul, Turkey
- Koç University School of Medicine Department of Psychiatry, Istanbul, Turkey
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Murat Kaçar
- Department of Child and Adolescent Psychiatry, Recep Tayyip Erdogan University School of Medicine, Rize, Turkey
| | - Mahmut Çakır
- Child Psychiatry Clinic, Health Sciences University, Amasya Research and Training Hospital, Amasya, Turkey
| | - Hasan Turan Karatepe
- Department of Psychiatry, Istanbul Medeniyet University, School of Medicine, Istanbul, Turkey
| | - Ümit Işık
- Department of Child and Adolescent Psychiatry, Yozgat State Hospital, Yozgat, Turkey
| | - Halil Kara
- Department of Child and Adolescent Psychiatry, Aksaray University Research and Training Hospital, Aksaray, Turkey
| | | | - Esra Yazıcı
- Department of Psychiatry, Sakarya University School of Medicine, Sakarya, Turkey
| | - Anıl Gündüz
- Health Sciences University, Haydarpaşa Numune Research and Training Hospital, Istanbul, Turkey
| | - Kader Semra Karataş
- Recep Tayyip Erdogan University School of Medicine Psychiatry Department, Rize, Turkey
| | - Figen Yavlal
- Department of Neurology, School of Medicine, Bahcesehir University, Istanbul, Turkey
- Department of Neurology, Bahcesehir University School of Medicine, Istanbul, Turkey
| | - Necati Uzun
- Department of Child and Adolescent Psychiatry, Elazığ Psychiatry Hospital, Elazığ, Turkey
| | - Ahmet Bulent Yazici
- Department of Psychiatry, Sakarya University School of Medicine, Sakarya, Turkey
| | - Şahin Bodur
- Health Sciences University, Gulhane Research and Training Hospital, Child and Adolescent Psychiatry Clinic, Ankara, Turkey
| | - Esma Akpınar Aslan
- Department of Psychiatry, Gaziosmanpaşa University School of Medicine, Tokat, Turkey
| | - Sedat Batmaz
- Department of Psychiatry, Gaziosmanpasa University School of Medicine, Tokat, Turkey
| | - Feyza Çelik
- Department of Psychiatry, Dumlupınar University School of Medicine, Evliya Çelebi Research and Training Hospital, Kütahya, Turkey
| | - Sadettin Burak Açıkel
- Dr. Sami Ulus Research and Training Hospital, Child and Adolescent Psychiatry Department, Ankara, Turkey
| | | | | | | | - Ömer Faruk Demirel
- Department of Psychiatry, Cerrahpaşa Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Serhat Çıtak
- Department of Psychiatry, Istanbul Medeniyet University, School of Medicine, Istanbul, Turkey
| | - Halime Tuna Çak
- Department of Child and Adolescent Psychiatry, Hacettepe University School of Medicine, Ankara, Turkey
| | - Abdül Baki Artık
- Department of Child and Adolescent Psychiatry, Hacettepe University School of Medicine, Ankara, Turkey
| | - Adnan Özçetin
- Department of Psychiatry, Duzce University School of Medicine, Duzce, Turkey
| | - Ilker Özdemir
- Giresun University Prof. Dr. A. İlhan Özdemir Research and Training Hospital, Giresun, Turkey
| | | | | | - Arif Çipil
- Health Sciences University, Haydarpaşa Numune Research and Training Hospital, Istanbul, Turkey
| | - Rukiye Ay
- Malatya Research and Training Hospital, Malatya, Turkey
| | - Ayşe Rodopman Arman
- Department of Child and Adolescent Psychiatry, Marmara University School of Medicine, Istanbul
| | - Kemal Utku Yazıcı
- Department of Child and Adolescent Psychiatry, Firat University School of Medicine, Elazig, Turkey
| | | | - İpek Perçinel Yazıcı
- Department of Child and Adolescent Psychiatry, Firat University School of Medicine, Elazig, Turkey
| | - Emel Kurt
- Psychiatry Clinic, Hisar Intercontinental Hospital, Istanbul, Turkey
| | - Anıl Şafak Kaçar
- Koc University, Research Center for Translational Medicine, Istanbul, Turkey
| | - Nurhan Erbil
