1
|
Cheraghi S, Mobaderi T, Mottaghi A, Movahedi Motlagh F, Taghizadeh S, Eghbali M. Genetic variants in the MC4R gene and risk of obesity/overweight: A systematic review and meta-analysis. Diabetes Obes Metab 2025. [PMID: 40302631 DOI: 10.1111/dom.16425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 05/02/2025]
Abstract
AIM Obesity is a significant health issue worldwide, progressing due to genetic factors and lifestyle. Melanocortin 4 receptor (MC4R) gene polymorphisms have been identified as a cause of overweight and obesity risk. The aim of this study was a comprehensive assessment of MC4R polymorphism effects on overweight/obesity risk. METHODS All retrieved literature from PubMed, Web of Science and Scopus according to PRISMA guidelines up to June 2022 was reviewed. Inclusion criteria are restricted to English-language, human case-control/cohort studies with genotype distributions of MC4R polymorphisms and their association with obesity and overweight in any geographic regions and age. The heterogeneity using the I-squared statistic (I2), the Q-test and Prediction Interval (PI) and publication bias using Begg's and Egger's tests were examined, and the pooled odds ratios in different genetic models were estimated using a random effect model. Subgroup analysis was performed by the geographic regions and age groups. Risk of bias for individual studies was not assessed. The review is limited by restricted racial diversity and exclusion of environmental factors, incomplete data and limited access to certain articles. This work received no specific funding, and the review was not prospectively registered. RESULTS In our study, 39 eligible studies with 43 697 overweight and obese cases and 52 272 normal weights were included. In mixed-age populations, rs17700633, rs17782313, rs11872992, rs12970134, rs2229616 and rs571312 were evaluated. The remarkable association was seen by rs17782313 and rs12970134 in the Homozygous model (OR = 1.73; 95% CI: 1.51, 1.98 and 1.74; 95% CI: 1.29; 2.35, respectively). In addition, rs17782313 and rs12970134 were found to be more strongly linked to overweight and obesity in Asian and European population groups, as determined by a subgroup analysis of the geographic regions. CONCLUSION The present study confirms the high association of rs17782313 and rs12970134 with obesity and overweight in all age groups and geographic regions. However, further functional studies and high-population research on other MC4R SNPs must validate their role.
Collapse
Affiliation(s)
- Sara Cheraghi
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Tofigh Mobaderi
- Minimally Invasive Surgery Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Azadeh Mottaghi
- Research Center for Prevention of Cardiovascular Diseases, Endocrinology & Metabolism, Institute of Endocrinology Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | | | - Sara Taghizadeh
- Translational Ophthalmology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Eghbali
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Jafari A, Khoshdooz S, Bafrani MA, Bakhshimoghaddam F, Abbasi H, Doaei S. Uncovering the Causal Link Between Obesity-Associated Genes and Multiple Sclerosis: A Systematic Literature Review. Brain Behav 2025; 15:e70439. [PMID: 40195065 PMCID: PMC11975544 DOI: 10.1002/brb3.70439] [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: 10/11/2024] [Revised: 02/25/2025] [Accepted: 03/02/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a multifaceted neurodegenerative disorder influenced by genetics and lifestyle. This systematic literature review investigates the role of six obesity-associated genes, including fat mass and obesity-associated (FTO), FAS apoptosis inhibitory molecule 2 (FAIM2), Niemann-Pick disease type C1-like 1 (NPC1), glucosamine-6-phosphate deaminase 2 (GNPDA2), melanocortin-4 receptor (MC4R), and brain-derived neurotrophic factor (BDNF) in the context of MS. METHODS A literature search was executed using Embase, Scopus, Cochrane, Web of Science, and PubMed databases from inception to July 2024. The related keywords employed during the search process are "fas apoptotic inhibitory molecule 2," "Niemann-Pick disease type C1," "fat mass and obesity-associated," "melanocortin-4 receptor," "brain-derived neurotrophic factor," "glucosamine-6-phosphate deaminase 2," and "multiple sclerosis." RESULTS Out of 2108 papers, 27 were entered into the present systematic literature review. The FTO gene may affect MS susceptibility through metabolic and inflammatory pathways. FAIM2 and NPC1 genes may contribute to MS pathogenesis, though their precise roles are still being elucidated. The GNPDA2 gene may have some connections with MS but requires further clarification. MC4R has demonstrated significant neuroprotective and anti-inflammatory effects, suggesting its potential impact on MS progression. BDNF plays a complex role in neuronal survival and repair and may influence the risk of MS. CONCLUSION Our findings demonstrated that obesity-related genes may have a significant impact on MS risk and disease course, revealing novel insights into the genetic underpinnings of MS.
Collapse
Affiliation(s)
- Ali Jafari
- Student Research Committee, Department of Community Nutrition, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research InstituteShahid Beheshti University of Medical SciencesTehranIran
- Systematic Review and Meta‐analysis Expert Group (SRMEG)Universal Scientific Education and Research Network (USERN)TehranIran
| | - Sara Khoshdooz
- Faculty of MedicineGuilan University of Medical ScienceRashtIran
| | | | - Farnush Bakhshimoghaddam
- Department of Nutrition, School of Allied Medical SciencesAhvaz Jundishapur University of Medical SciencesAhvazIran
| | - Hamid Abbasi
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
- Neurosciences Research CenterTabriz University of Medical SciencesTabrizIran
| | - Saeid Doaei
- Department of Community Nutrition, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research InstituteShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
3
|
Carniglia L, Turati J, Saba J, López Couselo F, Romero AC, Caruso C, Durand D, Lasaga M. Melanocortin-receptor 4 activation modulates proliferation and differentiation of rat postnatal hippocampal neural precursor cells. Neuropharmacology 2024; 257:110058. [PMID: 38960135 DOI: 10.1016/j.neuropharm.2024.110058] [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: 05/08/2024] [Revised: 06/28/2024] [Accepted: 06/30/2024] [Indexed: 07/05/2024]
Abstract
Postnatal hippocampal neurogenesis is essential for learning and memory. Hippocampal neural precursor cells (NPCs) can be induced to proliferate and differentiate into either glial cells or dentate granule cells. Notably, hippocampal neurogenesis decreases dramatically with age, partly due to a reduction in the NPC pool and a decrease in their proliferative activity. Alpha-melanocyte-stimulating hormone (α-MSH) improves learning, memory, neuronal survival and plasticity. Here, we used postnatally-isolated hippocampal NPCs from Wistar rat pups (male and female combined) to determine the role of the melanocortin analog [Nle4, D-Phe7]-α-MSH (NDP-MSH) in proliferation and fate acquisition of NPCs. Incubation of growth-factor deprived NPCs with 10 nM NDP-MSH for 6 days increased the proportion of Ki-67- and 5-bromo-2'-deoxyuridine (BrdU)-positive cells, compared to the control group, and these effects were blocked by the MC4R antagonist JKC-363. NDP-MSH also increased the proportion of glial fibrillar acidic protein (GFAP)/Ki-67, GFAP/sex-determining region Y-box2 (SOX2) and neuroepithelial stem cell protein (NESTIN)/Ki-67-double positive cells (type-1 and type-2 precursors). Finally, NDP-MSH induced peroxisome proliferator-activated receptor (PPAR)-γ protein expression, and co-incubation with the PPAR-γ inhibitor GW9662 prevented the effect of NDP-MSH on NPC proliferation and differentiation. Our results indicate that in vitro activation of MC4R in growth-factor-deprived postnatal hippocampal NPCs induces proliferation and promotes the relative expansion of the type-1 and type-2 NPC pool through a PPAR-γ-dependent mechanism. These results shed new light on the mechanisms underlying the beneficial effects of melanocortins in hippocampal plasticity and provide evidence linking the MC4R and PPAR-γ pathways in modulation of hippocampal NPC proliferation and differentiation.
Collapse
Affiliation(s)
- Lila Carniglia
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Paraguay 2155, piso 10, CP 1121, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Juan Turati
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Paraguay 2155, piso 10, CP 1121, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Julieta Saba
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Paraguay 2155, piso 10, CP 1121, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Federico López Couselo
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Paraguay 2155, piso 10, CP 1121, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Ana Clara Romero
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Paraguay 2155, piso 10, CP 1121, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Carla Caruso
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Paraguay 2155, piso 10, CP 1121, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Daniela Durand
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Paraguay 2155, piso 10, CP 1121, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Mercedes Lasaga
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Paraguay 2155, piso 10, CP 1121, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
4
|
Kupcova I, Danisovic L, Bernatova S, Harsanyi S. Analysis of Salivary Neuropeptides in Anxiety and Depression Using the Luminex MAGPIX® System. Cureus 2024; 16:e67984. [PMID: 39347144 PMCID: PMC11433651 DOI: 10.7759/cureus.67984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Anxiety and depressive disorders are highly prevalent mental health conditions, affecting millions worldwide. Advancements in neurobiology have identified the effects of various neuropeptides in modulating mood and stress responses. Some of the well-researched neuropeptides in plasma are oxytocin (OXT), alpha-melanocyte-stimulating hormone (alpha-MSH), beta-endorphin, neurotensin, and substance P. In this study, we used methods of liquid biopsy to acquire saliva samples to analyze the concentrations of neuropeptides associated with depression. METHODS The study was conducted in Bratislava, Slovakia, from January to June 2022. Participants were 20 subjects treated for depression and anxiety without medication; the control group consisted of 20 healthy individuals with no personal history of depression or anxiety. Salivary samples were collected using buccal swabs to measure the concentrations of the examined neuropeptides. Laboratory analysis was based on detecting fluorescent signals performed on the Luminex MAGPIX® System (Luminex Corporation, Austin, Texas). Means and standard deviations were calculated for individual neuropeptide levels. To determine if there are statistically significant differences in neuropeptide levels between individuals with and without depression, independent t-tests and a one-way ANOVA were conducted. RESULTS Our findings indicate a significant decrease in all studied neuropeptides in subjects compared to healthy controls. Reductions in mean levels were observed for OXT (7.3), alpha-MSH (3.9), beta-endorphin (2.9), neurotensin (15.1), and a 6.9-fold decrease for substance P. Alpha-MSH and beta-endorphin showed higher variability in measured levels within both groups. CONCLUSION The results of this study indicate that the levels of the studied salivary neuropeptides, OXT, alpha-MSH, beta-endorphin, neurotensin, and substance P, are statistically significantly reduced in individuals with depression compared to healthy controls.
Collapse
Affiliation(s)
- Ida Kupcova
- Institute of Medical Biology, Genetics, and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Bratislava, SVK
- Department of Psychiatry, Psychiatric Clinic, The University Hospital Brno, Brno, CZE
| | - Lubos Danisovic
- Institute of Medical Biology, Genetics, and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Bratislava, SVK
| | - Sona Bernatova
- Institute of Medical Biology, Genetics, and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Bratislava, SVK
| | - Stefan Harsanyi
- Institute of Medical Biology, Genetics, and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Bratislava, SVK
| |
Collapse
|
5
|
Wekwejt P, Wojda U, Kiryk A. Melanotan-II reverses memory impairment induced by a short-term HF diet. Biomed Pharmacother 2023; 165:115129. [PMID: 37478579 DOI: 10.1016/j.biopha.2023.115129] [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: 05/10/2023] [Revised: 06/12/2023] [Accepted: 07/02/2023] [Indexed: 07/23/2023] Open
Abstract
A high-fat (HF) diet has been shown to increase the risk of neurological impairments and neurodegenerative disorders. The melanotropins used in this study have been associated with diet-related disorders; however, there is an absence of studies on their effect on diet-induced neurobehavioral conditions. Here, we investigated the possible relationship among diet, Melanotan-II (MT-II) targeting melanotropin receptors, and the behavior of zebrafish (Danio rerio). Surprisingly, even a short-term HF diet lasting for ∼ 1 % of the zebrafish's life had a strong developmental effect. Zebrafish fed the HF diet showed an impairment in recognition memory, elevated anxiety levels, and reduced exploratory propensity after just three weeks compared to zebrafish fed the control diet. These HF diet-induced abnormalities were reversed by MT-II. Animals fed a HF diet and treated with MT-II demonstrated recognition memory, anxiety, and exploratory behavior similar to the control group. This study provides evidence that even a short-term HF diet has an impact on memory and emotions and is the first study to show that MT-II reverses these changes.
