1
|
Sun Q, Zhu J, Zhao X, Huang X, Qu W, Tang X, Ma D, Shu Q, Li X. Mettl3-m 6A-NPY axis governing neuron-microglia interaction regulates sleep amount of mice. Cell Discov 2025; 11:10. [PMID: 39905012 PMCID: PMC11794856 DOI: 10.1038/s41421-024-00756-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 11/13/2024] [Indexed: 02/06/2025] Open
Abstract
Sleep behavior is regulated by diverse mechanisms including genetics, neuromodulation and environmental signals. However, it remains completely unknown regarding the roles of epitranscriptomics in regulating sleep behavior. In the present study, we showed that the deficiency of RNA m6A methyltransferase Mettl3 in excitatory neurons specifically induces microglia activation, neuroinflammation and neuronal loss in thalamus of mice. Mettl3 deficiency remarkably disrupts sleep rhythm and reduces the amount of non-rapid eye movement sleep. We also showed that Mettl3 regulates neuropeptide Y (NPY) via m6A modification and Mettl3 conditional knockout (cKO) mice displayed significantly decreased expression of NPY in thalamus. In addition, the dynamic distribution pattern of NPY is observed during wake-sleep cycle in cKO mice. Ectopic expression of Mettl3 and NPY significantly inhibits microglia activation and neuronal loss in thalamus, and restores the disrupted sleep behavior of cKO mice. Collectively, our study has revealed the critical function of Mettl3-m6A-NPY axis in regulating sleep behavior.
Collapse
Affiliation(s)
- Qihang Sun
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jinpiao Zhu
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
- Department of Rehabilitation, Perioperative and Systems Medicine Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
| | - Xingsen Zhao
- Institute of Biotechnology, Xianghu Laboratory, Hangzhou, Zhejiang, China
| | - Xiaoli Huang
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Wenzheng Qu
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Xia Tang
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Daqing Ma
- Department of Rehabilitation, Perioperative and Systems Medicine Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
- Division of Anesthetics, Pain Medicine & Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK.
| | - Qiang Shu
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
| | - Xuekun Li
- Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
- The Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
- Binjiang Institute of Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Ren Y, Huang P, Zhang L, Tang Y, He S, Li H, Huang X, Ding Y, Liu L, Liu L, He X. Multi-omics landscape of childhood simple obesity: novel insights into pathogenesis and biomarkers discovery. Cell Biosci 2024; 14:145. [PMID: 39609876 PMCID: PMC11606102 DOI: 10.1186/s13578-024-01322-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/11/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND The increasing incidence of childhood obesity annually has led to a surge in physical and mental health risks, making it a significant global public health concern. This study aimed to discover novel biomarkers of childhood simple obesity through integrative multi-omics analysis, uncovering their potential connections and providing fresh research directions for the complex pathogenesis and treatment strategies. METHODS Transcriptome, untargeted metabolome, and 16 S rDNA sequencing were conducted on subjects to examine transcripts, metabolites in blood, and gut microflora in stool. RESULTS Transcriptomic analysis identified 599 differentially expressed genes (DEGs), of which 25 were immune-related genes, and participated in immune pathways such as antimicrobial peptides, neutrophil degranulation, and interferons. The optimal random forest model based on these genes exhibited an AUC of 0.844. The metabolomic analysis examined 71 differentially expressed metabolites (DEMs), including 12 immune-related metabolites. Notably, lauric acid showed an extremely strong positive correlation with BMI and showed a good discriminative power for obesity (AUC = 0.82). DEMs were found to be significantly enriched in four metabolic pathways, namely "Aminoacyl-tRNA biosynthesis", "Valine leucine and isoleucine biosynthesis, and Glycine", "Serine and threonine metabolism", and "Biosynthesis of unsaturated fatty acids". Microbiome analysis revealed 12 differential gut microbiotas (DGMs) at the phylum and genus levels, with p_Firmicutes dominating in the obese group and g_Escherichia-Shigella in the normal group. Subsequently, a Random Forest model was developed based on the DEMs, immune-related DEGs, and metabolites with an AUC value of 0.912. The 14 indicators identified by this model could potentially serve as a set of biomarkers for obesity. The analysis of the inter-omics correlation network found 233 pairs of significant correlations. DEGs BPIFA1, BPI, and SAA1, DEMs Dimethy(tetradecyl)amine, Deoxycholic acid, Pathalic anhydride, and DL-Alanine, and DGMs g_Intestinimonas and g_Turicibacter showed strong connectivity within the network, constituting a large proportion of interactions. CONCLUSION This study presents the first comprehensive description of the multi-omics characteristics of childhood simple obesity, recognizing promising biomarkers. Immune-related markers offer a new perspective for researching the immunological mechanisms underlying obesity and its associated complications. The revealed interactions among these biomarkers contribute to a deeper understanding the intricate biological regulatory networks associated with obesity.
Collapse
Affiliation(s)
- Yi Ren
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
- Department of Pediatrics, Haikou Hospital of the Maternal and Child Health, Haikou, 570100, China
- Department of Pediatrics, Hainan Modern Women and Children's. Medical, Haikou, 570100, China
| | - Peng Huang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Lu Zhang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Yufen Tang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Siyi He
- Department of Anesthesiology, Hainan General Hospital, Haikou, Hainan, 570311, China
- Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - HaiDan Li
- Department of Pediatrics, Hainan Women and Children's Medical Center, Hainan, 570100, China
| | - XiaoYan Huang
- Department of Pediatrics, Hainan Women and Children's Medical Center, Hainan, 570100, China
| | - Yan Ding
- Department of Dermatology, Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Lingjuan Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Liqun Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
| | - Xiaojie He
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Laboratory of Pediatric Nephrology, Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
| |
Collapse
|
3
|
Shapovalova K, Zorkina Y, Abramova O, Andryushchenko A, Chekhonin V, Kostyuk G. The Role of Neuropeptide Y in the Pathogenesis of Alzheimer's Disease: Diagnostic Significance and Neuroprotective Functions. Neurol Int 2024; 16:1318-1331. [PMID: 39585059 PMCID: PMC11587103 DOI: 10.3390/neurolint16060100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/24/2024] [Accepted: 10/30/2024] [Indexed: 11/26/2024] Open
Abstract
Background. Alzheimer's disease (AD) is one of the most common neurodegenerative diseases. It has been suggested that the factors that cause pathologic changes and lead to the development of AD may also include changes in certain neuropeptides. The implication of the neuropeptide (NPY) in the pathogenesis of AD and its potential therapeutic role is possible due to the following properties: involvement in adult neurogenesis, regulatory effects on the immune system, the inhibition of potential-dependent Ca2+ channels, and the reduction in glutamate excitotoxicity. The aim of our review was to summarize recent data on the role of NPY in AD development and to explore its potential as a biomarker and a possible therapeutic target. Materials and methods. We performed a systematic review of studies, for which we search using the keywords "Alzheimer's disease and neuropeptide Y", "Alzheimer's disease and NPY", "AD and NPY", "Neuropeptide Y and Neurodegenerative disease". Nineteen articles were included in the review. Results. The NPY levels in cerebrospinal fluid and plasma have been found to be reduced or unchanged in AD patients; however, these findings need to be confirmed in more recent studies. Data obtained in transgenic animal models support the role of NPY in AD pathogenesis. The neuroprotective effects of NPY have been demonstrated in vitro and in vivo in AD models. Conclusion. The findings may open new possibilities for using NPY as a diagnostic marker to detect AD at earlier stages of the disease or as a potential therapeutic target due to its neuroprotective properties.
Collapse
Affiliation(s)
- Ksenia Shapovalova
- Mental-Health Clinic No. 1 Named After N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (K.S.); (O.A.); (A.A.); (G.K.)
| | - Yana Zorkina
- Mental-Health Clinic No. 1 Named After N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (K.S.); (O.A.); (A.A.); (G.K.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky Per. 23, 119034 Moscow, Russia;
| | - Olga Abramova
- Mental-Health Clinic No. 1 Named After N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (K.S.); (O.A.); (A.A.); (G.K.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky Per. 23, 119034 Moscow, Russia;
| | - Alisa Andryushchenko
- Mental-Health Clinic No. 1 Named After N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (K.S.); (O.A.); (A.A.); (G.K.)
| | - Vladimir Chekhonin
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Kropotkinsky Per. 23, 119034 Moscow, Russia;
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Georgy Kostyuk
- Mental-Health Clinic No. 1 Named After N.A. Alekseev, Zagorodnoe Highway 2, 115191 Moscow, Russia; (K.S.); (O.A.); (A.A.); (G.K.)
- Department of Psychiatry, Federal State Budgetary Educational Institution of Higher Education “Moscow State University of Food Production”, Volokolamskoye Highway 11, 125080 Moscow, Russia
- Department of Mental Health, Faculty of Psychology, M. V. Lomonosov Moscow State University, 119991 Moscow, Russia
- Department of Psychiatry and Psychosomatics, I. M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| |
Collapse
|
4
|
Confino S, Wexler Y, Medvetzky A, Elazary Y, Ben-Moshe Z, Reiter J, Dor T, Edvardson S, Prag G, Harel T, Gothilf Y. A deleterious variant of INTS1 leads to disrupted sleep-wake cycles. Dis Model Mech 2024; 17:dmm050746. [PMID: 39189071 PMCID: PMC11381918 DOI: 10.1242/dmm.050746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/25/2024] [Indexed: 08/28/2024] Open
Abstract
Sleep disturbances are common among children with neurodevelopmental disorders. Here, we report a syndrome characterized by prenatal microcephaly, intellectual disability and severe disruption of sleep-wake cycles in a consanguineous family. Exome sequencing revealed homozygous variants (c.5224G>A and c.6506G>T) leading to the missense mutations E1742K and G2169V in integrator complex subunit 1 (INTS1), the core subunit of the Integrator complex. Conservation and structural analyses suggest that G2169V has a minor impact on the structure and function of the complex, while E1742K significantly alters a negatively charged conserved patch on the surface of the protein. The severe sleep-wake cycles disruption in human carriers highlights a new aspect of Integrator complex impairment. To further study INTS1 pathogenicity, we generated Ints1-deficient zebrafish lines. Mutant zebrafish larvae displayed abnormal circadian rhythms of locomotor activity and sleep, as is the case with the affected humans. Furthermore, Ints1-deficent larvae exhibited elevated levels of dopamine β-hydroxylase (dbh) mRNA in the locus coeruleus, a wakefulness-inducing brainstem center. Altogether, these findings suggest a significant, likely indirect, effect of INTS1 and the Integrator complex on maintaining circadian rhythms of locomotor activity and sleep homeostasis across vertebrates.
Collapse
Affiliation(s)
- Shir Confino
- School of Neurobiology, Biochemistry and Biophysics, Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Yair Wexler
- School of Neurobiology, Biochemistry and Biophysics, Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Adar Medvetzky
- School of Neurobiology, Biochemistry and Biophysics, Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Yotam Elazary
- School of Neurobiology, Biochemistry and Biophysics, Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Zohar Ben-Moshe
- School of Neurobiology, Biochemistry and Biophysics, Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Joel Reiter
- Pediatric Pulmonary & Sleep Unit, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Talya Dor
- ALYN - Children and Adolescent Rehabilitation Center, Jerusalem 9109002, Israel
| | - Simon Edvardson
- ALYN - Children and Adolescent Rehabilitation Center, Jerusalem 9109002, Israel
| | - Gali Prag
- School of Neurobiology, Biochemistry and Biophysics, Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Tamar Harel
- Department of Genetics, Hadassah Medical Center, Jerusalem 91120, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Yoav Gothilf
- School of Neurobiology, Biochemistry and Biophysics, Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv 6997801, Israel
| |
Collapse
|
5
|
Barbaresi P, Fabri M, Lorenzi T, Sagrati A, Morroni M. Intrinsic organization of the corpus callosum. Front Physiol 2024; 15:1393000. [PMID: 39035452 PMCID: PMC11259024 DOI: 10.3389/fphys.2024.1393000] [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] [Received: 02/28/2024] [Accepted: 05/16/2024] [Indexed: 07/23/2024] Open
Abstract
The corpus callosum-the largest commissural fiber system connecting the two cerebral hemispheres-is considered essential for bilateral sensory integration and higher cognitive functions. Most studies exploring the corpus callosum have examined either the anatomical, physiological, and neurochemical organization of callosal projections or the functional and/or behavioral aspects of the callosal connections after complete/partial callosotomy or callosal lesion. There are no works that address the intrinsic organization of the corpus callosum. We review the existing information on the activities that take place in the commissure in three sections: I) the topographical and neurochemical organization of the intracallosal fibers, II) the role of glia in the corpus callosum, and III) the role of the intracallosal neurons.
