1
|
Sánchez-Marín L, Jiménez-Castilla V, Flores-López M, Navarro JA, Gavito A, Blanco-Calvo E, Santín LJ, Pavón-Morón FJ, Rodríguez de Fonseca F, Serrano A. Sex-specific alterations in emotional behavior and neurotransmitter systems in LPA 1 receptor-deficient mice. Neuropharmacology 2025; 268:110325. [PMID: 39864586 DOI: 10.1016/j.neuropharm.2025.110325] [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: 10/31/2024] [Revised: 01/09/2025] [Accepted: 01/23/2025] [Indexed: 01/28/2025]
Abstract
Lysophosphatidic acid (LPA) and the endocannabinoid system (ECS) are critical lipid signaling pathways involved in emotional regulation and behavior. Despite their interconnected roles and shared metabolic pathways, the specific contributions of LPA signaling through the LPA1 receptor to stress-related disorders remain poorly understood. This study investigates the effects of LPA1 receptor deficiency on emotional behavior and neurotransmitter-related gene expression, with a focus on sex-specific differences, using maLPA1-null mice of both sexes. We hypothesized LPA1 receptor loss disrupts the interplay between LPA and the endocannabinoid 2-arachidonoylglycerol (2-AG) signaling, resulting in distinct behavioral and molecular alterations. maLPA1-null mice exhibited increased anxiety-like behaviors and altered stress-coping responses compared to wild-type counterparts, with more pronounced effects observed in females. Female mice also displayed higher corticosterone levels, though no genotype-related differences were observed. Plasma analyses revealed elevated LPA levels in maLPA1-null mice, suggesting a compensatory mechanism, and reduced 2-AG levels, indicating impaired ECS signaling. Gene expression profiling in the amygdala and medial prefrontal cortex showed significant alterations in the gene expression of key components of LPA and 2-AG signaling pathways, as well as neuropeptide systems such as corticotropin-releasing hormone (CRH) and neuropeptide Y (NPY). Glutamatergic signaling components also exhibited sex-specific variations. These findings suggest that LPA1 receptor deficiency impacts behavioral response and disrupts sex-specific neurotransmitter signaling, emphasizing the importance of LPA-ECS crosstalk in emotional regulation. This study provides insights into the molecular mechanisms underlying stress-related disorders such as depression and anxiety, which may inform the development of sex-specific therapeutic approaches.
Collapse
Affiliation(s)
- Laura Sánchez-Marín
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), 29590, Málaga, Spain; Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010, Málaga, Spain
| | - Violeta Jiménez-Castilla
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), 29590, Málaga, Spain; Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010, Málaga, Spain
| | - María Flores-López
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), 29590, Málaga, Spain; Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010, Málaga, Spain
| | - Juan A Navarro
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), 29590, Málaga, Spain; Servicio de Neurología, Hospital Regional Universitario de Málaga, 29010, Málaga, Spain
| | - Ana Gavito
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), 29590, Málaga, Spain; Servicio de Neurología, Hospital Regional Universitario de Málaga, 29010, Málaga, Spain
| | - Eduardo Blanco-Calvo
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Universidad de Málaga, 29010, Málaga, Spain
| | - Luis J Santín
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Universidad de Málaga, 29010, Málaga, Spain
| | - Francisco J Pavón-Morón
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), 29590, Málaga, Spain; Área del Corazón, Hospital Universitario Virgen de la Victoria de Málaga, 29010, Málaga, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Fernando Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), 29590, Málaga, Spain; Servicio de Neurología, Hospital Regional Universitario de Málaga, 29010, Málaga, Spain; Andalusian Network for Clinical and Translational Research in Neurology (NEURO-RECA), 29001, Malaga, Spain.
| | - Antonia Serrano
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), 29590, Málaga, Spain; Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010, Málaga, Spain.
| |
Collapse
|
2
|
Xiao Y, Liu C, Wang X, Li H, Wang L, Gou K, Liu X, Guan X, Zhou X, He X, Zhao Y, Tao L, Pan X, Jiang L, Chen Y, Liu H, Dai Y, Bu Q, Qin M, Zhu R, Chen B, Flores AD, Zhao Y, Cen X. Dysregulated glycerophospholipid metabolism in amygdala may mediate favipiravir-induced anxiety-like behaviors in mice. Front Pharmacol 2025; 16:1491150. [PMID: 40103591 PMCID: PMC11913839 DOI: 10.3389/fphar.2025.1491150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 01/20/2025] [Indexed: 03/20/2025] Open
Abstract
Favipiravir, the first RNA polymerase inhibitor approved to treat resistant influenza, has been reported to be associated with central nervous system (CNS) side effects, particularly anxiety-like behavior; nevertheless, the underlying mechanism remains largely unknown. In this study, we investigated the effect of favipiravir on the neurobehavior of mice, and combined lipidomics and transcriptomics analysis to explore the mechanism underlying this effect. In behavioral tests, the mice displayed anxiety-like behaviors after oral favipiravir administration (200 mg/kg) for 7 days continuously. By lipidomics analysis, we observed that favipiravir induced a dysregulation of glycerophospholipid metabolism in the amygdala. Moreover, favipiravir significantly reduced the mRNA level of glycerol-3-phosphate acyltransferase 2 (Gpat2), the rate-limiting enzyme of glycerophospholipid synthesis. Notably, favipiravir markedly reduced the levels of docosahexaenoic acid-enriched phosphatidylethanolamine or phosphatidylcholine (DHA-PE/PC) and arachidonic acid-enriched phosphatidylethanolamine or phosphatidylcholine (AA-PE/PC), two components of glycerophospholipids, in the amygdala. The increased expression of phospholipase A2 (Pla2) may attribute to the enhanced release of arachidonic acid (AA) from AA-PE/PC. Furthermore, favipiravir altered neurite morphology and reduced neurophysiological activity in amygdala neurons in vitro. Collectively, dysregulated glycerophospholipid metabolism in the amygdala may contribute to the adverse effect of favipiravir.
