1
|
Cankar N, Beschorner N, Tsopanidou A, Qvist FL, Colaço AR, Andersen M, Kjaerby C, Delle C, Lambert M, Mundt F, Weikop P, Jucker M, Mann M, Skotte NH, Nedergaard M. Sleep deprivation leads to non-adaptive alterations in sleep microarchitecture and amyloid-β accumulation in a murine Alzheimer model. Cell Rep 2024; 43:114977. [PMID: 39541211 DOI: 10.1016/j.celrep.2024.114977] [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: 03/06/2024] [Revised: 09/09/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Impaired sleep is a common aspect of aging and often precedes the onset of Alzheimer's disease. Here, we compare the effects of sleep deprivation in young wild-type mice and their APP/PS1 littermates, a murine model of Alzheimer's disease. After 7 h of sleep deprivation, both genotypes exhibit an increase in EEG slow-wave activity. However, only the wild-type mice demonstrate an increase in the power of infraslow norepinephrine oscillations, which are characteristic of healthy non-rapid eye movement sleep. Notably, the APP/PS1 mice fail to enhance norepinephrine oscillations 24 h after sleep deprivation, coinciding with an accumulation of cerebral amyloid-β protein. Proteome analysis of cerebrospinal fluid and extracellular fluid further supports these findings by showing altered protein clearance in APP/PS1 mice. We propose that the suppression of infraslow norepinephrine oscillations following sleep deprivation contributes to increased vulnerability to sleep loss and heightens the risk of developing amyloid pathology in early stages of Alzheimer's disease.
Collapse
Affiliation(s)
- Neža Cankar
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Natalie Beschorner
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Anastasia Tsopanidou
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Filippa L Qvist
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Ana R Colaço
- Proteomics Research Infrastructure, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Mie Andersen
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Celia Kjaerby
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Christine Delle
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Marius Lambert
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Filip Mundt
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pia Weikop
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Mathias Jucker
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Matthias Mann
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Department for Proteomics and Signal Transduction, Max-Planck Institute for Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Niels Henning Skotte
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Medical and Health Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark; Center for Translational Neuromedicine, University of Rochester Medical School, Elmwood Avenue 601, Rochester, NY 14642, USA.
| |
Collapse
|
2
|
Zhong X, Xu L, Wang L, Chen J, Gong X, Lian J, Gong J, Shao Y. Caffeine and modafinil modulate the effects of sleep deprivation on thalamic resting-state functional connectivity: A double-blind pilot study. Sleep Med 2024; 122:71-83. [PMID: 39137663 DOI: 10.1016/j.sleep.2024.08.007] [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: 02/04/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND Studies have found that the use of clinically approved caffeine and modafinil can alleviate cognitive impairment due to sleep deprivation (SD) to some extent. However, the neural mechanisms by which these two cognitive enhancers work to counteract the effects of SD on cognitive impairment remain unclear. METHODS A double-blind within-subjects experiment using resting-state functional magnetic resonance imaging (rs-fMRI) was designed. Participants underwent three 36-h SD trials, each of which involved taking 200 mg of caffeine, modafinil, or placebo at the 28th and 32 nd h of SD. Sixteen subregions of the thalamus were selected as the regions of interest and changes in functional connectivity (FC) between the thalamus and the other brain regions were explored after the participants took caffeine or modafinil. RESULTS The subjective sleepiness of the participants increased with the duration of SD. compared with placebo, modafinil and caffeine had insignificant effects on wakefulness or sleepiness. However, in terms of neural FC, we found varying degrees of attenuation or enhancement of the FC between the thalamus and other regions. Taking caffeine during SD weakened the FC between the right rostral temporal thalamus (rTtha) subregion and the left lingual gyrus compared with placebo. Caffeine enhanced the FC between three subregions of the thalamus, namely the left sensory thalamus, the left rTtha, and the right lateral pre-frontal thalamus, and the right inferior temporal, left orbitofrontal, and right superior occipital gyris. Modafinil weakened the FC between the right posterior parietal thalamus and left middle temporal gyrus, and enhanced the FC between the left medial pre-frontal thalamus, left rTtha, and right occipital thalamus and left middle frontal gyrus. CONCLUSIONS After 36 h of total SD, modafinil and caffeine administration enhanced or attenuated the time-domain correlations between various subregions of the thalamus and brain regions of the frontal and temporal lobes in healthy adults, compared with placebo. These results provide valuable evidence for further unraveling the neuropharmacological mechanisms of caffeine and modafinil, as well as important insights for exploring effective pharmacological intervention strategies against SD.
Collapse
Affiliation(s)
- Xiao Zhong
- School of Psychology, Beijing Sport University, Beijing, China
| | - Lin Xu
- School of Psychology, Beijing Sport University, Beijing, China
| | - Letong Wang
- School of Psychology, Beijing Sport University, Beijing, China
| | - Jie Chen
- School of Psychology, Beijing Sport University, Beijing, China
| | - Xinxin Gong
- School of Psychology, Beijing Sport University, Beijing, China
| | - Jie Lian
- School of Psychology, Beijing Sport University, Beijing, China
| | - Jingjing Gong
- School of Psychology, Beijing Sport University, Beijing, China; Department of Medical Psychology, Second Medical Center, General Hospital of the People's Liberation Army, China.
| | - Yongcong Shao
- School of Psychology, Beijing Sport University, Beijing, China.
| |
Collapse
|
3
|
Tian H, Ge K, Wang L, Gao P, Chen A, Wang F, Guo F, Wang F, Zhang Q. Advances in PGD2/PTGDR2 signaling pathway in tumors: A review. BIOMOLECULES & BIOMEDICINE 2024; 24:1055-1067. [PMID: 38704736 PMCID: PMC11378995 DOI: 10.17305/bb.2024.10485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/14/2024] [Accepted: 04/14/2024] [Indexed: 05/07/2024]
Abstract
Studies have shown that the prostaglandin (PG) family acts as an allergic inflammatory mediator in malignant diseases. Furthermore, prostaglandin E2 (PGE2) and its related receptors, as well as the prostaglandin D2 (PGD2)/PGD2 receptor (PTGDR2), play irreplaceable roles in tumorigenesis and anti-tumor therapy. Several experiments have demonstrated that PGD2 signaling through PTGDR2 not only directly inhibits cancer cell survival, proliferation, and migration but also reduces resistance toward conventional chemotherapeutic agents. Recent studies from our and other laboratories have shown that PGD2, its ligands, and related metabolites can significantly alter the tumor microenvironment (TME) by promoting the secretion of chemokines and cytokines, thereby inhibiting tumor progression. Additionally, reduced PGD2 expression has been associated with poor prognosis in patients with gastric, breast, lung, and pancreatic cancers, validating the preclinical findings and their clinical relevance. This review focuses on the current understanding of PGD2/PTGDR2 expression patterns and biological activity in cancer, proposing questions to guide the assessment of PGD2 and its receptors as potential targets for effective cancer therapies.
Collapse
Affiliation(s)
- Hengjin Tian
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu, China
| | - Kunpeng Ge
- Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu, China
| | - Lulu Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Peiyao Gao
- Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical University, Bengbu, China
| | - Amin Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Feifan Wang
- Department of Blood Transfusion, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Fangzheng Guo
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Bengbu, China
| | - FengChao Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Qiang Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| |
Collapse
|
4
|
Fu H, Wang S, Xu P, Feng Z, Pan S, Ge X. Early predictive value of lipocalin-type prostaglandin D synthase for 28-day mortality in cardiac arrest patients: study protocol for a prospective study. BMJ Open 2024; 14:e083136. [PMID: 38839386 PMCID: PMC11163600 DOI: 10.1136/bmjopen-2023-083136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 05/10/2024] [Indexed: 06/07/2024] Open
Abstract
INTRODUCTION Public training in cardiopulmonary resuscitation and treatment in emergency and intensive care unit have made tremendous progress. However, cardiac arrest remains a major health burden worldwide, with brain damage being a significant contributor to disability and mortality. Lipocalin-type prostaglandin D synthase (L-PGDS), which is mainly localised in the central nervous system, has been previously shown to inhibit postischemia neuronal apoptosis. Therefore, we aim to observe whether serum L-PGDS can serve as a potential biomarker and explore its role in determining the severity and prognosis of patients who have achieved restoration of spontaneous circulation (ROSC). METHODS AND ANALYSIS This is a prospective observational study. The participants (n = 60) who achieve ROSC will be distributed into two groups (non-survivor and survivor) based on 28-day survival. Healthy volunteers (n = 30) will be enrolled as controls. Each individual's relevant information will be extracted from Electronic Medical Record System in Xinhua Hospital, including demographic characteristics, clinical data, laboratory findings and so on. On days 1, 3 and 7 after ROSC, blood samples will be drawn and batch tested on the level of serum neuron-specific enolase, soluble protein 100β, L-PGDS, procalcitonin, tumour necrosis factor-alpha and interleukin-6. The cerebral performance category score was assessed on the 28th day after ROSC. ETHICS AND DISSEMINATION This study was performed with the approval of the Clinical Ethical Committee of Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (Approval No. XHEC-C-2023-130-1). The results will be published in a peer-reviewed journal. TRIAL REGISTRATION NUMBER Chinese Clinical Trial Registry (ChiCTR2300078564).
Collapse
Affiliation(s)
- Huimin Fu
- Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shangyuan Wang
- Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Peixian Xu
- Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhihui Feng
- Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shuming Pan
- Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaoli Ge
- Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Hao KX, Shen CY, Jiang JG. Sedative and hypnotic effects of Polygala tenuifolia willd. saponins on insomnia mice and their targets. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117618. [PMID: 38141791 DOI: 10.1016/j.jep.2023.117618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/14/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Polygala tenuifolia Willd. has been widely used in the treatment of cancer, forgetfulness, depression and other diseases. AIM OF REVIEW The purpose of this study was to investigate the sleep-enhancing effect and mechanism of P. tenuifolia saponins (PTS). MATERIALS AND METHODS The total saponin (YZ-I) and purified saponin (YZ-II) fractions were extracted and ICR mice model of insomnia was established by p-chlorophenylalanine (PCPA) induction to observe anxiety and depression behaviors. Effects of YZ-I and YZ-II on the levels of neurotransmitters, hormones, and inflammation cytokines were detected by ELISA, RT-qPCR and western blotting. RESULTS The results showed that YZ-I and YZ-II reduced the immobility time of mice and prolonged the sleep time of mice and significantly increased the concentrations of 5-HT, NE, PGD2, IL-1β and TNF-α. YZ-I and YZ-II regulated GABAARα2, GABAARα3, GAD65/67, 5-HT1A and 5-HT2A, while regulated the levels of inflammatory cytokines such as DPR, PGD2, iNOS and TNF-α to exert sedative and hypnotic effects. CONCLUSION PTS are mainly achieved sedative and hypnotic effects by altering serotonergic, GABAergic and immune systems, but the effects and mechanisms of action of YZ-I were different from YZ-II.
Collapse
Affiliation(s)
- Ke-Xin Hao
- College of Food and Bioengineering, South China University of Technology, Guangzhou, 510640, China
| | - Chun-Yan Shen
- College of Food and Bioengineering, South China University of Technology, Guangzhou, 510640, China; Southern Medical University, School of Traditional Chinese Medicine, Guangzhou 510515, China
| | - Jian-Guo Jiang
- College of Food and Bioengineering, South China University of Technology, Guangzhou, 510640, China.
| |
Collapse
|
6
|
Sang D, Lin K, Yang Y, Ran G, Li B, Chen C, Li Q, Ma Y, Lu L, Cui XY, Liu Z, Lv SQ, Luo M, Liu Q, Li Y, Zhang EE. Prolonged sleep deprivation induces a cytokine-storm-like syndrome in mammals. Cell 2023; 186:5500-5516.e21. [PMID: 38016470 DOI: 10.1016/j.cell.2023.10.025] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 08/17/2023] [Accepted: 10/25/2023] [Indexed: 11/30/2023]
Abstract
Most animals require sleep, and sleep loss induces serious pathophysiological consequences, including death. Previous experimental approaches for investigating sleep impacts in mice have been unable to persistently deprive animals of both rapid eye movement sleep (REMS) and non-rapid eye movement sleep (NREMS). Here, we report a "curling prevention by water" paradigm wherein mice remain awake 96% of the time. After 4 days of exposure, mice exhibit severe inflammation, and approximately 80% die. Sleep deprivation increases levels of prostaglandin D2 (PGD2) in the brain, and we found that elevated PGD2 efflux across the blood-brain-barrier-mediated by ATP-binding cassette subfamily C4 transporter-induces both accumulation of circulating neutrophils and a cytokine-storm-like syndrome. Experimental disruption of the PGD2/DP1 axis dramatically reduced sleep-deprivation-induced inflammation. Thus, our study reveals that sleep-related changes in PGD2 in the central nervous system drive profound pathological consequences in the peripheral immune system.