Collapse
Affiliation(s)
- Paolo Barbaresi
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Marche Polytechnic University, Ancona, Italy
| | - Mara Fabri
- Department of Life and Environmental Sciences, Marche Polytechnic University, Ancona, Italy
| | - Teresa Lorenzi
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Marche Polytechnic University, Ancona, Italy
| | - Andrea Sagrati
- Department of Life and Environmental Sciences, Marche Polytechnic University, Ancona, Italy
| | - Manrico Morroni
- Electron Microscopy Unit, Azienda Ospedaliero-Universitaria, Ancona, Italy
| |
Collapse
|
6
|
Pereira PA, Tavares M, Laires M, Mota B, Madeira MD, Paula-Barbosa MM, Cardoso A. Effects of Aging and Nerve Growth Factor on Neuropeptide Expression and Cholinergic Innervation of the Rat Basolateral Amygdala. BIOLOGY 2024; 13:155. [PMID: 38534426 DOI: 10.3390/biology13030155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 03/28/2024]
Abstract
The basolateral amygdala (BLA) contains interneurons that express neuropeptide Y (NPY) and vasoactive intestinal polypeptide (VIP), both of which are involved in the regulation of functions and behaviors that undergo deterioration with aging. There is considerable evidence that, in some brain areas, the expression of NPY and VIP might be modulated by acetylcholine. Importantly, the BLA is one of the brain regions that has one of the densest cholinergic innervations, which arise mainly from the basal forebrain cholinergic neurons. These cholinergic neurons depend on nerve growth factor (NGF) for their survival, connectivity, and function. Thus, in this study, we sought to determine if aging alters the densities of NPY- and VIP-positive neurons and cholinergic varicosities in the BLA and, in the affirmative, if those changes might rely on insufficient trophic support provided by NGF. The number of NPY-positive neurons was significantly reduced in aged rats, whereas the number of VIP-immunoreactive neurons was unaltered. The decreased NPY expression was fully reversed by the infusion of NGF in the lateral ventricle. The density of cholinergic varicosities was similar in adult and old rats. On the other hand, the density of cholinergic varicosities is significantly higher in old rats treated with NGF than in adult and old rats. Our results indicate a dissimilar resistance of different populations of BLA interneurons to aging. Furthermore, the present data also show that the BLA cholinergic innervation is particularly resistant to aging effects. Finally, our results also show that the reduced NPY expression in the BLA of aged rats can be related to changes in the NGF neurotrophic support.
Collapse
Affiliation(s)
- Pedro A Pereira
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Marta Tavares
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Miguel Laires
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Bárbara Mota
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Maria Dulce Madeira
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Manuel M Paula-Barbosa
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Armando Cardoso
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
7
|
Prochazkova P, Sonka K, Roubalova R, Jezkova J, Nevsimalova S, Buskova J, Merkova R, Dvorakova T, Prihodova I, Dostalova S, Tlaskalova-Hogenova H. Investigation of anti-neuronal antibodies and disparity in central hypersomnias. Sleep Med 2024; 113:220-231. [PMID: 38056084 DOI: 10.1016/j.sleep.2023.11.039] [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: 09/29/2023] [Revised: 11/06/2023] [Accepted: 11/25/2023] [Indexed: 12/08/2023]
Abstract
STUDY OBJECTIVES Microbial antigens can elicit an immune response leading to the production of autoantibodies cross-reacting with autoantigens. Still, their clinical significance in human sera in the context of brain diseases is unclear. Therefore, assessment of natural autoantibodies reacting with their neuropeptides may elucidate the autoimmune etiology of central hypersomnias. The study aims to determine whether serum autoantibody levels differ in patients with different types of central hypersomnias (narcolepsy type 1 and 2, NT1 and NT2; idiopathic hypersomnia, IH) and healthy controls and if the differences could suggest the participation of autoantibodies in disease pathogenesis. METHODS Sera from 91 patients with NT1, 27 with NT2, 46 with IH, and 50 healthy controls were examined for autoantibodies against assorted neuropeptides. Participants were screened using questionnaires related to sleep disorders, quality of life, and mental health conditions. In addition, serum biochemical parameters and biomarkers of microbial penetration through the intestinal wall were determined. RESULTS A higher prevalence of autoantibodies against neuropeptides was observed only for alpha-melanocytes-stimulating hormone (α-MSH) and neuropeptide glutamic acid-isoleucine (NEI), which differed slightly among diagnoses. Patients with both types of narcolepsy exhibited signs of microbial translocation through the gut barrier. According to the questionnaires, patients diagnosed with NT2 or IH had subjectively worse life quality than patients with NT1. Patients displayed significantly lower levels of bilirubin and creatinine and slightly higher alkaline phosphatase values than healthy controls. CONCLUSIONS Overall, serum anti-neuronal antibodies prevalence is rare, suggesting that their participation in the pathophysiology of concerned sleep disorders is insignificant. Moreover, their levels vary slightly between diagnoses indicating no major diagnostic significance.
Collapse
Affiliation(s)
- Petra Prochazkova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic.
| | - Karel Sonka
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Radka Roubalova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Janet Jezkova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic; First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Sona Nevsimalova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Jitka Buskova
- National Institute of Mental Health, Klecany, Czech Republic; Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Radana Merkova
- National Institute of Mental Health, Klecany, Czech Republic; Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Tereza Dvorakova
- National Institute of Mental Health, Klecany, Czech Republic; Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Iva Prihodova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Simona Dostalova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Helena Tlaskalova-Hogenova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
8
|
Luo W, Sun M, Wan J, Zhang Z, Huang J, Zhang J, Xiong W, Xia L, Xu P, Miao C, Zhang X, Liu M, Zhong J. Efficacy and safety of remimazolam tosilate versus propofol in patients undergoing day surgery: a prospective randomized controlled trial. BMC Anesthesiol 2023; 23:182. [PMID: 37237331 DOI: 10.1186/s12871-023-02092-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/14/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Remimazolam tosilate (RT) is a novel short-acting GABA (A) receptor agonist that has a rapid recovery from procedural sedation and can be fully reversed by flumazenil. To date, there have been relatively few articles comparing RT and propofol for general anesthesia. This study aimed to assess the efficacy and safety of RT with or without flumazenil compared with propofol in general anesthesia for day surgery. METHODS 115 patients scheduled for day surgery were randomized into three groups: RT (n = 39), RT + flumazenil (n = 38) and propofol (n = 38). The primary endpoints were anesthesia induction time and time until fully alert. Anesthesia success rate, bispectral index (BIS) values, injection pain, opioid and vasopressor dosages, postoperative recovery profiles and perioperative inflammatory and cognitive changes were assessed. Any adverse events were recorded. RESULTS Induction times were similar among the three groups (P = 0.437), but the median time until fully alert in patients treated with RT was longer than that of the propofol or RT + flumazenil groups (17.6 min vs. 12.3 min vs. 12.3 min, P < 0.001). The three groups had comparable postoperative recovery quality and inflammatory and cognitive state changes (P > 0.05). Smaller percentages of patients who received RT (26.3%) and RT + flumazenil (31.6%) developed hypotension during anesthesia maintenance compared with propofol (68.4%), and consequently less ephedrine (P < 0.001) and phenylephrine (P = 0.015) were needed in the RT group. Furthermore, serum triglyceride levels were lower (P < 0.001) and injection pain was much less frequent in the RT with or without flumazenil groups compared with the propofol group (5.3% vs. 0% vs. 18.4%). CONCLUSION RT permits rapid induction and comparable recovery profile compared with propofol in general anesthesia for day surgery, but has a prolonged recovery time without flumazenil. The safety profile of RT was superior to propofol in terms of hypotension and injection pain. TRIAL REGISTRATION The study was registered at Chinese Clinical Trial Registry http://www.chictr.org.cn/ (Registration date: 19/7/2021; Trial ID: ChiCTR2100048904).
Collapse
Affiliation(s)
- Wenchen Luo
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, 200032, People's Republic of China
| | - Minli Sun
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, 200032, People's Republic of China
| | - Jie Wan
- School of Anesthesiology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Zhenyu Zhang
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, 200032, People's Republic of China
| | - Jian Huang
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, 200032, People's Republic of China
| | - Jinlin Zhang
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, 200032, People's Republic of China
| | - Wanxia Xiong
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, 200032, People's Republic of China
| | - Lirong Xia
- Jiangsu Jiangyin People's Hospital, Jiangyin, Jiangsu, People's Republic of China
| | - Peiyao Xu
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, 200032, People's Republic of China
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, People's Republic of China
| | - Xuesong Zhang
- Department of Anesthesiology, Zhongshan Wusong Hospital Affiliated to Fudan University, Shanghai, People's Republic of China
| | - Mingyue Liu
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, 200032, People's Republic of China.
| | - Jing Zhong
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, 200032, People's Republic of China.
- Fudan Zhangjiang Institute, Shanghai, People's Republic of China.
- Department of Anesthesiology, Zhongshan Wusong Hospital Affiliated to Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
9
|
Efficacy and safety of Ciprofol for procedural sedation and anesthesia in non-operating room settings. J Clin Anesth 2023; 85:111047. [PMID: 36599219 DOI: 10.1016/j.jclinane.2022.111047] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/11/2022] [Accepted: 12/25/2022] [Indexed: 01/03/2023]
Abstract
STUDY OBJECTIVE Ciprofol, a novel intravenous anesthetic, provides rapid recovery in patients undergoing colonoscopy. We aimed to examine the efficacy and safety of ciprofol in comparison with propofol for sedation or anesthesia in non-operating room settings including endoscopic submucosal dissection, endoscopic retrograde cholangiopancreatography, and flexible bronchoscopy (FB). DESIGN Prospective, randomized, double-blind, parallel-group clinical trial. SETTING University-affiliated teaching hospital. PATIENTS We recruited 207 patients scheduled for an endoscopic procedure from October 2021 to December 2021. INTERVENTIONS Patients were randomized into three groups according to the dose during induction (n = 69 each): 1) ciprofol 6 mg/kg/h, 2) ciprofol 8 mg/kg/h, or 3) propofol 40 mg/kg/h. Ciprofol or propofol was administered throughout the procedure. MEASUREMENTS The primary outcome was the success rate of sedation or anesthesia for the procedures. Secondary outcomes included induction time, endoscope insertion time, recovery time, discharge time, incidence of drug-related adverse events (AEs), neurological and inflammatory outcomes. MAIN RESULTS The procedure success rates in the three groups were 100%. The induction time in the 6 (3.3 ± 1.0 min) and 8 mg/kg/h (2.9 ± 0.6 min) ciprofol groups was longer than that in the propofol group (2.5 ± 0.6 min) only in patients undergoing FB (p = 0.004). The time for patients to be fully alert and discharged from the post-anesthesia care unit was comparable across the three groups (p > 0.05). The incidence of drug-related AEs in the propofol and 6 and 8 mg/kg/h ciprofol groups was 84.1%, 76.8%, and 79.7%. No pain on injection was reported by ciprofol groups. Neurological outcomes and inflammatory responses were comparable among the three groups. CONCLUSIONS Ciprofol induced a level of sedation or anesthesia equivalent to that induced by propofol in non-operating room settings except for a prolonged induction time in patients undergoing FB. Ciprofol had a safety profile similar to that of propofol. No pain on injection was reported by ciprofol.