- Department of Biophysics, Hacettepe University School of Medicine, Ankara, Turkey
| | - Cana Aksoy Poyraz
- Department of Psychiatry, Istanbul University Cerrahpaşa School of Medicine, Istanbul, Turkey
| | | | - Berkan Şahin
- Iğdır State Hospital, Child and Adolescent Psychiatry Clinic, Iğdır, Turkey
| | - Özge Kılıç
- Department of Psychiatry, Koç University Hospital, Istanbul, Turkey
| | - Şenol Turan
- Department of Psychiatry, Istanbul University Cerrahpaşa School of Medicine, Istanbul, Turkey
| | - Memduha Aydın
- Department of Psychiatry, Selçuk University School of Medicine, Konya, Turkey
| | - Erkan Kuru
- Özel Boylam Psychiatry Hospital, Ankara, Turkey
| | - Abdullah Bozkurt
- Department of Child and Adolescent Psychiatry, Konya Research and Training Hospital, Konya, Turkey
| | - Hüseyin Güleç
- Erenköy Mental Health and Neurology Research and Training Hospital, Department of Psychiatry, Istanbul, Turkey
| | | | - Ali Emre Şevik
- Department of Psychiatry, Çanakkale 18 Mart University School of Medicine, Çanakkale, Türkiye
| | - Saliha Baykal
- Department of Child and Adolescent Psychiatry, Namık Kemal University School of Medicine, Tekirdağ, Turkey
| | - Yusuf Karaer
- Department of Child and Adolescent Psychiatry, Hacettepe University School of Medicine, Ankara, Turkey
| | - Omer Yanartaş
- Department of Psychiatry, Marmara Medical School, Istanbul, Turkiye
| | - Hatice Aksu
- Department of Child and Adolescent Psychiatry, Adnan Menderes University School of Medicine, Aydın, Turkey
| | - Serhat Ergün
- Department of Psychiatry, Marmara University Pendik Research and Training Hospital, Istanbul, Turkey
| | - Aynur Görmez
- Department of Child and Adolescent Psychiatry, Istanbul Medeniyet University School of Medicine, Istanbul, Turkey
| | - Mesut Yıldız
- Department of Psychiatry, School of Medicine, Marmara University, Istanbul, Turkey
| | - Sevda Bag
- Bakirkoy Research and Training Hospital for Psychiatry, Neurology and Neurosurgery, Istanbul, Turkey
| | | | - Mecit Caliskan
- Health Sciences University, Haydarpaşa Numune Research and Training Hospital, Istanbul, Turkey
| | - Alişan Burak Yaşar
- Health Sciences University, Haydarpaşa Numune Research and Training Hospital, Istanbul, Turkey
- Behavioral Sciences Institute, Istanbul, Turkey
| | - Emre Konuk
- Health Sciences University, Haydarpaşa Numune Research and Training Hospital, Istanbul, Turkey
- Behavioral Sciences Institute, Istanbul, Turkey
| | - Murat Altın
- Istinye University Hospital, Psychiatry Clinic, Istanbul, Turkey
| | - Serkut Bulut
- Psychiatry Clinic, Health Sciences University Sakarya Research and Training Hospital, Sakarya, Turkey
| | | | - Rıza Gökçer Tulacı
- Uşak University School of Medicine Research and Training Hospital, Uşak, Turkey
| | - Neşe Yorguner Küpeli
- Department of Psychiatry, Marmara University Pendik Research and Training Hospital, Istanbul, Turkey
| | - Necati Enver
- Department of Otolaryngology, Marmara University Pendik Research and Training Hospital, Istanbul, Turkey
| | - İlker Tasci
- Health Sciences University, Gulhane School of Medicine, Department of Internal Medicine, Ankara, Turkey
| | - Ayşe Sakallı Kani
- Marmara University Pendik Research and Training Hospital, Istanbul, Turkey
| | - Bülent Bahçeci
- Department of Psychiatry, Recep Tayyip Erdogan University, Rize, Turkey
| | | | | | - Gülşen Teksin Ünal
- Bakirkoy Prof. Dr. Mazhar Osman Research and Training Hospital for Psychiatry, Neurology, and Neurosurgery, Istanbul, Turkey
| | - Çiğdem Yektaş
- Duzce University School of Medicine, Department of Child and Adolescent Psychiatry, Duzce, Turkey
| | - Mehmet Hamdi Örüm