Collapse
Affiliation(s)
- Di Sang
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China; National Institute of Biological Sciences, Beijing, China
| | - Keteng Lin
- National Institute of Biological Sciences, Beijing, China; College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yini Yang
- Peking University School of Life Sciences, Beijing, China
| | - Guangdi Ran
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Bohan Li
- Peking-Tsinghua Center for Life Sciences, Beijing, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Chen Chen
- National Institute of Biological Sciences, Beijing, China
| | - Qi Li
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Yan Ma
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Lihui Lu
- National Institute of Biological Sciences, Beijing, China
| | - Xi-Yang Cui
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Zhibo Liu
- Peking-Tsinghua Center for Life Sciences, Beijing, China; Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Sheng-Qing Lv
- Department of Neurosurgery, Xinqiao Hospital, Chongqing, China
| | - Minmin Luo
- National Institute of Biological Sciences, Beijing, China; School of Life Sciences, Tsinghua University, Beijing, China; Chinese Institute for Brain Research, Beijing, China
| | - Qinghua Liu
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Yulong Li
- Peking University School of Life Sciences, Beijing, China; Peking-Tsinghua Center for Life Sciences, Beijing, China; State Key Laboratory of Membrane Biology, Beijing, China; PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Eric Erquan Zhang
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China.
| |
Collapse
|
7
|
Korkutata M, Lazarus M. Adenosine A 2A receptors and sleep. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 170:155-178. [PMID: 37741690 DOI: 10.1016/bs.irn.2023.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2023]
Abstract
Adenosine, a known endogenous somnogen, induces sleep via A1 and A2A receptors. In this chapter, we review the current knowledge regarding the role of the adenosine A2A receptor and its agonists, antagonists, and allosteric modulators in sleep-wake regulation. Although many adenosine A2A receptor agonists, antagonists, and allosteric modulators have been identified, only a few have been tested to see if they can promote sleep or wakefulness. In addition, the growing popularity of natural sleep aids has led to an investigation of natural compounds that may improve sleep by activating the adenosine A2A receptor. Finally, we discuss the potential therapeutic advantage of allosteric modulators of adenosine A2A receptors over classic agonists and antagonists for treating sleep and neurologic disorders.
Collapse
Affiliation(s)
- Mustafa Korkutata
- Department of Neurology, Division of Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| | - Michael Lazarus
- International Institute for Integrative Sleep Medicine (WPI-IIIS) and Institute of Medicine, University of Tsukuba, Tsukuba, Japan.
| |
Collapse
|
8
|
Varma M, Kaur A, Bhandari R, Kumar A, Kuhad A. Major depressive disorder (mdd): emerging immune targets at preclinical level. Expert Opin Ther Targets 2023; 27:479-501. [PMID: 37334668 DOI: 10.1080/14728222.2023.2225216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 06/11/2023] [Indexed: 06/20/2023]
Abstract
BACKGROUND Major depressive disorder is a mental health disorder that is characterized by a persistently low mood and loss of interest. MDD is affecting over 3.8% of the global population as a major health problem. Its etiology is complex, and involves the interaction between a number of factors, including genetic predisposition and the presence of environmental stresses. AREAS COVERED The role of the immune and inflammatory systems in depression has been gaining interest, with evidence suggesting the potential involvement of pro-inflammatory molecules like TNF, interleukins, prostaglandins, and other cytokines, among others, has been put forth. Along with this, the potential of agents, from NSAIDs to antibiotics, are being evaluated in therapy for depression. The current review will discuss emerging immune targets at the preclinical level. EXPERT OPINION With increasing evidence to show that immune and inflammatory mediators are implicated in MDD, increasing research toward their potential as drug targets is encouraged. At the same time, agents acting on these mediators and possessing anti-inflammatory potential are also being evaluated as future therapeutic options for MDD, and increasing focus toward non-conventional drugs which can act through these mechanisms is important as regards the future prospects of the use of anti-inflammatory agents in depression.
Collapse
Affiliation(s)
- Manasi Varma
- UGC-Centre of Advanced Study, University Institute of Pharmaceutical Sciences, Chandigarh, India
| | - Arshpreet Kaur
- UGC-Centre of Advanced Study, University Institute of Pharmaceutical Sciences, Chandigarh, India
| | - Ranjana Bhandari
- UGC-Centre of Advanced Study, University Institute of Pharmaceutical Sciences, Chandigarh, India
| | - Ashwani Kumar
- UGC-Centre of Advanced Study, University Institute of Pharmaceutical Sciences, Chandigarh, India
| | - Anurag Kuhad
- UGC-Centre of Advanced Study, University Institute of Pharmaceutical Sciences, Chandigarh, India
| |
Collapse
|
9
|
Almeida ZL, Brito RMM. Amyloid Disassembly: What Can We Learn from Chaperones? Biomedicines 2022; 10:3276. [PMID: 36552032 PMCID: PMC9776232 DOI: 10.3390/biomedicines10123276] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/14/2022] [Accepted: 09/26/2022] [Indexed: 12/23/2022] Open
Abstract
Protein aggregation and subsequent accumulation of insoluble amyloid fibrils with cross-β structure is an intrinsic characteristic of amyloid diseases, i.e., amyloidoses. Amyloid formation involves a series of on-pathway and off-pathway protein aggregation events, leading to mature insoluble fibrils that eventually accumulate in multiple tissues. In this cascade of events, soluble oligomeric species are formed, which are among the most cytotoxic molecular entities along the amyloid cascade. The direct or indirect action of these amyloid soluble oligomers and amyloid protofibrils and fibrils in several tissues and organs lead to cell death in some cases and organ disfunction in general. There are dozens of different proteins and peptides causing multiple amyloid pathologies, chief among them Alzheimer's, Parkinson's, Huntington's, and several other neurodegenerative diseases. Amyloid fibril disassembly is among the disease-modifying therapeutic strategies being pursued to overcome amyloid pathologies. The clearance of preformed amyloids and consequently the arresting of the progression of organ deterioration may increase patient survival and quality of life. In this review, we compiled from the literature many examples of chemical and biochemical agents able to disaggregate preformed amyloids, which have been classified as molecular chaperones, chemical chaperones, and pharmacological chaperones. We focused on their mode of action, chemical structure, interactions with the fibrillar structures, morphology and toxicity of the disaggregation products, and the potential use of disaggregation agents as a treatment option in amyloidosis.
Collapse
Affiliation(s)
| | - Rui M. M. Brito
- Chemistry Department and Coimbra Chemistry Centre—Institute of Molecular Sciences (CQC-IMS), University of Coimbra, 3004-535 Coimbra, Portugal
| |
Collapse
|
10
|
Horikami D, Fujii W, Aritake K, Murata T. L-PGDS Attenuates Acute Lung Injury by Prostaglandin D 2 in Both Dependent and Independent Ways. THE JOURNAL OF IMMUNOLOGY 2021; 207:2545-2550. [PMID: 34615734 DOI: 10.4049/jimmunol.2100394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 09/06/2021] [Indexed: 11/19/2022]
Abstract
Lipocalin-type PG D synthase (L-PGDS) has two roles: it can be a PGD synthase, or it can be a carrier protein of hydrophobic small molecules. In this study, we investigated the dual roles of L-PGDS in acute lung injury by using L-PGDS-deficient and point-mutated mice, which lack PGD2 producibility but maintain lipocalin ability. Hydrochloride (HCl) administration (0.1 M intratracheally for 6 h) caused hemorrhage and dysfunction in the wild-type (WT) mouse lung. These symptoms were accompanied by an increase in PGD2 production. Both deficiency and point mutation of L-PGDS aggravated the HCl-induced hemorrhage and dysfunction. Although both the gene modifications decreased PGD2 production, only L-PGDS-deficient mice, but not point mutation mice, lacked protein expressions of L-PGDS in the lungs. In the WT mice, HCl administration caused pulmonary edema, indexed as an increase in lung water content and protein leakage in bronchoalveolar lavage fluid. L-PGDS deficiency and point mutation similarly aggravated edema formation. HCl administration also stimulated mucin production and bronchoalveolar lavage fluid leukocyte infiltration in the WT mouse lungs. Of interest, L-PGDS deficiency, but not point mutation, exacerbated these manifestations. Consistently, only L-PGDS deficiency increased the mRNA expression of IL-33, which stimulates mucin production in the inflamed lung. These results show that L-PGDS attenuated HCl-induced acute lung injury progresses in two different ways: L-PGDS produced PGD2, which inhibited pulmonary edema formation, whereas its lipocalin ability decreased mucin formation and inflammatory cell infiltration in the inflamed lung.
Collapse
Affiliation(s)
- Daiki Horikami
- Department of Animal Radiology, Graduate School of Agricultural and Life Sciences, the University of Tokyo, Tokyo, Japan
| | - Wataru Fujii
- Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, the University of Tokyo, Tokyo, Japan; and
| | - Kosuke Aritake
- Laboratory of Chemical Pharmacology, Daiichi University of Pharmacy, Fukuoka, Japan
| | - Takahisa Murata
- Department of Animal Radiology, Graduate School of Agricultural and Life Sciences, the University of Tokyo, Tokyo, Japan;
| |
Collapse
|
11
|
Urade Y. Biochemical and Structural Characteristics, Gene Regulation, Physiological, Pathological and Clinical Features of Lipocalin-Type Prostaglandin D 2 Synthase as a Multifunctional Lipocalin. Front Physiol 2021; 12:718002. [PMID: 34744762 PMCID: PMC8569824 DOI: 10.3389/fphys.2021.718002] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/01/2021] [Indexed: 11/13/2022] Open
Abstract
Lipocalin-type prostaglandin (PG) D2 synthase (L-PGDS) catalyzes the isomerization of PGH2, a common precursor of the two series of PGs, to produce PGD2. PGD2 stimulates three distinct types of G protein-coupled receptors: (1) D type of prostanoid (DP) receptors involved in the regulation of sleep, pain, food intake, and others; (2) chemoattractant receptor-homologous molecule expressed on T helper type 2 cells (CRTH2) receptors, in myelination of peripheral nervous system, adipocyte differentiation, inhibition of hair follicle neogenesis, and others; and (3) F type of prostanoid (FP) receptors, in dexamethasone-induced cardioprotection. L-PGDS is the same protein as β-trace, a major protein in human cerebrospinal fluid (CSF). L-PGDS exists in the central nervous system and male genital organs of various mammals, and human heart; and is secreted into the CSF, seminal plasma, and plasma, respectively. L-PGDS binds retinoic acids and retinal with high affinities (Kd < 100 nM) and diverse small lipophilic substances, such as thyroids, gangliosides, bilirubin and biliverdin, heme, NAD(P)H, and PGD2, acting as an extracellular carrier of these substances. L-PGDS also binds amyloid β peptides, prevents their fibril formation, and disaggregates amyloid β fibrils, acting as a major amyloid β chaperone in human CSF. Here, I summarize the recent progress of the research on PGD2 and L-PGDS, in terms of its “molecular properties,” “cell culture studies,” “animal experiments,” and “clinical studies,” all of which should help to understand the pathophysiological role of L-PGDS and inspire the future research of this multifunctional lipocalin.