Collapse
|
10
|
Roles of Neuropeptides in Sleep-Wake Regulation. Int J Mol Sci 2022; 23:ijms23094599. [PMID: 35562990 PMCID: PMC9103574 DOI: 10.3390/ijms23094599] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/31/2022] [Accepted: 04/19/2022] [Indexed: 12/04/2022] Open
Abstract
Sleep and wakefulness are basic behavioral states that require coordination between several brain regions, and they involve multiple neurochemical systems, including neuropeptides. Neuropeptides are a group of peptides produced by neurons and neuroendocrine cells of the central nervous system. Like traditional neurotransmitters, neuropeptides can bind to specific surface receptors and subsequently regulate neuronal activities. For example, orexin is a crucial component for the maintenance of wakefulness and the suppression of rapid eye movement (REM) sleep. In addition to orexin, melanin-concentrating hormone, and galanin may promote REM sleep. These results suggest that neuropeptides play an important role in sleep–wake regulation. These neuropeptides can be divided into three categories according to their effects on sleep–wake behaviors in rodents and humans. (i) Galanin, melanin-concentrating hormone, and vasoactive intestinal polypeptide are sleep-promoting peptides. It is also noticeable that vasoactive intestinal polypeptide particularly increases REM sleep. (ii) Orexin and neuropeptide S have been shown to induce wakefulness. (iii) Neuropeptide Y and substance P may have a bidirectional function as they can produce both arousal and sleep-inducing effects. This review will introduce the distribution of various neuropeptides in the brain and summarize the roles of different neuropeptides in sleep–wake regulation. We aim to lay the foundation for future studies to uncover the mechanisms that underlie the initiation, maintenance, and end of sleep–wake states.
Collapse
|
11
|
Zhang F, Xiong Y, Qin F, Yuan J. Short Sleep Duration and Erectile Dysfunction: A Review of the Literature. Nat Sci Sleep 2022; 14:1945-1961. [PMID: 36325277 PMCID: PMC9621223 DOI: 10.2147/nss.s375571] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 10/14/2022] [Indexed: 11/05/2022] Open
Abstract
The meaning of sleep has puzzled people for millennia. In modern society, short sleep duration is becoming a global problem. It has been established that short sleep duration can increase the risk of several diseases, such as cardiovascular and metabolic diseases. Currently, a growing body of research has revealed a possible link between sleep disorders and erectile dysfunction (ED). However, the mechanisms linking short sleep duration and ED are largely unknown. Thus, we provide a review of clinical trials and animal studies. In this review, we propose putative pathways connecting short sleep duration and ED, including neuroendocrine pathways and molecular mechanisms, aiming to pave the way for future research. Meanwhile, the assessment and improvement of sleep quality should be recommended in the diagnosis and treatment of ED patients.
Collapse
Affiliation(s)
- Fuxun Zhang
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, People's Republic of China.,Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yang Xiong
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, People's Republic of China.,Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Feng Qin
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Jiuhong Yuan
- Andrology Laboratory, West China Hospital, Sichuan University, Chengdu, People's Republic of China.,Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
12
|
Toenders YJ, Laskaris L, Davey CG, Berk M, Milaneschi Y, Lamers F, Penninx BWJH, Schmaal L. Inflammation and depression in young people: a systematic review and proposed inflammatory pathways. Mol Psychiatry 2022; 27:315-327. [PMID: 34635789 DOI: 10.1038/s41380-021-01306-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/12/2021] [Accepted: 09/13/2021] [Indexed: 02/07/2023]
Abstract
Depression onset peaks during adolescence and young adulthood. Current treatments are only moderately effective, driving the search for novel pathophysiological mechanisms underlying youth depression. Inflammatory dysregulation has been shown in adults with depression, however, less is known about inflammation in youth depression. This systematic review identified 109 studies examining the association between inflammation and youth depression and showed subtle evidence for inflammatory dysregulation in youth depression. Longitudinal studies support the bidirectional association between inflammation and depression in youth. We hypothesise multiple inflammatory pathways contributing to depression. More research is needed on anti-inflammatory treatments, potentially tailored to individual symptom profiles.
Collapse
Affiliation(s)
- Yara J Toenders
- Orygen, Parkville, VIC, Australia.,Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Liliana Laskaris
- Orygen, Parkville, VIC, Australia.,Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Christopher G Davey
- Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia
| | - Michael Berk
- Orygen, Parkville, VIC, Australia.,Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia.,Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia.,IMPACT-the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, VIC, Australia.,Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam UMC, Department of Amsterdam Public Health Research Institute and Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Femke Lamers
- Department of Psychiatry, Amsterdam UMC, Department of Amsterdam Public Health Research Institute and Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam UMC, Department of Amsterdam Public Health Research Institute and Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Lianne Schmaal
- Orygen, Parkville, VIC, Australia. .,Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
13
|
Chen QC, Zhang Y. The Role of NPY in the Regulation of Bone Metabolism. Front Endocrinol (Lausanne) 2022; 13:833485. [PMID: 35273572 PMCID: PMC8902412 DOI: 10.3389/fendo.2022.833485] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 01/20/2022] [Indexed: 11/13/2022] Open
Abstract
Bone diseases are the leading causes of disability and severely compromised quality of life. Neuropeptide Y (NPY) is a multifunctional neuropeptide that participates in various physiological and pathological processes and exists in both the nerve system and bone tissue. In bone tissue, it actively participates in bone metabolism and disease progression through its receptors. Previous studies have focused on the opposite effects of NPY on bone formation and resorption through paracrine modes. In this review, we present a brief overview of the progress made in this research field in recent times in order to provide reference for further understanding the regulatory mechanism of bone physiology and pathological metabolism.
Collapse
Affiliation(s)
- Qing-Chang Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yan Zhang
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Yan Zhang,
| |
Collapse
|
14
|
Patthy Á, Murai J, Hanics J, Pintér A, Zahola P, Hökfelt TGM, Harkany T, Alpár A. Neuropathology of the Brainstem to Mechanistically Understand and to Treat Alzheimer's Disease. J Clin Med 2021; 10:jcm10081555. [PMID: 33917176 PMCID: PMC8067882 DOI: 10.3390/jcm10081555] [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: 03/15/2021] [Revised: 04/01/2021] [Accepted: 04/05/2021] [Indexed: 12/18/2022] Open
Abstract
Alzheimer’s disease (AD) is a devastating neurodegenerative disorder as yet without effective therapy. Symptoms of this disorder typically reflect cortical malfunction with local neurohistopathology, which biased investigators to search for focal triggers and molecular mechanisms. Cortex, however, receives massive afferents from caudal brain structures, which do not only convey specific information but powerfully tune ensemble activity. Moreover, there is evidence that the start of AD is subcortical. The brainstem harbors monoamine systems, which establish a dense innervation in both allo- and neocortex. Monoaminergic synapses can co-release neuropeptides either by precisely terminating on cortical neurons or, when being “en passant”, can instigate local volume transmission. Especially due to its early damage, malfunction of the ascending monoaminergic system emerges as an early sign and possible trigger of AD. This review summarizes the involvement and cascaded impairment of brainstem monoaminergic neurons in AD and discusses cellular mechanisms that lead to their dysfunction. We highlight the significance and therapeutic challenges of transmitter co-release in ascending activating system, describe the role and changes of local connections and distant afferents of brainstem nuclei in AD, and summon the rapidly increasing diagnostic window during the last few years.
Collapse
Affiliation(s)
- Ágoston Patthy
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
| | - János Murai
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
| | - János Hanics
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences, H-1094 Budapest, Hungary
| | - Anna Pintér
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
| | - Péter Zahola
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
| | - Tomas G. M. Hökfelt
- Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, 17165 Stockholm, Sweden; (T.G.M.H.); (T.H.)
| | - Tibor Harkany
- Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, 17165 Stockholm, Sweden; (T.G.M.H.); (T.H.)
- Center for Brain Research, Department of Molecular Neurosciences, Medical University of Vienna, 1090 Vienna, Austria
| | - Alán Alpár
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences, H-1094 Budapest, Hungary
- Correspondence:
| |
Collapse
|
15
|
Khoury S, Wang QP, Parisien M, Gris P, Bortsov AV, Linnstaedt SD, McLean SA, Tungate AS, Sofer T, Lee J, Louie T, Redline S, Kaunisto MA, Kalso EA, Munter HM, Nackley AG, Slade GD, Smith SB, Zaykin DV, Fillingim RB, Ohrbach R, Greenspan JD, Maixner W, Neely GG, Diatchenko L. Multi-ethnic GWAS and meta-analysis of sleep quality identify MPP6 as a novel gene that functions in sleep center neurons. Sleep 2021; 44:zsaa211. [PMID: 33034629 PMCID: PMC7953222 DOI: 10.1093/sleep/zsaa211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 08/28/2020] [Indexed: 11/14/2022] Open
Abstract
Poor sleep quality can have harmful health consequences. Although many aspects of sleep are heritable, the understandings of genetic factors involved in its physiology remain limited. Here, we performed a genome-wide association study (GWAS) using the Pittsburgh Sleep Quality Index (PSQI) in a multi-ethnic discovery cohort (n = 2868) and found two novel genome-wide loci on chromosomes 2 and 7 associated with global sleep quality. A meta-analysis in 12 independent cohorts (100 000 individuals) replicated the association on chromosome 7 between NPY and MPP6. While NPY is an important sleep gene, we tested for an independent functional role of MPP6. Expression data showed an association of this locus with both NPY and MPP6 mRNA levels in brain tissues. Moreover, knockdown of an orthologue of MPP6 in Drosophila melanogaster sleep center neurons resulted in decreased sleep duration. With convergent evidence, we describe a new locus impacting human variability in sleep quality through known NPY and novel MPP6 sleep genes.
Collapse
Affiliation(s)
- Samar Khoury
- The Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada
| | - Qiao-Ping Wang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Guangzhou, China
| | - Marc Parisien
- The Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada
| | - Pavel Gris
- The Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada
| | - Andrey V Bortsov
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University, Durham, NC
| | - Sarah D Linnstaedt
- Institute for Trauma Recovery and Department of Anesthesiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Samuel A McLean
- Institute for Trauma Recovery and Department of Anesthesiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Andrew S Tungate
- Institute for Trauma Recovery and Department of Anesthesiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Tamar Sofer
- Department of Medicine, Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Jiwon Lee
- Department of Medicine, Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Tin Louie
- Department of Biostatistics, University of Washington, Seattle, WA
| | - Susan Redline
- Department of Medicine, Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Mari Anneli Kaunisto
- Department of Diagnostics and Therapeutics, University of Helsinki, Helsinki, Finland
| | - Eija A Kalso
- Department of Diagnostics and Therapeutics, University of Helsinki, Helsinki, Finland
| | | | - Andrea G Nackley
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University, Durham, NC
| | - Gary D Slade
- School of dentistry, University of North Carolina Chapel Hill, Chapel Hill, NC
| | - Shad B Smith
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University, Durham, NC
| | - Dmitri V Zaykin
- Biostatistics and Computational Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | | | - Richard Ohrbach
- Department of Oral Diagnostic Services, University at Buffalo, Buffalo, NY
| | - Joel D Greenspan
- Department of Neural and Pain Sciences, Brotman Facial Pain Clinic, School of Dentistry and Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD
| | - William Maixner
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University, Durham, NC
| | - G Gregory Neely
- The Dr. John and Anne Chong Laboratory for Functional Genomics, Charles Perkins Centre and School of Life & Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Luda Diatchenko
- The Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada
| |
Collapse
|
16
|
Alcohol. Alcohol 2021. [DOI: 10.1016/b978-0-12-816793-9.00001-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
17
|
Aly J, Engmann O. The Way to a Human's Brain Goes Through Their Stomach: Dietary Factors in Major Depressive Disorder. Front Neurosci 2020; 14:582853. [PMID: 33364919 PMCID: PMC7750481 DOI: 10.3389/fnins.2020.582853] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
Globally, more than 250 million people are affected by depression (major depressive disorder; MDD), a serious and debilitating mental disorder. Currently available treatment options can have substantial side effects and take weeks to be fully effective. Therefore, it is important to find safe alternatives, which act more rapidly and in a larger number of patients. While much research on MDD focuses on chronic stress as a main risk factor, we here make a point of exploring dietary factors as a somewhat overlooked, yet highly promising approach towards novel antidepressant pathways. Deficiencies in various groups of nutrients often occur in patients with mental disorders. These include vitamins, especially members of the B-complex (B6, B9, B12). Moreover, an imbalance of fatty acids, such as omega-3 and omega-6, or an insufficient supply with minerals, including magnesium and zinc, are related to MDD. While some of them are relevant for the synthesis of monoamines, others play a crucial role in inflammation, neuroprotection and the synthesis of growth factors. Evidence suggests that when deficiencies return to normal, changes in mood and behavior can be, at least in some cases, achieved. Furthermore, supplementation with dietary factors (so called "nutraceuticals") may improve MDD symptoms even in the absence of a deficiency. Non-vital dietary factors may affect MDD symptoms as well. For instance, the most commonly consumed psychostimulant caffeine may improve behavioral and molecular markers of MDD. The molecular structure of most dietary factors is well known. Hence, dietary factors may provide important molecular tools to study and potentially help treat MDD symptoms. Within this review, we will discuss the role of dietary factors in MDD risk and symptomology, and critically discuss how they might serve as auxiliary treatments or preventative options for MDD.