Collapse
Affiliation(s)
- Patryk Wekwejt
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Urszula Wojda
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Anna Kiryk
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology, 3 Pasteur Street, 02-093 Warsaw, Poland.
| |
Collapse
|
6
|
Gebrie A. The melanocortin receptor signaling system and its role in neuroprotection against neurodegeneration: Therapeutic insights. Ann N Y Acad Sci 2023; 1527:30-41. [PMID: 37526975 DOI: 10.1111/nyas.15048] [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] [Indexed: 08/02/2023]
Abstract
The melanocortin signaling system consists of the melanocortin peptides, their distinctive receptors, accessory proteins, and endogenous antagonists. Melanocortin peptides are small peptide hormones that have been studied in a variety of physiological and pathological conditions. There are five types of melanocortin receptors, and they are distributed within the central nervous system and in several tissues of the periphery. The G protein-coupled melanocortin receptors typically signal through adenylyl cyclase and other downstream signaling pathways. Depending on the ligand, surface expression of melanocortin receptor, receptor occupancy period, related proteins, the type of cell, and other parameters, the signaling pathways are complicated and pleiotropic. While it is known that all five melanocortin receptors are coupled to Gs, they can also occasionally couple to Gq or Gi. Both direct and indirect neuroprotection are induced by the melanocortin signaling system. Targeting several of the components of the melanocortin signaling system (ligands, receptors, accessory proteins, signaling effectors, and regulators) may provide therapeutic opportunities. Activation of the melanocortin system improves different functional traits in neurodegenerative diseases. There is a potential for additional melanocortin system interventions by interfering with dimerization or dissociation. This review aims to discuss the melanocortin receptor signaling system and its role in neuroprotection, as well as its therapeutic potential.
Collapse
Affiliation(s)
- Alemu Gebrie
- Department of Biomedical Sciences, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
7
|
Wu CLS, Cioanca AV, Gelmi MC, Wen L, Di Girolamo N, Zhu L, Natoli R, Conway RM, Petsoglou C, Jager MJ, McCluskey PJ, Madigan MC. The multifunctional human ocular melanocortin system. Prog Retin Eye Res 2023; 95:101187. [PMID: 37217094 DOI: 10.1016/j.preteyeres.2023.101187] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/12/2023] [Accepted: 05/12/2023] [Indexed: 05/24/2023]
Abstract
Immune privilege in the eye involves physical barriers, immune regulation and secreted proteins that together limit the damaging effects of intraocular immune responses and inflammation. The neuropeptide alpha-melanocyte stimulating hormone (α-MSH) normally circulates in the aqueous humour of the anterior chamber and the vitreous fluid, secreted by iris and ciliary epithelium, and retinal pigment epithelium (RPE). α-MSH plays an important role in maintaining ocular immune privilege by helping the development of suppressor immune cells and by activating regulatory T-cells. α-MSH functions by binding to and activating melanocortin receptors (MC1R to MC5R) and receptor accessory proteins (MRAPs) that work in concert with antagonists, otherwise known as the melanocortin system. As well as controlling immune responses and inflammation, a broad range of biological functions is increasingly recognised to be orchestrated by the melanocortin system within ocular tissues. This includes maintaining corneal transparency and immune privilege by limiting corneal (lymph)angiogenesis, sustaining corneal epithelial integrity, protecting corneal endothelium and potentially enhancing corneal graft survival, regulating aqueous tear secretion with implications for dry eye disease, facilitating retinal homeostasis via maintaining blood-retinal barriers, providing neuroprotection in the retina, and controlling abnormal new vessel growth in the choroid and retina. The role of melanocortin signalling in uveal melanocyte melanogenesis however remains unclear compared to its established role in skin melanogenesis. The early application of a melanocortin agonist to downregulate systemic inflammation used adrenocorticotropic hormone (ACTH)-based repository cortisone injection (RCI), but adverse side effects including hypertension, edema, and weight gain, related to increased adrenal gland corticosteroid production, impacted clinical uptake. Compared to ACTH, melanocortin peptides that target MC1R, MC3R, MC4R and/or MC5R, but not adrenal gland MC2R, induce minimal corticosteroid production with fewer adverse systemic effects. Pharmacological advances in synthesising MCR-specific targeted peptides provide further opportunities for treating ocular (and systemic) inflammatory diseases. Following from these observations and a renewed clinical and pharmacological interest in the diverse biological roles of the melanocortin system, this review highlights the physiological and disease-related involvement of this system within human eye tissues. We also review the emerging benefits and versatility of melanocortin receptor targeted peptides as non-steroidal alternatives for inflammatory eye diseases such as non-infectious uveitis and dry eye disease, and translational applications in promoting ocular homeostasis, for example, in corneal transplantation and diabetic retinopathy.
Collapse
Affiliation(s)
- Chieh-Lin Stanley Wu
- School of Optometry and Vision Science, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia; Save Sight Institute and Ophthalmology, The Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Department of Optometry, Asia University, Taichung, Taiwan
| | - Adrian V Cioanca
- Save Sight Institute and Ophthalmology, The Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; John Curtin School of Medical Research, The Australian National University, ACT, Australia; ANU Medical School, The Australian National University, ACT, Australia
| | - Maria C Gelmi
- Department of Ophthalmology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Li Wen
- New South Wales Organ and Tissue Donation Service, Sydney Hospital and Sydney Eye Hospital, NSW, 2000, Australia
| | - Nick Di Girolamo
- School of Biomedical Sciences, Mechanisms of Disease and Translational Research, University of New South Wales, Sydney, Australia
| | - Ling Zhu
- Save Sight Institute and Ophthalmology, The Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Riccardo Natoli
- Save Sight Institute and Ophthalmology, The Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; John Curtin School of Medical Research, The Australian National University, ACT, Australia; ANU Medical School, The Australian National University, ACT, Australia
| | - R Max Conway
- Save Sight Institute and Ophthalmology, The Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Constantinos Petsoglou
- Save Sight Institute and Ophthalmology, The Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; New South Wales Organ and Tissue Donation Service, Sydney Hospital and Sydney Eye Hospital, NSW, 2000, Australia
| | - Martine J Jager
- Department of Ophthalmology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Peter J McCluskey
- Save Sight Institute and Ophthalmology, The Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Michele C Madigan
- School of Optometry and Vision Science, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia; Save Sight Institute and Ophthalmology, The Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.
| |
Collapse
|
8
|
Markov DD, Dolotov OV, Grivennikov IA. The Melanocortin System: A Promising Target for the Development of New Antidepressant Drugs. Int J Mol Sci 2023; 24:ijms24076664. [PMID: 37047638 PMCID: PMC10094937 DOI: 10.3390/ijms24076664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Major depression is one of the most prevalent mental disorders, causing significant human suffering and socioeconomic loss. Since conventional antidepressants are not sufficiently effective, there is an urgent need to develop new antidepressant medications. Despite marked advances in the neurobiology of depression, the etiology and pathophysiology of this disease remain poorly understood. Classical and newer hypotheses of depression suggest that an imbalance of brain monoamines, dysregulation of the hypothalamic-pituitary-adrenal axis (HPAA) and immune system, or impaired hippocampal neurogenesis and neurotrophic factors pathways are cause of depression. It is assumed that conventional antidepressants improve these closely related disturbances. The purpose of this review was to discuss the possibility of affecting these disturbances by targeting the melanocortin system, which includes adrenocorticotropic hormone-activated receptors and their peptide ligands (melanocortins). The melanocortin system is involved in the regulation of various processes in the brain and periphery. Melanocortins, including peripherally administered non-corticotropic agonists, regulate HPAA activity, exhibit anti-inflammatory effects, stimulate the levels of neurotrophic factors, and enhance hippocampal neurogenesis and neurotransmission. Therefore, endogenous melanocortins and their analogs are able to complexly affect the functioning of those body’s systems that are closely related to depression and the effects of antidepressants, thereby demonstrating a promising antidepressant potential.
Collapse
Affiliation(s)
- Dmitrii D. Markov
- National Research Center “Kurchatov Institute”, Kurchatov Sq. 2, 123182 Moscow, Russia
| | - Oleg V. Dolotov
- National Research Center “Kurchatov Institute”, Kurchatov Sq. 2, 123182 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Leninskie Gory, 119234 Moscow, Russia
| | - Igor A. Grivennikov
- National Research Center “Kurchatov Institute”, Kurchatov Sq. 2, 123182 Moscow, Russia
| |
Collapse
|
9
|
Hidese S, Yoshida F, Ishida I, Matsuo J, Hattori K, Kunugi H. Plasma neuropeptide levels in patients with schizophrenia, bipolar disorder, or major depressive disorder and healthy controls: A multiplex immunoassay study. Neuropsychopharmacol Rep 2023; 43:57-68. [PMID: 36414415 PMCID: PMC10009433 DOI: 10.1002/npr2.12304] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/27/2022] [Accepted: 11/10/2022] [Indexed: 11/24/2022] Open
Abstract
AIM We aimed to compare neuropeptide levels between patients with major psychiatric disorders and healthy controls and examine their association with symptoms and cognitive function. METHODS The participants were 149 patients with schizophrenia, 115 patients with bipolar disorder (BD), 186 unremitted patients with major depressive disorder (MDD), and 350 healthy controls. Psychiatric (schizophrenic, manic, and depressive) symptoms, sleep state, and cognitive (premorbid intelligence quotient, general cognitive, and memory) functions were evaluated. A multiplex immunoassay kit was used to measure cerebrospinal fluid (CSF) and plasma α-melanocyte-stimulating hormone (MSH), β-endorphin, neurotensin, oxytocin, and substance P levels. RESULTS The verification assay revealed that CSF α-MSH, β-endorphin, neurotensin, oxytocin, and substance P levels were too low to be reliably measured, while plasma α-MSH, β-endorphin, neurotensin, oxytocin, and substance P levels could be successfully measured. Plasma α-MSH, β-endorphin, neurotensin, oxytocin, and substance P levels were not significantly different between patients with schizophrenia, BD, or MDD and healthy controls. Plasma α-MSH, β-endorphin, neurotensin, oxytocin, and substance P levels were not significantly correlated with psychiatric symptom scores in patients with schizophrenia, BD, or MDD and cognitive function scores in patients or healthy controls. CONCLUSION Our data suggest that plasma neuropeptide levels do not elucidate the involvement of neuropeptides in the pathology of schizophrenia, BD, or MDD.
Collapse
Affiliation(s)
- Shinsuke Hidese
- Department of Psychiatry, Teikyo University School of Medicine, Itabashi-ku, Japan.,Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Fuyuko Yoshida
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Ikki Ishida
- Department of Psychiatry, Teikyo University School of Medicine, Itabashi-ku, Japan.,Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Junko Matsuo
- Department of Psychiatry, National Center Hospital, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Kotaro Hattori
- Medical Genome Center, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Hiroshi Kunugi
- Department of Psychiatry, Teikyo University School of Medicine, Itabashi-ku, Japan.,Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| |
Collapse
|
10
|
Li Q, Jiang B, Zhang Z, Huang Y, Xu Z, Chen X, Huang Y, Jian J, Yan Q. α-MSH is partially involved in the immunomodulation of Nile tilapia (Oreochromis niloticus) antibacterial immunity. FISH & SHELLFISH IMMUNOLOGY 2022; 131:929-938. [PMID: 36343851 DOI: 10.1016/j.fsi.2022.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 06/16/2023]
Abstract
α-Melanocyte-stimulating hormone (α-MSH) is a well-studied neuropeptide controlling skin and hair color. Besides, numerous immunomodulation roles of α-MSH were recorded in humans and mice. However, the regulatory effects of α-MSH in teleost immunity haven't been well elucidated. In this study, several precursor molecules of α-MSH (POMCs) and its receptors (MCRs) in Nile tilapia (Oreochromis niloticus) were characterized, and their expression characteristics and specific functions on antibacterial immunity were determined. Overall, POMCs and MCRs were principally detected in the brain, skin, and liver, and were remarkably promoted post Streptococcus agalactiae infection. However, tiny POMCs and MCRs were observed in tilapia immune organs (head kidney and spleen) or lymphocytes, and no evident immunomodulation effect was detected in vitro. Moreover, the in vivo challenge experiments revealed that α-MSH protects tilapia from bacterial infection by regulating responses in the brain and intestine. This study lays theoretical data for a deeper comprehension of the immunomodulation mechanisms of teleost α-MSH and the evolutional process of the vertebrate melanocortin system.