Collapse
Affiliation(s)
- Yuzhou Xiao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chunqi Liu
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaojie Wang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hongchun Li
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Liang Wang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Kun Gou
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xingchen Liu
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Xinqi Guan
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xia Zhou
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiumei He
- School of Life Sciences, Guangxi Normal University, Guilin, China
| | - Yue Zhao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Tao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaodan Pan
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Linhong Jiang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yaxing Chen
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Huan Liu
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yanping Dai
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Bu
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Meng Qin
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ruiming Zhu
- Chengdu Westchina Frontier Pharmatech, Co., Ltd., Chengdu, China
| | - Bo Chen
- Chengdu Westchina Frontier Pharmatech, Co., Ltd., Chengdu, China
| | - Angelo D Flores
- Department of Neuroscience, City University of Hong Kong, Kowloon, China
| | - Yinglan Zhao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaobo Cen
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Li F, Xie L, Xiao Q, Li J, Zhong H, Xu X, Tu J, Luo Q. Benzo[a]pyrene exposure induces anxiety-like behaviors in the mice through brain metabolic alterations. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 954:176215. [PMID: 39276998 DOI: 10.1016/j.scitotenv.2024.176215] [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: 07/17/2024] [Revised: 09/07/2024] [Accepted: 09/09/2024] [Indexed: 09/17/2024]
Abstract
The deleterious health impacts of polycyclic aromatic hydrocarbons (PAHs) on the population have been extensively substantiated and acknowledged. Mounting evidence underscores that PAH exposure is closely linked to an elevated risk of mental disorders, particularly in populations experiencing occupational and high-level exposure. In this study, we aimed to investigate the mechanisms underlying anxiety-like behaviors induced by different dosages of PAHs, with a concentrated focus on brain region-specific metabolic alterations in mice using various metabolomics approaches. Male C57BL/6 mice were exposed to benzo[a]pyrene (B[a]P), a typical PAH, through gavage at occupational exposure and EPA toxicologically relevant dosages (2.0 and 20.0 mg/kg/day) for 21 days, respectively. Behavioral assessments revealed that occupational exposure to B[a]P induced anxiety-like behaviors in C57BL/6 mice. Meanwhile, elevated serum norepinephrine and corticotropin-releasing hormone further confirmed the anxiety-inducing effects of B[a]P exposure. Metabolomics analysis uncovered dysregulation across various metabolic pathways following B[a]P exposure, encompassing brain neurotransmitter, organic acid, amino acid, lipid, fatty acid, and cholesterol. Anxiety levels and lipid metabolic abnormalities were notably exacerbated at the higher dosage, despite being only a 10-fold increase. Of particular significance, a decrease in lysophosphatidic acid (LPA) and lysophosphatidylserine (LPS) emerged as pivotal indicators of B[a]P neurotoxicity. Spatial-resolved metabolomics further demonstrated distinctive lipid and metabolite profiles across different brain subregions after exposure to B[a]P. Remarkably, alterations were specifically observed in the anxiety-related brain regions, such as the hippocampus, cortex, white matter, and thalamus, varying with exposure dosages. These findings underscore the significance of brain metabolic abnormalities in the development of mental disorders triggered by B[a]P exposure and highlight the need for establishing precise exposure limits of B[a]P to safeguard public mental health.
Collapse
Affiliation(s)
- Fang Li
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Li Xie
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen 518055, China; Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518067, China
| | - Qian Xiao
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen 518055, China
| | - Jingguang Li
- The Key Laboratory of Food Safety Risk Assessment, Ministry of Health, China National Center of Food Safety and Risk Assessment, Beijing 100021, China
| | - Huifang Zhong
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xirong Xu
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Tu
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China; CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Qian Luo
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
4
|
Li N, Li Y. Lysophosphatidic Acid (LPA) and Its Receptors in Mood Regulation: A Systematic Review of the Molecular Mechanisms and Therapeutic Potential. Int J Mol Sci 2024; 25:7440. [PMID: 39000547 PMCID: PMC11242315 DOI: 10.3390/ijms25137440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
Mood disorders affect over 300 million individuals worldwide, often characterized by their chronic and refractory nature, posing significant threats to patient life. There has been a notable increase in mood disorders among American adolescents and young adults, with a rising number of suicide attempts and fatalities, highlighting a growing association between mood disorders and suicidal outcomes. Dysregulation within the neuroimmune-endocrine system is now recognized as one of the fundamental biological mechanisms underlying mood and mood disorders. Lysophosphatidic acid (LPA), a novel mediator of mood behavior, induces anxiety-like and depression-like phenotypes through its receptors LPA1 and LPA5, regulating synaptic neurotransmission and plasticity. Consequently, LPA has garnered substantial interest in the study of mood regulation. This study aimed to elucidate the molecular mechanisms of lysophosphatidic acid and its receptors, along with LPA receptor ligands, in mood regulation and to explore their potential therapeutic efficacy in treating mood disorders. A comprehensive literature search was conducted using the PubMed and Web of Science databases, identifying 208 articles through keyword searches up to June 2024. After excluding duplicates, irrelevant publications, and those restricted by open access limitations, 21 scientific papers were included in this review. The findings indicate that LPA/LPA receptor modulation could be beneficial in treating mood disorders, suggesting that pharmacological agents or gintonin, an extract from ginseng, may serve as effective therapeutic strategies. This study opens new avenues for future research into how lysophosphatidic acid and its receptors, as well as lysophosphatidic acid receptor ligands, influence emotional behavior in animals and humans.