Collapse
Affiliation(s)
- Janine Aly
- Faculty of Medicine, Friedrich Schiller Universität, Jena, Germany
| | - Olivia Engmann
- Institute for Human Genetics, Jena University Hospital, Jena, Germany
| |
Collapse
|
18
|
Méndez-Couz M, Manahan-Vaughan D, Silva AP, González-Pardo H, Arias JL, Conejo NM. Metaplastic contribution of neuropeptide Y receptors to spatial memory acquisition. Behav Brain Res 2020; 396:112864. [PMID: 32827566 DOI: 10.1016/j.bbr.2020.112864] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/21/2022]
Abstract
Neuropeptide Y (NPY) is highly abundant in the brain and is released as a co-transmitter with plasticity-related neurotransmitters such as glutamate, GABA and noradrenaline. Functionally, its release is associated with appetite, anxiety, and stress regulation. NPY acting on Y2 receptors (Y2R), facilitates fear extinction, suggesting a role in associative memory. Here, we explored to what extent NPY action at Y2R contributes to hippocampus-dependent spatial memory and found that dorsal intrahippocampal receptor antagonism improved spatial reference memory acquired in a water maze in rats, without affecting anxiety levels, or spontaneous motor activity. Water maze training resulted in an increase of Y2R, but not Y1R expression in the hippocampus. By contrast, in the prefrontal cortex there was a decrease in Y2R, and an increase of Y1R expression. Our results indicate that neuropeptide Y2R are significantly involved in hippocampus-dependent spatial memory and that receptor expression is dynamically regulated by this learning experience. Effects are consistent with a metaplastic contribution of NPY receptors to cumulative spatial learning.
Collapse
Affiliation(s)
- Marta Méndez-Couz
- Laboratory of Neuroscience, Department of Psychology, Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Spain; Ruhr University Bochum, Medical Faculty, Dept. Neurophysiology, Bochum, Germany.
| | | | - Ana Paula Silva
- Laboratory of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine and Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal
| | - Héctor González-Pardo
- Laboratory of Neuroscience, Department of Psychology, Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Spain
| | - Jorge Luis Arias
- Laboratory of Neuroscience, Department of Psychology, Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Spain
| | - Nélida María Conejo
- Laboratory of Neuroscience, Department of Psychology, Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Spain
| |
Collapse
|
19
|
Koob GF, Colrain IM. Alcohol use disorder and sleep disturbances: a feed-forward allostatic framework. Neuropsychopharmacology 2020; 45:141-165. [PMID: 31234199 PMCID: PMC6879503 DOI: 10.1038/s41386-019-0446-0] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 06/09/2019] [Accepted: 06/13/2019] [Indexed: 11/09/2022]
Abstract
The development of alcohol use disorder (AUD) involves binge or heavy drinking to high levels of intoxication that leads to compulsive intake, the loss of control in limiting intake, and a negative emotional state when alcohol is removed. This cascade of events occurs over an extended period within a three-stage cycle: binge/intoxication, withdrawal/negative affect, and preoccupation/anticipation. These three heuristic stages map onto the dysregulation of functional domains of incentive salience/habits, negative emotional states, and executive function, mediated by the basal ganglia, extended amygdala, and frontal cortex, respectively. Sleep disturbances, alterations of sleep architecture, and the development of insomnia are ubiquitous in AUD and also map onto the three stages of the addiction cycle. During the binge/intoxication stage, alcohol intoxication leads to a faster sleep onset, but sleep quality is poor relative to nights when no alcohol is consumed. The reduction of sleep onset latency and increase in wakefulness later in the night may be related to the acute effects of alcohol on GABAergic systems that are associated with sleep regulation and the effects on brain incentive salience systems, such as dopamine. During the withdrawal/negative affect stage, there is a decrease in slow-wave sleep and some limited recovery in REM sleep when individuals with AUD stop drinking. Limited recovery of sleep disturbances is seen in AUD within the first 30 days of abstinence. The effects of withdrawal on sleep may be related to the loss of alcohol as a positive allosteric modulator of GABAA receptors, a decrease in dopamine function, and the overactivation of stress neuromodulators, including hypocretin/orexin, norepinephrine, corticotropin-releasing factor, and cytokines. During the preoccupation/anticipation stage, individuals with AUD who are abstinent long-term present persistent sleep disturbances, including a longer latency to fall asleep, more time awake during the night, a decrease in slow-wave sleep, decreases in delta electroencephalogram power and evoked delta activity, and an increase in REM sleep. Glutamatergic system dysregulation that is observed in AUD is a likely substrate for some of these persistent sleep disturbances. Sleep pathology contributes to AUD pathology, and vice versa, possibly as a feed-forward drive to an unrecognized allostatic load that drives the addiction process.
Collapse
Affiliation(s)
- George F Koob
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 6700B Rockledge Drive, Room 1209, MSC 6902, Bethesda, MD, 20892-6902, USA.
- National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, 20892-6902, USA.
| | - Ian M Colrain
- SRI Biosciences, SRI International, Menlo Park, CA, USA
- Melbourne School of Psychological Sciences, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
20
|
Hofmann S, Bellmann-Sickert K, Beck-Sickinger AG. Chemical modification of neuropeptide Y for human Y1 receptor targeting in health and disease. Biol Chem 2019; 400:299-311. [PMID: 30653463 DOI: 10.1515/hsz-2018-0364] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/17/2018] [Indexed: 12/14/2022]
Abstract
As a very abundant neuropeptide in the brain and widely distributed peptide hormone in the periphery, neuropeptide Y (NPY) appears to be a multisignaling key peptide. Together with peptide YY, pancreatic polypeptide and the four human G protein-coupled receptor subtypes hY1R, hY2R, hY4R and hY5R it forms the NPY/hYR multiligand/multireceptor system, which is involved in essential physiological processes as well as in human diseases. In particular, NPY-induced hY1R signaling plays a central role in the regulation of food intake and stress response as well as in obesity, mood disorders and cancer. Thus, several hY1R-preferring NPY analogs have been developed as versatile tools to unravel the complex NPY/hY1R signaling in health and disease. Further, these peptides provide basic lead structures for the development of innovative drugs. Here, the current research is summarized focusing on the development of differently sized hY1R-preferring NPY analogs as well as their advances with respect to hY1R profiling, potential therapeutic applications and targeted cancer imaging and therapy. Finally, major limitations and innovative strategies for next generation hY1R-preferring NPY analogs are addressed.
Collapse
Affiliation(s)
- Sven Hofmann
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, D-04103 Leipzig, Germany
| | - Kathrin Bellmann-Sickert
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, D-04103 Leipzig, Germany
| | - Annette G Beck-Sickinger
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstr. 34, D-04103 Leipzig, Germany
| |
Collapse
|
21
|
Yamakawa GR, Weerawardhena H, Eyolfson E, Griep Y, Antle MC, Mychasiuk R. Investigating the Role of the Hypothalamus in Outcomes to Repetitive Mild Traumatic Brain Injury: Neonatal Monosodium Glutamate Does Not Exacerbate Deficits. Neuroscience 2019; 413:264-278. [DOI: 10.1016/j.neuroscience.2019.06.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 05/29/2019] [Accepted: 06/15/2019] [Indexed: 12/20/2022]
|
22
|
Juneau ZC, Stonemetz JM, Toma RF, Possidente DR, Heins RC, Vecsey CG. Optogenetic activation of short neuropeptide F (sNPF) neurons induces sleep in Drosophila melanogaster. Physiol Behav 2019; 206:143-156. [PMID: 30935941 PMCID: PMC6520144 DOI: 10.1016/j.physbeh.2019.03.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 01/18/2019] [Accepted: 03/28/2019] [Indexed: 01/31/2023]
Abstract
Sleep abnormalities have widespread and costly public health consequences, yet we have only a rudimentary understanding of the events occurring at the cellular level in the brain that regulate sleep. Several key signaling molecules that regulate sleep across taxa come from the family of neuropeptide transmitters. For example, in Drosophila melanogaster, the neuropeptide Y (NPY)-related transmitter short neuropeptide F (sNPF) appears to promote sleep. In this study, we utilized optogenetic activation of neuronal populations expressing sNPF to determine the causal effects of precisely timed activity in these cells on sleep behavior. Combining sNPF-GAL4 and UAS-Chrimson transgenes allowed us to activate sNPF neurons using red light. We found that activating sNPF neurons for as little as 3 s at a time of day when most flies were awake caused a rapid transition to sleep that persisted for another 2+ hours following the stimulation. Changing the timing of red light stimulation to times of day when flies were already asleep caused the control flies to wake up (due to the pulse of light), but the flies in which sNPF neurons were activated stayed asleep through the light pulse, and then showed further increases in sleep at later points when they would have normally been waking up. Video recording of individual fly responses to short-term (0.5-20 s) activation of sNPF neurons demonstrated a clear light duration-dependent decrease in movement during the subsequent 4-min period. These results provide supportive evidence that sNPF-producing neurons promote long-lasting increases in sleep, and show for the first time that even brief periods of activation of these neurons can cause changes in behavior that persist after cessation of activation. We have also presented evidence that sNPF neuron activation produces a homeostatic sleep drive that can be dissipated at times long after the neurons were stimulated. Future studies will determine the specific roles of sub-populations of sNPF-producing neurons, and will also assess how sNPF neurons act in concert with other neuronal circuits to control sleep.
Collapse
Affiliation(s)
- Zoe Claire Juneau
- Neuroscience Program, Skidmore College, 815 N. Broadway, Saratoga Springs, NY 12866, United States of America
| | - Jamie M Stonemetz
- Neuroscience Program, Skidmore College, 815 N. Broadway, Saratoga Springs, NY 12866, United States of America
| | - Ryan F Toma
- Neuroscience Program, Skidmore College, 815 N. Broadway, Saratoga Springs, NY 12866, United States of America
| | - Debra R Possidente
- Neuroscience Program, Skidmore College, 815 N. Broadway, Saratoga Springs, NY 12866, United States of America
| | - R Conor Heins
- Biology Department, Swarthmore College, 500 College Avenue, Swarthmore, PA 19081, United States of America
| | - Christopher G Vecsey
- Neuroscience Program, Skidmore College, 815 N. Broadway, Saratoga Springs, NY 12866, United States of America; Biology Department, Swarthmore College, 500 College Avenue, Swarthmore, PA 19081, United States of America.
| |
Collapse
|
23
|
In vivo cell type-specific CRISPR knockdown of dopamine beta hydroxylase reduces locus coeruleus evoked wakefulness. Nat Commun 2018; 9:5211. [PMID: 30523254 PMCID: PMC6283864 DOI: 10.1038/s41467-018-07566-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 09/13/2018] [Indexed: 11/08/2022] Open
Abstract
Locus coeruleus (LC) neurons in the brainstem have long been associated with attention and arousal. Optogenetic stimulation of LC-NE neurons induces immediate sleep-to-wake transitions. However, LC neurons also secrete other neurotransmitters in addition to NE. To interrogate the role of NE derived from the LC in regulating wakefulness, we applied in vivo cell type-specific CRISPR/Cas9 technology to disrupt the dopamine beta hydroxylase (dbh) gene selectively in adult LC-NE neurons. Unilateral dbh gene disruption abolished immediate arousal following optogenetic stimulation of LC. Bilateral LC-specific dbh disruption significantly reduced NE concentration in LC projection areas and reduced wake length even in the presence of salient stimuli. These results suggest that NE may be crucial for the awakening effect of LC stimulation and serve as proof-of-principle that CRISPR gene editing in adult neurons can be used to interrogate gene function within genetically-defined neuronal circuitry associated with complex behaviors.