Collapse
Affiliation(s)
- Qi Li
- Fisheries College, Jimei University, Xiamen, China; College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Baijian Jiang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Zhiqiang Zhang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Yongxiong Huang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Zhou Xu
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Xinjin Chen
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China
| | - Yu Huang
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China
| | - Jichang Jian
- College of Fishery, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China.
| | - Qingpi Yan
- Fisheries College, Jimei University, Xiamen, China.
| |
Collapse
|
11
|
Hussein SZ, Abdalla MA. Serum levels of alpha-melanocyte stimulating hormone, vitamin D, calcium, phosphorus and magnesium in COVID-19 patients. UKRAINIAN BIOCHEMICAL JOURNAL 2021. [DOI: 10.15407/ubj93.06.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
12
|
Zhao J, Jiang L, Uehara M, Banouni N, Al Dulaijan BS, Azzi J, Ichimura T, Li X, Jarolim P, Fiorina P, Tullius SG, Madsen JC, Kasinath V, Abdi R. ACTH treatment promotes murine cardiac allograft acceptance. JCI Insight 2021; 6:e143385. [PMID: 34236047 PMCID: PMC8410061 DOI: 10.1172/jci.insight.143385] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 06/02/2021] [Indexed: 12/13/2022] Open
Abstract
Heart transplantation is the optimal therapy for patients with end-stage heart disease, but its long-term outcome remains inadequate. Recent studies have highlighted the importance of the melanocortin receptors (MCRs) in inflammation, but how MCRs regulate the balance between alloreactive T cells and Tregs, and whether they impact chronic heart transplant rejection, is unknown. Here, we found that Tregs express MC2R, and MC2R expression was highest among all MCRs by Tregs. Our data indicate that adrenocorticotropic hormone (ACTH), the sole ligand for MC2R, promoted the formation of Tregs by increasing the expression of IL-2Rα (CD25) in CD4+ T cells and activation of STAT5 in CD4+CD25+ T cells. ACTH treatment also improved the survival of heart allografts and increased the formation of Tregs in CD28KO mice. ACTH treatment synergized with the tolerogenic effect of CTLA-4–Ig, resulting in long-term survival of heart allografts and an increase in intragraft Tregs. ACTH administration also demonstrated higher prolongation of heart allograft survival in transgenic mouse recipients with both complete KO and conditional KO of PI3Kγ in T cells. Finally, ACTH treatment reduced chronic rejection markedly. These data demonstrate that ACTH treatment improved heart transplant outcomes, and this effect correlated with an increase in Tregs.
Collapse
Affiliation(s)
- Jing Zhao
- Transplantation Research Center.,Renal Division, and
| | - Liwei Jiang
- Transplantation Research Center.,Renal Division, and
| | - Mayuko Uehara
- Transplantation Research Center.,Renal Division, and
| | - Naima Banouni
- Transplantation Research Center.,Renal Division, and
| | | | - Jamil Azzi
- Transplantation Research Center.,Renal Division, and
| | | | - Xiaofei Li
- Transplantation Research Center.,Renal Division, and
| | - Petr Jarolim
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Paolo Fiorina
- Department of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,International Center for Type 1 Diabetes, Centro di Ricerca Pediatrica Romeo ed Enrica Invernizzi, Dipartimento di Scienze Biomediche e Cliniche "L. Sacco", Università di Milano, Milan, Italy.,Endocrinology Division, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Stefan G Tullius
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Joren C Madsen
- Center for Transplantation Sciences, Department of Surgery, and.,Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Reza Abdi
- Transplantation Research Center.,Renal Division, and
| |
Collapse
|
13
|
Review: Pro-inflammatory cytokines and hypothalamic inflammation: implications for insufficient feed intake of transition dairy cows. Animal 2020; 14:s65-s77. [PMID: 32024569 PMCID: PMC7003138 DOI: 10.1017/s1751731119003124] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Improvements in feed intake of dairy cows entering the early lactation period potentially decrease the risk of metabolic disorders, but before developing approaches targeting the intake level, mechanisms controlling and dysregulating energy balance and feed intake need to be understood. This review focuses on different inflammatory pathways interfering with the neuroendocrine system regulating feed intake of periparturient dairy cows. Subacute inflammation in various peripheral organs often occurs shortly before or after calving and is associated with increased pro-inflammatory cytokine levels. These cytokines are released into the circulation and sensed by neurons located in the hypothalamus, the key brain region regulating energy balance, to signal reduction in feed intake. Besides these peripheral humoral signals, glia cells in the brain may produce pro-inflammatory cytokines independent of peripheral inflammation. Preliminary results show intensive microglia activation in early lactation, suggesting their involvement in hypothalamic inflammation and the control of feed intake of dairy cows. On the other hand, pro-inflammatory cytokine-induced activation of the vagus nerve transmits signalling to the brain, but this pathway seems not exclusively necessary to signal feed intake reduction. Yet, less studied in dairy cows so far, the endocannabinoid system links inflammation and the hypothalamic control of feed intake. Distinct endocannabinoids exert anti-inflammatory action but also stimulate the posttranslational cleavage of neuronal proopiomelanocortin towards β-endorphin, an orexigen promoting feed intake. Plasma endocannabinoid concentrations and hypothalamic β-endorphin levels increase from late pregnancy to early lactation, but less is known about the regulation of the hypothalamic endocannabinoid system during the periparturient period of dairy cows. Dietary fatty acids may modulate the formation of endocannabinoids, which opens new avenues to improve metabolic health and immune status of dairy cows.
Collapse
|
14
|
Moscowitz AE, Asif H, Lindenmaier LB, Calzadilla A, Zhang C, Mirsaeidi M. The Importance of Melanocortin Receptors and Their Agonists in Pulmonary Disease. Front Med (Lausanne) 2019; 6:145. [PMID: 31316990 PMCID: PMC6610340 DOI: 10.3389/fmed.2019.00145] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/10/2019] [Indexed: 12/13/2022] Open
Abstract
Melanocortin agonists are ancient neuropeptides that have steroidogenesis and anti-inflammatory properties. They activate melanocortin receptors (MCR), a family of five seven-transmembrane G-protein coupled receptors. MC1R and MC3R are mainly involved in immunomodulatory effects. Adrenocorticotropin hormone (ACTH) and alpha-Melanocortin stimulating hormone (α-MSH) reduce pro-inflammatory cytokines in several pulmonary inflammatory disorders including asthma, sarcoidosis, and the acute respiratory distress syndrome. They have also been shown to reduce fibrogenesis in animal models with pulmonary fibrosis. By understanding the functions of MCR in macrophages, T-helper cell type 1, and T-helper cell type 17, we may uncover the mechanism of action of melanocortin agonists in sarcoidosis. Further translational and clinical research is needed to define the role of ACTH and α-MSH in pulmonary diseases.
Collapse
Affiliation(s)
| | - Huda Asif
- Division of Pulmonary and Critical Care, University of Miami, Miami, FL, United States
| | | | - Andrew Calzadilla
- Division of Pulmonary and Critical Care, University of Miami, Miami, FL, United States
| | - Chongxu Zhang
- Section of Pulmonary, Miami VA Healthcare System, Miami, FL, United States
| | - Mehdi Mirsaeidi
- Division of Pulmonary and Critical Care, University of Miami, Miami, FL, United States.,Section of Pulmonary, Miami VA Healthcare System, Miami, FL, United States
| |
Collapse
|
15
|
Chen S, Zuo Y, Huang L, Sherchan P, Zhang J, Yu Z, Peng J, Zhang J, Zhao L, Doycheva D, Liu F, Zhang JH, Xia Y, Tang J. The MC 4 receptor agonist RO27-3225 inhibits NLRP1-dependent neuronal pyroptosis via the ASK1/JNK/p38 MAPK pathway in a mouse model of intracerebral haemorrhage. Br J Pharmacol 2019; 176:1341-1356. [PMID: 30811584 DOI: 10.1111/bph.14639] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 01/07/2019] [Accepted: 02/05/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Inflammasome-mediated pyroptosis is an important neuronal cell death mechanism. Previous studies reported that activation of melanocortin MC4 receptor exerted neuroprotection in several neurological diseases. Here, we have investigated the role of MC4 receptor activation with RO27-3225 in suppressing neuronal pyroptosis after experimental intracerebral haemorrhage (ICH) and the underlying mechanism. EXPERIMENTAL APPROACH One hundred and sixty-nine male CD1 mice were used. ICH was induced by injection of bacterial collagenase into the right-side basal ganglia. RO27-3225, a selective agonist of MC4 receptor, was injected intraperitoneally at 1 hr after ICH. To elucidate the underlying mechanism, we used the specific MC4 receptor antagonist HS024 and NQDI-1, a specific inhibitor of the apoptosis signalling-regulating kinase 1 (ASK1). Neurological tests, Western blot, Fluoro-Jade C, TUNEL, and immunofluorescence staining were conducted. KEY RESULTS Expression of MC4 receptor and the NOD-like receptor family, pyrin domain containing 1 (NLRP1) inflammasome in brain were increased after ICH. RO27-3225 treatment decreased neuronal pyroptosis and neurobehavioural deficits at 24 and 72 hr after ICH. RO27-3225 reduced the expression of p-ASK1, p-JNK, p-p38 MAPK, NLRP1 inflammasome, cleaved caspase-1, and IL-1β after ICH. HS024 pretreatment prevented the effects of RO27-3225. Similar to RO27-3225, NQDI-1 alone improved neurological functions and down-regulated ASK1/JNK/p38MAPK expression after ICH. CONCLUSIONS AND IMPLICATIONS RO27-3225 suppressed NLRP1-dependent neuronal pyroptosis and improved neurological function, possibly mediated by activation of MC4 receptor and inhibition of ASK1/JNK/p38 MAPK signalling pathways, after experimental ICH in mice. The MC4 receptor may be a promising therapeutic target for the management of ICH.
Collapse
Affiliation(s)
- Shengpan Chen
- Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou, China.,Department of Physiology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Yuchun Zuo
- Department of Physiology, School of Medicine, Loma Linda University, Loma Linda, CA, USA.,Department of Neurosurgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Lei Huang
- Department of Physiology, School of Medicine, Loma Linda University, Loma Linda, CA, USA.,Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA, USA
| | - Prativa Sherchan
- Department of Physiology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Jian Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhengtao Yu
- Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou, China
| | - Jianhua Peng
- Department of Physiology, School of Medicine, Loma Linda University, Loma Linda, CA, USA.,Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Junyi Zhang
- Department of Physiology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Lianhua Zhao
- Department of Physiology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Desislava Doycheva
- Department of Physiology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Fei Liu
- Department of Neurosurgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - John H Zhang
- Department of Physiology, School of Medicine, Loma Linda University, Loma Linda, CA, USA.,Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA, USA
| | - Ying Xia
- Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou, China
| | - Jiping Tang
- Department of Physiology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
16
|
Wang W, Guo DY, Lin YJ, Tao YX. Melanocortin Regulation of Inflammation. Front Endocrinol (Lausanne) 2019; 10:683. [PMID: 31649620 PMCID: PMC6794349 DOI: 10.3389/fendo.2019.00683] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/19/2019] [Indexed: 12/18/2022] Open
Abstract
Adrenocorticotropic hormone (ACTH), and α-, β-, and γ-melanocyte-stimulating hormones (α-, β-, γ-MSH), collectively known as melanocortins, together with their receptors (melanocortin receptors), are components of an ancient modulatory system. The clinical use of ACTH in the treatment of rheumatoid arthritis started in 1949, originally thought that the anti-inflammatory action was through hypothalamus-pituitary-adrenal axis and glucocorticoid-dependent. Subsequent decades have witnessed extensive attempts in unraveling the physiology and pharmacology of the melanocortin system. It is now known that ACTH, together with α-, β-, and γ-MSHs, also possess glucocorticoid-independent anti-inflammatory and immunomodulatory effects by activating the melanocortin receptors expressed in the brain or peripheral immune cells. This review will briefly introduce the melanocortin system and highlight the action of melanocortins in the regulation of immune functions from in vitro, in vivo, preclinical, and clinical studies. The potential therapeutic use of melanocortins are also summarized.