Collapse
Affiliation(s)
- Nan Li
- School of Competitive Sports, Beijing Sport University, Beijing 100084, China
| | - Yanchun Li
- China Institute of Sports and Health Science, Beijing Sport University, Beijing 100084, China
- Beijing Key Laboratory of Sports Performance and Skill Assessment, Beijing 100084, China
- Key Laboratory for Performance Training & Recovery of General Administration of Sport, Beijing 100084, China
| |
Collapse
|
5
|
Requena-Ocaña N, Flores-López M, García-Marchena N, Pavón-Morón FJ, Pedraza C, Wallace A, Castilla-Ortega E, Rodríguez de Fonseca F, Serrano A, Araos P. Plasma Lysophosphatidic Acid Concentrations in Sex Differences and Psychiatric Comorbidity in Patients with Cocaine Use Disorder. Int J Mol Sci 2023; 24:15586. [PMID: 37958570 PMCID: PMC10649657 DOI: 10.3390/ijms242115586] [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: 09/25/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
We have recently reported sex differences in the plasma concentrations of lysophosphatidic acid (LPA) and alterations in LPA species in patients with alcohol and cocaine use disorders. Preclinical evidence suggests a main role of lysophosphatidic acid (LPA) signaling in anxiogenic responses and drug addiction. To further explore the potential role of the LPA signaling system in sex differences and psychiatric comorbidity in cocaine use disorder (CUD), we conducted a cross-sectional study with 88 patients diagnosed with CUD in outpatient treatment and 60 healthy controls. Plasma concentrations of total LPA and LPA species (16:0, 18:0, 18:1, 18:2 and 20:4) were quantified and correlated with cortisol and tryptophan metabolites [tryptophan (TRP), serotonin (5-HT), kynurenine (KYN), quinolinic acid (QUIN) and kynurenic acid (KYNA)]. We found sexual dimorphism for the total LPA and most LPA species in the control and CUD groups. The total LPA and LPA species were not altered in CUD patients compared to the controls. There was a significant correlation between 18:2 LPA and age at CUD diagnosis (years) in the total sample, but total LPA, 16:0 LPA and 18:2 LPA correlated with age at onset of CUD in male patients. Women with CUD had more comorbid anxiety and eating disorders, whereas men had more cannabis use disorders. Total LPA, 18:0 LPA and 20:4 LPA were significantly decreased in CUD patients with anxiety disorders. Both 20:4 LPA and total LPA were significantly higher in women without anxiety disorders compared to men with and without anxiety disorders. Total LPA and 16:0 LPA were significantly decreased in CUD patients with childhood ADHD. Both 18:1 LPA and 20:4 LPA were significantly augmented in CUD patients with personality disorders. KYNA significantly correlated with total LPA, 16:0 LPA and 18:2 LPA species, while TRP correlated with the 18:1 LPA species. Our results demonstrate that LPA signaling is affected by sex and psychiatric comorbidity in CUD patients, playing an essential role in mediating their anxiety symptoms.
Collapse
Affiliation(s)
- Nerea Requena-Ocaña
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), 29590 Málaga, Spain; (N.R.-O.); (M.F.-L.); (F.J.P.-M.); (C.P.); (P.A.)
- Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain;
| | - María Flores-López
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), 29590 Málaga, Spain; (N.R.-O.); (M.F.-L.); (F.J.P.-M.); (C.P.); (P.A.)
- Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain;
| | - Nuria García-Marchena
- Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain;
- Departamento de Psicobiología y Metodología en Ciencias del Comportamiento, Facultad de Psicología, Universidad Complutense de Madrid, 28223 Madrid, Spain
| | - Francisco J. Pavón-Morón
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), 29590 Málaga, Spain; (N.R.-O.); (M.F.-L.); (F.J.P.-M.); (C.P.); (P.A.)
- Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain;
- Unidad de Gestión Clínica del Corazón, Hospital Universitario Virgen de la Victoria de Málaga, 29010 Málaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Carmen Pedraza
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), 29590 Málaga, Spain; (N.R.-O.); (M.F.-L.); (F.J.P.-M.); (C.P.); (P.A.)
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Universidad de Málaga, 29010 Málaga, Spain; (A.W.); (E.C.-O.)
| | - Agustín Wallace
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Universidad de Málaga, 29010 Málaga, Spain; (A.W.); (E.C.-O.)
| | - Estela Castilla-Ortega
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Universidad de Málaga, 29010 Málaga, Spain; (A.W.); (E.C.-O.)
| | - Fernando Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), 29590 Málaga, Spain; (N.R.-O.); (M.F.-L.); (F.J.P.-M.); (C.P.); (P.A.)
- Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain;
| | - Antonia Serrano
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), 29590 Málaga, Spain; (N.R.-O.); (M.F.-L.); (F.J.P.-M.); (C.P.); (P.A.)
- Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain;
| | - Pedro Araos
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), 29590 Málaga, Spain; (N.R.-O.); (M.F.-L.); (F.J.P.-M.); (C.P.); (P.A.)
- Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain;
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Facultad de Psicología, Universidad de Málaga, 29010 Málaga, Spain; (A.W.); (E.C.-O.)
| |
Collapse
|
6
|
Moreno-Fernández RD, Sampedro-Piquero P, Gómez-Salas FJ, Nieto-Quero A, Estivill-Torrús G, Rodríguez de Fonseca F, Santín LJ, Pedraza C. Social avoidance and altered hypothalamic-pituitary-adrenal axis in a mouse model of anxious depression: The role of LPA 1 receptor. Behav Brain Res 2023; 455:114681. [PMID: 37741054 DOI: 10.1016/j.bbr.2023.114681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/03/2023] [Accepted: 09/20/2023] [Indexed: 09/25/2023]
Abstract
Anxious depression is a prevalent disease with devastating consequences. Despite the lack of knowledge about the neurobiological basis of this subtype of depression, recently our group has identified a relationship between the LPA1 receptor, one of the six characterized G protein-coupled receptors (LPA1-6) for lysophosphatidic acid, with a mixed depressive-anxiety phenotype. Dysfunctional social behaviors, which have been related to increased activation of the hypothalamus-pituitary-adrenal (HPA) axis, are key symptoms of depression and are even more prominent in patients with comorbid anxiety and depressive disorders. Social behavior and HPA functioning were assessed in animals lacking the LPA1 receptor. For these purposes, we first examined social behaviors in wild-type and LPA1 receptor-null mice. In addition, a dexamethasone (DEX) suppression test was carried out. maLPA1-null mice exhibited social avoidance, a blunted response to DEX administration and an impaired circadian rhythm of corticosterone levels, which are features that are consistently dysregulated in many mental illnesses including anxious depression. Here, we have strengthened the previous experimental evidence for maLPA1-null mice to represent a good animal model of anxious depression, providing an opportunity to explore new therapeutic targets for the treatment of mood disorders, particularly this subtype of depression.