- Department of Psychiatry, Adiyaman University School of Medicine, Adiyaman, Turkey
| | - Erol Göka
- SBÜ Ankara Numune Eğitim ve Araştırma Hastanesi
| | - Şakir Gıca
- Bakirkoy Prof. Dr. Mazhar Osman Research and Training Hospital for Psychiatry, Neurology, and Neurosurgery, Istanbul, Turkey
| | - Özge Şahmelikoğlu
- Bakirkoy Prof. Dr. Mazhar Osman Research and Training Hospital for Psychiatry, Neurology, and Neurosurgery, Istanbul, Turkey
| | - Gülser Şenses Dinç
- Department of Child and Adolescent Psychiatry, Ankara Children’s Hematology Oncology Research and Training Hospital, Ankara Turkey
| | - Serpil Erşan
- Cumhuriyet University Advanced Technology Research and Application Center, Sivas, Turkey
| | - Erdal Erşan
- Sivas Numune Hospital, Community Mental Health Center, Sivas, Turkey
| | - Mehmet Fatih Ceylan
- Department of Child and Adolescent Psychiatry, Yıldırım Beyazıt University School of Medicine, Ankara, Turkey
| | - Selma Tural Hesapçıoğlu
- Department of Child and Adolescent Psychiatry, Yıldırım Beyazıt University School of Medicine, Ankara, Turkey
| | - Mustafa Solmaz
- Health Sciences University Bagcilar Research and Training Hospital, Department of Psychiatry, Istanbul, Turkey
- Bakirkoy Prof. Mazhar Osman Training and Research Hospital for Psychiatry, Neurology, and Neurosurgery, Forensic Psychiatry Unit, Istanbul, Turkey
| | - Yasin Hasan Balcioglu
- Health Sciences University Bagcilar Research and Training Hospital, Department of Psychiatry, Istanbul, Turkey
- Bakirkoy Prof. Mazhar Osman Training and Research Hospital for Psychiatry, Neurology, and Neurosurgery, Forensic Psychiatry Unit, Istanbul, Turkey
| | | | - Musa Tosun
- Istanbul University Cerrahpaşa School of Medicine, Department of Child and Adolescent Psychiatry, Istanbul, Turkey
| | - Nihal Yurteri
- Duzce University School of Medicine, Department of Child and Adolescent Psychiatry, Duzce, Turkey
| | - Sevinc Ulusoy
- Bakirkoy Prof. Dr. Mazhar Osman Research and Training Hospital for Psychiatry and Neurology, Istanbul, Turkey
| | | | - Yüksel Kivrak
- Department of Psychiatry, Kafkas University School of Medicine, Kars, Turkey
| | - Vahdet Görmez
- Bezmialem Vakif University, Department of Child and Adolescent Psychiatry, Istanbul, Turkey
| |
Collapse
|
39
|
Cleverdon ER, McGovern-Gooch KR, Hougland JL. The octanoylated energy regulating hormone ghrelin: An expanded view of ghrelin's biological interactions and avenues for controlling ghrelin signaling. Mol Membr Biol 2017; 33:111-124. [PMID: 29143554 DOI: 10.1080/09687688.2017.1388930] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Ghrelin is a small peptide hormone that requires a unique post-translational modification, serine octanoylation, to bind and activate the GHS-R1a receptor. Initially demonstrated to stimulate hunger and appetite, ghrelin-dependent signaling is implicated in a variety of neurological and physiological processes influencing diseases such as diabetes, obesity, and Prader-Willi syndrome. In addition to its cognate receptor, recent studies have revealed ghrelin interacts with a range of binding partners within the bloodstream. Defining the scope of ghrelin's interactions within the body, understanding how these interactions work in concert to modulate ghrelin signaling, and developing molecular tools for controlling ghrelin signaling are essential for exploiting ghrelin for therapeutic effect. In this review, we discuss recent findings regarding the biological effects of ghrelin signaling, outline binding partners that control ghrelin trafficking and stability in circulation, and summarize the current landscape of inhibitors targeting ghrelin octanoylation.