Collapse
Affiliation(s)
- Yoshihiro Urade
- Center for Supporting Pharmaceutical Education, Daiichi University of Pharmacy, Fukuoka, Japan.,Isotope Science Center, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
12
|
Fréchette L, Degrandmaison J, Binda C, Boisvert M, Côté L, Michaud T, Lalumière MP, Gendron L, Parent JL. Identification of the interactome of the DP1 receptor for Prostaglandin D 2: Regulation of DP1 receptor signaling and trafficking by IQGAP1. Biochim Biophys Acta Gen Subj 2021; 1865:129969. [PMID: 34352343 DOI: 10.1016/j.bbagen.2021.129969] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 07/19/2021] [Accepted: 07/25/2021] [Indexed: 01/16/2023]
Abstract
BACKGROUND Mechanisms governing localization, trafficking and signaling of G protein-coupled receptors (GPCRs) are critical in cell function. Protein-protein interactions are determinant in these processes. However, there are very little interacting proteins known to date for the DP1 receptor for prostaglandin D2. METHODS We performed LC-MS/MS analyses of the DP1 receptor interactome in HEK293 cells. To functionally validate our LC-MS/MS data, we studied the implications of the interaction with the IQGAP1 scaffold protein in the trafficking and signaling of DP1. RESULTS In addition to expected interacting proteins such as heterotrimeric G protein subunits, we identified proteins involved in signaling, trafficking, and folding localized in various cell compartments. Endogenous DP1-IQGAP1 co-immunoprecipitation was observed in colon cancer HT-29 cells. The interaction was augmented by DP1 agonist activation in HEK293 cells and GST-pulldown assays showed that IQGAP1 binds to intracellular loops 2 and 3 of DP1. Co-localization of the two proteins was observed by confocal microscopy at the cell periphery and in intracellular vesicles in the basal state. PGD2 treatment resulted in the redistribution of the DP1-IQGAP1 co-localization in the perinuclear vicinity. DP1 receptor internalization was promoted by overexpression of IQGAP1, while it was diminished by IQGAP1 knockdown with DsiRNAs. DP1-mediated ERK1/2 activation was augmented and sustained overtime by overexpression of IQGAP1 when compared to DP1 expressed alone. IQGAP1 knockdown decreased ERK1/2 activation by DP1 stimulation. Interestingly, ERK1/2 signaling by DP1 was increased when IQGAP2 was silenced, while it was impaired by IQGAP3 knockdown. CONCLUSIONS Our findings define the putative DP1 interactome, a patho-physiologically important receptor, and validated the interaction with IQGAP1 in DP1 function. Our data also reveal that IQGAP proteins may differentially regulate GPCR signaling. GENERAL SIGNIFICANCE The identified putative DP1-interacting proteins open multiple lines of research in DP1 and GPCR biology in various cell compartments.
Collapse
Affiliation(s)
- Louis Fréchette
- Département de Médecine, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Institut de Pharmacologie de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Centre de recherche du Centre Hospitalier de l'Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Jade Degrandmaison
- Département de Médecine, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Institut de Pharmacologie de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Centre de recherche du Centre Hospitalier de l'Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Chantal Binda
- Département de Médecine, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Institut de Pharmacologie de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Centre de recherche du Centre Hospitalier de l'Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Marilou Boisvert
- Département de Médecine, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Institut de Pharmacologie de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Centre de recherche du Centre Hospitalier de l'Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Laurie Côté
- Département de Médecine, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Institut de Pharmacologie de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Centre de recherche du Centre Hospitalier de l'Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Thomas Michaud
- Département de Médecine, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Institut de Pharmacologie de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Centre de recherche du Centre Hospitalier de l'Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Marie-Pier Lalumière
- Département de Médecine, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Institut de Pharmacologie de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Centre de recherche du Centre Hospitalier de l'Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Louis Gendron
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Département d'Anesthésiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Institut de Pharmacologie de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Centre de recherche du Centre Hospitalier de l'Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Jean-Luc Parent
- Département de Médecine, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Institut de Pharmacologie de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Centre de recherche du Centre Hospitalier de l'Université de Sherbrooke, Sherbrooke, Québec, Canada.
| |
Collapse
|
13
|
Sultan ZW, Jaeckel ER, Krause BM, Grady SM, Murphy CA, Sanders RD, Banks MI. Electrophysiological signatures of acute systemic lipopolysaccharide-induced inflammation: potential implications for delirium science. Br J Anaesth 2021; 126:996-1008. [PMID: 33648701 PMCID: PMC8132883 DOI: 10.1016/j.bja.2020.12.040] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Novel preventive therapies are needed for postoperative delirium, which especially affects older patients. A mouse model is presented that captures inflammation-associated cortical slow wave activity (SWA) observed in patients, allowing exploration of the mechanistic role of prostaglandin-adenosine signalling. METHODS EEG and cortical cytokine measurements (interleukin 6, monocyte chemoattractant protein-1) were obtained from adult and aged mice. Behaviour, SWA, and functional connectivity were assayed before and after systemic administration of lipopolysaccharide (LPS)+piroxicam (cyclooxygenase inhibitor) or LPS+caffeine (adenosine receptor antagonist). To avoid the confounder of inflammation-driven changes in movement which alter SWA and connectivity, electrophysiological recordings were classified as occurring during quiescence or movement, and propensity score matching was used to match distributions of movement magnitude between baseline and post-LPS administration. RESULTS LPS produces increases in cortical cytokines and behavioural quiescence. In movement-matched data, LPS produces increases in SWA (likelihood-ratio test: χ2(4)=21.51, P<0.001), but not connectivity (χ2(4)=6.39, P=0.17). Increases in SWA associate with interleukin 6 (P<0.001) and monocyte chemoattractant protein-1 (P=0.001) and are suppressed by piroxicam (P<0.001) and caffeine (P=0.046). Aged animals compared with adult animals show similar LPS-induced SWA during movement, but exaggerated cytokine response and increased SWA during quiescence. CONCLUSIONS Cytokine-SWA correlations during wakefulness are consistent with observations in patients with delirium. Absence of connectivity effects after accounting for movement changes suggests decreased connectivity in patients is a biomarker of hypoactivity. Exaggerated effects in quiescent aged animals are consistent with increased hypoactive delirium in older patients. Prostaglandin-adenosine signalling may link inflammation to neural changes and hence delirium.
Collapse
Affiliation(s)
- Ziyad W Sultan
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Elizabeth R Jaeckel
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Bryan M Krause
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Sean M Grady
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Caitlin A Murphy
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Robert D Sanders
- Specialty of Anaesthetics, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Department of Anaesthetics, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Matthew I Banks
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
14
|
Voss M, Kotrba J, Gaffal E, Katsoulis-Dimitriou K, Dudeck A. Mast Cells in the Skin: Defenders of Integrity or Offenders in Inflammation? Int J Mol Sci 2021; 22:ijms22094589. [PMID: 33925601 PMCID: PMC8123885 DOI: 10.3390/ijms22094589] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/23/2021] [Accepted: 04/25/2021] [Indexed: 12/13/2022] Open
Abstract
Mast cells (MCs) are best-known as key effector cells of immediate-type allergic reactions that may even culminate in life-threatening anaphylactic shock syndromes. However, strategically positioned at the host–environment interfaces and equipped with a plethora of receptors, MCs also play an important role in the first-line defense against pathogens. Their main characteristic, the huge amount of preformed proinflammatory mediators embedded in secretory granules, allows for a rapid response and initiation of further immune effector cell recruitment. The same mechanism, however, may account for detrimental overshooting responses. MCs are not only detrimental in MC-driven diseases but also responsible for disease exacerbation in other inflammatory disorders. Focusing on the skin as the largest immune organ, we herein review both beneficial and detrimental functions of skin MCs, from skin barrier integrity via host defense mechanisms to MC-driven inflammatory skin disorders. Moreover, we emphasize the importance of IgE-independent pathways of MC activation and their role in sustained chronic skin inflammation and disease exacerbation.
Collapse
Affiliation(s)
- Martin Voss
- Medical Faculty, Institute for Molecular and Clinical Immunology, Otto-Von-Guericke-University Magdeburg, 39120 Magdeburg, Germany; (M.V.); (J.K.); (K.K.-D.)
| | - Johanna Kotrba
- Medical Faculty, Institute for Molecular and Clinical Immunology, Otto-Von-Guericke-University Magdeburg, 39120 Magdeburg, Germany; (M.V.); (J.K.); (K.K.-D.)
| | - Evelyn Gaffal
- Laboratory for Experimental Dermatology, Department of Dermatology, University Hospital Magdeburg, 39120 Magdeburg, Germany;
| | - Konstantinos Katsoulis-Dimitriou
- Medical Faculty, Institute for Molecular and Clinical Immunology, Otto-Von-Guericke-University Magdeburg, 39120 Magdeburg, Germany; (M.V.); (J.K.); (K.K.-D.)
| | - Anne Dudeck
- Medical Faculty, Institute for Molecular and Clinical Immunology, Otto-Von-Guericke-University Magdeburg, 39120 Magdeburg, Germany; (M.V.); (J.K.); (K.K.-D.)
- Health Campus Immunology, Infectiology and Inflammation, Otto-Von-Guericke-University Magdeburg, 39120 Magdeburg, Germany
- Correspondence:
| |
Collapse
|
15
|
Wang P, Li Q, Dong X, An H, Li J, Zhao L, Yan H, Aritake K, Huang Z, Strohl KP, Urade Y, Zhang J, Han F. Lipocalin-type prostaglandin D synthase levels increase in patients with narcolepsy and idiopathic hypersomnia. Sleep 2021; 44:zsaa234. [PMID: 33175978 DOI: 10.1093/sleep/zsaa234] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 08/31/2020] [Indexed: 12/11/2022] Open
Abstract
STUDY OBJECTIVES Excessive daytime sleepiness (EDS) is a frequent cause for consultation and a defining symptom of narcolepsy and idiopathic hypersomnia (IH). The associated mechanisms remain unclear. Lipocalin-type prostaglandin D synthase (LPGDS) is a plausible sleep-inducing candidate. This study is to compare cerebral spinal fluid (CSF) and serum LPGDS levels in patients group with hypersomnia of central origin, including those with narcolepsy type 1 (NT1) and type 2 (NT2) and IH, to those in healthy controls (Con). METHODS Serum LPGDS, CSF LPGDS, and CSF hypocretin-1(Hcrt-1) levels were measured by ELISA in 122 narcolepsy patients (106 NT1 and 16 NT2), 27 IH, and 51Con. RESULTS LPGDS levels in CSF (p = 0.02) and serum (p < 0.001) were 22%-25% lower in control subjects than in patients with EDS complaints, including NT1, NT2, and IH. In contrast to significant differences in CSF Hcrt-1 levels, CSF L-PGDS levels and serum L-PGDS were comparable among NT1, NT2, and IH (p > 0.05), except for slightly lower serum LPGDS in IH than in NT1 (p = 0.01). Serum L-PGDS correlated modestly and negatively to sleep latency on MSLT (r = -0.227, p = 0.007) in hypersomnia subjects. CONCLUSIONS As a somnogen-producing enzyme, CSF/serum LPGDS may serve as a new biomarker for EDS of central origin and imply a common pathogenetic association, but would complement rather than replaces orexin markers.
Collapse
Affiliation(s)
- Peipei Wang
- Department of Pulmonary Medicine, Peking University People's Hospital, Beijing, China
- Sleep and Psychosomatic Medicine Center, The Third People's Hospital of Hainan Province, Sanya, Hainan, China
| | - Qinghua Li
- Department of Pulmonary Medicine, Peking University People's Hospital, Beijing, China
| | - Xiaosong Dong
- Department of Pulmonary Medicine, Peking University People's Hospital, Beijing, China
| | - Haiyan An
- Department of Anesthesia, Peking University People's Hospital, Beijing, China
| | - Jing Li
- Department of Pulmonary Medicine, Peking University People's Hospital, Beijing, China
| | - Long Zhao
- Department of Pulmonary Medicine, Peking University People's Hospital, Beijing, China
| | - Han Yan
- Department of Pulmonary Medicine, Peking University People's Hospital, Beijing, China
| | - Kosuke Aritake
- Laboratory of Chemical Pharmacology, Daiichi University of Pharmacy, Minami-ku, Fukuoka, Japan
| | - Zhili Huang
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Kingman P Strohl
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Case Western Reserve University, and Cleveland Louis Stokes VA Medical Center, Cleveland, OH
| | - Yoshihiro Urade
- Isotope Science Center, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Jun Zhang
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Fang Han
- Department of Pulmonary Medicine, Peking University People's Hospital, Beijing, China
| |
Collapse
|
16
|
Dorsey A, de Lecea L, Jennings KJ. Neurobiological and Hormonal Mechanisms Regulating Women's Sleep. Front Neurosci 2021; 14:625397. [PMID: 33519372 PMCID: PMC7840832 DOI: 10.3389/fnins.2020.625397] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/22/2020] [Indexed: 01/22/2023] Open
Abstract
Sleep is crucial for optimal well-being, and sex differences in sleep quality have significant implications for women's health. We review the current literature on sex differences in sleep, such as differences in objective and subjective sleep measures and their relationship with aging. We then discuss the convincing evidence for the role of ovarian hormones in regulating female sleep, and survey how these hormones act on a multitude of brain regions and neurochemicals to impact sleep. Lastly, we identify several important areas in need of future research to narrow the knowledge gap and improve the health of women and other understudied populations.