Collapse
Affiliation(s)
| | - P Sampedro-Piquero
- Departamento de Psicología Biológica y de la Salud. Facultad de Psicología. Universidad Autónoma de Madrid. Madrid, Spain
| | - F J Gómez-Salas
- Departamento de Psicobiologia y Metodologia en las CC, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Malaga, Malaga, Spain
| | - A Nieto-Quero
- Departamento de Psicobiologia y Metodologia en las CC, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Malaga, Malaga, Spain; Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Malaga, Spain
| | - G Estivill-Torrús
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Malaga, Spain
| | - F Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Malaga, Spain; Unidad Clínica de Neurociencias, Hospital Regional Universitario de Málaga, Spain
| | - L J Santín
- Departamento de Psicobiologia y Metodologia en las CC, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Malaga, Malaga, Spain; Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Malaga, Spain
| | - C Pedraza
- Departamento de Psicobiologia y Metodologia en las CC, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Malaga, Malaga, Spain; Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Malaga, Spain.
| |
Collapse
|
7
|
Ren Z, Hou J, Li W, Tang Y, Wang M, Ding R, Liu S, Fu Y, Mai Y, Xia J, Zuo W, Zhou LH, Ye JH, Fu R. LPA1 receptors in the lateral habenula regulate negative affective states associated with alcohol withdrawal. Neuropsychopharmacology 2023; 48:1567-1578. [PMID: 37059867 PMCID: PMC10516930 DOI: 10.1038/s41386-023-01582-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/17/2023] [Accepted: 03/30/2023] [Indexed: 04/16/2023]
Abstract
The role of lysophosphatidic acid (LPA) signaling in psychiatric disorders and drug abuse is significant. LPA receptors are widely expressed in the central nervous system, including the lateral habenula (LHb). Recent studies suggest that LHb is involved in a negative emotional state during alcohol withdrawal, which can lead to relapse. The current study examines the role of LHb LPA signaling in the negative affective state associated with alcohol withdrawal. Adult male Long-Evans rats were trained to consume either alcohol or water for eight weeks. At 48 h of withdrawal, alcohol-drinking rats showed anxiety- and depression-like symptoms, along with a significant increase in LPA signaling and related neuronal activation molecules, including autotaxin (ATX, Enpp2), LPA receptor 1/3 (LPA1/3), βCaMKII, and c-Fos. However, there was a decrease in lipid phosphate phosphatase-related protein type 4 (LPPR4) in the LHb. Intra-LHb infusion of the LPA1/3 receptor antagonist ki-16425 or PKC-γ inhibitor Go-6983 reduced the abnormal behaviors and elevated relapse-like ethanol drinking. It also normalized high LPA1/3 receptors and enhanced AMPA GluA1 phosphorylation in Ser831 and GluA1/GluA2 ratio. Conversely, selective activation of LPA1/3 receptors by intra-LHb infusion of 18:1 LPA induced negative affective states and upregulated βCaMKII-AMPA receptor phosphorylation in Naive rats, which were reversed by pretreatment with intra-LHb Go-6983. Our findings suggest that disturbances in LPA signaling contribute to adverse affective disorders during alcohol withdrawal, likely through PKC-γ/βCaMKII-linked glutamate signaling. Targeting LPA may therefore be beneficial for individuals suffering from alcohol use disorders.
Collapse
Affiliation(s)
- Zhiheng Ren
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-sen University, Shenzhen, Guangdong, 518106, China
| | - Jiawei Hou
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-sen University, Shenzhen, Guangdong, 518106, China
| | - Wenfu Li
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-sen University, Shenzhen, Guangdong, 518106, China
| | - Ying Tang
- Department of Biology, School of Life Science, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
- Basic and Clinical Medicine Teaching Laboratory, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518100, China
| | - Molin Wang
- Basic and Clinical Medicine Teaching Laboratory, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518100, China
| | - Ruxuan Ding
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-sen University, Shenzhen, Guangdong, 518106, China
| | - Songlin Liu
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-sen University, Shenzhen, Guangdong, 518106, China
| | - Yixin Fu
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-sen University, Shenzhen, Guangdong, 518106, China
| | - Yunlin Mai
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-sen University, Shenzhen, Guangdong, 518106, China
| | - Jianxun Xia
- Department of Basic Medical Sciences, Yunkang School of Medicine and Health, Nanfang College, Guangzhou, Guangdong, 510970, China
| | - Wanhong Zuo
- Department of Anesthesiology, Pharmacology, Physiology & Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, 07103, USA
| | - Li-Hua Zhou
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-sen University, Shenzhen, Guangdong, 518106, China
| | - Jiang-Hong Ye
- Department of Anesthesiology, Pharmacology, Physiology & Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ, 07103, USA.
| | - Rao Fu
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-sen University, Shenzhen, Guangdong, 518106, China.
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, 518106, China.
| |
Collapse
|
8
|
Nieto-Quero A, Infantes-López MI, Zambrana-Infantes E, Chaves-Peña P, Gavito AL, Munoz-Martin J, Tabbai S, Márquez J, Rodríguez de Fonseca F, García-Fernández MI, Santín LJ, Pedraza C, Pérez-Martín M. Unveiling the Secrets of the Stressed Hippocampus: Exploring Proteomic Changes and Neurobiology of Posttraumatic Stress Disorder. Cells 2023; 12:2290. [PMID: 37759512 PMCID: PMC10527244 DOI: 10.3390/cells12182290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/28/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Intense stress, especially traumatic stress, can trigger disabling responses and in some cases even lead to the development of posttraumatic stress disorder (PTSD). PTSD is heterogeneous, accompanied by a range of distress symptoms and treatment-resistant disorders that may be associated with a number of other psychopathologies. PTSD is a very heterogeneous disorder with different subtypes that depend on, among other factors, the type of stressor that provokes it. However, the neurobiological mechanisms are poorly understood. The study of early stress responses may hint at the way PTSD develops and improve the understanding of the neurobiological mechanisms involved in its onset, opening the opportunity for possible preventive treatments. Proteomics is a promising strategy for characterizing these early mechanisms underlying the development of PTSD. The aim of the work was to understand how exposure to acute and intense stress using water immersion restraint stress (WIRS), which could be reminiscent of natural disaster, may induce several PTSD-associated symptoms and changes in the hippocampal proteomic profile. The results showed that exposure to WIRS induced behavioural symptoms and corticosterone levels reminiscent of PTSD. Moreover, the expression profiles of hippocampal proteins at 1 h and 24 h after stress were deregulated in favour of increased inflammation and reduced neuroplasticity, which was validated by histological studies and cytokine determination. Taken together, these results suggest that neuroplastic and inflammatory dysregulation may be a therapeutic target for the treatment of post-traumatic stress disorders.