Collapse
Affiliation(s)
| | | | - James L Hougland
- a Department of Chemistry , Syracuse University , Syracuse , NY , USA
| |
Collapse
|
40
|
Higgs S, Spetter MS, Thomas JM, Rotshtein P, Lee M, Hallschmid M, Dourish CT. Interactions between metabolic, reward and cognitive processes in appetite control: Implications for novel weight management therapies. J Psychopharmacol 2017; 31:1460-1474. [PMID: 29072515 PMCID: PMC5700796 DOI: 10.1177/0269881117736917] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Traditional models of appetite control have emphasised the role of parallel homeostatic and hedonic systems, but more recently the distinction between independent homeostatic and hedonic systems has been abandoned in favour of a framework that emphasises the cross talk between the neurochemical substrates of the two systems. In addition, evidence has emerged more recently, that higher level cognitive functions such as learning, memory and attention play an important role in everyday appetite control and that homeostatic signals also play a role in cognition. Here, we review this evidence and present a comprehensive model of the control of appetite that integrates cognitive, homeostatic and reward mechanisms. We discuss the implications of this model for understanding the factors that may contribute to disordered patterns of eating and suggest opportunities for developing more effective treatment approaches for eating disorders and weight management.
Collapse
Affiliation(s)
- Suzanne Higgs
- 1 School of Psychology, University of Birmingham, Birmingham, UK
| | | | - Jason M Thomas
- 2 Department of Psychology, Aston University, Birmingham, UK
| | - Pia Rotshtein
- 1 School of Psychology, University of Birmingham, Birmingham, UK
| | - Michelle Lee
- 3 Department of Psychology, Swansea University, Swansea, UK
| | - Manfred Hallschmid
- 4 Institute for Medical Psychology and Behavioural Neurobiology, University Tübingen, Tübingen, Germany
- 6 Institute for Diabetes Research and Metabolic Diseases, University of Tübingen, Tübingen, Germany
| | | |
Collapse
|
41
|
Westfall S, Lomis N, Kahouli I, Dia SY, Singh SP, Prakash S. Microbiome, probiotics and neurodegenerative diseases: deciphering the gut brain axis. Cell Mol Life Sci 2017; 74:3769-3787. [PMID: 28643167 PMCID: PMC11107790 DOI: 10.1007/s00018-017-2550-9] [Citation(s) in RCA: 339] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 05/05/2017] [Accepted: 05/29/2017] [Indexed: 02/07/2023]
Abstract
The gut microbiota is essential to health and has recently become a target for live bacterial cell biotherapies for various chronic diseases including metabolic syndrome, diabetes, obesity and neurodegenerative disease. Probiotic biotherapies are known to create a healthy gut environment by balancing bacterial populations and promoting their favorable metabolic action. The microbiota and its respective metabolites communicate to the host through a series of biochemical and functional links thereby affecting host homeostasis and health. In particular, the gastrointestinal tract communicates with the central nervous system through the gut-brain axis to support neuronal development and maintenance while gut dysbiosis manifests in neurological disease. There are three basic mechanisms that mediate the communication between the gut and the brain: direct neuronal communication, endocrine signaling mediators and the immune system. Together, these systems create a highly integrated molecular communication network that link systemic imbalances with the development of neurodegeneration including insulin regulation, fat metabolism, oxidative markers and immune signaling. Age is a common factor in the development of neurodegenerative disease and probiotics prevent many harmful effects of aging such as decreased neurotransmitter levels, chronic inflammation, oxidative stress and apoptosis-all factors that are proven aggravators of neurodegenerative disease. Indeed patients with Parkinson's and Alzheimer's diseases have a high rate of gastrointestinal comorbidities and it has be proposed by some the management of the gut microbiota may prevent or alleviate the symptoms of these chronic diseases.
Collapse
Affiliation(s)
- Susan Westfall
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada
| | - Nikita Lomis
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada
- Department of Experimental Medicine, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada
| | - Imen Kahouli
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada
- Department of Experimental Medicine, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada
| | - Si Yuan Dia
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada
| | - Surya Pratap Singh
- Department of Biochemistry, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada.
- Department of Experimental Medicine, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada.