Collapse
Affiliation(s)
| | | | - Kimberly J. Jennings
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|
17
|
Chen L, Li S, Zhou Y, Liu T, Cai A, Zhang Z, Xu F, Manyande A, Wang J, Peng M. Neuronal mechanisms of adenosine A 2A receptors in the loss of consciousness induced by propofol general anesthesia with functional magnetic resonance imaging. J Neurochem 2020; 156:1020-1032. [PMID: 32785947 DOI: 10.1111/jnc.15146] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/20/2020] [Accepted: 07/30/2020] [Indexed: 01/04/2023]
Abstract
Propofol is the most common intravenous anesthetic agent for induction and maintenance of anesthesia, and has been used clinically for more than 30 years. However, the mechanism by which propofol induces loss of consciousness (LOC) remains largely unknown. The adenosine A2A receptor (A2A R) has been extensively proven to have an effect on physiological sleep. It is, therefore, important to investigate the role of A2A R in the induction of LOC using propofol. In the present study, the administration of the highly selective A2A R agonist (CGS21680) and antagonist (SCH58261) was utilized to investigate the function of A2A R under general anesthesia induced by propofol by means of animal behavior studies, resting-state magnetic resonance imaging and c-Fos immunofluorescence staining approaches. Our results show that CGS21680 significantly prolonged the duration of LOC induced by propofol, increased the c-Fos expression in nucleus accumbens (NAc) and suppressed the functional connectivity of NAc-dorsal raphe nucleus (DR) and NAc-cingulate cortex (CG). However, SCH58261 significantly shortened the duration of LOC induced by propofol, decreased the c-Fos expression in NAc, increased the c-Fos expression in DR, and elevated the functional connectivity of NAc-DR and NAc-CG. Collectively, our findings demonstrate the important roles played by A2A R in the LOC induced by propofol and suggest that the neural circuit between NAc-DR maybe controlled by A2A R in the mechanism of anesthesia induced by propofol.
Collapse
Affiliation(s)
- Lei Chen
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China.,Center of Brain Science, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Shuang Li
- Center of Brain Science, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Ying Zhou
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China
| | - Taotao Liu
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Aoling Cai
- Center of Brain Science, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, PR China
| | - Zongze Zhang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China
| | - Fuqiang Xu
- Center of Brain Science, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, PR China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, P. R. China.,Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Anne Manyande
- School of Human and Social Sciences, University of West London, London, UK
| | - Jie Wang
- Center of Brain Science, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, PR China
| | - Mian Peng
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, P.R. China
| |
Collapse
|
18
|
Xu W, Ding W, Sheng H, Lu D, Xu X, Xu B. Dexamethasone Suppresses Radicular Pain Through Targeting the L-PGDS/PI3K/Akt Pathway in Rats With Lumbar Disc Herniation. Pain Pract 2020; 21:64-74. [PMID: 32640501 DOI: 10.1111/papr.12934] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/21/2020] [Accepted: 07/02/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE Lumbar disc herniation (LDH) is a frequently occurring disease with unknown etiology, which makes treatment a challenge. The aim of this study was to analyze the effects of dexamethasone on LDH and elucidate the underlying mechanisms. GENERAL METHODS An LDH rat model was established by nucleus pulposus implantation. The activity of the lipocalin type prostaglandin D synthase (L-PGDS)/phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) axis was evaluated by Western blotting. Paw withdrawal threshold and paw withdrawal latency were assessed by the Von Frey hairs method and the thermal dolorimeter of Hargreaves, respectively. The 21-point Basso-Beattie-Bresnahan scale was used to assess the locomotor function of rats. Pathological changes in the affected region were analyzed by hematoxylin-eosin staining. Immunofluorescence was used to measure the expression of microtubule-associated protein (MAP-2). FINDINGS Lumbar disc herniation markedly increased thermo-mechanical allodynia and induced dorsal root ganglion (DRG) degeneration by inactivating the L-PGS/PI3K/Akt pathway. Dexamethasone restored the L-PGDS/PI3K/Akt pathway and relieved LDH-induced thermo-mechanical allodynia. Furthermore, overexpression and knockdown of L-PGDS respectively attenuated and worsened LDH-triggered thermo-mechanical allodynia and tissue degeneration by modulating the PI3K/Akt pathway. Pretreatment with dexamethasone partially abrogated the effect of L-PGDS knockdown through PI3K/Akt activation. CONCLUSIONS Dexamethasone relieves LDH-mediated radicular pain by exerting anti-inflammatory effects and reducing the suppression of L-PGDS induced by LDH. Meanwhile, the activity of the PI3K/Akt pathway was decreased, possibly due to the attenuated inflammation induced by dexamethasone. Our results revealed the underlying mechanism of dexamethasone, which might be helpful in reducing the side effects of dexamethasone and provide more focused therapy in LDH.
Collapse
Affiliation(s)
- Weixing Xu
- Department of Orthopaedics, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Weiguo Ding
- Department of Orthopaedics, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Hongfeng Sheng
- Department of Orthopaedics, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Di Lu
- Department of Orthopaedics, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Xinwei Xu
- Department of Orthopaedics, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Bin Xu
- Department of Orthopaedics, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| |
Collapse
|
19
|
GGA3 interacts with L-type prostaglandin D synthase and regulates the recycling and signaling of the DP1 receptor for prostaglandin D2 in a Rab4-dependent mechanism. Cell Signal 2020; 72:109641. [DOI: 10.1016/j.cellsig.2020.109641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/15/2020] [Accepted: 04/15/2020] [Indexed: 12/21/2022]
|
20
|
Amyloid β chaperone - lipocalin-type prostaglandin D synthase acts as a peroxidase in the presence of heme. Biochem J 2020; 477:1227-1240. [PMID: 32271881 PMCID: PMC7148433 DOI: 10.1042/bcj20190536] [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: 07/24/2019] [Revised: 02/07/2020] [Accepted: 02/12/2020] [Indexed: 12/01/2022]
Abstract
The extracellular transporter, lipocalin-type prostaglandin D synthase (L-PGDS) binds to heme and heme metabolites with high affinity. It has been reported that L-PGDS protects neuronal cells against apoptosis induced by exposure to hydrogen peroxide. Our study demonstrates that when human WT L-PGDS is in complex with heme, it exhibits a strong peroxidase activity thus behaving as a pseudo-peroxidase. Electron paramagnetic resonance studies confirm that heme in the L-PGDS–heme complex is hexacoordinated with high-spin Fe(III). NMR titration of heme in L-PGDS points to hydrophobic interaction between heme and several residues within the β-barrel cavity of L-PGDS. In addition to the transporter function, L-PGDS is a key amyloid β chaperone in human cerebrospinal fluid. The presence of high levels of bilirubin and its derivatives, implicated in Alzheimer's disease, by binding to L-PGDS may reduce its chaperone activity. Nevertheless, our ThT binding assay establishes that heme and heme metabolites do not significantly alter the neuroprotective chaperone function of L-PGDS. Guided by NMR data we reconstructed the heme L-PGDS complex using extensive molecular dynamics simulations providing a platform for mechanistic interpretation of the catalytic and transporting functions and their modulation by secondary ligands like Aβ peptides and heme metabolites.
Collapse
|
21
|
Kannaian B, Sharma B, Phillips M, Chowdhury A, Manimekalai MSS, Adav SS, Ng JTY, Kumar A, Lim S, Mu Y, Sze SK, Grüber G, Pervushin K. Abundant neuroprotective chaperone Lipocalin-type prostaglandin D synthase (L-PGDS) disassembles the Amyloid-β fibrils. Sci Rep 2019; 9:12579. [PMID: 31467325 PMCID: PMC6715741 DOI: 10.1038/s41598-019-48819-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 08/06/2019] [Indexed: 01/08/2023] Open
Abstract
Misfolding of Amyloid β (Aβ) peptides leads to the formation of extracellular amyloid plaques. Molecular chaperones can facilitate the refolding or degradation of such misfolded proteins. Here, for the first time, we report the unique ability of Lipocalin-type Prostaglandin D synthase (L-PGDS) protein to act as a disaggregase on the pre-formed fibrils of Aβ(1-40), abbreviated as Aβ40, and Aβ(25-35) peptides, in addition to inhibiting the aggregation of Aβ monomers. Furthermore, our proteomics results indicate that L-PGDS can facilitate extraction of several other proteins from the insoluble aggregates extracted from the brain of an Alzheimer's disease patient. In this study, we have established the mode of binding of L-PGDS with monomeric and fibrillar Aβ using Nuclear Magnetic Resonance (NMR) Spectroscopy, Small Angle X-ray Scattering (SAXS), and Transmission Electron Microscopy (TEM). Our results confirm a direct interaction between L-PGDS and monomeric Aβ40 and Aβ(25-35), thereby inhibiting their spontaneous aggregation. The monomeric unstructured Aβ40 binds to L-PGDS via its C-terminus, while the N-terminus remains free which is observed as a new domain in the L-PGDS-Aβ40 complex model.
Collapse
Affiliation(s)
- Bhuvaneswari Kannaian
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Bhargy Sharma
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Margaret Phillips
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Anup Chowdhury
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Malathy S S Manimekalai
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Sunil S Adav
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
- Singapore Phenome Centre, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 636921, Singapore
| | - Justin T Y Ng
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Ambrish Kumar
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637459, Singapore
| | - Sierin Lim
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, 637459, Singapore
| | - Yuguang Mu
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Siu K Sze
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Gerhard Grüber
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Konstantin Pervushin
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore.
| |
Collapse
|
22
|
Ahmad AS, Ottallah H, Maciel CB, Strickland M, Doré S. Role of the L-PGDS-PGD2-DP1 receptor axis in sleep regulation and neurologic outcomes. Sleep 2019; 42:zsz073. [PMID: 30893431 PMCID: PMC6559173 DOI: 10.1093/sleep/zsz073] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 02/16/2019] [Indexed: 12/18/2022] Open
Abstract
To meet the new challenges of modern lifestyles, we often compromise a good night's sleep. In preclinical models as well as in humans, a chronic lack of sleep is reported to be among the leading causes of various physiologic, psychologic, and neurocognitive deficits. Thus far, various endogenous mediators have been implicated in inter-regulatory networks that collectively influence the sleep-wake cycle. One such mediator is the lipocalin-type prostaglandin D2 synthase (L-PGDS)-Prostaglandin D2 (PGD2)-DP1 receptor (L-PGDS-PGD2-DP1R) axis. Findings in preclinical models confirm that DP1R are predominantly expressed in the sleep-regulating centers. This finding led to the discovery that the L-PGDS-PGD2-DP1R axis is involved in sleep regulation. Furthermore, we showed that the L-PGDS-PGD2-DP1R axis is beneficial in protecting the brain from ischemic stroke. Protein sequence homology was also performed, and it was found that L-PGDS and DP1R share a high degree of homology between humans and rodents. Based on the preclinical and clinical data thus far pertaining to the role of the L-PGDS-PGD2-DP1R axis in sleep regulation and neurologic conditions, there is optimism that this axis may have a high translational potential in human therapeutics. Therefore, here the focus is to review the regulation of the homeostatic component of the sleep process with a special focus on the L-PGDS-PGD2-DP1R axis and the consequences of sleep deprivation on health outcomes. Furthermore, we discuss whether the pharmacological regulation of this axis could represent a tool to prevent sleep disturbances and potentially improve outcomes, especially in patients with acute brain injuries.