Collapse
Affiliation(s)
- Andrea Nieto-Quero
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, 29010 Malaga, Spain; (A.N.-Q.); (E.Z.-I.); (S.T.); (L.J.S.)
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma Bionand, 29590 Malaga, Spain; (M.I.I.-L.); (A.L.G.); (J.M.); (F.R.d.F.); (M.I.G.-F.)
| | - María Inmaculada Infantes-López
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma Bionand, 29590 Malaga, Spain; (M.I.I.-L.); (A.L.G.); (J.M.); (F.R.d.F.); (M.I.G.-F.)
- Departamento de Biología Celular, Genética y Fisiología, Universidad de Málaga, 29010 Malaga, Spain; (P.C.-P.); (J.M.-M.)
| | - Emma Zambrana-Infantes
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, 29010 Malaga, Spain; (A.N.-Q.); (E.Z.-I.); (S.T.); (L.J.S.)
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma Bionand, 29590 Malaga, Spain; (M.I.I.-L.); (A.L.G.); (J.M.); (F.R.d.F.); (M.I.G.-F.)
| | - Patricia Chaves-Peña
- Departamento de Biología Celular, Genética y Fisiología, Universidad de Málaga, 29010 Malaga, Spain; (P.C.-P.); (J.M.-M.)
| | - Ana L. Gavito
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma Bionand, 29590 Malaga, Spain; (M.I.I.-L.); (A.L.G.); (J.M.); (F.R.d.F.); (M.I.G.-F.)
| | - Jose Munoz-Martin
- Departamento de Biología Celular, Genética y Fisiología, Universidad de Málaga, 29010 Malaga, Spain; (P.C.-P.); (J.M.-M.)
| | - Sara Tabbai
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, 29010 Malaga, Spain; (A.N.-Q.); (E.Z.-I.); (S.T.); (L.J.S.)
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma Bionand, 29590 Malaga, Spain; (M.I.I.-L.); (A.L.G.); (J.M.); (F.R.d.F.); (M.I.G.-F.)
| | - Javier Márquez
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma Bionand, 29590 Malaga, Spain; (M.I.I.-L.); (A.L.G.); (J.M.); (F.R.d.F.); (M.I.G.-F.)
- Departamento de Biología Molecular y Bioquímica, Canceromics Lab, Universidad de Málaga, 29010 Malaga, Spain
| | - Fernando Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma Bionand, 29590 Malaga, Spain; (M.I.I.-L.); (A.L.G.); (J.M.); (F.R.d.F.); (M.I.G.-F.)
| | - María Inmaculada García-Fernández
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma Bionand, 29590 Malaga, Spain; (M.I.I.-L.); (A.L.G.); (J.M.); (F.R.d.F.); (M.I.G.-F.)
- Departamento de Fisiología Humana, Histología Humana, Anatomía Patológica y Educación Física y Deportiva, Universidad de Málaga, 29010 Malaga, Spain
| | - Luis J. Santín
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, 29010 Malaga, Spain; (A.N.-Q.); (E.Z.-I.); (S.T.); (L.J.S.)
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma Bionand, 29590 Malaga, Spain; (M.I.I.-L.); (A.L.G.); (J.M.); (F.R.d.F.); (M.I.G.-F.)
| | - Carmen Pedraza
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, 29010 Malaga, Spain; (A.N.-Q.); (E.Z.-I.); (S.T.); (L.J.S.)
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma Bionand, 29590 Malaga, Spain; (M.I.I.-L.); (A.L.G.); (J.M.); (F.R.d.F.); (M.I.G.-F.)
| | - Margarita Pérez-Martín
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma Bionand, 29590 Malaga, Spain; (M.I.I.-L.); (A.L.G.); (J.M.); (F.R.d.F.); (M.I.G.-F.)
- Departamento de Biología Celular, Genética y Fisiología, Universidad de Málaga, 29010 Malaga, Spain; (P.C.-P.); (J.M.-M.)
| |
Collapse
|
9
|
Gao X, Wang Z, Du L. Glial Cells and Itch: Possible Targets for Novel Antipruritic Therapies. ACS Chem Neurosci 2023; 14:331-339. [PMID: 36655585 DOI: 10.1021/acschemneuro.2c00638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Glial cells, which are the non-neuronal cells of the nervous system, play essential roles in brain development, homeostasis, and diseases. Glial cells have attracted attention because of their active involvement in many neurological disorders. In recent years, substantial progress has been made in our understanding of the roles of glial cells in the pathogenesis of itch. Mechanistically, central and peripheral glial cells modulate acute and chronic pruritus via different mechanisms. In this review, we present the current knowledge about the involvement of glial cells in the modulation of itch processing and the mechanism of glial cell activation under itch stimuli. Targeting glial cells may provide novel approaches for itch therapy.