Collapse
Affiliation(s)
- Wei Wang
- Department of Clinical Laboratory, Xiamen Huli Guoyu Clinic, Co., Ltd., Xiamen, China
| | - Dong-Yu Guo
- Department of Clinical Laboratory, Xiamen Huli Guoyu Clinic, Co., Ltd., Xiamen, China
- *Correspondence: Dong-Yu Guo
| | - Yue-Jun Lin
- Department of Clinical Laboratory, Xiamen Huli Guoyu Clinic, Co., Ltd., Xiamen, China
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
- Ya-Xiong Tao
| |
Collapse
|
17
|
Blough B, Namjoshi O. Small Molecule Neuropeptide S and Melanocortin 4 Receptor Ligands as Potential Treatments for Substance Use Disorders. Handb Exp Pharmacol 2019; 258:61-87. [PMID: 31628605 DOI: 10.1007/164_2019_313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There is a vital need for novel approaches and biological targets for drug discovery and development. Treatment strategies for substance use disorders (SUDs) to date have been mostly ineffective other than substitution-like therapeutics. Two such targets are the peptide G-protein-coupled receptors neuropeptide S (NPS) and melanocortin 4 (MC4). Preclinical evidence suggests that antagonists, inverse agonists, or negative allosteric modulators of these receptors might be novel therapeutics for SUDs. NPS is a relatively unexplored receptor with high potential for treating SUD. MC4 has a strong link to early-onset obesity, and emerging evidence suggests significant overlap between food-maintained and drug-maintained behaviors making MC4 an intriguing target for SUD. This chapter provides an overview of the literature in relation to the roles of NPS and MC4 in drug-seeking behaviors and then provides a medicinal chemistry-based survey of the small molecule ligands for each receptor.
Collapse
Affiliation(s)
- Bruce Blough
- Center for Drug Discovery, RTI International, Research Triangle Park, NC, USA.
| | - Ojas Namjoshi
- Center for Drug Discovery, RTI International, Research Triangle Park, NC, USA
| |
Collapse
|
18
|
Feng G, Feng J, Zhang S, Tong Y, Zhang Q, Yang X, Zhang H. Altered levels of α-melanocyte stimulating hormone in cerebrospinal fluid and plasma of patients with traumatic brain injury. Brain Res 2018; 1696:22-30. [PMID: 29859146 DOI: 10.1016/j.brainres.2018.05.044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 05/07/2018] [Accepted: 05/27/2018] [Indexed: 10/16/2022]
Abstract
Traumatic brain injury (TBI) is closely associated with marked inflammation. Although alpha-Melanocyte-Stimulating Hormone (α-MSH) exerts powerful anti-inflammatory effects, changes in endogenous α-MSH levels following TBI remain poorly understood. We investigated the changes of α-MSH levels in the cerebrospinal fluid (CSF) and plasma of post-TBI patients and the association of these changes with the severity of TBI and inflammation. TBI severity was assessed by the GCS coma scale from which, patients were separated into three groups. Clinical data were collected on days 1, 3, 5, and 7 including levels of α-MSH, tumor necrosis factor (TNF-α), and intracranial pressure (ICP). α-MSH levels in CSF steadily increased for one week (peak at day 5) but plasma α-MSH decreased and remained low. These changes were more substantial in the Severe Group of TBI with lower GCS. TNF-α levels were similarly increased in both CSF and plasma (peak at day 3). In the early phase of TBI elevated TNF-α and ICP dominated, and CSF α-MSH displayed a slow and insufficient increase. In later phases of TBI, TNF-α and ICP levels were alleviated concordantly with sustained increases in central α-MSH, wherein an anti-inflammatory environment might predominate. The relationship between plasma α-MSH and TNF-α showed significant negative correlation, and the relationship between CSF α-MSH and TNF-α showed significant positive correlation with a two-day lag. In conclusion, plasma α-MSH levels decreased, but CSF levels increased slowly following TBI. These changes were more substantial in severe patients with a lower GCS. Increases in central α-MSH paralleled alleviation of inflammation.
Collapse
Affiliation(s)
- Guilong Feng
- Department of Emergency, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Jie Feng
- Department of Emergency, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Shaojun Zhang
- Department of Endocrinology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Yuping Tong
- Department of Emergency, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Qiang Zhang
- Department of Emergency, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaoming Yang
- Department of Emergency, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Huaping Zhang
- Molecular Biology Laboratories, Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
19
|
Liu G, Li M, Saeed M, Xu Y, Ren Q, Sun C. αMSH inhibits adipose inflammation via reducing FoxOs transcription and blocking Akt/JNK pathway in mice. Oncotarget 2018; 8:47642-47654. [PMID: 28514752 PMCID: PMC5564594 DOI: 10.18632/oncotarget.17465] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 04/07/2017] [Indexed: 12/21/2022] Open
Abstract
Alpha melanocyte stimulating hormone (αMSH) abates inflammation in multiple tissues, while Forkhead box proteins O (FoxOs) stimulate inflammatory cascade. However, the relationship between αMSH and FoxOs in adipose inflammation remains unclear. In this study, we used LPS-induced inflammation model, attempted to interpret the function of αMSH in inflammation and the interactions with FoxOs. Results indicated that upon inflammatory situation, the secretion of αMSH and the expression of its receptor MC5R were greatly decreased, but FoxOs expressions were elevated. After the treatment with αMSH, LPS-induced adipose inflammation together with FoxOs expressions was significantly reduced. Conversely, when Foxo1, Foxo3a or Foxo4 overexpressed in αMSH treated inflammatory mouse model, all the anti-inflammatory impacts of αMSH were found disappeared. We further studied the mechanisms by which αMSH exerts its anti-inflammatory impacts and how FoxOs reverse αMSH's function. Foxo4 was found as a negative regulator for MC5R transcription in αMSH inhibited inflammation. Moreover, a negative role was found of αMSH in regulating both Akt and JNK signal pathways by observing the enhanced the anti-inflammatory impacts of pathway-specific inhibitors with αMSH treatment. Our findings demonstrate αMSH plays a key role in the prevention of adipose inflammation and inflammatory diseases by down-regulating Akt/JNK signal pathway and negatively interacting with FoxOs, which brings up αMSH as a novel candidate factor in the adipose anti-inflammation process in obesity.
Collapse
Affiliation(s)
- Guannv Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Meihang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Muhammad Saeed
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yatao Xu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Qian Ren
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Chao Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| |
Collapse
|
20
|
Kreutzer C, Peters S, Schulte DM, Fangmann D, Türk K, Wolff S, van Eimeren T, Ahrens M, Beckmann J, Schafmayer C, Becker T, Kerby T, Rohr A, Riedel C, Heinsen FA, Degenhardt F, Franke A, Rosenstiel P, Zubek N, Henning C, Freitag-Wolf S, Dempfle A, Psilopanagioti A, Petrou-Papadaki H, Lenk L, Jansen O, Schreiber S, Laudes M. Hypothalamic Inflammation in Human Obesity Is Mediated by Environmental and Genetic Factors. Diabetes 2017; 66:2407-2415. [PMID: 28576837 DOI: 10.2337/db17-0067] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 05/24/2017] [Indexed: 11/13/2022]
Abstract
Obesity is associated with hypothalamic inflammation (HI) in animal models. In the current study, we examined the mediobasal hypothalamus (MBH) of 57 obese human subjects and 54 age- and sex- matched nonobese control subjects by MRI and analyzed the T2 hyperintensity as a measure of HI. Obese subjects exhibited T2 hyperintensity in the left but not the right MBH, which was strongly associated with systemic low-grade inflammation. MRS revealed the number of neurons in the left hypothalamic region to be similar in obese versus control subjects, suggesting functional but not structural impairment due to the inflammatory process. To gain mechanistic insights, we performed nutritional analysis and 16S rDNA microbiome sequencing, which showed that high-fat diet induces reduction of Parasutterella sp. in the gut, which is significantly correlated with MBH T2 hyperintensity. In addition to these environmental factors, we found subjects carrying common polymorphisms in the JNK or the MC4R gene to be more susceptible to HI. Finally, in a subgroup analysis, bariatric surgery had no effect on MBH T2 hyperintensity despite inducing significant weight loss and improvement of peripheral insulin sensitivity. In conclusion, obesity in humans is associated with HI and disturbances in the gut-brain axis, which are influenced by both environmental and genetic factors.
Collapse
Affiliation(s)
| | - Sönke Peters
- Department of Radiology and Neuroradiology, University of Kiel, Kiel, Germany
| | | | | | - Kathrin Türk
- Department of Medicine 1, University of Kiel, Kiel, Germany
| | - Stephan Wolff
- Department of Radiology and Neuroradiology, University of Kiel, Kiel, Germany
| | - Thilo van Eimeren
- Department of Nuclear Medicine, University of Cologne, Cologne, Germany
| | - Markus Ahrens
- Department of General, Visceral, Thoracic, Transplantation, and Pediatric Surgery, University of Kiel, Kiel, Germany
| | - Jan Beckmann
- Department of General, Visceral, Thoracic, Transplantation, and Pediatric Surgery, University of Kiel, Kiel, Germany
| | - Clemens Schafmayer
- Department of General, Visceral, Thoracic, Transplantation, and Pediatric Surgery, University of Kiel, Kiel, Germany
| | - Thomas Becker
- Department of General, Visceral, Thoracic, Transplantation, and Pediatric Surgery, University of Kiel, Kiel, Germany
| | - Tina Kerby
- Department of Radiology and Neuroradiology, University of Kiel, Kiel, Germany
| | - Axel Rohr
- Department of Radiology and Neuroradiology, University of Kiel, Kiel, Germany
| | - Christian Riedel
- Department of Radiology and Neuroradiology, University of Kiel, Kiel, Germany
| | | | - Frauke Degenhardt
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | - Nana Zubek
- Department of Agricultural Politics, University of Kiel, Kiel, Germany
| | - Christian Henning
- Department of Agricultural Politics, University of Kiel, Kiel, Germany
| | - Sandra Freitag-Wolf
- Institute of Medical Informatics and Statistics, University of Kiel, Kiel, Germany
| | - Astrid Dempfle
- Institute of Medical Informatics and Statistics, University of Kiel, Kiel, Germany
| | | | - Helen Petrou-Papadaki
- Department of Anatomy, Histology and Embryology, University of Patras, Patras, Greece
| | - Lennart Lenk
- Institute for Experimental Cancer Research, University of Kiel, Kiel, Germany
| | - Olav Jansen
- Department of Radiology and Neuroradiology, University of Kiel, Kiel, Germany
| | - Stefan Schreiber
- Department of Medicine 1, University of Kiel, Kiel, Germany
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | | |
Collapse
|
21
|
Central Modulation of Neuroinflammation by Neuropeptides and Energy-Sensing Hormones during Obesity. BIOMED RESEARCH INTERNATIONAL 2017; 2017:7949582. [PMID: 28913358 PMCID: PMC5587954 DOI: 10.1155/2017/7949582] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/19/2017] [Accepted: 07/24/2017] [Indexed: 02/06/2023]
Abstract
Central nervous system (CNS) senses energy homeostasis by integrating both peripheral and autonomic signals and responding to them by neurotransmitters and neuropeptides release. Although it is previously considered an immunologically privileged organ, we now know that this is not so. Cells belonging to the immune system, such as B and T lymphocytes, can be recruited into the CNS to face damage or infection, in addition to possessing resident immunological cells, called microglia. In this way, positive energy balance during obesity promotes an inflammatory state in the CNS. Saturated fatty acids from the diet have been pointed out as powerful candidates to trigger immune response in peripheral system and in the CNS. However, how central immunity communicates to peripheral immune response remains to be clarified. Recently there has been a great interest in the neuropeptides, POMC derived peptides, ghrelin, and leptin, due to their capacity to suppress or induce inflammatory responses in the brain, respectively. These may be potential candidates to treat different pathologies associated with autoimmunity and inflammation. In this review, we will discuss the role of lipotoxicity associated with positive energy balance during obesity in proinflammatory response in microglia, B and T lymphocytes, and its modulation by neuropeptides.