Collapse
Affiliation(s)
- Abdullah Shafique Ahmad
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL
- McKnight Brain Institute, University of Florida, Gainesville, FL
| | - Haneen Ottallah
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL
- McKnight Brain Institute, University of Florida, Gainesville, FL
| | - Carolina B Maciel
- Department of Neurology, University of Florida College of Medicine, Gainesville, FL
| | - Michael Strickland
- Division of Biology and Biomedical Sciences, Washington University in Saint Louis, Saint Louis, MO
| | - Sylvain Doré
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL
- McKnight Brain Institute, University of Florida, Gainesville, FL
- Department of Psychiatry, University of Florida, Gainesville, FL
- Department of Pharmaceutics, University of Florida, Gainesville, FL
- Department of Psychology, University of Florida, Gainesville, FL
- Department of Neuroscience, University of Florida, Gainesville, FL
| |
Collapse
|
23
|
Horikami D, Toya N, Kobayashi K, Omori K, Nagata N, Murata T. L-PGDS-derived PGD2
attenuates acute lung injury by enhancing endothelial barrier formation. J Pathol 2019; 248:280-290. [DOI: 10.1002/path.5253] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 12/31/2018] [Accepted: 02/04/2019] [Indexed: 11/08/2022]
Affiliation(s)
- Daiki Horikami
- Department of Animal Radiology; Graduate School of Agriculture and Life Sciences, The University of Tokyo; Tokyo Japan
| | - Naoki Toya
- Department of Animal Radiology; Graduate School of Agriculture and Life Sciences, The University of Tokyo; Tokyo Japan
| | - Koji Kobayashi
- Department of Animal Radiology; Graduate School of Agriculture and Life Sciences, The University of Tokyo; Tokyo Japan
| | - Keisuke Omori
- Department of Animal Radiology; Graduate School of Agriculture and Life Sciences, The University of Tokyo; Tokyo Japan
| | - Nanae Nagata
- Department of Animal Radiology; Graduate School of Agriculture and Life Sciences, The University of Tokyo; Tokyo Japan
| | - Takahisa Murata
- Department of Animal Radiology; Graduate School of Agriculture and Life Sciences, The University of Tokyo; Tokyo Japan
| |
Collapse
|
24
|
Tsubosaka Y, Maehara T, Imai D, Nakamura T, Kobayashi K, Nagata N, Fujii W, Murata T. Hematopoietic prostaglandin D synthase–derived prostaglandin D
2
ameliorates adjuvant‐induced joint inflammation in mice. FASEB J 2019; 33:6829-6837. [DOI: 10.1096/fj.201802153r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Yoshiki Tsubosaka
- Department of Animal RadiologyGraduate School of Agricultural and Life SciencesThe University of TokyoTokyoJapan
| | - Toko Maehara
- Department of Animal RadiologyGraduate School of Agricultural and Life SciencesThe University of TokyoTokyoJapan
| | - Daiki Imai
- Department of Animal RadiologyGraduate School of Agricultural and Life SciencesThe University of TokyoTokyoJapan
| | - Tatsuro Nakamura
- Department of Animal RadiologyGraduate School of Agricultural and Life SciencesThe University of TokyoTokyoJapan
| | - Koji Kobayashi
- Department of Animal RadiologyGraduate School of Agricultural and Life SciencesThe University of TokyoTokyoJapan
| | - Nanae Nagata
- Department of Animal RadiologyGraduate School of Agricultural and Life SciencesThe University of TokyoTokyoJapan
| | - Wataru Fujii
- Department of Applied GeneticsGraduate School of Agriculture and Life SciencesThe University of TokyoTokyoJapan
| | - Takahisa Murata
- Department of Animal RadiologyGraduate School of Agricultural and Life SciencesThe University of TokyoTokyoJapan
| |
Collapse
|
25
|
Zhang Z, Liu WY, Diao YP, Xu W, Zhong YH, Zhang JY, Lazarus M, Liu YY, Qu WM, Huang ZL. Superior Colliculus GABAergic Neurons Are Essential for Acute Dark Induction of Wakefulness in Mice. Curr Biol 2019; 29:637-644.e3. [PMID: 30713103 DOI: 10.1016/j.cub.2018.12.031] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 11/12/2018] [Accepted: 12/17/2018] [Indexed: 01/05/2023]
Abstract
Sleep is regulated by homeostatic process and circadian clock. Light indirectly modulates sleep by entraining the circadian clock to the solar day. Light can also influence sleep independent of photo-entrainment [1]. An acute light exposure could induce sleep, and an acute dark pulse could increase wakefulness in nocturnal animals [1, 2]. The photoreceptors and cell types in the retina that mediate light and dark effects on sleep are well characterized [1-4]. A few studies have explored the brain region involved in acute light induction of sleep. Fos expression and nonspecific lesions suggest that the superior colliculus (SC) may play a role in acute light induction of sleep [2, 5]. In contrast, the brain area and neural circuits mediating acute dark induction of wakefulness are unknown. Here, we demonstrated that retina ganglion cells (RGCs) had direct innervations on the GABAergic neurons in the mouse SC, and the activities of these cells were inhibited by an acute dark pulse, but not influenced by a light pulse. Moreover, ablating SC GABAergic neurons abolished the acute dark induction of wakefulness, but not light induction of sleep. Based on optogenetic and electrophysiological experiments, we found that SC GABAergic neurons formed monosynaptic functional connections with dopaminergic neurons in the ventral tegmental area (VTA). Selective lesions of VTA dopaminergic cells totally abolished acute dark induction of wakefulness without affecting the light induction of sleep. Collectively, our findings uncover a fundamental role for a retinal-SC GABAergic-VTA dopaminergic circuit in acute dark induction of wakefulness and indicate that the dark and light signals affect sleep-wake behaviors through distinct pathways.
Collapse
Affiliation(s)
- Ze Zhang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Wen-Ying Liu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yu-Pu Diao
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Wei Xu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yu-Heng Zhong
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jia-Yi Zhang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Michael Lazarus
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Yuan-Yuan Liu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Wei-Min Qu
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| | - Zhi-Li Huang
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
26
|
Korkutata M, Saitoh T, Cherasse Y, Ioka S, Duo F, Qin R, Murakoshi N, Fujii S, Zhou X, Sugiyama F, Chen JF, Kumagai H, Nagase H, Lazarus M. Enhancing endogenous adenosine A2A receptor signaling induces slow-wave sleep without affecting body temperature and cardiovascular function. Neuropharmacology 2019; 144:122-132. [DOI: 10.1016/j.neuropharm.2018.10.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 10/10/2018] [Accepted: 10/14/2018] [Indexed: 01/20/2023]
|
27
|
Cherasse Y, Aritake K, Oishi Y, Kaushik MK, Korkutata M, Urade Y. The Leptomeninges Produce Prostaglandin D 2 Involved in Sleep Regulation in Mice. Front Cell Neurosci 2018; 12:357. [PMID: 30364224 PMCID: PMC6193105 DOI: 10.3389/fncel.2018.00357] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/24/2018] [Indexed: 11/13/2022] Open
Abstract
Injection of nanomolar amounts of prostaglandin D2 (PGD2) into the rat brain has dose and time-dependent somnogenic effects, and the PGD2-induced sleep is indistinguishable from physiologic sleep. Sleep-inducing PGD2 is produced in the brain by lipocalin-type PGD2 synthase (LPGDS). Three potential intracranial sources of LPGDS have been identified: oligodendrocytes, choroid plexus, and leptomeninges. We aimed at the identification of the site of synthesis of somnogenic PGD2 and therefore, generated a transgenic mouse line with the LPGDS gene amenable to conditional deletion using Cre recombinase (flox-LPGDS mouse). To identify the cell type responsible for producing somnogenic PGD2, we engineered animals lacking LPGDS expression specifically in oligodendrocytes (OD-LPGDS KO), choroid plexus (CP-LPGDS KO), or leptomeninges (LM-LPGDS KO). We measured prostaglandins and LPGDS concentrations together with PGD synthase activity in the brain of these mice. While the LPGDS amount and PGD synthase activity were drastically reduced in the OD- and LM-LPGDS KO mice, they were unchanged in the CP-LPGDS KO mice compared with control animals. We then recorded electroencephalograms, electromyograms, and locomotor activity to measure sleep in 10-week-old mice with specific knockdown of LPGDS in each of the three targets. Using selenium tetrachloride, a specific PGDS inhibitor, we demonstrated that sleep is inhibited in OD-LPGDS and CP-LPGDS KO mice, but not in the LM-LPGDS KO mice. We concluded that somnogenic PGD2 is produced primarily by the leptomeninges, and not by oligodendrocytes or choroid plexus.
Collapse
Affiliation(s)
- Yoan Cherasse
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Kosuke Aritake
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Yo Oishi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Mahesh K Kaushik
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Mustafa Korkutata
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan.,Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Japan
| | - Yoshihiro Urade
- The University of Tokyo Hospital, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
28
|
Wang YQ, Zhang MQ, Li R, Qu WM, Huang ZL. The Mutual Interaction Between Sleep and Epilepsy on the Neurobiological Basis and Therapy. Curr Neuropharmacol 2018; 16:5-16. [PMID: 28486925 PMCID: PMC5771383 DOI: 10.2174/1570159x15666170509101237] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 07/11/2017] [Accepted: 04/27/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Sleep and epilepsy are mutually related in a complex, bidirectional manner. However, our understanding of this relationship remains unclear. RESULTS The literatures of the neurobiological basis of the interactions between sleep and epilepsy indicate that non rapid eye movement sleep and idiopathic generalized epilepsy share the same thalamocortical networks. Most of neurotransmitters and neuromodulators such as adenosine, melatonin, prostaglandin D2, serotonin, and histamine are found to regulate the sleep-wake behavior and also considered to have antiepilepsy effects; antiepileptic drugs, in turn, also have effects on sleep. Furthermore, many drugs that regulate the sleep-wake cycle can also serve as potential antiseizure agents. The nonpharmacological management of epilepsy including ketogenic diet, epilepsy surgery, neurostimulation can also influence sleep. CONCLUSION In this paper, we address the issues involved in these phenomena and also discuss the various therapies used to modify them.
Collapse
Affiliation(s)
| | | | - Rui Li
- Department of Pharmacology and Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation
Center for Brain Science, Fudan University, Shanghai200032, P.R. China
| | - Wei-Min Qu
- Department of Pharmacology and Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation
Center for Brain Science, Fudan University, Shanghai200032, P.R. China
| | - Zhi-Li Huang
- Department of Pharmacology and Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences; State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation
Center for Brain Science, Fudan University, Shanghai200032, P.R. China
| |
Collapse
|
29
|
Omori K, Morikawa T, Kunita A, Nakamura T, Aritake K, Urade Y, Fukayama M, Murata T. Lipocalin-type prostaglandin D synthase-derived PGD 2 attenuates malignant properties of tumor endothelial cells. J Pathol 2017; 244:84-96. [PMID: 29124765 DOI: 10.1002/path.4993] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 08/10/2017] [Accepted: 09/12/2017] [Indexed: 11/06/2022]
Abstract
Endothelial cells (ECs) are a key component of the tumor microenvironment. They have abnormal characteristics compared to the ECs in normal tissues. Here, we found a marked increase in lipocalin-type prostaglandin D synthase (L-PGDS) mRNA (Ptgds) expression in ECs isolated from mouse melanoma. Immunostaining of mouse melanoma revealed expression of L-PGDS protein in the ECs. In situ hybridization also showed L-PGDS (PTGDS) mRNA expression in the ECs of human melanoma and oral squamous cell carcinoma. In vitro experiments showed that stimulation with tumor cell-derived IL-1 and TNF-α increased L-PGDS mRNA expression and its product prostaglandin D2 (PGD2 ) in human normal ECs. We also investigated the contribution of L-PGDS-PGD2 to tumor growth and vascularization. Systemic or EC-specific deficiency of L-PGDS accelerated the growth of melanoma in mice, whereas treatment with an agonist of the PGD2 receptor, DP1 (BW245C, 0.1 mg/kg, injected intraperitoneally twice daily), attenuated it. Morphological and in vivo studies showed that endothelial L-PGDS deficiency resulted in functional changes of tumor ECs such as accelerated vascular hyperpermeability, angiogenesis, and endothelial-to-mesenchymal transition (EndMT) in tumors, which in turn reduced tumor cell apoptosis. These observations suggest that tumor cell-derived inflammatory cytokines increase L-PGDS expression and subsequent PGD2 production in the tumor ECs. This PGD2 acts as a negative regulator of the tumorigenic changes in tumor ECs. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Keisuke Omori
- Department of Animal Radiology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Teppei Morikawa
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akiko Kunita
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tatsuro Nakamura
- Department of Animal Radiology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Kosuke Aritake
- Intemational Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yoshihiro Urade
- Intemational Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Japan
| | - Masashi Fukayama
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takahisa Murata
- Department of Animal Radiology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
30
|
Saitoh A, Tominaga H, Ogawa Y, Irukayama-Tomobe Y, Yamada M, Yanagisawa M, Nagase H. Effects of the delta opioid receptor agonist KNT-127 on electroencephalographic activity in mice. Pharmacol Rep 2017; 70:350-354. [PMID: 29477045 DOI: 10.1016/j.pharep.2017.08.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 08/16/2017] [Accepted: 08/25/2017] [Indexed: 11/15/2022]
Abstract
BACKGROUND We previously reported that the novel selective delta opioid receptor (DOP) agonist KNT-127 did not cause convulsions in mice, whereas the prototype DOP agonist SNC80 did. Previous studies have reported that SNC80 caused electroencephalographic (EEG) disturbances in rodents. However, whether KNT-127 affects EEG responses is unknown. Therefore, the present study aimed to compare the effect of KNT-127 on EEG responses with that of SNC80 in mice. METHODS For behavioral experiments, male C57BL6/J mice were injected intraperitoneally with either KNT-127 (30 mg/kg) or SNC80 (30 mg/kg) and monitored for convulsions and subsequent catalepsy-like behavior for 10 min immediately after drug treatment. For EEG recording experiments, EEG electrodes were implanted into the right hemisphere. EEG signals exceeding twice the baseline amplitude were defined as seizure spikes. RESULTS KNT-127 did not induce convulsive or catalepsy-like behaviors in mice and did not result in seizure spikes, while significantly higher EEG power density was observed at 2 Hz. In contrast, SNC80 administration resulted in convulsive behaviors, seizure spikes, and significantly higher EEG power density between 2 and 10 Hz in mice. CONCLUSIONS In this study, we clearly demonstrated that KNT-127 administration induces neither convulsive effects nor seizure spikes in mice. We propose that KNT-127 should be considered a candidate compound for the development of improved DOP-based psychotropic drug that lack the convulsive properties.