Collapse
Affiliation(s)
- Xinyi Gao
- School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhifei Wang
- School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lixia Du
- Department of Biochemistry, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
10
|
Neyama H, Nishiyori M, Cui Y, Watanabe Y, Ueda H. Lysophosphatidic acid receptor type-1 mediates brain activation in micro-Positron Emission Tomography analysis in a fibromyalgia-like mouse model. Eur J Neurosci 2022; 56:4224-4233. [PMID: 35666711 DOI: 10.1111/ejn.15729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 05/30/2022] [Indexed: 11/26/2022]
Abstract
The intermittent cold stress-induced generalized pain response mimics the pathophysiological and pharmacotherapeutic features reported for fibromyalgia patients, including the presence of chronic generalized pain and female dominance. In addition, the intermittent cold stress-induced generalized pain is abolished in lysophosphatidic acid receptor type-1 knockout mice, as reported in many cases of neuropathic pain models. This study aimed to identify the brain loci involved in the intermittent cold stress generalized pain response and test their dependence on the lysophosphatidic acid receptor type-1. Positron emission tomography analyses using 2-deoxy-2-[18 F]fluoro-D-glucose in the presence of a pain stimulus showed that intermittent cold stress causes a significant increase in uptake in the ipsilateral regions, including the salience networking-related anterior cingulate cortex and insular cortex and the cognition-related hippocampus. A significant decrease was observed in the default mode network-related posterior cingulate cortex. Almost these intermittent cold stress-induced changes were abolished in lysophosphatidic acid receptor type-1 knockout mice. There results suggest that the intermittent cold stress-induced generalized pain response is mediated by the lysophosphatidic acid receptor type-1 in specific brain loci related to salience networking and cognition, which may lead to further developments in the treatment of fibromyalgia.
Collapse
Affiliation(s)
- Hiroyuki Neyama
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan.,Laboratory for Biofunction Dynamics Imaging, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Michiko Nishiyori
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan
| | - Yilong Cui
- Laboratory for Biofunction Dynamics Imaging, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Yasuyoshi Watanabe
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Hiroshi Ueda
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Institute of Biomedical Sciences, Nagasaki, Japan.,Laboratory for the Study of Pain, Research Institute for Production Development, Kyoto, Japan
| |
Collapse
|
11
|
Endle H, Horta G, Stutz B, Muthuraman M, Tegeder I, Schreiber Y, Snodgrass IF, Gurke R, Liu ZW, Sestan-Pesa M, Radyushkin K, Streu N, Fan W, Baumgart J, Li Y, Kloss F, Groppa S, Opel N, Dannlowski U, Grabe HJ, Zipp F, Rácz B, Horvath TL, Nitsch R, Vogt J. AgRP neurons control feeding behaviour at cortical synapses via peripherally derived lysophospholipids. Nat Metab 2022; 4:683-692. [PMID: 35760867 PMCID: PMC9940119 DOI: 10.1038/s42255-022-00589-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 05/17/2022] [Indexed: 01/13/2023]
Abstract
Phospholipid levels are influenced by peripheral metabolism. Within the central nervous system, synaptic phospholipids regulate glutamatergic transmission and cortical excitability. Whether changes in peripheral metabolism affect brain lipid levels and cortical excitability remains unknown. Here, we show that levels of lysophosphatidic acid (LPA) species in the blood and cerebrospinal fluid are elevated after overnight fasting and lead to higher cortical excitability. LPA-related cortical excitability increases fasting-induced hyperphagia, and is decreased following inhibition of LPA synthesis. Mice expressing a human mutation (Prg-1R346T) leading to higher synaptic lipid-mediated cortical excitability display increased fasting-induced hyperphagia. Accordingly, human subjects with this mutation have higher body mass index and prevalence of type 2 diabetes. We further show that the effects of LPA following fasting are under the control of hypothalamic agouti-related peptide (AgRP) neurons. Depletion of AgRP-expressing cells in adult mice decreases fasting-induced elevation of circulating LPAs, as well as cortical excitability, while blunting hyperphagia. These findings reveal a direct influence of circulating LPAs under the control of hypothalamic AgRP neurons on cortical excitability, unmasking an alternative non-neuronal route by which the hypothalamus can exert a robust impact on the cortex and thereby affect food intake.
Collapse
Affiliation(s)
- Heiko Endle
- Department of Molecular and Translational Neuroscience of Anatomy II, University of Cologne, Cologne, Germany
- Cluster of Excellence-Cellular Stress Response in Aging-Associated Diseases, Center of Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Guilherme Horta
- Focus Program Translational Neuroscience, Johannes Gutenberg-University, Mainz, Germany
- Translational Animal Research Center, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Institute for Microscopic Anatomy and Neurobiology, Johannes Gutenberg-University, Mainz, Germany
| | - Bernardo Stutz
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Muthuraman Muthuraman
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Goethe-University, Frankfurt am Main, Germany
| | - Yannick Schreiber
- Fraunhofer Institute for Translational Medicine and Pharmacology and Fraunhofer Cluster of Excellence for Immune Mediated Diseases, Frankfurt am Main, Germany
| | - Isabel Faria Snodgrass
- Fraunhofer Institute for Translational Medicine and Pharmacology and Fraunhofer Cluster of Excellence for Immune Mediated Diseases, Frankfurt am Main, Germany
| | - Robert Gurke
- Fraunhofer Institute for Translational Medicine and Pharmacology and Fraunhofer Cluster of Excellence for Immune Mediated Diseases, Frankfurt am Main, Germany
| | - Zhong-Wu Liu
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Matija Sestan-Pesa
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Konstantin Radyushkin
- Focus Program Translational Neuroscience, Johannes Gutenberg-University, Mainz, Germany
- Translational Animal Research Center, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Nora Streu
- Focus Program Translational Neuroscience, Johannes Gutenberg-University, Mainz, Germany
| | - Wei Fan
- Focus Program Translational Neuroscience, Johannes Gutenberg-University, Mainz, Germany
| | - Jan Baumgart
- Translational Animal Research Center, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Yan Li
- Transfer Group Antiinfectives, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Florian Kloss
- Transfer Group Antiinfectives, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Sergiu Groppa
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Nils Opel
- Institute of Translational Psychiatry, Westfälische Wilhelms University, Münster, Germany
| | - Udo Dannlowski
- Institute of Translational Psychiatry, Westfälische Wilhelms University, Münster, Germany
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Bence Rácz
- Department of Anatomy and Histology, University of Veterinary Medicine, Budapest, Hungary
| | - Tamas L Horvath
- Cluster of Excellence-Cellular Stress Response in Aging-Associated Diseases, Center of Molecular Medicine Cologne, University of Cologne, Cologne, Germany.