Collapse
|
22
|
Myung G, Nelson WW, McMahon MA. Effects of Repository Corticotropin Injection on Medication Use in Patients With Rheumatologic Conditions: A Claims Data Study. J Pharm Technol 2017; 33:151-155. [PMID: 34861677 DOI: 10.1177/8755122517709825] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background: Currently, specific studies identifying how repository corticotropin injection (RCI) is used in rheumatologic conditions are lacking. This is a first step to familiarize the trends of demographics using RCI as well as other medication use. Objective: RCI may produce anti-inflammatory as well as immune-modulatory effects. The purpose of this study is to examine the demographics of those who use RCI and the change in medication use, specifically prednisone, after RCI initiation. Method: This study used the Symphony Health Solutions (SHA) Claims database from 2008 to 2015. International Classification of Disease, Ninth Revision, codes were used to identify rheumatologic conditions including rheumatoid arthritis, systemic lupus erythematosus, dermatomyositis, and polymyositis. Information including RCI dose and concomitant medication uses was also obtained. Results: A total of 2749 patients with rheumatologic conditions receiving RCI were investigated for demographic information, and a total of 1048 patients with rheumatologic conditions on RCI were examined for medication use. The use of nonsteroidal anti-inflammatory drugs, disease-modifying anti-rheumatic drugs, and biologics overall decreased significantly in all 3 rheumatologic conditions except biologics in dermatomyositis/polymyositis. In addition, mean prednisone dose before and after RCI use significantly decreased one quarter (12 weeks) after RCI initiation. Conclusion: Claims-based study on RCI use indicates that RCI use might reduce use of prednisone, disease-modifying anti-rheumatic drugs, and other biologics. Further prospective study is needed.
Collapse
|
23
|
Melanocortin Receptor-4 Gene Polymorphisms in Glioblastoma Patients Treated with Concomitant Radio-Chemotherapy. Mol Neurobiol 2017; 55:1396-1404. [DOI: 10.1007/s12035-017-0414-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 01/18/2017] [Indexed: 10/20/2022]
|
24
|
Singh RK, Kumar P, Mahalingam K. Molecular genetics of human obesity: A comprehensive review. C R Biol 2017; 340:87-108. [PMID: 28089486 DOI: 10.1016/j.crvi.2016.11.007] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 10/03/2016] [Accepted: 11/10/2016] [Indexed: 12/25/2022]
Abstract
Obesity and its related health complications is a major problem worldwide. Hypothalamus and their signalling molecules play a critical role in the intervening and coordination with energy balance and homeostasis. Genetic factors play a crucial role in determining an individual's predisposition to the weight gain and being obese. In the past few years, several genetic variants were identified as monogenic forms of human obesity having success over common polygenic forms. In the context of molecular genetics, genome-wide association studies (GWAS) approach and their findings signified a number of genetic variants predisposing to obesity. However, the last couple of years, it has also been noticed that alterations in the environmental and epigenetic factors are one of the key causes of obesity. Hence, this review might be helpful in the current scenario of molecular genetics of human obesity, obesity-related health complications (ORHC), and energy homeostasis. Future work based on the clinical discoveries may play a role in the molecular dissection of genetic approaches to find more obesity-susceptible gene loci.
Collapse
Affiliation(s)
- Rajan Kumar Singh
- Department of Bio-Medical Sciences, School of Biosciences and Technology, VIT University, 632014 Vellore, India
| | - Permendra Kumar
- Department of Bio-Medical Sciences, School of Biosciences and Technology, VIT University, 632014 Vellore, India
| | - Kulandaivelu Mahalingam
- Department of Bio-Medical Sciences, School of Biosciences and Technology, VIT University, 632014 Vellore, India.
| |
Collapse
|
25
|
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
|
26
|
Multiple beneficial effects of melanocortin MC 4 receptor agonists in experimental neurodegenerative disorders: Therapeutic perspectives. Prog Neurobiol 2016; 148:40-56. [PMID: 27916623 DOI: 10.1016/j.pneurobio.2016.11.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 11/22/2016] [Accepted: 11/28/2016] [Indexed: 12/13/2022]
Abstract
Melanocortin peptides induce neuroprotection in acute and chronic experimental neurodegenerative conditions. Melanocortins likewise counteract systemic responses to brain injuries. Furthermore, they promote neurogenesis by activating critical signaling pathways. Melanocortin-induced long-lasting improvement in synaptic activity and neurological performance, including learning and memory, sensory-motor orientation and coordinated limb use, has been consistently observed in experimental models of acute and chronic neurodegeneration. Evidence indicates that the neuroprotective and neurogenic effects of melanocortins, as well as the protection against systemic responses to a brain injury, are mediated by brain melanocortin 4 (MC4) receptors, through an involvement of the vagus nerve. Here we discuss the targets and mechanisms underlying the multiple beneficial effects recently observed in animal models of neurodegeneration. We comment on the potential clinical usefulness of melanocortin MC4 receptor agonists as neuroprotective and neuroregenerative agents in ischemic stroke, subarachnoid hemorrhage, traumatic brain injury, spinal cord injury, and Alzheimer's disease.
Collapse
|
27
|
[Nle4, D-Phe7]-α-MSH Inhibits Toll-Like Receptor (TLR)2- and TLR4-Induced Microglial Activation and Promotes a M2-Like Phenotype. PLoS One 2016; 11:e0158564. [PMID: 27359332 PMCID: PMC4928783 DOI: 10.1371/journal.pone.0158564] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 06/19/2016] [Indexed: 12/22/2022] Open
Abstract
α-melanocyte stimulating hormone (α-MSH) is an anti-inflammatory peptide, proved to be beneficial in many neuroinflammatory disorders acting through melanocortin receptor 4 (MC4R). We previously determined that rat microglial cells express MC4R and that NDP-MSH, an analog of α-MSH, induces PPAR-γ expression and IL-10 release in these cells. Given the great importance of modulation of glial activation in neuroinflammatory disorders, we tested the ability of NDP-MSH to shape microglial phenotype and to modulate Toll-like receptor (TLR)-mediated inflammatory responses. Primary rat cultured microglia were stimulated with NDP-MSH followed by the TLR2 agonist Pam3CSK4 or the TLR4 agonist LPS. NDP-MSH alone induced expression of the M2a/M2c marker Ag1 and reduced expression of the M2b marker Il-4rα and of the LPS receptor Tlr4. Nuclear translocation of NF-κB subunits p65 and c-Rel was induced by LPS and these effects were partially prevented by NDP-MSH. NDP-MSH reduced LPS- and Pam3CSK4-induced TNF-α release but did not affect TLR-induced IL-10 release. Also, NDP-MSH inhibited TLR2-induced HMGB1 translocation from nucleus to cytoplasm and TLR2-induced phagocytic activity. Our data show that NDP-MSH inhibits TLR2- and TLR4-mediated proinflammatory mechanisms and promotes microglial M2-like polarization, supporting melanocortins as useful tools for shaping microglial activation towards an alternative immunomodulatory phenotype.
Collapse
|
28
|
Gómez-SanMiguel AB, Villanúa MÁ, Martín AI, López-Calderón A. D-TRP(8)-γMSH Prevents the Effects of Endotoxin in Rat Skeletal Muscle Cells through TNFα/NF-KB Signalling Pathway. PLoS One 2016; 11:e0155645. [PMID: 27177152 PMCID: PMC4866687 DOI: 10.1371/journal.pone.0155645] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 05/02/2016] [Indexed: 02/07/2023] Open
Abstract
Sepsis induces anorexia and muscle wasting secondary to an increase in muscle proteolysis. Melanocyte stimulating hormones (MSH) is a family of peptides that have potent anti-inflammatory effects. Melanocortin receptor-3 (MC3-R) has been reported as the predominant anti-inflammatory receptor for melanocortins. The aim of this work was to analyse whether activation of MC3-R, by administration of its agonist D-Trp(8)-γMSH, is able to modify the response of skeletal muscle to inflammation induced by lipopolysaccharide endotoxin (LPS) or TNFα. Adult male rats were injected with 250 μg/kg LPS and/or 500 μg/kg D-Trp(8)-γMSH 17:00 h and at 8:00 h the following day, and euthanized 4 hours afterwards. D-Trp(8)-γMSH decreased LPS-induced anorexia and prevented the stimulatory effect of LPS on hypothalamic IL-1β, COX-2 and CRH as well as on serum ACTH and corticosterone. Serum IGF-I and its expression in liver and gastrocnemius were decreased in rats injected with LPS, but not in those that also received D-Trp(8)-γMSH. However, D-Trp(8)-γMSH was unable to modify the effect of LPS on IGFBP-3. In the gastrocnemius D-Trp(8)-γMSH blocked LPS-induced decrease in pAkt, pmTOR, MHC I and MCH II, as well as the increase in pNF-κB(p65), FoxO1, FoxO3, LC3b, Bnip-3, Gabarap1, atrogin-1, MuRF1 and in LC3a/b lipidation. In L6 myotube cultures, D-Trp(8)-γMSH was able to prevent TNFα-induced increase of NF-κB(p65) phosphorylation and decrease of Akt phosphorylation as well as of IGF-I and MHC I expression. These data suggest that MC3-R activation prevents the effect of endotoxin on skeletal wasting by modifying inflammation, corticosterone and IGF-I responses and also by directly acting on muscle cells through the TNFα/NF-κB(p65) pathway.
Collapse
Affiliation(s)
- Ana Belén Gómez-SanMiguel
- Department of Physiology, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - María Ángeles Villanúa
- Department of Physiology, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Ana Isabel Martín
- Department of Physiology, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Asunción López-Calderón
- Department of Physiology, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
- * E-mail:
| |
Collapse
|
29
|
Loram LC, Culp ME, Connolly-Strong EC, Sturgill-Koszycki S. Melanocortin peptides: potential targets in systemic lupus erythematosus. Inflammation 2015; 38:260-71. [PMID: 25323206 PMCID: PMC4312383 DOI: 10.1007/s10753-014-0029-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune disease resulting in loss of self-tolerance with multiple organs, such as the kidney, skin, joints, and the central nervous system (CNS), being targeted. Numerous immunosuppressant therapies are currently being used for the treatment of SLE, but their clinical utility is somewhat variable because of the clinical heterogeneity. Melanocortins are a family of peptides derived from the common precursor protein pro-opiomelanocortin. These multifunctional peptides activate five subtypes of melanocortin receptors expressed on immune, skin, muscle, bone, and kidney cells and cells within the CNS. Melanocortin peptides have demonstrated a variety of biologic actions including immunomodulation, melanogenesis, and renoprotection. This review aims to introduce the melanocortin system and explore the mechanisms by which they may be beneficial in diseases such as SLE.