Collapse
Affiliation(s)
- Akiyoshi Saitoh
- Department of Neuropsychopharmacology, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Hiromu Tominaga
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yasuhiro Ogawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yoko Irukayama-Tomobe
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Mitsuhiko Yamada
- Department of Neuropsychopharmacology, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hiroshi Nagase
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan.
| |
Collapse
|
31
|
Adenosine A 2A receptor mediates hypnotic effects of ethanol in mice. Sci Rep 2017; 7:12678. [PMID: 28978989 PMCID: PMC5627250 DOI: 10.1038/s41598-017-12689-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 09/13/2017] [Indexed: 12/25/2022] Open
Abstract
Ethanol has extensive effects on sleep and daytime alertness, causing premature disability and death. Adenosine, as a potent sleep-promoting substance, is involved in many cellular and behavioral responses to ethanol. However, the mechanisms of hypnotic effects of ethanol remain unclear. In this study, we investigated the role of adenosine in ethanol-induced sleep using C57BL/6Slac mice, adenosine A2A receptor (A2AR) knockout mice, and their wild-type littermates. The results showed that intraperitoneal injection of ethanol (3.0 g/kg) at 21:00 decreased the latency to non-rapid eye movement (NREM) sleep and increased the duration of NREM sleep for 5 h. Ethanol dose-dependently increased NREM sleep, which was consistent with decreases in wakefulness in C57BL/6Slac mice compared with their own control. Caffeine (5, 10, or 15 mg/kg), a nonspecific adenosine receptor antagonist, dose-dependently and at high doses completely blocked ethanol-induced NREM sleep when administered 30 min prior to (but not after) ethanol injection. Moreover, ethanol-induced NREM sleep was completely abolished in A2AR knockout mice compared with wild-type mice. These findings strongly indicate that A2AR is a key receptor for the hypnotic effects of ethanol, and pretreatment of caffeine might be a strategy to counter the hypnotic effects of ethanol.
Collapse
|
32
|
|
33
|
Jandl K, Heinemann A. The therapeutic potential of CRTH2/DP2 beyond allergy and asthma. Prostaglandins Other Lipid Mediat 2017; 133:42-48. [PMID: 28818625 PMCID: PMC7612073 DOI: 10.1016/j.prostaglandins.2017.08.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 08/04/2017] [Accepted: 08/07/2017] [Indexed: 12/12/2022]
Abstract
Prostaglandin (PG) D2 has been in the focus of research for quite a long time, but its biological effects and its roles in human disease are still not fully characterized. When in 2001 a second major PGD2 receptor termed chemoattractant receptor homologue expressed on Th2 cells (CRTH2; alternative name DP2) was discovered, diverse investigations started to shed more light on the complex and often controversial actions of the prostaglandin. With various immunomodulating effects, such as induction of migration, activation, and cytokine release of leukocytes observed both in vivo and in vitro, CRTH2 has emerged as a promising target for the treatment of allergic diseases. However, with more and more research being performed on CRTH2, it has also become clear that its biological actions are far more diverse than expected at the beginning. In this review, we aim to summarize the roles that PGD2 - and CRTH2 in particular - might play in diseases of the central nervous system, kidney, intestine, lung, hair and skin, bone and cartilage, and in cancer. Based on current data we propose that blocking CRTH2 might be a potential therapeutic approach to numerous conditions beyond classical allergic diseases and asthma.
Collapse
Affiliation(s)
- Katharina Jandl
- Institute for Experimental and Clinical Pharmacology, Medical University Graz, Austria; Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Akos Heinemann
- Institute for Experimental and Clinical Pharmacology, Medical University Graz, Austria; BioTechMed Graz, Austria.
| |
Collapse
|
34
|
Chu C, Wei H, Zhu W, Shen Y, Xu Q. Decreased Prostaglandin D2 Levels in Major Depressive Disorder Are Associated with Depression-Like Behaviors. Int J Neuropsychopharmacol 2017; 20:731-739. [PMID: 28582515 PMCID: PMC5581486 DOI: 10.1093/ijnp/pyx044] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 06/01/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Prostaglandin (PG) D2 is the most abundant prostaglandin in the mammalian brain. The physiological and pharmacological actions of PGD2 in the central nervous system seem to be associated with some of the symptoms exhibited by patients with major depressive disorder. Previous studies have found that PGD2 synthase was decreased in the cerebrospinal fluid of major depressive disorder patients. We speculated that there may be a dysregulation of PGD2 levels in major depressive disorder. METHODS Ultra-performance liquid chromatography-tandem mass spectrometry coupled with a stable isotopic-labeled internal standard was used to determine PGD2 levels in the plasma of major depressive disorder patients and in the brains of depressive mice. A total of 32 drug-free major depressive disorder patients and 30 healthy controls were recruited. An animal model of depression was constructed by exposing mice to 5 weeks of chronic unpredictable mild stress. To explore the role of PGD2 in major depressive disorder, selenium tetrachloride was administered to simulate the change in PGD2 levels in mice. RESULTS Mice exposed to chronic unpredictable mild stress exhibited depression-like behaviors, as indicated by reduced sucrose preference and increased immobility time in the forced swimming test. PGD2 levels in the plasma of major depressive disorder patients and in the brains of depressive mice were both decreased compared with their corresponding controls. Further inhibiting PGD2 production in mice resulted in an increased immobility time in the forced swimming test that could be reversed by imipramine. CONCLUSION Decreased PGD2 levels in major depressive disorder are associated with depression-like behaviors.
Collapse
Affiliation(s)
- Cuilin Chu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Hui Wei
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Wanwan Zhu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yan Shen
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Qi Xu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Correspondence: Qi Xu, PhD, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 5 Dong Dan San Tiao, Dong Cheng District, Beijing, China 100005 ()
| |
Collapse
|
35
|
Zhang Z, Wang HJ, Wang DR, Qu WM, Huang ZL. Red light at intensities above 10 lx alters sleep-wake behavior in mice. LIGHT, SCIENCE & APPLICATIONS 2017; 6:e16231. [PMID: 30167247 PMCID: PMC6062196 DOI: 10.1038/lsa.2016.231] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 09/16/2016] [Accepted: 09/21/2016] [Indexed: 05/10/2023]
Abstract
Sleep is regulated by two mechanisms: the homeostatic process and the circadian clock. Light affects sleep and alertness by entraining the circadian clock, and acutely inducing sleep/alertness, in a manner mediated by intrinsically photosensitive retinal ganglion cells. Because intrinsically photosensitive retinal ganglion cells are believed to be minimally sensitive to red light, which is widely used for illumination to reduce the photic disturbance to nocturnal animals during the dark phase. However, the appropriate intensity of the red light is unknown. In the present study, we recorded electroencephalograms and electromyograms of freely moving mice to investigate the effects of red light emitted by light-emitting diodes at different intensities and for different durations on the sleep-wake behavior of mice. White light was used as a control. Unexpectedly, red light exerted potent sleep-inducing effects and changed the sleep architecture in terms of the duration and number of sleep episodes, the stage transition, and the EEG power density when the intensity was >20 lx. Subsequently, we lowered the light intensity and demonstrated that red light at or below 10 lx did not affect sleep-wake behavior. White light markedly induced sleep and disrupted sleep architecture even at an intensity as low as 10 lx. Our findings highlight the importance of limiting the intensity of red light (⩽10 lx) to avoid optical influence in nocturnal behavioral experiments, particularly in the field of sleep and circadian research.
Collapse
|
36
|
Bellieni CV, Vannuccini S, Petraglia F. Is fetal analgesia necessary during prenatal surgery? J Matern Fetal Neonatal Med 2017; 31:1241-1245. [PMID: 28337942 DOI: 10.1080/14767058.2017.1311860] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Fetal pain and fetal anesthesia are still matter of debate: some authors hypothesize that several intrauterine endocrine neuroinhibitors (ENIn) anesthetize the fetus, keeping it in a constant state of sleep, and making pharmacological fetal anesthesia useless for fetal surgery, while others argue fetal pain is possible and shoud be prevented with fetal anesthesy. AIM To retrieve evidences about fetal pain, fetal arousability and about the level of sedation induced by the ENIn, in order to assess the necessity of direct fetal anesthesia during prenatal fetal surgery. METHODS We performed a careful literature review (1990-2016) on fetal arousability, and on the possibility that ENIn at the average fetal blood levels induce actual anesthesia. We retrieved the papers that fulfilled the research criteria, with particular attention to the second half of pregnancy, the period when most fetal surgery is performed. RESULTS Fetuses are awake about 10% of the total time in the last gestational weeks, and they can be aroused by external stimuli. ENIn have not an anesthetic effect at normal fetal values, but only when they areartificialy injected at high doses; their blood levels in the last trimester of average pregnancies are not dissimilar either in the fetus or in the mother. CONCLUSIONS During the second half of the pregnancy, external stimuli can awake the fetuses, although they spend most of the time in sleeping state; the presence of ENIn is absolutely not enough to guarantee an effective anesthesia during surgery. Thus, direct fetal analgesia/anesthesia is mandatory, though further studies on its possible drawbacks are necessary.
Collapse
Affiliation(s)
- Carlo V Bellieni
- a Neonatal Intensive Care Unit , University Hospital of Siena , Siena , Italy
| | - Silvia Vannuccini
- b Department of Molecular and Developmental Medicine, Division of Obstetrics and Gynecology , University of Siena , Siena , Italy
| | - Felice Petraglia
- b Department of Molecular and Developmental Medicine, Division of Obstetrics and Gynecology , University of Siena , Siena , Italy
| |
Collapse
|
37
|
Zhang BJ, Huang ZL, Chen JF, Urade Y, Qu WM. Adenosine A 2A receptor deficiency attenuates the somnogenic effect of prostaglandin D 2 in mice. Acta Pharmacol Sin 2017; 38:469-476. [PMID: 28112177 DOI: 10.1038/aps.2016.140] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 11/11/2016] [Indexed: 01/03/2023]
Abstract
Prostaglandin D2 (PGD2) is one of the most potent endogenous sleep promoting substances. PGD2 activates the PGD2 receptor (DPR) and increases the extracellular level of adenosine in wild-type (WT) mice but not DPR knockout (KO) mice, suggesting that PGD2-induced sleep is DPR-dependent, and adenosine may be the signaling molecule that mediates the somnogenic effect of PGD2. The aim of this study was to determine the involvement of the adenosine A2A receptor (A2AR) in PGD2-induced sleep. We infused PGD2 into the lateral ventricle of WT and A2AR KO mice between 20:00 and 2:00 for 6 h, and electroencephalograms and electromyograms were simultaneously recorded. In WT mice, PGD2 infusion dose-dependently increased non-rapid eye movement (non-REM, NREM) sleep, which was 139.1%, 145.0% and 202.7% as large as that of vehicle-treated mice at doses of 10, 20 and 50 pmol/min, respectively. PGD2 infusion at doses of 20 and 50 pmol/min also increased REM sleep during the 6-h PGD2 infusion and 4-h post-dosing periods in WT mice to 148.9% and 166.7%, respectively. In A2AR KO mice, however, PGD2 infusion at 10 pmol/min did not change the sleep profile, whereas higher doses at 20 and 50 pmol/min increased the NREM sleep during the 6-h PGD2 infusion to 117.5% and 155.6%, respectively, but did not change the sleep in the post-dosing period. Moreover, PGD2 infusion at 50 pmol/min significantly increased the episode number in both genotypes but only enhanced the episode duration in WT mice. The results demonstrate that PGD2-induced sleep in mice is mediated by both adenosine A2AR-dependent and -independent systems.