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA.
- Department of Anatomy and Histology, University of Veterinary Medicine, Budapest, Hungary.
| | - Robert Nitsch
- Institute for Translational Neuroscience, Westfälische Wilhelms University, Münster, Germany.
| | - Johannes Vogt
- Department of Molecular and Translational Neuroscience of Anatomy II, University of Cologne, Cologne, Germany.
- Cluster of Excellence-Cellular Stress Response in Aging-Associated Diseases, Center of Molecular Medicine Cologne, University of Cologne, Cologne, Germany.
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany.
| |
Collapse
|
12
|
Hoshino Y, Okuno T, Saigusa D, Kano K, Yamamoto S, Shindou H, Aoki J, Uchida K, Yokomizo T, Ito N. Lysophosphatidic acid receptor 1/3 antagonist inhibits the activation of satellite glial cells and reduces acute nociceptive responses. FASEB J 2022; 36:e22236. [PMID: 35218596 DOI: 10.1096/fj.202101678r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/10/2022] [Accepted: 02/14/2022] [Indexed: 02/06/2023]
Abstract
Lysophosphatidic acid (LPA) exerts various biological activities through six characterized G protein-coupled receptors (LPA1-6 ). While LPA-LPA1 signaling contributes toward the demyelination and retraction of C-fiber and induces neuropathic pain, the effects of LPA-LPA1 signaling on acute nociceptive pain is uncertain. This study investigated the role of LPA-LPA1 signaling in acute nociceptive pain using the formalin test. The pharmacological inhibition of the LPA-LPA1 axis significantly attenuated formalin-induced nociceptive behavior. The LPA1 mRNA was expressed in satellite glial cells (SGCs) in dorsal root ganglion (DRG) and was particularly abundant in SGCs surrounding large DRG neurons, which express neurofilament 200. Treatment with LPA1/3 receptor (LPA1/3 ) antagonist inhibited the upregulation of glial markers and inflammatory cytokines in DRG following formalin injection. The LPA1/3 antagonist also attenuated phosphorylation of extracellular signal-regulated kinase, especially in SGCs and cyclic AMP response element-binding protein in the dorsal horn following formalin injection. LPA amounts after formalin injection to the footpad were quantified by liquid chromatography/tandem mass spectrometry, and LPA levels were found to be increased in the innervated DRGs. Our results indicate that LPA produced in the innervated DRGs promotes the activation of SGCs through LPA1 , increases the sensitivity of primary neurons, and modulates pain behavior. These results facilitate our understanding of the pathology of acute nociceptive pain and demonstrate the possibility of the LPA1 on SGCs as a novel target for acute pain control.
Collapse
Affiliation(s)
- Yoko Hoshino
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Anesthesiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshiaki Okuno
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Daisuke Saigusa
- Laboratory of Biomedical and Analytical Sciences, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan.,Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
| | - Kuniyuki Kano
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Shota Yamamoto
- Department of Lipid Signaling, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hideo Shindou
- Department of Lipid Signaling, National Center for Global Health and Medicine, Tokyo, Japan.,Department of Lipid Medical Science, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Junken Aoki
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kanji Uchida
- Department of Anesthesiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takehiko Yokomizo
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Nobuko Ito
- Department of Anesthesiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
13
|
Wang H, Long T, Zhang H, Li M, Sun Q, Zhai X, Sun L. Anti-fibrosis Attributes; UHPLC-MS/MS-Based pharmacokinetics profiling of a novel ATX inhibitor with excellent vivo efficacy in rat. Biomed Chromatogr 2021; 36:e5301. [PMID: 34928514 DOI: 10.1002/bmc.5301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/28/2021] [Accepted: 11/29/2021] [Indexed: 11/08/2022]
Abstract
3,4-Difluorobenzyl(1-ethyl-5-(4-((4-hydroxypiperidin-1-yl)-methyl)thiazol-2-yl)-1H-indol-3-yl)carbamate (NAI59), a small molecule with outstanding therapeutic effectiveness to anti-pulmonary fibrosis, is being developed as an autotaxin inhibitor candidate compound. To evaluate the pharmacokinetics and plasma protein binding of NAI59, a UPLC-MS/MS method was developed to quantity NAI59 in plasma and phosphate-buffered saline. The calibration curve linearity ranged from 9.95 ng·mL-1 to 1990.00 ng·mL-1 in plasma. The accuracy was -6.8%-5.9%, and the intra- and inter-day precision were within 15%. The matrix effect and recovery were within the criteria, as well as dilution integrity. The chromatographic and mass spectrometric conditions were also feasible to determine PBS samples, and it's proved that this method had good precision and accuracy in the range from 9.95 ng·mL-1 to 497.50 ng·mL-1 in PBS. It's the first time to determine the pharmacokinetics, absolute bioavailability, and plasma protein binding of NAI59 in rats by this established method. As a result, the pharmacokinetic profiles of NAI59 showed a dose-dependent relationship after oral administration, and the absolute bioavailability in rats was 6.3%. In addition, the results of protein binding showed that the combining capacity of NAI59 with plasma protein attained 90% and increased with the increase of drug concentration.