Collapse
|
24
|
Hypothalamic Neurons that Regulate Feeding Can Influence Sleep/Wake States Based on Homeostatic Need. Curr Biol 2018; 28:3736-3747.e3. [PMID: 30471995 DOI: 10.1016/j.cub.2018.09.055] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 07/10/2018] [Accepted: 09/25/2018] [Indexed: 12/22/2022]
Abstract
Eating and sleeping represent two mutually exclusive behaviors that satisfy distinct homeostatic needs. Because an animal cannot eat and sleep at the same time, brain systems that regulate energy homeostasis are likely to influence sleep/wake behavior. Indeed, previous studies indicate that animals adjust sleep cycles around periods of food need and availability. Furthermore, hormones that affect energy homeostasis also affect sleep/wake states: the orexigenic hormone ghrelin promotes wakefulness, and the anorexigenic hormones leptin and insulin increase the duration of slow-wave sleep. However, whether neural populations that regulate feeding can influence sleep/wake states is unknown. The hypothalamic arcuate nucleus contains two neuronal populations that exert opposing effects on energy homeostasis: agouti-related protein (AgRP)-expressing neurons detect caloric need and orchestrate food-seeking behavior, whereas activity in pro-opiomelanocortin (POMC)-expressing neurons induces satiety. We tested the hypotheses that AgRP neurons affect sleep homeostasis by promoting states of wakefulness, whereas POMC neurons promote states of sleep. Indeed, optogenetic or chemogenetic stimulation of AgRP neurons in mice promoted wakefulness while decreasing the quantity and integrity of sleep. Inhibition of AgRP neurons rescued sleep integrity in food-deprived mice, highlighting the physiological importance of AgRP neuron activity for the suppression of sleep by hunger. Conversely, stimulation of POMC neurons promoted sleep states and decreased sleep fragmentation in food-deprived mice. Interestingly, we also found that sleep deprivation attenuated the effects of AgRP neuron activity on food intake and wakefulness. These results indicate that homeostatic feeding neurons can hierarchically affect behavioral outcomes, depending on homeostatic need.
Collapse
|
25
|
Neuropeptide Y impairs the acquisition of conditioned defeat in Syrian hamsters. Neurosci Lett 2018; 690:214-218. [PMID: 30312751 DOI: 10.1016/j.neulet.2018.09.049] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/10/2018] [Accepted: 09/25/2018] [Indexed: 12/12/2022]
Abstract
Recent evidence indicates that Neuropeptide Y (NPY) may function as a potent anxiolytic as well as a resilience factor that can insulate the brain from the effects of stress. However, most of these studies have utilized physical stressors such as shock or restraint. In the present study, we use an ethologically-based model in Syrian hamsters (Mesocricetus auratus) called Conditioned Defeat (CD) to investigate whether NPY can ameliorate the effect of social defeat stress. In the CD model, a male Syrian hamster is socially defeated by a larger, more aggressive conspecific. Subsequently, when paired with a smaller, non-aggressive intruder (NAI) in its own home cage, changes in its behavioral repertoire occur, including a reduction in aggression and chemosensory (social) investigation, and a concomitant increase in submissive behaviors. In Experiment 1, hamsters were infused intracerebroventricularly (icv) with NPY prior to social defeat, and 24-hours later, hamsters were exposed to a NAI. Results indicate that NPY significantly reduced submissive/defensive behaviors in socially defeated hamsters compared to control animals. In Experiment 2, we examined whether this effect was mediated by the NPY Y1 receptor. Subjects were first pre-treated with the Y1 receptor antagonist BIBP 3226 or vehicle, followed by NPY and then socially defeated. Upon testing with a NAI 24-hours later, pretreatment with BIBP 3226 failed to block the NPY effect compared to controls. These results demonstrate that NPY may function as an important resilience factor in socially defeated hamsters, but that these effects are not mediated by the Y1 receptor.
Collapse
|
26
|
Balkan B, Pogun S. Nicotinic Cholinergic System in the Hypothalamus Modulates the Activity of the Hypothalamic Neuropeptides During the Stress Response. Curr Neuropharmacol 2018; 16:371-387. [PMID: 28730966 PMCID: PMC6018196 DOI: 10.2174/1570159x15666170720092442] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 07/14/2017] [Accepted: 07/18/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The hypothalamus harbors high levels of cholinergic neurons and axon terminals. Nicotinic acetylcholine receptors, which play an important role in cholinergic neurotransmission, are expressed abundantly in the hypothalamus. Accumulating evidence reveals a regulatory role for nicotine in the regulation of the stress responses. The present review will discuss the hypothalamic neuropeptides and their interaction with the nicotinic cholinergic system. The anatomical distribution of the cholinergic neurons, axon terminals and nicotinic receptors in discrete hypothalamic nuclei will be described. The effect of nicotinic cholinergic neurotransmission and nicotine exposure on hypothalamic-pituitaryadrenal (HPA) axis regulation at the hypothalamic level will be analyzed in view of the different neuropeptides involved. METHODS Published research related to nicotinic cholinergic regulation of the HPA axis activity at the hypothalamic level is reviewed. RESULTS The nicotinic cholinergic system is one of the major modulators of the HPA axis activity. There is substantial evidence supporting the regulation of hypothalamic neuropeptides by nicotinic acetylcholine receptors. However, most of the studies showing the nicotinic regulation of hypothalamic neuropeptides have employed systemic administration of nicotine. Additionally, we know little about the nicotinic receptor distribution on neuropeptide-synthesizing neurons in the hypothalamus and the physiological responses they trigger in these neurons. CONCLUSION Disturbed functioning of the HPA axis and hypothalamic neuropeptides results in pathologies such as depression, anxiety disorders and obesity, which are common and significant health problems. A better understanding of the nicotinic regulation of hypothalamic neuropeptides will aid in drug development and provide means to cope with these diseases. Considering that nicotine is also an abused substance, a better understanding of the role of the nicotinic cholinergic system on the HPA axis will aid in developing improved therapeutic strategies for smoking cessation.
Collapse
Affiliation(s)
- Burcu Balkan
- Center for Brain Research, Ege University, Bornova, Izmir, Turkey.,Department of Physiology, School of Medicine, Ege University, Bornova, Izmir, Turkey
| | - Sakire Pogun
- Center for Brain Research, Ege University, Bornova, Izmir, Turkey
| |
Collapse
|
27
|
Quirk S, Hopkins MM, Bureau H, Lusk RJ, Allen C, Hernandez R, Bain DL. Mutational Analysis of Neuropeptide Y Reveals Unusual Thermal Stability Linked to Higher-Order Self-Association. ACS OMEGA 2018; 3:2141-2154. [PMID: 29619413 PMCID: PMC5876621 DOI: 10.1021/acsomega.7b01949] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 02/08/2018] [Indexed: 06/08/2023]
Abstract
Neuropeptide Y (NPY) is a 36-residue peptide, abundant in the central and peripheral nervous system. The peptide interacts with membrane-bound receptors to control processes such as food intake, vasoconstriction, and memory retention. The N-terminal polyproline sequence of NPY folds back onto a C-terminal α-helix to form a hairpin structure. The hairpin undergoes transient unfolding to allow the monomer to interact with its target membranes and receptors and to form reversible dimers in solution. Using computational, functional, and biophysical approaches, we characterized the role of two conserved tyrosines (Y20 and Y27) located within the hydrophobic core of the hairpin fold. Successive mutation of the tyrosines to more hydrophobic phenylalanines increased the thermal stability of NPY and reduced functional activity, consistent with computational studies predicting a more stable hairpin structure. However, mutant stability was high relative to wild-type: melting temperatures increased by approximately 20 °C for the single mutants (Y20F and Y27F) and by 30 °C for the double mutant (Y20F + Y27F). These findings suggested that the mutations were not just simply enhancing hairpin structure stability, but might also be driving self-association to dimer. Using analytical ultracentrifugation, we determined that the mutations indeed increased self-association, but shifted the equilibrium toward hexamer-like species. Notably, these latter species were not unique to the NPY mutants, but were found to preexist at low levels in the wild-type population. Collectively, the findings indicate that NPY self-association is more complex than previously recognized and that the ensemble of NPY quaternary states is tunable by modulating hairpin hydrophobicity.
Collapse
Affiliation(s)
- Stephen Quirk
- Archeus
Bioscience, 7094 Peachtree
Industrial Blvd., Norcross, Georgia 30071, United
States
| | - Mandi M. Hopkins
- Department
of Pharmaceutical Sciences, University of
Colorado Anschutz Medical Campus, 12850 E Montview Blvd., Aurora, Colorado 80045, United
States
| | - Hailey Bureau
- Center
for Computational and Molecular Science and Technology, School of
Chemistry and Biochemistry, Georgia Institute
of Technology, 901 Atlantic
Dr, Atlanta, Georgia 30332, United States
| | - Ryan J. Lusk
- Department
of Pharmaceutical Sciences, University of
Colorado Anschutz Medical Campus, 12850 E Montview Blvd., Aurora, Colorado 80045, United
States
| | - Caley Allen
- Department
of Chemistry, Johns Hopkins University, 3400 N Charles Street, Baltimore, Maryland 21218, United States
| | - Rigoberto Hernandez
- Center
for Computational and Molecular Science and Technology, School of
Chemistry and Biochemistry, Georgia Institute
of Technology, 901 Atlantic
Dr, Atlanta, Georgia 30332, United States
- Department
of Chemistry, Johns Hopkins University, 3400 N Charles Street, Baltimore, Maryland 21218, United States
| | - David L. Bain
- Department
of Pharmaceutical Sciences, University of
Colorado Anschutz Medical Campus, 12850 E Montview Blvd., Aurora, Colorado 80045, United
States
| |
Collapse
|
28
|
Huang H, Zhu Y, Eliot MN, Knopik VS, McGeary JE, Carskadon MA, Hart AC. Combining Human Epigenetics and Sleep Studies in Caenorhabditis elegans: A Cross-Species Approach for Finding Conserved Genes Regulating Sleep. Sleep 2018; 40:3738764. [PMID: 28431118 DOI: 10.1093/sleep/zsx063] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Study Objectives We aimed to test a combined approach to identify conserved genes regulating sleep and to explore the association between DNA methylation and sleep length. Methods We identified candidate genes associated with shorter versus longer sleep duration in college students based on DNA methylation using Illumina Infinium HumanMethylation450 BeadChip arrays. Orthologous genes in Caenorhabditis elegans were identified, and we examined whether their loss of function affected C. elegans sleep. For genes whose perturbation affected C. elegans sleep, we subsequently undertook a small pilot study to re-examine DNA methylation in an independent set of human participants with shorter versus longer sleep durations. Results Eighty-seven out of 485,577 CpG sites had significant differential methylation in young adults with shorter versus longer sleep duration, corresponding to 52 candidate genes. We identified 34 C. elegans orthologs, including NPY/flp-18 and flp-21, which are known to affect sleep. Loss of five additional genes alters developmentally timed C. elegans sleep (B4GALT6/bre-4, DOCK180/ced-5, GNB2L1/rack-1, PTPRN2/ida-1, ZFYVE28/lst-2). For one of these genes, ZFYVE28 (also known as hLst2), the pilot replication study again found decreased DNA methylation associated with shorter sleep duration at the same two CpG sites in the first intron of ZFYVE28. Conclusions Using an approach that combines human epigenetics and C. elegans sleep studies, we identified five genes that play previously unidentified roles in C. elegans sleep. We suggest sleep duration in humans may be associated with differential DNA methylation at specific sites and that the conserved genes identified here likely play roles in C. elegans sleep and in other species.