Collapse
|
38
|
Zhang BJ, Shao SR, Aritake K, Takeuchi A, Urade Y, Huang ZL, Lazarus M, Qu WM. Interleukin-1β induces sleep independent of prostaglandin D 2 in rats and mice. Neuroscience 2017; 340:258-267. [DOI: 10.1016/j.neuroscience.2016.09.053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/01/2016] [Accepted: 09/08/2016] [Indexed: 01/28/2023]
|
39
|
Sagawa Y, Sato M, Sakai N, Chikahisa S, Chiba S, Maruyama T, Yamamoto J, Nishino S. Wake-promoting effects of ONO-4127Na, a prostaglandin DP1 receptor antagonist, in hypocretin/orexin deficient narcoleptic mice. Neuropharmacology 2016; 110:268-276. [PMID: 27474349 DOI: 10.1016/j.neuropharm.2016.07.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 07/11/2016] [Accepted: 07/12/2016] [Indexed: 11/28/2022]
Abstract
Prostaglandin (PG)D2 is an endogenous sleep substance, and a series of animal studies reported that PGD2 or PGD2 receptor (DP1) agonists promote sleep, while DP1 antagonists promote wakefulness. This suggests the possibility of use of PG DP1 antagonists as wake-promoting compounds. We therefore evaluated the wake-promoting effects of ONO-4127Na, a DP1 antagonist, in a mouse model of narcolepsy (i.e., orexin/ataxin-3 transgenic mice) and compared those to effects of modafinil. ONO-4127Na perfused in the basal forebrain (BF) area potently promoted wakefulness in both wild type and narcoleptic mice, and the wake-promoting effects of ONO-4127Na at 2.93 × 10(-4) M roughly corresponded to those of modafinil at 100 mg/kg (p.o.). The wake promoting effects of ONO-4127Na was observed both during light and dark periods, and much larger effects were seen during the light period when mice slept most of the time. ONO-4127Na, when perfused in the hypothalamic area, had no effects on sleep. We further demonstrated that wake-promoting effects of ONO-4127Na were abolished in DP1 KO mice, confirming that the wake-promoting effect of ONO-4127Na is mediated by blockade of the PG DP1 receptors located in the BF area. ONO-4127Na reduced DREM, an EEG/EMG assessment of behavioral cataplexy in narcoleptic mice, suggesting that ONO-4127Na is likely to have anticataplectic effects. DP1 antagonists may be a new class of compounds for the treatment of narcolepsy-cataplexy, and further studies are warranted.
Collapse
Affiliation(s)
- Yohei Sagawa
- Sleep and Circadian Neurobiology Laboratory, Stanford University, United States; Department of Neuropsychiatry, Akita University Graduate School of Medicine, Japan
| | - Masatoshi Sato
- Sleep and Circadian Neurobiology Laboratory, Stanford University, United States; Department of Neuropsychiatry, Akita University Graduate School of Medicine, Japan
| | - Noriaki Sakai
- Sleep and Circadian Neurobiology Laboratory, Stanford University, United States
| | - Sachiko Chikahisa
- Sleep and Circadian Neurobiology Laboratory, Stanford University, United States; Department of Integrative Physiology, Institute of Health Biosciences, The University of Tokushima Graduate School, Japan
| | - Shintaro Chiba
- Sleep and Circadian Neurobiology Laboratory, Stanford University, United States; Department of Otorhinolaryngology, Jikei University School of Medicine, Japan
| | - Takashi Maruyama
- Sleep and Circadian Neurobiology Laboratory, Stanford University, United States
| | - Junki Yamamoto
- Minase Research Institute, Ono Pharmaceutical Co., Ltd., Osaka, Japan
| | - Seiji Nishino
- Sleep and Circadian Neurobiology Laboratory, Stanford University, United States.
| |
Collapse
|
40
|
Oishi Y, Takata Y, Taguchi Y, Kohtoh S, Urade Y, Lazarus M. Polygraphic Recording Procedure for Measuring Sleep in Mice. J Vis Exp 2016:e53678. [PMID: 26863349 PMCID: PMC4781694 DOI: 10.3791/53678] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Recording of the epidural electroencephalogram (EEG) and electromyogram (EMG) in small animals, like mice and rats, has been pivotal to study the homeodynamics and circuitry of sleep-wake regulation. In many laboratories, a cable-based sleep recording system is used to monitor the EEG and EMG in freely behaving mice in combination with computer software for automatic scoring of the vigilance states on the basis of power spectrum analysis of EEG data. A description of this system is detailed herein. Steel screws are implanted over the frontal cortical area and the parietal area of 1 hemisphere for monitoring EEG signals. In addition, EMG activity is monitored by the bilateral placement of wires in both neck muscles. Non-rapid eye movement (Non-REM; NREM) sleep is characterized by large, slow brain waves with delta activity below 4 Hz in the EEG, whereas a shift from low-frequency delta activity to a rapid low-voltage EEG in the theta range between 6 and 10 Hz can be observed at the transition from NREM to REM sleep. By contrast, wakefulness is identified by low- to moderate-voltage brain waves in the EEG trace and significant EMG activity.
Collapse
Affiliation(s)
- Yo Oishi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba
| | - Yohko Takata
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba
| | - Yujiro Taguchi
- Public Sector/Medical Solutions, Kissei Comtech Co., Ltd
| | - Sayaka Kohtoh
- Public Sector/Medical Solutions, Kissei Comtech Co., Ltd
| | - Yoshihiro Urade
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba
| | - Michael Lazarus
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba;
| |
Collapse
|
41
|
Abstract
Cortical electroencephalographic activity arises from corticothalamocortical interactions, modulated by wake-promoting monoaminergic and cholinergic input. These wake-promoting systems are regulated by hypothalamic hypocretin/orexins, while GABAergic sleep-promoting nuclei are found in the preoptic area, brainstem and lateral hypothalamus. Although pontine acetylcholine is critical for REM sleep, hypothalamic melanin-concentrating hormone/GABAergic cells may "gate" REM sleep. Daily sleep-wake rhythms arise from interactions between a hypothalamic circadian pacemaker and a sleep homeostat whose anatomical locus has yet to be conclusively defined. Control of sleep and wakefulness involves multiple systems, each of which presents vulnerability to sleep/wake dysfunction that may predispose to physical and/or neuropsychiatric disorders.
Collapse
Affiliation(s)
- Michael D Schwartz
- Biosciences Division, Center for Neuroscience, SRI International, 333 Ravenswood Avenue, Menlo Park, CA 94025, USA
| | - Thomas S Kilduff
- Biosciences Division, Center for Neuroscience, SRI International, 333 Ravenswood Avenue, Menlo Park, CA 94025, USA.
| |
Collapse
|
42
|
Cherasse Y, Saito H, Nagata N, Aritake K, Lazarus M, Urade Y. Zinc-containing yeast extract promotes nonrapid eye movement sleep in mice. Mol Nutr Food Res 2015; 59:2087-93. [PMID: 26105624 DOI: 10.1002/mnfr.201500082] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 05/01/2015] [Accepted: 06/16/2015] [Indexed: 12/12/2022]
Abstract
Zinc is an essential trace element for humans and animals, being located, among other places, in the synaptic vesicles of cortical glutamatergic neurons and hippocampal mossy fibers in the brain. Extracellular zinc has the potential to interact with and modulate many different synaptic targets, including glutamate and GABA receptors. Because of the central role of these neurotransmitters in brain activity, we examined in this study the sleep-promoting activity of zinc by monitoring locomotor activity and electroencephalogram after its administration to mice. Zinc-containing yeast extract (40 and 80 mg/kg) dose dependently increased the total amount of nonrapid eye movement sleep and decreased the locomotor activity. However, this preparation did not change the amount of rapid eye movement sleep or show any adverse effects such as rebound of insomnia during a period of 24 h following the induction of sleep; whereas the extracts containing other divalent cations (manganese, iron, and copper) did not decrease the locomotor activity. This is the first evidence that zinc can induce sleep. Our data open the way to new types of food supplements designed to improve sleep.
Collapse
Affiliation(s)
- Yoan Cherasse
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Hitomi Saito
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan.,Fujifilm Corporation, Tokyo, Japan
| | - Nanae Nagata
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Kosuke Aritake
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Michael Lazarus
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Yoshihiro Urade
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
43
|
Oishi Y, Yoshida K, Scammell TE, Urade Y, Lazarus M, Saper CB. The roles of prostaglandin E2 and D2 in lipopolysaccharide-mediated changes in sleep. Brain Behav Immun 2015; 47:172-7. [PMID: 25532785 PMCID: PMC4468012 DOI: 10.1016/j.bbi.2014.11.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 11/25/2014] [Accepted: 11/29/2014] [Indexed: 12/26/2022] Open
Abstract
When living organisms become sick as a result of a bacterial infection, a suite of brain-mediated responses occur, including fever, anorexia and sleepiness. Systemic administration of lipopolysaccharide (LPS), a common constituent of bacterial cell walls, increases body temperature and non-rapid eye movement (NREM) sleep in animals and induces the production of pro-inflammatory prostaglandins (PGs). PGE2 is the principal mediator of fever, and both PGE2 and PGD2 regulate sleep-wake behavior. The extent to which PGE2 and PGD2 are involved in the effect of LPS on NREM sleep remains to be clarified. Therefore, we examined LPS-induced changes in body temperature and NREM sleep in mice with nervous system-specific knockouts (KO) for the PGE2 receptors type EP3 or EP4, in mice with total body KO of microsomal PGE synthase-1 or the PGD2 receptor type DP, and in mice treated with the cyclooxygenase (COX) inhibitor meloxicam. We observed that LPS-induced NREM sleep was slightly attenuated in mice lacking EP4 receptors in the nervous system, but was not affected in any of the other KO mice or in mice pretreated with the COX inhibitor. These results suggest that the effect of LPS on NREM sleep is partially dependent on PGs and is likely mediated mainly by other pro-inflammatory substances. In addition, our data show that the main effect of LPS on body temperature is hypothermia in the absence of nervous system EP3 receptors or in the presence of a COX inhibitor.
Collapse
Affiliation(s)
- Yo Oishi
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States,International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Kyoko Yoshida
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Thomas E. Scammell
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Yoshihiro Urade
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan,Department of Molecular Behavioral Biology, Osaka Bioscience Institute, Osaka, Japan
| | - Michael Lazarus
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan; Department of Molecular Behavioral Biology, Osaka Bioscience Institute, Suita, Osaka, Japan.
| | - Clifford B. Saper
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States,Corresponding authors. Address: International Institute for Integrative Sleep Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan. Tel.: + 81 29 853 3681 (M. Lazarus). Address: Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, United States. Tel.: + 1 617 667 2622 (C.B. Saper). , (M. Lazarus), (C.B. Saper)
| |
Collapse
|
44
|
Huang ZL, Zhang Z, Qu WM. Roles of adenosine and its receptors in sleep-wake regulation. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 119:349-71. [PMID: 25175972 DOI: 10.1016/b978-0-12-801022-8.00014-3] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
This chapter summarizes the current knowledge about the role of adenosine in the sleep-wake regulation with a focus on adenosine in the brain, regulation of adenosine levels, adenosine receptors, and manipulations of the adenosine system by the use of pharmacological and molecular biological tools. Adenosine is neither stored nor released as a classical neurotransmitter and is thought to be formed inside cells or on their surface, mostly by breakdown of adenine nucleotides. The extracellular level of adenosine increases in the cortex and basal forebrain (BF) during prolonged wakefulness and decreases during the sleep-recovery period. Therefore, adenosine is proposed to act as a homeostatic regulator of sleep. The endogenous somnogen prostaglandin (PG) D2 increases the extracellular level of adenosine under the subarachnoid space of the BF and promotes physiological sleep. There are four adenosine receptor subtypes: adenosine A1 receptor (R, A1R), A2AR, A2BR, and A3R. Both the A1R and the A2AR have been reported to be involved in sleep induction. The A2AR plays an important role in the somnogenic effects of PGD2. Activation of A2AR by its agonist infused into the brain potently increases sleep and the arousal effect of caffeine, an A1R and A2AR antagonist, was shown to be dependent on the A2AR. On the other hand, inhibition of wake-promoting neurons via the A1R also mediates the sleep-inducing effects of adenosine, whereas activation of A1R in the lateral preoptic area induces wakefulness. These findings indicate that A2AR plays a predominant role in sleep induction, whereas A1R regulates the sleep-wake cycle in a site-dependent manner.