| |
Collapse
|
42
|
Yoneyama-Hirozane M, Deguchi K, Hirakawa T, Ishii T, Odani T, Matsui J, Nakano Y, Imahashi K, Takakura N, Chisaki I, Takekawa S, Sakamoto J. Identification and Characterization of a New Series of Ghrelin O-Acyl Transferase Inhibitors. SLAS DISCOVERY 2017; 23:154-163. [PMID: 28846466 DOI: 10.1177/2472555217727097] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Ghrelin O-acyl transferase (GOAT; MBOAT4) catalyzes O-acylation at serine-3 of des-acyl ghrelin. Acyl ghrelin is secreted by stomach X/A-like cells and plays a role in appetite and metabolism. Therefore, GOAT has been expected to be a novel antiobesity target because it is responsible for acyl ghrelin production. Here, we report homogeneous time-resolved fluorescence (HTRF) and enzyme-linked immunosorbent assay (ELISA) methods utilizing human GOAT-expressing microsomes as a novel high-throughput assay system for the discovery of hit compounds and optimization of lead compounds. Hit compounds exemplified by compound A (2-[(2,4-dichlorobenzyl)sulfanyl]-1,3-benzoxazole-5-carboxylic acid) were identified by high-throughput screening using the HTRF assay and confirmed to have GOAT inhibitory activity using the ELISA. Based on the hit compound information, the novel lead compound (compound B, (4-chloro-6-{[2-methyl-6-(trifluoromethyl)pyridin-3-yl]methoxy}-1-benzothiophen-3-yl)acetic acid) was synthesized and exhibited potent GOAT inhibition with oral bioavailability. Both the hit compound and lead compound showed octanoyl-CoA competitive inhibitory activity. Moreover, these two compounds decreased acyl ghrelin production in the stomach of mice after their oral administration. These novel findings demonstrate that GOAT is a druggable target, and its inhibitors are promising antiobesity drugs.
Collapse
Affiliation(s)
| | - Kohei Deguchi
- 1 Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Takeshi Hirakawa
- 1 Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Tsuyoshi Ishii
- 1 Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Tomoyuki Odani
- 1 Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Junji Matsui
- 1 Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Yoshihide Nakano
- 1 Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Kenichi Imahashi
- 1 Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | | | - Ikumi Chisaki
- 1 Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Shiro Takekawa
- 1 Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Junichi Sakamoto
- 1 Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| |
Collapse
|
43
|
The components of somatostatin and ghrelin systems are altered in neuroendocrine lung carcinoids and associated to clinical-histological features. Lung Cancer 2017; 109:128-136. [PMID: 28577942 DOI: 10.1016/j.lungcan.2017.05.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 05/02/2017] [Accepted: 05/07/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Lung carcinoids (LCs) are rare tumors that comprise 1-5% of lung malignancies but represent 20-30% of neuroendocrine tumors. Their incidence is progressively increasing and a better characterization of these tumors is required. Alterations in somatostatin (SST)/cortistatin (CORT) and ghrelin systems have been associated to development/progression of various endocrine-related cancers, wherein they may become useful diagnostic, prognostic and therapeutic biomarkers. OBJECTIVES We aimed to evaluate the expression levels of ghrelin and SST/CORT system components in LCs, as well as to explore their putative relationship with histological/clinical characteristics. PATIENTS AND METHODS An observational retrospective study was performed; 75 LC patients with clinical/histological characteristics were included. Samples from 46 patients were processed to isolate mRNA from tumor and adjacent non-tumor region, and the expression levels of SST/CORT and ghrelin systems components, determined by quantitative-PCR, were compared to those of 7 normal lung tissues. RESULTS Patient cohort was characterized by mean age 53±15 years, 48% males, 34% with tobacco exposure; 71.4/28.6% typical/atypical carcinoids, 21.7% incidental tumors, 4.3% functioning tumors, 17.7% with metastasis. SST/CORT and ghrelin system components were expressed at variable levels in a high proportion of tumors, as well as in adjacent non-tumor tissues, while a lower proportion of normal lung samples also expressed these molecules. A gradation was observed from normal non-neoplastic lung tissues, non-tumor adjacent tissue and LCs, being SST, sst4, sst5, GHS-R1a and GHS-R1b overexpressed in tumor tissue compared to normal tissue. Importantly, several SST/CORT and ghrelin system components displayed significant correlations with relevant clinical parameters, such as necrosis, peritumoral and vascular invasion, or metastasis. CONCLUSION Altogether, these data reveal a prominent, widespread expression of key SST/CORT/ghrelin system components in LCs, where they display clinical-histological correlations, which could provide novel, valuable markers for NET patient management.