Collapse
Affiliation(s)
- Huiyan Huang
- Department of Neuroscience, Brown University, Providence, RI
| | - Yong Zhu
- Department of Environmental Health Sciences, Yale University School of Public Health, New Haven, CT
| | - Melissa N Eliot
- Department of Epidemiology, Brown University, Providence, RI
| | - Valerie S Knopik
- Division of Behavioral Genetics, Department of Psychiatry, Rhode Island Hospital, Providence, RI.,Department of Psychiatry and Human Behavior, Brown University, Providence, RI
| | - John E McGeary
- Division of Behavioral Genetics, Department of Psychiatry, Rhode Island Hospital, Providence, RI.,Department of Psychiatry and Human Behavior, Brown University, Providence, RI.,Providence Veterans Affairs Medical Center, Providence, RI
| | - Mary A Carskadon
- Department of Psychiatry and Human Behavior, Brown University, Providence, RI.,E.P. Bradley Hospital Sleep Research Laboratory, Providence, RI.,Center for Sleep Research, University of South Australia, Adelaide, Australia
| | - Anne C Hart
- Department of Neuroscience, Brown University, Providence, RI
| |
Collapse
|
29
|
Sleeping Beauty? Developmental Timing, Sleep, and the Circadian Clock in Caenorhabditis elegans. ADVANCES IN GENETICS 2017; 97:43-80. [PMID: 28838356 DOI: 10.1016/bs.adgen.2017.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The genetics toolkit is pretty successful in drilling down into minutiae. The big challenge is to integrate the information from this specialty as well as those of biochemistry, physiology, behavior, and anatomy to explain how fundamental biological processes really work. Sleep, the circadian clock and development all qualify as overarching processes that encompass levels from molecule to behavior as part of their known mechanisms. They overlap each other, such that understanding the mechanisms of one can lead to insights into one of the others. In this essay, we consider how the experimental approaches and findings relating to Caenorhabditis elegans development and lethargus on one hand, and to the circadian clock and sleep in higher organisms on the other, could complement and enhance one another.
Collapse
|
30
|
Lee EY, Hwang YG, Lee HS. Hypothalamic neuronal origin of neuropeptide Y (NPY) or cocaine- and amphetamine-regulated transcript (CART) fibers projecting to the tuberomammillary nucleus of the rat. Brain Res 2017; 1657:16-28. [DOI: 10.1016/j.brainres.2016.11.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/17/2016] [Accepted: 11/21/2016] [Indexed: 12/25/2022]
|
31
|
Eshragh J, Dhruva A, Paul SM, Cooper BA, Mastick J, Hamolsky D, Levine JD, Miaskowski C, Kober KM. Associations Between Neurotransmitter Genes and Fatigue and Energy Levels in Women After Breast Cancer Surgery. J Pain Symptom Manage 2017; 53:67-84.e7. [PMID: 27720787 PMCID: PMC5191954 DOI: 10.1016/j.jpainsymman.2016.08.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 07/19/2016] [Accepted: 08/03/2016] [Indexed: 12/16/2022]
Abstract
CONTEXT Fatigue is a common problem in oncology patients. Less is known about decrements in energy levels and the mechanisms that underlie both fatigue and energy. OBJECTIVES In patients with breast cancer, variations in neurotransmitter genes between lower and higher fatigue latent classes and between the higher and lower energy latent classes were evaluated. METHODS Patients completed assessments before and monthly for six months after surgery. Growth mixture modeling was used to identify distinct latent classes for fatigue severity and energy levels. Thirty candidate genes involved in various aspects of neurotransmission were evaluated. RESULTS Eleven single-nucleotide polymorphisms or haplotypes (i.e., ADRB2 rs1042718, BDNF rs6265, COMT rs9332377, CYP3A4 rs4646437, GALR1 rs949060, GCH1 rs3783642, NOS1 rs9658498, NOS1 rs2293052, NPY1R Haplotype A04, SLC6A2 rs17841327, and 5HTTLPR + rs25531 in SLC6A4) were associated with latent class membership for fatigue. Seven single-nucleotide polymorphisms or haplotypes (i.e., NOS1 rs471871, SLC6A1 rs2675163, SLC6A1 Haplotype D01, SLC6A2 rs36027, SLC6A3 rs37022, SLC6A4 rs2020942, and TAC1 rs2072100) were associated with latent class membership for energy. Three of 13 genes (i.e., NOS1, SLC6A2, and SLC6A4) were associated with latent class membership for both fatigue and energy. CONCLUSIONS Molecular findings support the hypothesis that fatigue and energy are distinct, yet related symptoms. Results suggest that a large number of neurotransmitters play a role in the development and maintenance of fatigue and energy levels in breast cancer patients.
Collapse
Affiliation(s)
- Jasmine Eshragh
- School of Nursing, University of California, San Francisco, California, USA
| | - Anand Dhruva
- School of Medicine, University of California, San Francisco, California, USA
| | - Steven M Paul
- School of Nursing, University of California, San Francisco, California, USA
| | - Bruce A Cooper
- School of Nursing, University of California, San Francisco, California, USA
| | - Judy Mastick
- School of Nursing, University of California, San Francisco, California, USA
| | - Deborah Hamolsky
- School of Nursing, University of California, San Francisco, California, USA
| | - Jon D Levine
- School of Medicine, University of California, San Francisco, California, USA
| | | | - Kord M Kober
- School of Nursing, University of California, San Francisco, California, USA.
| |
Collapse
|
32
|
Moriya S, Soga T, Wong DW, Parhar IS. Transcriptome composition of the preoptic area in mid-age and escitalopram treatment in male mice. Neurosci Lett 2016; 622:67-71. [PMID: 27113202 DOI: 10.1016/j.neulet.2016.04.052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 04/16/2016] [Accepted: 04/22/2016] [Indexed: 01/18/2023]
Abstract
The decrease in serotonergic neurotransmission during aging can increase the risk of neuropsychiatric diseases such as depression in elderly population and decline the reproductive system. Therefore, it is important to understand the age-associated molecular mechanisms of brain aging. In this study, the effect of aging and chronic escitalopram (antidepressant) treatment to admit mice was investigated by comparing transcriptomes in the preoptic area (POA) which is a key nucleus for reproduction. In the mid-aged brain, the immune system-related genes were increased and hormone response-related genes were decreased. In the escitalopram treated brains, transcription-, granule cell proliferation- and vasoconstriction-related genes were increased and olfactory receptors were decreased. Since homeostasis and neuroprotection-related genes were altered in both of mid-age and escitalopram treatment, these genes could be important for serotonin related physiologies in the POA.
Collapse
Affiliation(s)
- Shogo Moriya
- Brain Research Institutes, Monash University Malaysia, Bandar Sunway, Selangor 47500, Malaysia.
| | - Tomoko Soga
- Brain Research Institutes, Monash University Malaysia, Bandar Sunway, Selangor 47500, Malaysia
| | - Dutt Way Wong
- Brain Research Institutes, Monash University Malaysia, Bandar Sunway, Selangor 47500, Malaysia
| | - Ishwar S Parhar
- Brain Research Institutes, Monash University Malaysia, Bandar Sunway, Selangor 47500, Malaysia
| |
Collapse
|
33
|
Palavra F, Almeida L, Ambrósio AF, Reis F. Obesity and brain inflammation: a focus on multiple sclerosis. Obes Rev 2016; 17:211-24. [PMID: 26783119 DOI: 10.1111/obr.12363] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Revised: 10/25/2015] [Accepted: 11/09/2015] [Indexed: 02/06/2023]
Abstract
The increase in prevalence of obesity in industrialized societies is an indisputable fact. However, the apparent passive role played by adipocytes, in pathophysiological terms, has been gradually substituted by a metabolically active performance, relevant to many biochemical mechanisms that may contribute to a chronic low-grade inflammatory status, which increasingly imposes itself as a key feature of obesity. This chronic inflammatory status will have to be integrated into the complex equation of many diseases in which inflammation plays a crucial role. Multiple sclerosis (MS) is a chronic inflammatory condition typically confined to the central nervous system, and many work has been produced to find possible points of contact between the biology of this immune-mediated disease and obesity. So far, clinical data are not conclusive, but many biochemical features have been recently disclosed. Brain inflammation has been implicated in some of the mechanisms that lead to obesity, which has also been recognized as an important player in inducing some degree of immune dysfunction. In this review, we collected evidence that allows establishing bridges between obesity and MS. After considering epidemiological controversies, we will focus on possible shared mechanisms, as well as on the potential contributions that disease-modifying drugs may have on this apparent relationship of mutual interference.
Collapse
Affiliation(s)
- F Palavra
- Laboratory of Pharmacology & Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute for Biomedical Imaging and Life Sciences (CNC.IBILI) Research Consortium, University of Coimbra, Coimbra, Portugal
| | - L Almeida
- Laboratory of Pharmacology & Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - A F Ambrósio
- Laboratory of Pharmacology & Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute for Biomedical Imaging and Life Sciences (CNC.IBILI) Research Consortium, University of Coimbra, Coimbra, Portugal.,Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, Portugal
| | - F Reis
- Laboratory of Pharmacology & Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute for Biomedical Imaging and Life Sciences (CNC.IBILI) Research Consortium, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
34
|
Abstract
Sleep and energy balance are essential for health. The two processes act in concert to regulate central and peripheral homeostasis. During sleep, energy is conserved due to suspended activity, movement, and sensory responses, and is redirected to restore and replenish proteins and their assemblies into cellular structures. During wakefulness, various energy-demanding activities lead to hunger. Thus, hunger promotes arousal, and subsequent feeding, followed by satiety that promotes sleep via changes in neuroendocrine or neuropeptide signals. These signals overlap with circuits of sleep-wakefulness, feeding, and energy expenditure. Here, we will briefly review the literature that describes the interplay between the circadian system, sleep-wake, and feeding-fasting cycles that are needed to maintain energy balance and a healthy metabolic profile. In doing so, we describe the neuroendocrine, hormonal/peptide signals that integrate sleep and feeding behavior with energy metabolism.