Collapse
Affiliation(s)
- Zhi-Li Huang
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology, Institute of Brain Science, Shanghai Medical College of Fudan University, Shanghai, China.
| | - Ze Zhang
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology, Institute of Brain Science, Shanghai Medical College of Fudan University, Shanghai, China
| | - Wei-Min Qu
- Department of Pharmacology, State Key Laboratory of Medical Neurobiology, Institute of Brain Science, Shanghai Medical College of Fudan University, Shanghai, China.
| |
Collapse
|
45
|
Nieves A, Garza LA. Does prostaglandin D2 hold the cure to male pattern baldness? Exp Dermatol 2015; 23:224-7. [PMID: 24521203 DOI: 10.1111/exd.12348] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2014] [Indexed: 12/17/2022]
Abstract
Lipids in the skin are the most diverse in the entire human body. Their bioactivity in health and disease is underexplored. Prostaglandin D2 has recently been identified as a factor which is elevated in the bald scalp of men with androgenetic alopecia (AGA) and has the capacity to decrease hair lengthening. An enzyme which synthesizes it, prostaglandin D2 synthase (PTGDS or lipocalin-PGDS), is hormone responsive in multiple other organs. PGD2 has two known receptors, GPR44 and PTGDR. GPR44 was found to be necessary for the decrease in hair growth by PGD2 . This creates an exciting opportunity to perhaps create novel treatments for AGA, which inhibit the activity of PTGDS, PGD2 or GPR44. This review discusses the current knowledge surrounding PGD2 , and future steps needed to translate these findings into novel therapies for patients with AGA.
Collapse
Affiliation(s)
- Ashley Nieves
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
46
|
Mazari AMA, Hegazy UM, Mannervik B. Identification of new inhibitors for human hematopoietic prostaglandin D2 synthase among FDA-approved drugs and other compounds. Chem Biol Interact 2015; 229:91-9. [PMID: 25603235 DOI: 10.1016/j.cbi.2015.01.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 11/22/2014] [Accepted: 01/08/2015] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Hematopoietic prostaglandin D2 synthase (HPGDS) is a member of the Sigma class glutathione transferases (GSTs) catalyzing the isomerization of prostaglandin H2 to prostaglandin D2, a mediator of allergy and inflammation responses. Selective inhibitors of human HPGDS are expected to be of therapeutic importance in relieving symptoms related to allergy and asthma. Hence, a collection of diverse FDA-approved compounds was screened for potential novel applications as inhibitors of HPGDS. METHODS The catalytic activity of purified HPGDS was used for inhibition studies in vitro. RESULTS Our inhibition studies revealed 23 compounds as effective inhibitors of HPGDS with IC50 values in the low micromolar range. Erythrosine sodium, suramin, tannic acid and sanguinarine sulfate were characterized with IC50 values of 0.2, 0.3, 0.4, and 0.6 μM, respectively. Kinetic inhibition analysis showed that erythrosine sodium is a nonlinear competitive inhibitor of HPGDS, while suramin, tannic acid and sanguinarine sulfate are linear competitive inhibitors. CONCLUSION The results show that certain FDA-approved compounds may have pharmacological effects not previously realized that warrant further consideration in their clinical use.
Collapse
Affiliation(s)
- Aslam M A Mazari
- Department of Neurochemistry, Stockholm University, SE-10691 Stockholm, Sweden
| | - Usama M Hegazy
- Molecular Biology Department, Genetic Engineering and Biotechnology Division, National Research Centre, Dokki, 12311 Cairo, Egypt
| | - Bengt Mannervik
- Department of Neurochemistry, Stockholm University, SE-10691 Stockholm, Sweden.
| |
Collapse
|
47
|
Soliman E, Henderson KL, Danell AS, Van Dross R. Arachidonoyl-ethanolamide activates endoplasmic reticulum stress-apoptosis in tumorigenic keratinocytes: Role of cyclooxygenase-2 and novel J-series prostamides. Mol Carcinog 2015; 55:117-30. [PMID: 25557612 DOI: 10.1002/mc.22257] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 11/03/2015] [Accepted: 11/05/2015] [Indexed: 11/12/2022]
Abstract
Non-melanoma skin cancer and other epithelial tumors overexpress cyclooxygenase-2 (COX-2), differentiating them from normal cells. COX-2 metabolizes arachidonic acid to prostaglandins including, the J-series prostaglandins, which induce apoptosis by mechanisms including endoplasmic reticulum (ER) stress. Arachidonoyl-ethanolamide (AEA) is a cannabinoid that causes apoptosis in diverse tumor types. Previous studies from our group demonstrated that AEA was metabolized by COX-2 to J-series prostaglandins. Thus, the current study examines the role of COX-2, J-series prostaglandins, and ER stress in AEA-induced apoptosis. In tumorigenic keratinocytes that overexpress COX-2, AEA activated the PKR-like ER kinase (PERK), inositol requiring kinase-1 (IRE1), and activating transcription factor-6 (ATF6) ER stress pathways and the ER stress apoptosis-associated proteins, C/EBP homologous protein-10 (CHOP10), caspase-12, and caspase-3. Using an ER stress inhibitor, it was determined that ER stress was required for AEA-induced apoptosis. To evaluate the role of COX-2 in ER stress-apoptosis, HaCaT keratinocytes with low endogenous COX-2 expression were transfected with COX-2 cDNA or an empty vector and AEA-induced ER stress-apoptosis occurred only in the presence of COX-2. Moreover, LC-MS analysis showed that the novel prostaglandins, 15-deoxyΔ(12,14) PGJ2 -EA and Δ(12) PGJ2 /PGJ2-EA, were synthesized from AEA. These findings suggest that AEA will be selectively toxic in tumor cells that overexpress COX-2 due to the metabolism of AEA by COX-2 to J-series prostaglandin-ethanolamides (prostamides). Hence, AEA may be an ideal topical agent for the elimination of malignancies that overexpress COX-2.
Collapse
Affiliation(s)
- Eman Soliman
- Brody School of Medicine, Pharmacology and Toxicology, East Carolina University, Greenville, NC
| | - Kate L Henderson
- Department of Chemistry, East Carolina University, Greenville, NC
| | - Allison S Danell
- Department of Chemistry, East Carolina University, Greenville, NC
| | - Rukiyah Van Dross
- Brody School of Medicine, Pharmacology and Toxicology, East Carolina University, Greenville, NC
| |
Collapse
|
48
|
Rossitto M, Ujjan S, Poulat F, Boizet-Bonhoure B. Multiple roles of the prostaglandin D2 signaling pathway in reproduction. Reproduction 2015; 149:R49-58. [DOI: 10.1530/rep-14-0381] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Prostaglandins signaling molecules are involved in numerous physiological processes. They are produced by several enzyme-limited reactions upon fatty acids, which are catalyzed by two cyclooxygenases and prostaglandin synthases. In particular, the prostaglandins E2(PGE2), D2(PGD2), and F2(PGF2α) have been shown to be involved in female reproductive mechanisms. Furthermore, widespread expression of lipocalin- and hematopoietic-PGD2synthases in the male reproductive tract supports the purported roles of PGD2in the development of both embryonic and adult testes, sperm maturation, and spermatogenesis. In this review, we summarize the putative roles of PGD2signaling and the roles of both PGD2synthases in testicular formation and function. We review the data reporting the involvement of PGD2signaling in the differentiation of Sertoli and germ cells of the embryonic testis. Furthermore, we discuss the roles of lipocalin-PGD2synthase in steroidogenesis and spermatogenesis, in terms of lipid molecule transport and PGD2production. Finally, we discuss the hypothesis that PGD2signaling may be affected in certain reproductive diseases, such as infertility, cryptorchidism, and testicular cancer.
Collapse
|
49
|
Prostaglandin D2 synthase/GPR44: a signaling axis in PNS myelination. Nat Neurosci 2014; 17:1682-92. [PMID: 25362470 DOI: 10.1038/nn.3857] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 10/07/2014] [Indexed: 12/18/2022]
Abstract
Neuregulin 1 type III is processed following regulated intramembrane proteolysis, which allows communication from the plasma membrane to the nucleus. We found that the intracellular domain of neuregulin 1 type III upregulated the prostaglandin D2 synthase (L-pgds, also known as Ptgds) gene, which, together with the G protein-coupled receptor Gpr44, forms a previously unknown pathway in PNS myelination. Neuronal L-PGDS is secreted and produces the PGD2 prostanoid, a ligand of Gpr44. We found that mice lacking L-PGDS were hypomyelinated. Consistent with this, specific inhibition of L-PGDS activity impaired in vitro myelination and caused myelin damage. Furthermore, in vivo ablation and in vitro knockdown of glial Gpr44 impaired myelination. Finally, we identified Nfatc4, a key transcription factor for myelination, as one of the downstream effectors of PGD2 activity in Schwann cells. Thus, L-PGDS and Gpr44 are previously unknown components of an axo-glial interaction that controls PNS myelination and possibly myelin maintenance.
Collapse
|
50
|
Moniot B, Ujjan S, Champagne J, Hirai H, Aritake K, Nagata K, Dubois E, Nidelet S, Nakamura M, Urade Y, Poulat F, Boizet-Bonhoure B. Prostaglandin D2 acts through the Dp2 receptor to influence male germ cell differentiation in the foetal mouse testis. Development 2014; 141:3561-71. [DOI: 10.1242/dev.103408] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Through intercellular signalling, the somatic compartment of the foetal testis is able to program primordial germ cells to undergo spermatogenesis. Fibroblast growth factor 9 and several members of the transforming growth factor β superfamily are involved in this process in the foetal testis, counteracting the induction of meiosis by retinoic acid and activating germinal mitotic arrest. Here, using in vitro and in vivo approaches, we show that prostaglandin D2 (PGD2), which is produced through both L-Pgds and H-Pgds enzymatic activities in the somatic and germ cell compartments of the foetal testis, plays a role in mitotic arrest in male germ cells by activating the expression and nuclear localization of the CDK inhibitor p21Cip1 and by repressing pluripotency markers. We show that PGD2 acts through its Dp2 receptor, at least in part through direct effects in germ cells, and contributes to the proper differentiation of male germ cells through the upregulation of the master gene Nanos2. Our data identify PGD2 signalling as an early pathway that acts in both paracrine and autocrine manners, and contributes to the differentiation of germ cells in the foetal testis.
Collapse
Affiliation(s)
- Brigitte Moniot
- Genetic and Development department, Institute of Human Genetics, CNRS UPR1142, Montpellier 34094, Cedex 05, France
| | - Safdar Ujjan
- Genetic and Development department, Institute of Human Genetics, CNRS UPR1142, Montpellier 34094, Cedex 05, France
| | - Julien Champagne
- Genetic and Development department, Institute of Human Genetics, CNRS UPR1142, Montpellier 34094, Cedex 05, France
| | - Hiroyuki Hirai
- Department of Advanced Technology and Development, BML, Matoba, Kawagoe, Saitama 350-1101, Japan
| | - Kosuke Aritake
- Department of Molecular Behavioral Biology, Osaka Bioscience Institute, Osaka 565-0874, Japan
| | - Kinya Nagata
- Department of Advanced Technology and Development, BML, Matoba, Kawagoe, Saitama 350-1101, Japan
| | - Emeric Dubois
- Plateforme MGX, Functional Genomic Institute, CNRS UMR 5203 – INSERM U 661, Montpellier 34094, Cedex 05, France
| | - Sabine Nidelet
- Plateforme MGX, Functional Genomic Institute, CNRS UMR 5203 – INSERM U 661, Montpellier 34094, Cedex 05, France
| | - Masataka Nakamura
- Human Gene Sciences Center, Tokyo Medical and Dental University, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yoshihiro Urade
- Department of Molecular Behavioral Biology, Osaka Bioscience Institute, Osaka 565-0874, Japan
| | - Francis Poulat
- Genetic and Development department, Institute of Human Genetics, CNRS UPR1142, Montpellier 34094, Cedex 05, France
| | - Brigitte Boizet-Bonhoure
- Genetic and Development department, Institute of Human Genetics, CNRS UPR1142, Montpellier 34094, Cedex 05, France
| |
Collapse
|