Collapse
|
44
|
Colldén G, Tschöp MH, Müller TD. Therapeutic Potential of Targeting the Ghrelin Pathway. Int J Mol Sci 2017; 18:ijms18040798. [PMID: 28398233 PMCID: PMC5412382 DOI: 10.3390/ijms18040798] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/03/2017] [Accepted: 04/06/2017] [Indexed: 02/07/2023] Open
Abstract
Ghrelin was discovered in 1999 as the endogenous ligand of the growth-hormone secretagogue receptor 1a (GHSR1a). Since then, ghrelin has been found to exert a plethora of physiological effects that go far beyond its initial characterization as a growth hormone (GH) secretagogue. Among the numerous well-established effects of ghrelin are the stimulation of appetite and lipid accumulation, the modulation of immunity and inflammation, the stimulation of gastric motility, the improvement of cardiac performance, the modulation of stress, anxiety, taste sensation and reward-seeking behavior, as well as the regulation of glucose metabolism and thermogenesis. Due to a variety of beneficial effects on systems’ metabolism, pharmacological targeting of the endogenous ghrelin system is widely considered a valuable approach to treat metabolic complications, such as chronic inflammation, gastroparesis or cancer-associated anorexia and cachexia. The aim of this review is to discuss and highlight the broad pharmacological potential of ghrelin pathway modulation for the treatment of anorexia, cachexia, sarcopenia, cardiopathy, neurodegenerative disorders, renal and pulmonary disease, gastrointestinal (GI) disorders, inflammatory disorders and metabolic syndrome.
Collapse
Affiliation(s)
- Gustav Colldén
- Institute for Diabetes and Obesity & Helmholtz Diabetes Center, Helmholtz Zentrum München German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany.
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity & Helmholtz Diabetes Center, Helmholtz Zentrum München German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany.
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 80333 Munich, Germany.
| | - Timo D Müller
- Institute for Diabetes and Obesity & Helmholtz Diabetes Center, Helmholtz Zentrum München German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany.
- Institute for Diabetes and Obesity (IDO), Business Campus Garching-Hochbrück, Parkring 13, 85748 Garching, Germany.
| |
Collapse
|
45
|
Rikkunshito prevents paclitaxel-induced peripheral neuropathy through the suppression of the nuclear factor kappa B (NFκB) phosphorylation in spinal cord of mice. PLoS One 2017; 12:e0171819. [PMID: 28182729 PMCID: PMC5300261 DOI: 10.1371/journal.pone.0171819] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 01/26/2017] [Indexed: 12/15/2022] Open
Abstract
Peripheral neuropathy is the major side effect caused by paclitaxel, a microtubule-binding antineoplastic drug. Paclitaxel-induced peripheral neuropathy causes a long-term negative impact on the patient's quality of life. However, the mechanism underlying paclitaxel-induced peripheral neuropathy is still unknown, and there is no established treatment. Ghrelin is known to attenuate thermal hyperalgesia and mechanical allodynia in chronic constriction injury of the sciatic nerve, and inhibit the activation of nuclear factor kappa B (NFκB) in the spinal dorsal horn. Rikkunshito (RKT), a kampo medicine, increases the secretion of ghrelin in rodents and humans. Thus, RKT may attenuate paclitaxel-induced peripheral neuropathy by inhibiting phosphorylated NFκB (pNFκB) in the spinal cord. We found that paclitaxel dose-dependently induced mechanical hyperalgesia in mice. Paclitaxel increased the protein levels of spinal pNFκB, but not those of spinal NFκB. NFκB inhibitor attenuated paclitaxel-induced mechanical hyperalgesia suggesting that the activation of NFκB mediates paclitaxel-induced hyperalgesia. RKT dose-dependently attenuated paclitaxel-induced mechanical hyperalgesia. Ghrelin receptor antagonist reversed the RKT-induced attenuation of paclitaxel-induced mechanical hyperalgesia. RKT inhibited the paclitaxel-induced increase in the protein levels of spinal pNFκB. Taken together, the present study indicates that RKT exerts an antihyperalgesic effect in paclitaxel-induced neuropathic pain by suppressing the activation of spinal NFκB.
Collapse
|
46
|
Neuropeptides and Microglial Activation in Inflammation, Pain, and Neurodegenerative Diseases. Mediators Inflamm 2017; 2017:5048616. [PMID: 28154473 PMCID: PMC5244030 DOI: 10.1155/2017/5048616] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 11/26/2016] [Accepted: 12/05/2016] [Indexed: 12/15/2022] Open
Abstract
Microglial cells are responsible for immune surveillance within the CNS. They respond to noxious stimuli by releasing inflammatory mediators and mounting an effective inflammatory response. This is followed by release of anti-inflammatory mediators and resolution of the inflammatory response. Alterations to this delicate process may lead to tissue damage, neuroinflammation, and neurodegeneration. Chronic pain, such as inflammatory or neuropathic pain, is accompanied by neuroimmune activation, and the role of glial cells in the initiation and maintenance of chronic pain has been the subject of increasing research over the last two decades. Neuropeptides are small amino acidic molecules with the ability to regulate neuronal activity and thereby affect various functions such as thermoregulation, reproductive behavior, food and water intake, and circadian rhythms. Neuropeptides can also affect inflammatory responses and pain sensitivity by modulating the activity of glial cells. The last decade has witnessed growing interest in the study of microglial activation and its modulation by neuropeptides in the hope of developing new therapeutics for treating neurodegenerative diseases and chronic pain. This review summarizes the current literature on the way in which several neuropeptides modulate microglial activity and response to tissue damage and how this modulation may affect pain sensitivity.