Collapse
Affiliation(s)
- Lisa Carole Loram
- Mallinckrodt Pharmaceuticals (formerly Questcor), 26118 Research Road, Hayward, CA, 94545, USA
| | | | | | | |
Collapse
|
30
|
Liu GS, Wu JC, Tsai HE, Dusting GJ, Chan EC, Wu CS, Tai MH. Proopiomelanocortin gene delivery induces apoptosis in melanoma through NADPH oxidase 4-mediated ROS generation. Free Radic Biol Med 2014; 70:14-22. [PMID: 24412703 DOI: 10.1016/j.freeradbiomed.2013.12.024] [Citation(s) in RCA: 11] [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: 08/26/2013] [Revised: 12/03/2013] [Accepted: 12/21/2013] [Indexed: 12/31/2022]
Abstract
Hypoxia in the tumor microenvironment triggers differential signaling pathways for tumor survival. In this study, we characterize the involvement of hypoxia and reactive oxygen species (ROS) generation in the antineoplastic mechanism of proopiomelanocortin (POMC) gene delivery in a mouse B16-F10 melanoma model in vivo and in vitro. Histological analysis revealed increased TUNEL-positive cells and enhanced hypoxic activities in melanoma treated with adenovirus encoding POMC (Ad-POMC) but not control vector. Because the apoptotic cells were detected mainly in regions distant from blood vessels, it was hypothesized that POMC therapy might render melanoma cells vulnerable to hypoxic insult. Using a hypoxic chamber or cobalt chloride (CoCl2), we showed that POMC gene delivery elicited apoptosis and caspase-3 activation in cultured B16-F10 cells only under hypoxic conditions. The apoptosis induced by POMC gene delivery was associated with elevated ROS generation in vitro and in vivo. Blocking ROS generation using the antioxidant N-acetyl-l-cysteine abolished the apoptosis and caspase-3 activities induced by POMC gene delivery and hypoxia. We further showed that POMC-derived melanocortins, including α-MSH, β-MSH, and ACTH, but not γ-MSH, contributed to POMC-induced apoptosis and ROS generation during hypoxia. To elucidate the source of ROS generation, application of the NADPH oxidase inhibitor diphenyleneiodonium attenuated α-MSH-induced apoptosis and ROS generation, implicating the proapoptotic role of NADPH oxidase in POMC action. Of the NADPH oxidase isoforms, only Nox4 was expressed in B16-F10 cells, and Nox4 was also elevated in Ad-POMC-treated melanoma tissues. Silencing Nox4 gene expression with Nox4 siRNA suppressed the stimulatory effect of α-MSH-induced ROS generation and cell apoptosis during hypoxia. In summary, we demonstrate that POMC gene delivery suppressed melanoma growth by inducing apoptosis, which was at least partly dependent on Nox4 upregulation.
Collapse
Affiliation(s)
- Guei-Sheung Liu
- Centre for Eye Research Australia, Melbourne, VIC 3002, Australia; Department of Ophthalmology, University of Melbourne, Melbourne, VIC 3002, Australia
| | - Jian-Ching Wu
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung 804, Taiwan
| | - Han-En Tsai
- Institute of Biomedical Science, and National Sun Yat-sen University, Kaohsiung 804, Taiwan
| | - Gregory J Dusting
- Centre for Eye Research Australia, Melbourne, VIC 3002, Australia; Department of Ophthalmology, University of Melbourne, Melbourne, VIC 3002, Australia
| | - Elsa C Chan
- Centre for Eye Research Australia, Melbourne, VIC 3002, Australia; Department of Ophthalmology, University of Melbourne, Melbourne, VIC 3002, Australia
| | - Chieh-Shan Wu
- Department of Dermatology, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
| | - Ming-Hong Tai
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung 804, Taiwan; Institute of Biomedical Science, and National Sun Yat-sen University, Kaohsiung 804, Taiwan; Center for Neuroscience, National Sun Yat-sen University, Kaohsiung 804, Taiwan.
| |
Collapse
|
31
|
Qu H, Li J, Chen W, Li Y, Jiang Q, Jiang H, Huo J, Zhao Z, Liu B, Zhang Q. Differential expression of the melanocortin-4 receptor in male and female C57BL/6J mice. Mol Biol Rep 2014; 41:3245-56. [DOI: 10.1007/s11033-014-3187-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 01/20/2014] [Indexed: 01/09/2023]
|
32
|
Temp FR, Santos AC, Marafiga JR, Jesse AC, Lenz QF, Oliveira SM, Guerra GP, Scimonelli TN, Mello CF. Alpha melanocyte stimulating hormone (α-MSH) does not modify pentylenetetrazol- and pilocarpine-induced seizures. Life Sci 2013; 93:723-31. [DOI: 10.1016/j.lfs.2013.09.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 08/09/2013] [Accepted: 09/04/2013] [Indexed: 12/30/2022]
|
33
|
Caruso C, Carniglia L, Durand D, Scimonelli TN, Lasaga M. Astrocytes: new targets of melanocortin 4 receptor actions. J Mol Endocrinol 2013; 51:R33-50. [PMID: 23881919 DOI: 10.1530/jme-13-0064] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Astrocytes exert a wide variety of functions with paramount importance in brain physiology. After injury or infection, astrocytes become reactive and they respond by producing a variety of inflammatory mediators that help maintain brain homeostasis. Loss of astrocyte functions as well as their excessive activation can contribute to disease processes; thus, it is important to modulate reactive astrocyte response. Melanocortins are peptides with well-recognized anti-inflammatory and neuroprotective activity. Although melanocortin efficacy was shown in systemic models of inflammatory disease, mechanisms involved in their effects have not yet been fully elucidated. Central anti-inflammatory effects of melanocortins and their mechanisms are even less well known, and, in particular, the effects of melanocortins in glial cells are poorly understood. Of the five known melanocortin receptors (MCRs), only subtype 4 is present in astrocytes. MC4R has been shown to mediate melanocortin effects on energy homeostasis, reproduction, inflammation, and neuroprotection and, recently, to modulate astrocyte functions. In this review, we will describe MC4R involvement in anti-inflammatory, anorexigenic, and anti-apoptotic effects of melanocortins in the brain. We will highlight MC4R action in astrocytes and discuss their possible mechanisms of action. Melanocortin effects on astrocytes provide a new means of treating inflammation, obesity, and neurodegeneration, making them attractive targets for therapeutic interventions in the CNS.
Collapse
Affiliation(s)
- Carla Caruso
- School of Medicine, Biomedical Research Institute (UBA-CONICET), University of Buenos Aires, Paraguay 2155 piso 10, 1121ABG Buenos Aires, Argentina IFEC (CONICET) Department of Pharmacology, School of Chemistry, National University of Córdoba, Córdoba, Argentina
| | | | | | | | | |
Collapse
|
34
|
Spaccapelo L, Galantucci M, Neri L, Contri M, Pizzala R, D'Amico R, Ottani A, Sandrini M, Zaffe D, Giuliani D, Guarini S. Up-regulation of the canonical Wnt-3A and Sonic hedgehog signaling underlies melanocortin-induced neurogenesis after cerebral ischemia. Eur J Pharmacol 2013; 707:78-86. [PMID: 23535605 DOI: 10.1016/j.ejphar.2013.03.030] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 03/05/2013] [Accepted: 03/07/2013] [Indexed: 12/31/2022]
Abstract
In experimental cerebral ischemia, melanocortin MC4 receptor agonists induce neuroprotection and neurogenesis with subsequent long-lasting functional recovery. Here we investigated the molecular mechanisms underlying melanocortin-induced neurogenesis. Gerbils were subjected to transient global cerebral ischemia, then they were treated every 12 h, and until sacrifice, with 5-bromo-2'-deoxyuridine (BrdU; to label proliferating cells), and the melanocortin analog [Nle(4),d-Phe(7)]α-melanocyte-stimulating hormone (NDP-α-MSH) or saline. NDP-α-MSH increased hippocampus dentate gyrus (DG) expression of Wnt-3A, β-catenin, Sonic hedgehog (Shh), Zif268, interleukin-10 (IL-10) and doublecortin (DCX), as detected at days 3, 6 and 10 after the ischemic insult. Further, an elevated number of BrdU immunoreactive cells was found at days 3 and 10, and an improved histological picture with reduced neuronal loss at day 10, associated with learning and memory recovery. Pharmacological blockade of the Wnt-3A/β-catenin and Shh pathways, as well as of melanocortin MC4 receptors, prevented all effects of NDP-α-MSH. These data indicate that, in experimental brain ischemia, treatment with melanocortins acting at MC4 receptors induces neural stem/progenitor cell proliferation in the DG by promptly and effectively triggering the canonical Wnt-3A/β-catenin and Shh signaling pathways. Activation of these pathways is associated with up-regulation of the repair factor Zif268 and the neurogenesis facilitating factor IL-10, and it seems to address mainly toward a neuronal fate, as indicated by the increase in DCX positive cells.
Collapse
Affiliation(s)
- Luca Spaccapelo
- Department of Biomedical, Metabolic and Neural Sciences, Section of Pharmacology and Molecular Medicine, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Carniglia L, Durand D, Caruso C, Lasaga M. Effect of NDP-α-MSH on PPAR-γ and -β expression and anti-inflammatory cytokine release in rat astrocytes and microglia. PLoS One 2013; 8:e57313. [PMID: 23468969 PMCID: PMC3582497 DOI: 10.1371/journal.pone.0057313] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 01/21/2013] [Indexed: 12/25/2022] Open
Abstract
Brain inflammation plays a central role in numerous brain pathologies. Microglia and astrocytes are the main effector cells that become activated when an inflammatory process takes place within the central nervous system. α-melanocyte-stimulating hormone (α-MSH) is a neuropeptide with proven anti-inflammatory properties. It binds with highest affinity to the melanocortin receptor 4 (MC4R), which is present in astrocytes and upon activation triggers anti-inflammatory pathways. The aim of this research was to identify anti-inflammatory mediators that may participate in the immunomodulatory effects of melanocortins in glial cells. Since peroxisome proliferator-activated receptors (PPARs) have recently been implicated in the modulation of inflammation, we investigated the effect of an α-MSH analog, [Nle(4), D-Phe(7)]-α-MSH (NDP-α-MSH), on PPAR-β and PPAR-γ gene and protein expression in rat primary astrocytes and microglia. We initially demonstrated that rat primary microglia express MC4R and showed that treatment with NDP-α-MSH increases PPAR-γ protein levels and strongly decreases PPAR-β levels in both astrocytes and microglia. We also showed that extracellular signal-regulated kinase 1/2 (ERK1/2)-mediated signaling is partially involved in these effects in a cell-specific fashion. Finally, we showed that NDP-α-MSH stimulates the release of the anti-inflammatory cytokines IL-10 and TGF-β from microglia and astrocytes, respectively. The presented data suggest a role for IL-10 and TGF-β in the protective action of melanocortins and a connection between MC4R pathway and that of the nuclear receptor PPAR-γ. This is the first report providing evidence that MC4R is expressed in rat primary microglia and that melanocortins modulate PPAR levels in glial cells. Our findings provide new insights into the mechanisms underlying the activation of glial MC4R and open perspectives for new therapeutic strategies for the treatment of inflammation-mediated brain diseases.