Collapse
Affiliation(s)
- Charu Shukla
- Department of Psychiatry, VA Boston Healthcare System, Harvard Medical School, West Roxbury, MA, USA
| | - Radhika Basheer
- Department of Psychiatry, VA Boston Healthcare System, Harvard Medical School, West Roxbury, MA, USA
| |
Collapse
|
35
|
Singh C, Oikonomou G, Prober DA. Norepinephrine is required to promote wakefulness and for hypocretin-induced arousal in zebrafish. eLife 2015; 4:e07000. [PMID: 26374985 PMCID: PMC4606453 DOI: 10.7554/elife.07000] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 09/14/2015] [Indexed: 12/12/2022] Open
Abstract
Pharmacological studies in mammals suggest that norepinephrine (NE) plays an important role in promoting arousal. However, the role of endogenous NE is unclear, with contradicting reports concerning the sleep phenotypes of mice lacking NE due to mutation of dopamine β-hydroxylase (dbh). To investigate NE function in an alternative vertebrate model, we generated dbh mutant zebrafish. In contrast to mice, these animals exhibit dramatically increased sleep. Surprisingly, despite an increase in sleep, dbh mutant zebrafish have a reduced arousal threshold. These phenotypes are also observed in zebrafish treated with small molecules that inhibit NE signaling, suggesting that they are caused by the lack of NE. Using genetic overexpression of hypocretin (Hcrt) and optogenetic activation of hcrt-expressing neurons, we also find that NE is important for Hcrt-induced arousal. These results establish a role for endogenous NE in promoting arousal and indicate that NE is a critical downstream effector of Hcrt neurons. DOI:http://dx.doi.org/10.7554/eLife.07000.001 Although the neural circuits that regulate sleep and wakefulness have yet to be fully identified, the importance of at least two brain regions is well established. These are the hypothalamus, a structure deep within the brain that controls a number of basic activities including hunger, thirst and sleep; and the brainstem, which connects the brain with the spinal cord. Specific neurons within the hypothalamus and brainstem regulate the sleep–wake cycle by signaling to one another using chemicals called neurotransmitters and neuropeptides. Throughout the day, some hypothalamic neurons release a neuropeptide called hypocretin, which helps maintain wakefulness. Hypocretin acts on neurons within the brainstem and causes them to release other neurotransmitters that promote wakefulness. However, the identity of these molecules is unclear. One candidate is norepinephrine. Drugs that enhance the effects of norepinephrine increase wakefulness, whereas those that block norepinephrine signaling promote sleep. Despite this, mice that have been genetically modified to lack the enzyme that produces norepinephrine exhibit relatively normal sleep. This may be because in mammals, norepinephrine also has important roles outside the brain, thus complicating the effects of this genetic modification on behavior. Alternatively, while zebrafish that lack norepinephrine are healthy, mice containing this modification die early in development. Treating these mice with a specific drug allows them to survive, but might affect their behavior. To clarify the role of norepinephrine and its interaction with hypocretin, Singh, Oikonomou and Prober created a new animal model by genetically modifying zebrafish. In contrast to mice, zebrafish that were unable to make norepinephrine slept more than normal fish, although they were also lighter sleepers and were more prone to being startled. A genetic modification that increases hypocretin signaling induces insomnia; Singh, Oikonomou and Prober found that this occurs only in animals with normal levels of norepinephrine. Thus, these experiments indicate that hypocretin does indeed promote wakefulness though norepinephrine. The work of Singh, Oikonomou and Prober has clarified the role of norepinephrine in regulating the sleep–wake cycle. These findings could help in the development of drugs that target the neurons that make hypocretin, which may improve treatments for sleep disorders. DOI:http://dx.doi.org/10.7554/eLife.07000.002
Collapse
Affiliation(s)
- Chanpreet Singh
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Grigorios Oikonomou
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - David A Prober
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| |
Collapse
|
36
|
Rohwedder A, Selcho M, Chassot B, Thum AS. Neuropeptide F neurons modulate sugar reward during associative olfactory learning ofDrosophilalarvae. J Comp Neurol 2015; 523:2637-64. [DOI: 10.1002/cne.23873] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 07/19/2015] [Accepted: 07/28/2015] [Indexed: 01/29/2023]
Affiliation(s)
- Astrid Rohwedder
- Department of Biology; University of Fribourg; Fribourg Switzerland
| | - Mareike Selcho
- Department of Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter; University of Würzburg; Würzburg Germany
| | - Bérénice Chassot
- Department of Biology; University of Fribourg; Fribourg Switzerland
| | - Andreas S. Thum
- Department of Biology; University of Fribourg; Fribourg Switzerland
- Department of Biology; University of Konstanz; Konstanz Germany
- Zukunftskolleg; University of Konstanz; Konstanz Germany
| |
Collapse
|
37
|
Expression of mRNA in the frontal cortex and hypothalamus in a rat model of acute carbon dioxide poisoning. Leg Med (Tokyo) 2015; 19:101-6. [PMID: 26257316 DOI: 10.1016/j.legalmed.2015.07.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 07/24/2015] [Accepted: 07/25/2015] [Indexed: 11/20/2022]
Abstract
Acute carbon dioxide (CO2) poisoning causes no specific features that are revealed upon autopsy, and the pathophysiological mechanism of this syndrome is unclear. To address this issue, in the present study, we exposed rats to CO2 concentrations ranging from 10% to 60% and determined the effects on mRNA expression. According to the results of Gene Ontology (GO) and cluster analyses of microarrays data, we selected the following genes for further analysis: alkylglycerone phosphate synthase (Agps), hypocretin (Hcrt), tyrosine hydroxylase (Th), heat shock protein beta 2 (Hspb2), and opioid receptor delta 1 (Oprd1) expressed in the frontal cortex and renin (Ren), pancreatic polypeptide (Ppy), corticotropin releasing hormone receptor 2 (Crhr2), carbonic anhydrase 1 (Car1), and hypocretin receptor 1 (Hcrtr1) expressed in the hypothalamus. We found significant differences between the expression levels of Agps and Hspb2 mRNAs in the frontal cortex and that of Ppy, Crhr2 mRNAs in the hypothalamus in the presence of high concentrations of CO2. Further investigation of these genes may clarify the pathophysiology of acute CO2 poisoning and facilitate the development of novel forensic tests that can diagnose the cause of death.
Collapse
|
38
|
Abstract
Sleep and wake are fundamental behavioral states whose molecular regulation remains mysterious. Brain states and body functions change dramatically between sleep and wake, are regulated by circadian and homeostatic processes, and depend on the nutritional and emotional condition of the animal. Sleep-wake transitions require the coordination of several brain regions and engage multiple neurochemical systems, including neuropeptides. Neuropeptides serve two main functions in sleep-wake regulation. First, they represent physiological states such as energy level or stress in response to environmental and internal stimuli. Second, neuropeptides excite or inhibit their target neurons to induce, stabilize, or switch between sleep-wake states. Thus, neuropeptides integrate physiological subsystems such as circadian time, previous neuron usage, energy homeostasis, and stress and growth status to generate appropriate sleep-wake behaviors. We review the roles of more than 20 neuropeptides in sleep and wake to lay the foundation for future studies uncovering the mechanisms that underlie the initiation, maintenance, and exit of sleep and wake states.
Collapse
Affiliation(s)
- Constance Richter
- Department of Molecular and Cellular Biology, Center for Brain Science, Division of Sleep Biology, Harvard University, Cambridge, Massachusetts 02138; ,
| | | | | |
Collapse
|
39
|
Koh K, Hamada A, Hamada Y, Yanase M, Sakaki M, Someya K, Narita M, Kuzumaki N, Ikegami D, Sakai H, Iseki M, Inada E, Narita M. Possible involvement of activated locus coeruleus–noradrenergic neurons in pain-related sleep disorders. Neurosci Lett 2015; 589:200-6. [DOI: 10.1016/j.neulet.2014.12.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 11/29/2014] [Accepted: 12/01/2014] [Indexed: 11/27/2022]
|
40
|
Lee JS, Lee EY, Lee HS. Hypothalamic, feeding/arousal-related peptidergic projections to the paraventricular thalamic nucleus in the rat. Brain Res 2015; 1598:97-113. [DOI: 10.1016/j.brainres.2014.12.029] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 11/06/2014] [Accepted: 12/11/2014] [Indexed: 11/25/2022]
|
41
|
Nagy S, Tramm N, Sanders J, Iwanir S, Shirley IA, Levine E, Biron D. Homeostasis in C. elegans sleep is characterized by two behaviorally and genetically distinct mechanisms. eLife 2014; 3:e04380. [PMID: 25474127 PMCID: PMC4273442 DOI: 10.7554/elife.04380] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 12/03/2014] [Indexed: 12/12/2022] Open
Abstract
Biological homeostasis invokes modulatory responses aimed at stabilizing internal conditions. Using tunable photo- and mechano-stimulation, we identified two distinct categories of homeostatic responses during the sleep-like state of Caenorhabditis elegans (lethargus). In the presence of weak or no stimuli, extended motion caused a subsequent extension of quiescence. The neuropeptide Y receptor homolog, NPR-1, and an inhibitory neuropeptide known to activate it, FLP-18, were required for this process. In the presence of strong stimuli, the correlations between motion and quiescence were disrupted for several minutes but homeostasis manifested as an overall elevation of the time spent in quiescence. This response to strong stimuli required the function of the DAF-16/FOXO transcription factor in neurons, but not that of NPR-1. Conversely, response to weak stimuli did not require the function of DAF-16/FOXO. These findings suggest that routine homeostatic stabilization of sleep may be distinct from homeostatic compensation following a strong disturbance. DOI:http://dx.doi.org/10.7554/eLife.04380.001 The regenerative properties of sleep are required by all animals, with even the simplest animal, the nematode Caenorhabditis elegans, displaying a sleep-like state called lethargus. During development, nematodes must pass through four larval stages en route to adulthood, and the end of each stage is preceded by a period of lethargus lasting 2 to 3 hr. Human sleep is divided into distinct stages that recur in a prescribed order throughout the night. Nematodes, on the other hand, simply experience alternating periods of activity and stillness as they sleep. Nevertheless, in both species, any disruptions to sleep automatically lead to adjustments of the rest of the sleep cycle to compensate for the disturbance and to ensure that the organism gets an adequate amount of sleep overall. To date, it has been assumed that a single mechanism is responsible for adjusting the sleep cycle after any disturbance, regardless of its severity. However, Nagy, Tramm, Sanders et al. now show that this is not the case in C. elegans. Sleeping nematodes that were lightly disturbed by exposing them to light or to vibrations—causing them to briefly increase their activity levels—compensated for the disturbance by lengthening their next inactive period. By contrast, worms that were vigorously agitated by stronger vibrations showed a different response: the alternating pattern of stillness and activity was disrupted for several minutes, followed by an overall increase in the length of time spent in the stillness phase. Experiments using genetically modified worms revealed that these two responses involve distinct molecular pathways. A signaling molecule called neuropeptide Y affects the response to minor sleep disruptions, whereas a transcription factor called DAF-16/FOXO is involved in the corresponding role after major disruptions. Given that neuropeptide Y has already been implicated in sleep regulation in humans and flies, it is not implausible that similar mechanisms may occur in response to disturbances of our own sleep. DOI:http://dx.doi.org/10.7554/eLife.04380.002
Collapse
Affiliation(s)
- Stanislav Nagy
- Institute for Biophysical Dynamics, University of Chicago, Chicago, United States
| | - Nora Tramm
- Department of Physics, University of Chicago, Chicago, United States
| | - Jarred Sanders
- Committee on Genetics, Genomics, and Systems Biology, University of Chicago, Chicago, United States
| | - Shachar Iwanir
- Department of Physics, University of Chicago, Chicago, United States
| | - Ian A Shirley
- Department of Physics, University of Chicago, Chicago, United States
| | - Erel Levine
- Department of Physics, Harvard University, Cambridge, United States
| | - David Biron
- Institute for Biophysical Dynamics, University of Chicago, Chicago, United States
| |
Collapse
|
42
|
Pearce WJ. In cerebrovascular circadian rhythms, EETs keep the beat. Focus on "Rhythmic expression of cytochrome P450 epoxygenases CYP4x1 and CYP2c11 in the rat brain and vasculature". Am J Physiol Cell Physiol 2014; 307:C986-8. [PMID: 25273881 DOI: 10.1152/ajpcell.00327.2014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- William J Pearce
- Loma Linda University School of Medicine, Loma Linda, California
| |
Collapse
|
43
|
When time stands still: an integrative review on the role of chronodisruption in posttraumatic stress disorder. Curr Opin Psychiatry 2014; 27:385-92. [PMID: 25023884 DOI: 10.1097/yco.0000000000000079] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW The human circadian system creates and maintains cellular and systemic rhythmicity essential to homeostasis. Loss of circadian rhythmicity fundamentally affects the neuroendocrine, immune and autonomic system, similar to chronic stress and, thus, may play a central role in the development of stress-related disorders. This article focuses on the role of circadian misalignment in the pathophysiology of posttraumatic stress disorder (PTSD). RECENT FINDINGS Sleep disruption is a core feature of PTSD supporting the important supraordinate pathophysiological role of circadian system in PTSD. Furthermore, direct and indirect human and animal PTSD research suggests circadian system linked neuroendocrine, immune, metabolic and autonomic dysregulation with blunted diurnal rhythms, specific sleep pattern pathologies and cognitive deficits, as well as endocannabinoid and neuropeptide Y system alterations and altered circadian gene expression, linking circadian misalignment to PTSD pathophysiology. SUMMARY PTSD development is associated with chronodisruption findings. Evaluation and treatment of sleep and circadian disruption should be the first steps in PTSD management. State-of-the-art methods of circadian rhythm assessment should be applied to bridge the gap between clinical significance and limited understanding of the relationship between traumatic stress, sleep and circadian system.