Collapse
|
47
|
Jiao Q, Du X, Li Y, Gong B, Shi L, Tang T, Jiang H. The neurological effects of ghrelin in brain diseases: Beyond metabolic functions. Neurosci Biobehav Rev 2016; 73:98-111. [PMID: 27993602 DOI: 10.1016/j.neubiorev.2016.12.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 12/01/2016] [Accepted: 12/10/2016] [Indexed: 02/08/2023]
Abstract
Ghrelin, a peptide released by the stomach that plays a major role in regulating energy metabolism, has recently been shown to have effects on neurobiological behaviors. Ghrelin enhances neuronal survival by reducing apoptosis, alleviating inflammation and oxidative stress, and accordingly improving mitochondrial function. Ghrelin also stimulates the proliferation, differentiation and migration of neural stem/progenitor cells (NS/PCs). Additionally, the ghrelin is benefit for the recovery of memory, mood and cognitive dysfunction after stroke or traumatic brain injury. Because of its neuroprotective and neurogenic roles, ghrelin may be used as a therapeutic agent in the brain to combat neurodegenerative disease. In this review, we highlight the pre-clinical evidence and the proposed mechanisms underlying the role of ghrelin in physiological and pathological brain function.
Collapse
Affiliation(s)
- Qian Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China.
| | - Xixun Du
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China.
| | - Yong Li
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China.
| | - Bing Gong
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China.
| | - Limin Shi
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China.
| | - Tingting Tang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China.
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, China.
| |
Collapse
|
48
|
Kunath N, Müller NCJ, Tonon M, Konrad BN, Pawlowski M, Kopczak A, Elbau I, Uhr M, Kühn S, Repantis D, Ohla K, Müller TD, Fernández G, Tschöp M, Czisch M, Steiger A, Dresler M. Ghrelin modulates encoding-related brain function without enhancing memory formation in humans. Neuroimage 2016; 142:465-473. [PMID: 27402596 DOI: 10.1016/j.neuroimage.2016.07.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 06/07/2016] [Accepted: 07/06/2016] [Indexed: 01/24/2023] Open
Abstract
Ghrelin regulates energy homeostasis in various species and enhances memory in rodent models. In humans, the role of ghrelin in cognitive processes has yet to be characterized. Here we show in a double-blind randomized crossover design that acute administration of ghrelin alters encoding-related brain activity, however does not enhance memory formation in humans. Twenty-one healthy young male participants had to memorize food- and non-food-related words presented on a background of a virtual navigational route while undergoing fMRI recordings. After acute ghrelin administration, we observed decreased post-encoding resting state fMRI connectivity between the caudate nucleus and the insula, amygdala, and orbitofrontal cortex. In addition, brain activity related to subsequent memory performance was modulated by ghrelin. On the next day, however, no differences were found in free word recall or cued location-word association recall between conditions; and ghrelin's effects on brain activity or functional connectivity were unrelated to memory performance. Further, ghrelin had no effect on a cognitive test battery comprising tests for working memory, fluid reasoning, creativity, mental speed, and attention. In conclusion, in contrast to studies with animal models, we did not find any evidence for the potential of ghrelin acting as a short-term cognitive enhancer in humans.