Collapse
Affiliation(s)
- Lila Carniglia
- Instituto de Investigaciones Biomédicas, School of Medicine, University of Buenos Aires – CONICET, Buenos Aires, Argentina
| | - Daniela Durand
- Instituto de Investigaciones Biomédicas, School of Medicine, University of Buenos Aires – CONICET, Buenos Aires, Argentina
| | - Carla Caruso
- Instituto de Investigaciones Biomédicas, School of Medicine, University of Buenos Aires – CONICET, Buenos Aires, Argentina
| | - Mercedes Lasaga
- Instituto de Investigaciones Biomédicas, School of Medicine, University of Buenos Aires – CONICET, Buenos Aires, Argentina
| |
Collapse
|
36
|
Muceniece R, Dambrova M. Melanocortins in brain inflammation: the role of melanocortin receptor subtypes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 681:61-70. [PMID: 21222260 DOI: 10.1007/978-1-4419-6354-3_5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The melanocortins (MC) are released from neurons and paracrine cells in the CNS where they are involved in important physiological functions, including regulation of body temperature and immune responses. MC bind to melanocortin receptors, a class of cell surface G-protein-coupled receptors. Of the five subtypes of MC receptors that have been cloned in mammals, the MC1, MC3, MC4 and MC5 receptors are expressed in brain tissues. Expression of MC receptors in both brain cells and cells of the immune system suggests direct involvement of MC in regulation of inflammatory processes in the brain. The binding of MC to MC receptors induces activation of adenylate cyclase, increase in intracellular cAMP level and, consequently, inhibition of the nuclear transcription factor kappaB (NF-κB) signalling. Inflammatory processes contribute to development of severe CNS diseases, both in acute and chronic conditions. Thus far, the anti-inflammatory effects of MC in the CNS have been mainly studied using peptides that are relatively unselective for individual MC receptor subtypes. Consequently, these studies do not allow identification of specific MC receptor(s) involved in the regulation of inflammatory processes. However, recently synthesized ligands selective for individual MC receptors indicated that both MC4 and MC3 agonists are promising anti-inflammatory agents in treatment of brain inflammation.
Collapse
Affiliation(s)
- Ruta Muceniece
- Faculty of Medicine, University of Latvia, Sarlotes St. 1a, Riga, LV-1001, Latvia.
| | | |
Collapse
|
37
|
Melanocortins and the cholinergic anti-inflammatory pathway. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 681:71-87. [PMID: 21222261 DOI: 10.1007/978-1-4419-6354-3_6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Experimental evidence indicates that small concentrations of inflammatory molecules produced by damaged tissues activate afferent signals through ascending vagus nerve fibers, that act as the sensory arm of an "inflammatory reflex". The subsequent activation of vagal efferent fibers, which represent the motor arm of the inflammatory reflex, rapidly leads to acetylcholine release in organs of the reticuloendothelial system. Acetylcholine interacts with α7 subunit-containing nicotinic receptors in tissue macrophages and other immune cells and rapidly inhibits the synthesis/release of tumor necrosis factor-α and other inflammatory cytokines. This neural anti-inflammatory response called "cholinergic anti-inflammatory pathway" is fast and integrated through the central nervous system. Preclinical studies are in progress, with the aim to develop therapeutic agents able to activate the cholinergic anti-inflammatory pathway. Melanocortin peptides bearing the adrenocorticotropin/α-melanocyte-stimulating hormone sequences exert a protective and life-saving effect in animals and humans in conditions of circulatory shock. These neuropeptides are likewise protective in other severe hypoxic conditions, such as prolonged respiratory arrest, myocardial ischemia, renal ischemia and ischemic stroke, as well as in experimental heart transplantation. Moreover, experimental evidence indicates that melanocortins reverse circulatory shock, prevent myocardial ischemia/reperfusion damage and exert neuroprotection against ischemic stroke through activation of the cholinergic anti-inflammatory pathway. This action occurs via stimulation of brain melanocortin MC3/MC4 receptors. Investigations that determine the molecular mechanisms of the cholinergic anti-inflammatory pathway activation could help design of superselective activators of this pathway.
Collapse
|
38
|
Giuliani D, Minutoli L, Ottani A, Spaccapelo L, Bitto A, Galantucci M, Altavilla D, Squadrito F, Guarini S. Melanocortins as potential therapeutic agents in severe hypoxic conditions. Front Neuroendocrinol 2012; 33:179-93. [PMID: 22531139 DOI: 10.1016/j.yfrne.2012.04.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Revised: 03/26/2012] [Accepted: 04/09/2012] [Indexed: 01/14/2023]
Abstract
Melanocortin peptides with the adrenocorticotropin/melanocyte-stimulating hormone (ACTH/MSH) sequences and synthetic analogs have protective and life-saving effects in experimental conditions of circulatory shock, myocardial ischemia, ischemic stroke, traumatic brain injury, respiratory arrest, renal ischemia, intestinal ischemia and testicular ischemia, as well as in experimental heart transplantation. Moreover, melanocortins improve functional recovery and stimulate neurogenesis in experimental models of cerebral ischemia. These beneficial effects of ACTH/MSH-like peptides are mostly mediated by brain melanocortin MC(3)/MC(4) receptors, whose activation triggers protective pathways that counteract the main ischemia/reperfusion-related mechanisms of damage. Induction of signaling pathways and other molecular regulators of neural stem/progenitor cell proliferation, differentiation and integration seems to be the key mechanism of neurogenesis stimulation. Synthesis of stable and highly selective agonists at MC(3) and MC(4) receptors could provide the potential for development of a new class of drugs for a novel approach to management of severe ischemic diseases.
Collapse
Affiliation(s)
- Daniela Giuliani
- Department of Biomedical Sciences, Section of Pharmacology, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Caruso C, Carniglia L, Durand D, Gonzalez PV, Scimonelli TN, Lasaga M. Melanocortin 4 receptor activation induces brain-derived neurotrophic factor expression in rat astrocytes through cyclic AMP-protein kinase A pathway. Mol Cell Endocrinol 2012; 348:47-54. [PMID: 21803120 DOI: 10.1016/j.mce.2011.07.036] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 06/28/2011] [Accepted: 07/12/2011] [Indexed: 02/07/2023]
Abstract
Melanocortin 4 receptors (MC4R) are mainly expressed in the brain. We previously showed that the anti-inflammatory action of α-melanocyte-stimulating hormone (α-MSH) in rat hypothalamus and in cultured astrocytes involved MC4R activation. However, MC4R mechanisms of action remain undetermined. Since brain-derived neurotrophic factor (BDNF) may be mediating MC4R hypothalamic anorexigenic actions, we determined melanocortin effects on BDNF expression in rat cultured astrocytes and certain mechanisms involved in MC4R signaling. α-MSH and its analogue NDP-MSH, induced production of cAMP in astrocytes. This effect was completely blocked by the MC4R antagonist, HS024. We found that NDP-MSH increased BDNF mRNA and protein levels in astrocytes. The effect of NDP-MSH on BDNF expression was abolished by the adenylate cyclase inhibitor SQ22536, and decreased by the PKA inhibitor Rp-cAMP. Since melanocortins are immunomodulators, we investigated their actions with bacterial lipopolysaccharide (LPS) and interferon-γ (IFN-γ) stimulus. Although both α-MSH and LPS+IFN-γ increased cAMP responding element binding protein (CREB) activation, LPS+IFN-γ did not modify BDNF expression. On the other hand, α-MSH did not modify basal or LPS+IFN-γ-induced nuclear factor-κB activation. Our results show for the first time that MC4R activation in astrocytes induces BDNF expression through cAMP-PKA-CREB pathway without involving NF-κB.
Collapse
Affiliation(s)
- Carla Caruso
- Research Institute for Reproduction, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
40
|
Cerebellum-specific and age-dependent expression of an endogenous retrovirus with intact coding potential. Retrovirology 2011; 8:82. [PMID: 21992658 PMCID: PMC3207890 DOI: 10.1186/1742-4690-8-82] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Accepted: 10/12/2011] [Indexed: 12/13/2022] Open
Abstract
Background Endogenous retroviruses (ERVs), including murine leukemia virus (MuLV) type-ERVs (MuLV-ERVs), are presumed to occupy ~10% of the mouse genome. In this study, following the identification of a full-length MuLV-ERV by in silico survey of the C57BL/6J mouse genome, its distribution in different mouse strains and expression characteristics were investigated. Results Application of a set of ERV mining protocols identified a MuLV-ERV locus with full coding potential on chromosome 8 (named ERVmch8). It appears that ERVmch8 shares the same genomic locus with a replication-incompetent MuLV-ERV, called Emv2; however, it was not confirmed due to a lack of relevant annotation and Emv2 sequence information. The ERVmch8 sequence was more prevalent in laboratory strains compared to wild-derived strains. Among 16 different tissues of ~12 week-old female C57BL/6J mice, brain homogenate was the only tissue with evident expression of ERVmch8. Further ERVmch8 expression analysis in six different brain compartments and four peripheral neuronal tissues of C57BL/6J mice revealed no significant expression except for the cerebellum in which the ERVmch8 locus' low methylation status was unique compared to the other brain compartments. The ERVmch8 locus was found to be surrounded by genes associated with neuronal development and/or inflammation. Interestingly, cerebellum-specific ERVmch8 expression was age-dependent with almost no expression at 2 weeks and a plateau at 6 weeks. Conclusions The ecotropic ERVmch8 locus on the C57BL/6J mouse genome was relatively undermethylated in the cerebellum, and its expression was cerebellum-specific and age-dependent.
Collapse
|
41
|
Pain sensitivity and experimentally induced sensitisation in red haired females. Scand J Pain 2011; 2:3-6. [DOI: 10.1016/j.sjpain.2010.08.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Accepted: 08/27/2010] [Indexed: 11/21/2022]
Abstract
Abstract
Introduction and aim
Pain sensitivity has been linked to the melanocortin-1 receptor (MC1R) gene. A mutation in MC1R can result in pale skin and red hair in humans and may modulate pain responses in general. Human studies have shown that women with non-functional MC1R’s were sensitive to experimental induced cold and heat pain. A study demonstrated that females with red hair required higher dose of anesthesia than females with dark hair to experience analgesia to electrical stimulation. Moreover, women expressing non-functional MC1Rs display greater analgesia from opioid analgesia. If redheads in general respond differently to pain and analgesics, this is of clinical importance. The aim of this study was therefore to investigate pain sensitivity and experimentally induced sensitisation in red haired females.
Method
Twenty healthy females with pale skin and red hair (mean age 32 years, range 20–55) and 20 healthy females with blond/dark hair (mean age 31 years, range 20–51) participated in this study. The pain tolerance thresholds to heat and pressure stimulation were determined. Hyperalgesia was induced experimentally by applying 0.075% topical capsaicin cream for 30 min. The secondary pin-prick hyperalgesic area was estimated with a calibrated filament (von Frey hair, 15 g) and the area of allodynia by a soft brush. This was done 0, 30, 60, and 90 min after cream removal.
Results
Neither heat nor pressure pain tolerance thresholds were changed in the two groups. The secondary pin-prick hyperalgesic areas were significantly smaller for red haired females than blond/dark haired females (P = 0.014). There were no significant differences in the allodynic areas.
Discussion
As the secondary hyperalgesic response evoked by topical capsaicin is a central phenomenon, the observed smaller pin-prick hyperalgesic area in the red haired females could indicate a central role of MCRs in development or maintenance of hyperalgesia. Central involvement of MC1Rs or dysfunction of peripheral MC1Rs activating central MC4Rs has been suggested to influence pain sensitivity. The difference observed between red haired and non-red haired females may have implications for pain management regimens as compounds interacting with sensitisation such as NMDA-antagonists or alpha-2-delta-ligands may exert different types of action in people with MC1R mutation.
Conclusion
The present study showed that red haired females were less sensitive to topical capsaicin induced pin-prick hyperalgesia compared with blond/dark haired females.
Implications
The smaller hyperalgesic area in redheads could be a manifestation of central anti-hyperalgesic involvement of MCRs and could have an influence on the treatment of pain as well as in studies investigating anti-hyperalgesic drugs.