Collapse
|
44
|
Angelucci F, Gelfo F, Fiore M, Croce N, Mathé AA, Bernardini S, Caltagirone C. The effect of neuropeptide Y on cell survival and neurotrophin expression in in-vitro models of Alzheimer's disease. Can J Physiol Pharmacol 2014; 92:621-30. [PMID: 25026432 DOI: 10.1139/cjpp-2014-0099] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a disorder characterized by the accumulation of abnormally folded protein fragments in neurons, i.e., β-amyloid (Aβ) and tau protein, leading to cell death. Several neuropeptides present in the central nervous system (CNS) are believed to be involved in the pathophysiology of AD. Among them, neuropeptide Y (NPY), a small peptide widely distributed throughout the brain, has generated interest because of its role in neuroprotection against excitotoxicity in animal models of AD. In addition, it has been shown that NPY modulates neurogenesis. Interestingly, these latter effects are similar to those elicited by neurotrophins, which are critical molecules for the function and survival of neurons that degenerate during the course of AD. In this review we summarize the evidence for the involvement of NPY and neurotrophins in AD pathogenesis, and the similarity between them in CNS neurons. Finally, we recapitulate our recent in-vitro evidence for the involvement of neurotrophin nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) in the neuroprotective effect elicited by NPY in AD neuron-like models (neuroblastoma cells or primary cultures exposed to toxic concentrations of Aβ's pathogenic fragment 25-35), and propose a putative mechanism based on NPY-induced inhibition of voltage-dependent Ca(2+) influx in pre- and post-synaptic neurons.
Collapse
Affiliation(s)
- Francesco Angelucci
- a Department of Clinical and Behavioural Neurology, IRCCS Santa Lucia Foundation, Via Ardeatina 354, 00142 Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
45
|
Vecsey CG, Pírez N, Griffith LC. The Drosophila neuropeptides PDF and sNPF have opposing electrophysiological and molecular effects on central neurons. J Neurophysiol 2014; 111:1033-45. [PMID: 24353297 PMCID: PMC3949227 DOI: 10.1152/jn.00712.2013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 12/12/2013] [Indexed: 12/26/2022] Open
Abstract
Neuropeptides have widespread effects on behavior, but how these molecules alter the activity of their target cells is poorly understood. We employed a new model system in Drosophila melanogaster to assess the electrophysiological and molecular effects of neuropeptides, recording in situ from larval motor neurons, which transgenically express a receptor of choice. We focused on two neuropeptides, pigment-dispersing factor (PDF) and small neuropeptide F (sNPF), which play important roles in sleep/rhythms and feeding/metabolism. PDF treatment depolarized motor neurons expressing the PDF receptor (PDFR), increasing excitability. sNPF treatment had the opposite effect, hyperpolarizing neurons expressing the sNPF receptor (sNPFR). Live optical imaging using a genetically encoded fluorescence resonance energy transfer (FRET)-based sensor for cyclic AMP (cAMP) showed that PDF induced a large increase in cAMP, whereas sNPF caused a small but significant decrease in cAMP. Coexpression of pertussis toxin or RNAi interference to disrupt the G-protein Gαo blocked the electrophysiological responses to sNPF, showing that sNPFR acts via Gαo signaling. Using a fluorescent sensor for intracellular calcium, we observed that sNPF-induced hyperpolarization blocked spontaneous waves of activity propagating along the ventral nerve cord, demonstrating that the electrical effects of sNPF can cause profound changes in natural network activity in the brain. This new model system provides a platform for mechanistic analysis of how neuropeptides can affect target cells at the electrical and molecular level, allowing for predictions of how they regulate brain circuits that control behaviors such as sleep and feeding.
Collapse
Affiliation(s)
- Christopher G Vecsey
- National Center for Behavioral Genomics, Volen National Center for Complex Systems and Department of Biology, Brandeis University, Waltham, Massachusetts
| | | | | |
Collapse
|
46
|
Shang Y, Donelson NC, Vecsey CG, Guo F, Rosbash M, Griffith LC. Short neuropeptide F is a sleep-promoting inhibitory modulator. Neuron 2013; 80:171-83. [PMID: 24094110 DOI: 10.1016/j.neuron.2013.07.029] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2013] [Indexed: 01/10/2023]
Abstract
To advance the understanding of sleep regulation, we screened for sleep-promoting cells and identified neurons expressing neuropeptide Y-like short neuropeptide F (sNPF). Sleep induction by sNPF meets all relevant criteria. Rebound sleep following sleep deprivation is reduced by activation of sNPF neurons, and flies experience negative sleep rebound upon cessation of sNPF neuronal stimulation, indicating that sNPF provides an important signal to the sleep homeostat. Only a subset of sNPF-expressing neurons, which includes the small ventrolateral clock neurons, is sleep promoting. Their release of sNPF increases sleep consolidation in part by suppressing the activity of wake-promoting large ventrolateral clock neurons, and suppression of neuronal firing may be the general response to sNPF receptor activation. sNPF acutely increases sleep without altering feeding behavior, which it affects only on a much longer time scale. The profound effect of sNPF on sleep indicates that it is an important sleep-promoting molecule.
Collapse
Affiliation(s)
- Yuhua Shang
- National Center for Behavioral Genomics and Department of Biology, Brandeis University, Waltham, MA 02454, USA
| | | | | | | | | | | |
Collapse
|
47
|
Wang W, Li Q, Pan Y, Zhu D, Wang L. Influence of hypercapnia on the synthesis of neuropeptides and their receptors in murine brain. Respirology 2013; 18:102-7. [PMID: 22882587 DOI: 10.1111/j.1440-1843.2012.02245.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND AND OBJECTIVE Sleep disorders are a complicated and major public health concern affecting millions of individuals. Obstructive sleep apnoea (OSA) is a common but still under-recognized disease which can cause intermittent nocturnal hypercapnia. Neuropeptides play critical roles in neurotransmission, acting as transmitters or modulators. Results from recent studies have implicated several neuropeptides in sleep and breathing regulation, including orexin, neuropeptides Y and galanin. Therefore, the present study aimed to evaluate the influence of hypercapnia on these neuropeptides and their receptors in order to assess their potential role in the pathogenesis of OSA. METHODS Fifteen C57BL/6J mice were randomly divided into three groups and exposed to moderate hypercapnia (5% CO(2) with balanced room air), or severe hypercapnia (10% CO(2) with balanced room air) or room air for 3 h (9:00-12:00 h), respectively. Immediately following exposure the brainstem and hypothalamus were excised for real-time reverse transcription polymerase chain reaction and western blot analyses. RESULTS In the hypothalamus gene expression including galanin, orexin and neuropeptide Y receptor 1 (NPYR1) was downregulated by hypercapnia. However, protein and mRNA levels of orexin-A receptor were upregulated by severe hypercapnia. In the brainstem only NPYR1 mRNA expression was decreased in moderate hypercapnia compared with that in severe hypercapnia. CONCLUSIONS These findings suggest that hypercapnia can affect these neuropeptides and their receptors, especially the orexin and orexin-A receptor. The potential relationships between these peptides and OSA are worthy of further investigation.
Collapse
Affiliation(s)
- Wei Wang
- Institute of Stomatology, Department of Orthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | | | | | | | | |
Collapse
|
48
|
Griffith LC. Neuromodulatory control of sleep in Drosophila melanogaster: integration of competing and complementary behaviors. Curr Opin Neurobiol 2013; 23:819-23. [PMID: 23743247 DOI: 10.1016/j.conb.2013.05.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 05/07/2013] [Accepted: 05/14/2013] [Indexed: 12/11/2022]
Abstract
The transition between wake and sleep states is characterized by rapid and generalized changes in both sensory and motor processing. Sleep is antagonistic to the expression of important behaviors, like feeding, reproduction and learning whose relative importance to an individual will depend on its circumstances at that moment. An understanding of how the decision to sleep is affected by these other drives and how this process is coordinated across the entire brain remains elusive. Neuromodulation is an important regulatory feature of many behavioral circuits and the reconfiguring of these circuits by modulators can have both long-term and short-term consequences. Drosophila melanogaster has become an important model system for understanding the molecular and genetic bases of behaviors and in recent years neuromodulatory systems have been shown to play a major role in regulation of sleep and other behaviors in this organism. The fly, with its increasingly well-defined behavioral circuitry and powerful genetic tools, is a system poised to provide new insight into the complex issue of how neuromodulation can coordinate situation-specific behavioral needs with the brain's arousal state.
Collapse
Affiliation(s)
- Leslie C Griffith
- Department of Biology, National Center for Behavioral Genomics and Volen Center for Complex Systems, Brandeis University, MS008, 415 South Street, Waltham, MA 02454-9110, USA.
| |
Collapse
|
49
|
Cavas M, Scesa G, Navarro JF. Effects of MPEP, a selective metabotropic glutamate mGlu5 ligand, on sleep and wakefulness in the rat. Prog Neuropsychopharmacol Biol Psychiatry 2013; 40:18-25. [PMID: 23022670 DOI: 10.1016/j.pnpbp.2012.09.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 09/18/2012] [Accepted: 09/18/2012] [Indexed: 10/27/2022]
Abstract
Metabotropic glutamate receptors (mGlu) have been implicated in the regulation of physiological and behavioral processes. Pharmacological evidence involves group I mGlu receptors in the regulation of emotional states and antagonism of these receptors has been proposed as a novel class of anxiolytic drugs having also antidepressant effects. Here, the effects of mGlu5 receptor selective modulation on sleep and wake states are explored. 32 male Wistar rats were implanted with electrodes for recording sleep and wake states. 2-Methyl-6-(phenylethynyl)pyridine hydrochloride (MPEP hydrochloride, 5, 10, and 20 mg/kg, i.p.), a potent, selective and systemically active mGlu5 receptor negative allosteric modulator, or vehicle was administered 1 h after the beginning of the light period. Sleep recordings were conducted for 3 h. MPEP (5, 10, and 20 mg/kg) significantly suppressed rapid eye movement (REM) sleep, decreasing the number of episodes and mean episode duration, and increased its latency. A reduction of light and deep slow wave sleep (SWS) latency was observed in the groups receiving 10 or 20 mg/kg, increasing latency to first wakefulness episode. 10 mg/kg of MPEP also increased non rapid eye movement sleep (NREM). The present results suggest that mGlu5 receptors might be involved in sleep regulation, more specifically in REM sleep, and drugs that block these receptors could potentially benefit the treatment of pathologies were REM sleep is enhanced.
Collapse
Affiliation(s)
- María Cavas
- Department of Psychobiology, Faculty of Psychology, Campus de Teatinos s/n, University of Málaga, 29071 Málaga, Spain.
| | | | | |
Collapse
|
50
|
Masliukov PM, Konovalov VV, Emanuilov AI, Nozdrachev AD. Development of neuropeptide Y-containing neurons in sympathetic ganglia of rats. Neuropeptides 2012; 46:345-52. [PMID: 22964363 DOI: 10.1016/j.npep.2012.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 07/22/2012] [Accepted: 08/02/2012] [Indexed: 12/27/2022]
Abstract
Expression of neuropeptide Y (NPY) in the sympathetic ganglia was investigated by immunohistochemistry and tract tracing. The distribution of NPY immunoreactivity (IR) was studied in the superior cervical ganglion (SCG), stellate ganglion (SG) and celiac ganglion (CG) from rats of different ages (newborn, 10-day-old, 20-day-old, 30-day-old, 2-month-old, 6-month-old, 24-month-old). We observed that the percentage of NPY-IR neuronal profiles increased during early postnatal development. In the SCG and SG, the percentage of NPY-IR profiles enlarged in the first month of life from 43±3.6% (SCG) and 46±3.8% (SG) until 64±4.1% (SCG) and 58±3.5% (SG). The percentage of NPY-IR profiles in the CG increased during the period between 20days (65±3.8%) and 30days (82±5.1%) of animals' life and did not change in further development. In newborn and 10-day-old rats, a large portion of NPY-IR neurons was also calbindin D28K (CB)-IR in all sympathetic ganglia. The proportion of CB-IR substantially decreased during next 10days in the SCG, SG and CG. NPY-IR was approximately present in a half of the postganglionic neurons innervating muscle vessels of the neck and forearm, and the percentage of labeled NPY-IR profiles did not change during the development. Only single Ki67-IR neurons were also NPY-IR in the SCG, SG and CG in newborns and not in older animals. No NPY+/caspase 3+IR neurons were observed. Finally, the process of morphological changes in the size and percentages of NPY-IR profiles is complete in rats by the first month of life.
Collapse
Affiliation(s)
- Petr M Masliukov
- Department of Normal Physiology, Yaroslavl State Medical Academy, ul. Revolucionnaya, 5, Yaroslavl 150000, Russia.
| | | | | | | |
Collapse
|