Collapse
Affiliation(s)
- N Kunath
- Max Planck Institute of Psychiatry, Munich, Germany
| | - N C J Müller
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - M Tonon
- Max Planck Institute of Psychiatry, Munich, Germany
| | - B N Konrad
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - M Pawlowski
- Max Planck Institute of Psychiatry, Munich, Germany
| | - A Kopczak
- Max Planck Institute of Psychiatry, Munich, Germany
| | - I Elbau
- Max Planck Institute of Psychiatry, Munich, Germany
| | - M Uhr
- Max Planck Institute of Psychiatry, Munich, Germany
| | - S Kühn
- Max Planck Institute for Human Development, Berlin, Germany
| | - D Repantis
- Charité - Universitätsmedizin Berlin, Department of Psychiatry and Psychotherapy, CBF, Berlin, Germany
| | - K Ohla
- German Institute for Human Nutrition, Potsdam-Rehbrücke, Germany
| | - T D Müller
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, Munich, Germany; Department of Medicine, Technische Universität München, Munich, Germany
| | - G Fernández
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - M Tschöp
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, Munich, Germany; Department of Medicine, Technische Universität München, Munich, Germany
| | - M Czisch
- Max Planck Institute of Psychiatry, Munich, Germany
| | - A Steiger
- Max Planck Institute of Psychiatry, Munich, Germany
| | - M Dresler
- Max Planck Institute of Psychiatry, Munich, Germany; Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijmegen, The Netherlands.
| |
Collapse
|
49
|
Physiological roles for butyrylcholinesterase: A BChE-ghrelin axis. Chem Biol Interact 2016; 259:271-275. [PMID: 26915976 DOI: 10.1016/j.cbi.2016.02.013] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 02/10/2016] [Accepted: 02/16/2016] [Indexed: 11/24/2022]
Abstract
Butyrylcholinesterase (BChE) has long been regarded as an "orphan enzyme" with no specific physiological role other than to metabolize exogenous bioactive esters in the diet or in medicines. Human beings with genetic mutations that eliminate all BChE activity appear completely normal, and BChE-knockout mice have been described as "lacking a phenotype" except for faster weight gain on high-fat diets. However, our recent studies with viral gene transfer of BChE in mice reveal that BChE hydrolyzes the so-called "hunger hormone," ghrelin, at a rate which strongly affects the circulating levels of this peptide hormone. This action has important consequences for weight gain and fat metabolism. Surprisingly, it also impacts emotional behaviors such as aggression. Overexpression of BChE leads to low ghrelin levels in the blood stream and reduces aggression and social stress in mice. Under certain circumstances these combined effects contribute to increased life-span in group-housed animals. These findings may generalize to humans, as recent clinical studies by multiple investigators indicate that, among patients with severe cardiovascular disease, longevity correlates with increasing levels of plasma BChE activity.
Collapse
|
50
|
Neuroprotective effects of metformin against Aβ-mediated inhibition of long-term potentiation in rats fed a high-fat diet. Brain Res Bull 2016; 121:178-85. [PMID: 26861514 DOI: 10.1016/j.brainresbull.2016.02.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 01/31/2016] [Accepted: 02/04/2016] [Indexed: 11/24/2022]
Abstract
Metformin (Met) is used to treat neurodegenerative disorders such as Alzheimer's disease (AD). Conversely, high-fat diets (HFD) have been shown to increase AD risk. In this study, we investigated the neuroprotective effects of Met on β-amyloid (Aβ)-induced impairments in hippocampal synaptic plasticity in AD model rats that were fed a HFD. In this study, 32 adult male Wistar rats were randomly assigned to four groups: group I (control group, regular diet); group II (HFD+vehicle); group III (HFD+Aβ); or group IV (Met+HFD+Aβ). Rats fed a HFD were injected with Aβ to induce AD, allowed to recover, and treated with Met for 8 weeks. The rats were then anesthetized with intraperitoneal injections of urethane and placed in a stereotaxic apparatus for surgery, electrode implantation, and field potential recording. In vivo electrophysiological recordings were then performed to measure population spike (PS) amplitude and excitatory postsynaptic potential (EPSP) slope in the hippocampal dentate gyrus. Long-term potentiation (LTP) was induced by high-frequency stimulation of the perforant pathway. Blood samples were then collected to measure plasma levels of triglycerides, low-density lipoproteins, very low-density lipoprotein, and cholesterol. After induction of LTP, PS amplitude and EPSP slope were significantly decreased in Aβ-injected rats fed a HFD compared to vehicle-injected animals or untreated animals that were fed a normal diet. Met treatment of Aβ-injected rats significantly attenuated these decreases, suggesting that Met decreased the effects of Aβ on LTP. These findings suggest that Met treatment is neuroprotective against the detrimental effects of Aβ and HFDs on hippocampal synaptic plasticity.
Collapse
|