Collapse
|
42
|
Machado I, González P, Schiöth HB, Lasaga M, Scimonelli TN. α-Melanocyte-stimulating hormone (α-MSH) reverses impairment of memory reconsolidation induced by interleukin-1 beta (IL-1 beta) hippocampal infusions. Peptides 2010; 31:2141-4. [PMID: 20688120 DOI: 10.1016/j.peptides.2010.07.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Revised: 07/15/2010] [Accepted: 07/15/2010] [Indexed: 12/14/2022]
Abstract
Interleukin-1 beta (IL-1β) significantly influences cognitive processes. Treatments which raise the level of IL-1β in the brain impair memory consolidation in contextual fear conditioning. However, the effect of IL-1β on memory reconsolidation has not yet been established. The melanocortin α-melanocyte-stimulating hormone (α-MSH) exerts potent anti-inflammatory actions by antagonizing the effect of proinflammatory cytokines. Five subtypes of melanocortin receptors (MC1R-MC5R) have been identified, of which MC3R and MC4R are predominant in the central nervous system. The present experiments show that the injection of IL-1β (5 ng/0.25 μl) in dorsal hippocampus up to 30 min after re-exposition to the context decreases freezing during the contextual fear test. Impairment of memory reconsolidation was reversed by treatment with α-MSH (0.05 μg/0.25 μl). Administration of the MC4 receptor antagonist HS014 (0.5 μg/0.25 μl) blocked the effect of α-MSH. These results suggest that IL-1β may influence memory reconsolidation and that activation of central MC4R could lead to improve cognitive performance.
Collapse
Affiliation(s)
- Ivana Machado
- IFEC CONICET Departamento Farmacología, Facultad de Ciencias Químicas, UNC, Córdoba, Argentina
| | | | | | | | | |
Collapse
|
43
|
Caruso C, Sanchez M, Durand D, de la Cruz Perez M, Gonzalez PV, Lasaga M, Scimonelli TN. α-Melanocyte-stimulating hormone modulates lipopolysaccharide plus interferon-γ-induced tumor necrosis factor-α expression but not tumor necrosis factor-α receptor expression in cultured hypothalamic neurons. J Neuroimmunol 2010; 227:52-9. [DOI: 10.1016/j.jneuroim.2010.06.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Revised: 06/09/2010] [Accepted: 06/10/2010] [Indexed: 01/09/2023]
|
44
|
Abstract
The melanocortin-4 receptor (MC4R) was cloned in 1993 by degenerate PCR; however, its function was unknown. Subsequent studies suggest that the MC4R might be involved in regulating energy homeostasis. This hypothesis was confirmed in 1997 by a series of seminal studies in mice. In 1998, human genetic studies demonstrated that mutations in the MC4R gene can cause monogenic obesity. We now know that mutations in the MC4R are the most common monogenic form of obesity, with more than 150 distinct mutations reported thus far. This review will summarize the studies on the MC4R, from its cloning and tissue distribution to its physiological roles in regulating energy homeostasis, cachexia, cardiovascular function, glucose and lipid homeostasis, reproduction and sexual function, drug abuse, pain perception, brain inflammation, and anxiety. I will then review the studies on the pharmacology of the receptor, including ligand binding and receptor activation, signaling pathways, as well as its regulation. Finally, the pathophysiology of the MC4R in obesity pathogenesis will be reviewed. Functional studies of the mutant MC4Rs and the therapeutic implications, including small molecules in correcting binding and signaling defect, and their potential as pharmacological chaperones in rescuing intracellularly retained mutants, will be highlighted.
Collapse
Affiliation(s)
- Ya-Xiong Tao
- Department of Anatomy, Physiology, and Pharmacology, Auburn University, Alabama 36849-5519, USA.
| |
Collapse
|
45
|
Fridmanis D, Petrovska R, Kalnina I, Slaidina M, Peculis R, Schiöth HB, Klovins J. Identification of domains responsible for specific membrane transport and ligand specificity of the ACTH receptor (MC2R). Mol Cell Endocrinol 2010; 321:175-83. [PMID: 20206229 DOI: 10.1016/j.mce.2010.02.032] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Revised: 02/23/2010] [Accepted: 02/23/2010] [Indexed: 12/26/2022]
Abstract
The adrenocorticotropic hormone (ACTH) receptor has highly specific membrane expression that is limited to adrenal cells; in other cell types the polypeptide fails to be transported to the cell surface. Unlike other evolutionarily related members of the melanocortin receptor family (MC1R-MC5R) that recognize different melanocortin peptides, ACTHR (MC2R) binds only ACTH. We used a mutagenesis approach involving systematic construction of chimeric ACTHR/MC4R receptors to identify the domains determining the selectivity of ACTHR membrane transport and ACTH binding. In total 15 chimeric receptors were created by replacement of selected domains of human ACTHR with the corresponding regions of human MC4R. We developed an analytical method to accurately quantify cell-membrane localization of recombinant receptors fused with enhanced green fluorescent protein by confocal fluorescence microscopy. The chimeric receptors were also tested for their ability to bind ACTH (1-24) and the melanocyte-stimulating hormone (MSH) analog, Nle4, DPhe7-alpha-MSH, and to induce a cAMP response. Our results indicate that substitution of the MC4R N-terminal segment with the homologous segment of ACTHR significantly decreased membrane transport. We also identified another signal localized in the third and fourth transmembrane regions as the main determinant of ACTHR intracellular retention. In addition, we found that the fourth and fifth transmembrane domains of the ACTHR are involved in ACTH binding selectivity. We discuss the mechanisms involved in bypassing these arrest signals via an interaction with melanocortin 2 receptor accessory protein (MRAP) and the possible mechanisms that determine the high ligand-binding specificity of ACTHR.
Collapse
|
46
|
Shariat-Madar B, Kolte D, Verlangieri A, Shariat-Madar Z. Prolylcarboxypeptidase (PRCP) as a new target for obesity treatment. Diabetes Metab Syndr Obes 2010; 3:67-78. [PMID: 20694162 PMCID: PMC2916657 DOI: 10.2147/dmsott.s7290] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Recently, we serendipitously discovered that mice with the deficiency of the enzyme prolylcarboxypeptidase (PRCP) have elevated alpha-melanocyte-stimulating hormone (alpha-MSH) levels which lead to decreased food intake and weight loss. This suggests that PRCP is an endogenous inactivator of alpha-MSH and an appetite stimulant. Since a modest weight loss can have the most profound influence on reducing cardiovascular risk factors, the inhibitors of PRCP would be emerging as a possible alternative for pharmacotherapy in high-risk patients with obesity and obesity-related disorders. The discovery of a new biological activity of PRCP in the PRCP-deficient mice and studies of alpha-MSH function indicate the importance and complexity of the hypothalamic pro-opiomelanocortin (POMC) system in altering food intake. Identifying a role for PRCP in regulating alpha-MSH in the brain may be a critical step in enhancing our understanding of how the brain controls food intake and body weight. In light of recent findings, the potential role of PRCP in regulating fuel homeostasis is critically evaluated. Further studies of the role of PRCP in obesity are much needed.
Collapse
Affiliation(s)
- B Shariat-Madar
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor MI, USA
| | - D Kolte
- School of Pharmacy, Department of Pharmacology, University of Mississippi, University, MS, USA
| | - A Verlangieri
- School of Pharmacy, Department of Pharmacology, University of Mississippi, University, MS, USA
| | - Z Shariat-Madar
- School of Pharmacy, Department of Pharmacology, University of Mississippi, University, MS, USA
- Correspondence: Zia Shariat-Madar, University of Mississippi, 219 B, Faser Hall, University, MS 38677–1848, USA, Tel +662 915 5150, Fax +662 915 5148, Email
| |
Collapse
|
47
|
Blagaić V, Houra K, Turčić P, Štambuk N, Konjevoda P, Boban-Blagaić A, Kelava T, Kos M, Aralica G, Čulo F. The influence of alpha-, beta-, and gamma-melanocyte stimulating hormone on acetaminophen induced liver lesions in male CBA mice. Molecules 2010; 15:1232-41. [PMID: 20335976 PMCID: PMC6257183 DOI: 10.3390/molecules15031232] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Revised: 03/02/2010] [Accepted: 03/03/2010] [Indexed: 11/18/2022] Open
Abstract
Research over the past decade has indicated that melanocortin peptides are potent inhibitors of inflammation and a promising source of new anti-inflammatory and cytoprotective therapies. The purpose of the present paper is to compare protective effects of α-, β-, and γ-melanocyte stimulating hormone on acetaminophen induced liver lesions in male CBA mice. Acetaminophen was applied intragastrically in a dose of 150 mg/kg, and tested substances were applied intraperitoneally 1 hour before acetaminophen. Mice were sacrificed after 24 hours and intensity of liver injury was estimated by measurement of plasma transaminase activity (AST and ALT) and histopathological grading of lesions. It was found that α-, β-, and γ-MSH decrease intensity of lesions by both criteria in a dose-dependent manner.
Collapse
Affiliation(s)
- Vladimir Blagaić
- University Hospital “Sveti Duh”, Sveti Duh 64, 10000 Zagreb, Croatia; E-Mail: (V.B.)
| | - Karlo Houra
- University Hospital “Sestre milosrdnice”, Vinogradska cesta 29, 10000 Zagreb, Croatia; E-Mails: (K.H.); (M.K.)
| | - Petra Turčić
- Faculty of Pharmacy and Biochemistry, University of Zagreb, Domagojeva 2, 10000 Zagreb, Croatia; E-Mail: (P.T.)
| | - Nikola Štambuk
- Ruđer Bošković Institute, Bijenička cesta 54, 10002 Zagreb, Croatia
- Authors to whom correspondence should be addressed; E-Mails: (N.Š.); (P.K.)
| | - Paško Konjevoda
- Ruđer Bošković Institute, Bijenička cesta 54, 10002 Zagreb, Croatia
- Authors to whom correspondence should be addressed; E-Mails: (N.Š.); (P.K.)
| | - Alenka Boban-Blagaić
- Department of Pharmacology, School of Medicine, University of Zagreb, Šalata 11, 10000 Zagreb, Croatia; E-Mail: (A.B.-B.)
| | - Tomislav Kelava
- Department of Physiology, School of Medicine, University of Zagreb, Šalata 3, 10000 Zagreb, Croatia; E-Mails: (T.K.); (F.Č.)
| | - Marina Kos
- University Hospital “Sestre milosrdnice”, Vinogradska cesta 29, 10000 Zagreb, Croatia; E-Mails: (K.H.); (M.K.)
| | - Gorana Aralica
- University Hospital Dubrava, Avenija Gojka Šuška 6, 10000 Zagreb, Croatia; E-Mail: (G.A.)
| | - Filip Čulo
- Department of Physiology, School of Medicine, University of Zagreb, Šalata 3, 10000 Zagreb, Croatia; E-Mails: (T.K.); (F.Č.)
| |
Collapse
|
48
|
Mountjoy KG. Distribution and function of melanocortin receptors within the brain. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 681:29-48. [PMID: 21222258 DOI: 10.1007/978-1-4419-6354-3_3] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Biological responses to pro-opiomelanocortin (POMC)-derived peptides administered in the brain were documented in the 1950s but their molecular mechanisms of action only began to be resolved with the mapping of melanocortin receptor subtypes to specific brain regions in the 1990s. Out of the five melanocortin receptor subtypes, MC3R and MC4R are widely recognised as 'neural' melanocortin receptors. In situ hybridization anatomical mapping of these receptor subtypes to distinct hypothalamic nuclei first indicated their roles in energy homeostasis, roles that were later confirmed with the obese phenotypes exhibited by Mc3R and Mc4R knockout mice. It is perhaps less well known however, that all five melanocortin receptor subtypes have been detected in developing and/or adult brains of various species. This chapter provides a comprehensive summary of the detection and mapping of each melanocortin receptor subtype in mammalian, chicken and fish brains and relates the sites of expression to functions that are either known or proposed for each receptor subtype.
Collapse
Affiliation(s)
- Kathleen G Mountjoy
- Departments of Physiology and Molecular Medicine and Pathology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, 1023, New Zealand.
| |
Collapse
|
49
|
Eves PC, Haycock JW. Melanocortin Signalling Mechanisms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 681:19-28. [DOI: 10.1007/978-1-4419-6354-3_2] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
50
|
What is the connection between red hair and Tourette syndrome? Med Hypotheses 2009; 73:849-53. [DOI: 10.1016/j.mehy.2009.03.052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2009] [Revised: 02/23/2009] [Accepted: 03/27/2009] [Indexed: 11/17/2022]
|