Collapse
Affiliation(s)
- Hongjin Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Tengfei Long
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Hao Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Meng Li
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Qi Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Xin Zhai
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, China
| | - Lixin Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
14
|
Rivera R, Williams NA, Kennedy GG, Sánchez-Pavón P, Chun J. Generation of an Lpar1-EGFP Fusion Knock-in Transgenic Mouse Line. Cell Biochem Biophys 2021; 79:619-627. [PMID: 34652685 PMCID: PMC8551097 DOI: 10.1007/s12013-021-01033-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2021] [Indexed: 10/25/2022]
Abstract
Lysophosphatidic acid (LPA) is a lysophospholipid that acts as an extracellular signal through the activation of cognate G protein-coupled receptors (GPCRs). There are six known LPA receptors (LPA1-6). The first such receptor, LPA1, was identified in the embryonic brain and has been studied extensively for gene expression throughout the body, including through studies of receptor-null mice. However, identifying receptor protein expression in situ and in vivo within living cells and tissues has been difficult because of biologically low receptor expression and variable antibody specificity. To visualize native LPA1 receptor expression in situ, we generated a knock-in mouse produced by homologous recombination in murine embryonic stem (ES) cells to replace a wildtype Lpar1 allele with a mutant allele created by in-frame fusion of EGFP to the 4th exon of Lpar1 (Lpar1-EGFP knock-in allele). Homozygous knock-in mice appeared normal and the expected mendelian ratios of knock-in allele transmission were present in females and males. Histological assessments of the fetal and adult central nervous system (CNS) demonstrated expression patterns that were consistent with prior in situ hybridization studies. This new mouse line will be useful for studies of LPA1 in the developing and adult CNS, as well as other tissues, and for receptor assessments in living tissues and disease models.
Collapse
Affiliation(s)
- Richard Rivera
- Translational Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Nyssa A Williams
- Translational Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Grace G Kennedy
- Translational Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Paloma Sánchez-Pavón
- Translational Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Jerold Chun
- Translational Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
15
|
Puigdomenech-Poch M, Martínez-Muriana A, Andrés-Benito P, Ferrer I, Chun J, López-Vales R. Dual Role of Lysophosphatidic Acid Receptor 2 (LPA 2) in Amyotrophic Lateral Sclerosis. Front Cell Neurosci 2021; 15:600872. [PMID: 33841099 PMCID: PMC8026865 DOI: 10.3389/fncel.2021.600872] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 03/01/2021] [Indexed: 11/13/2022] Open
Abstract
Lysophosphatidic acid (LPA) is a pleiotropic extracellular lipid mediator with many physiological functions that signal through six known G protein-coupled receptors (LPA1-6). In the central nervous system (CNS), LPA mediates a wide range of effects including neural progenitor cell physiology, neuronal cell death, axonal retraction, and inflammation. Since inflammation is a hallmark of most neurological conditions, we hypothesized that LPA could be involved in the physiopathology of amyotrophic lateral sclerosis (ALS). We found that LPA2 RNA was upregulated in post-mortem spinal cord samples of ALS patients and in the sciatic nerve and skeletal muscle of SOD1G93A mouse, the most widely used ALS mouse model. To assess the contribution of LPA2 to ALS, we generated a SOD1G93A mouse that was deficient in Lpar2. This animal revealed that LPA2 signaling accelerates disease onset and neurological decline but, unexpectedly, extended the lifespan. To gain insights into the early harmful actions of LPA2 in ALS, we studied the effects of this receptor in the spinal cord, peripheral nerve, and skeletal muscle of ALS mice. We found that LPA2 gene deletion increased microglial activation but did not contribute to motoneuron death, astrogliosis, degeneration, and demyelination of motor axons. However, we observed that Lpar2 deficiency protected against muscle atrophy. Moreover, we also found the deletion of Lpar2 reduced the invasion of macrophages into the skeletal muscle of SOD1G93A mice, linking LPA2 signaling with muscle inflammation and atrophy in ALS. Overall, these results suggest for the first time that LPA2 contributes to ALS, and its genetic deletion results in protective actions at the early stages of the disease but shortens survival thereafter.
Collapse
Affiliation(s)
- Maria Puigdomenech-Poch
- Departament de Biologia Cellular, Fisiologia i Immunologia, Institut de Neurociències, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Bellaterra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Anna Martínez-Muriana
- Departament de Biologia Cellular, Fisiologia i Immunologia, Institut de Neurociències, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Pol Andrés-Benito
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Departament de Patologia i Terapèutica Experimental, Hospital Universitari de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Universitat de Barcelona, Barcelona, Spain
| | - Isidre Ferrer
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Departament de Patologia i Terapèutica Experimental, Hospital Universitari de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Universitat de Barcelona, Barcelona, Spain
| | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Rubèn López-Vales
- Departament de Biologia Cellular, Fisiologia i Immunologia, Institut de Neurociències, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, Bellaterra, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
16
|
Birgbauer E. Lysophosphatidic Acid Signalling in Nervous System Development and Function. Neuromolecular Med 2021; 23:68-85. [PMID: 33151452 PMCID: PMC11420905 DOI: 10.1007/s12017-020-08630-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/30/2020] [Indexed: 02/06/2023]
Abstract
One class of molecules that are now coming to be recognized as essential for our understanding of the nervous system are the lysophospholipids. One of the major signaling lysophospholipids is lysophosphatidic acid, also known as LPA. LPA activates a variety of G protein-coupled receptors (GPCRs) leading to a multitude of physiological responses. In this review, I describe our current understanding of the role of LPA and LPA receptor signaling in the development and function of the nervous system, especially the central nervous system (CNS). In addition, I highlight how aberrant LPA receptor signaling may underlie neuropathological conditions, with important clinical application.
Collapse
Affiliation(s)
- Eric Birgbauer
- Department of Biology, Winthrop University, Rock Hill, SC, USA.
| |
Collapse
|
17
|
Interface of Phospholipase Activity, Immune Cell Function, and Atherosclerosis. Biomolecules 2020; 10:biom10101449. [PMID: 33076403 PMCID: PMC7602611 DOI: 10.3390/biom10101449] [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: 09/12/2020] [Revised: 09/30/2020] [Accepted: 10/13/2020] [Indexed: 12/16/2022] Open
Abstract
Phospholipases are a family of lipid-altering enzymes that can either reduce or increase bioactive lipid levels. Bioactive lipids elicit signaling responses, activate transcription factors, promote G-coupled-protein activity, and modulate membrane fluidity, which mediates cellular function. Phospholipases and the bioactive lipids they produce are important regulators of immune cell activity, dictating both pro-inflammatory and pro-resolving activity. During atherosclerosis, pro-inflammatory and pro-resolving activities govern atherosclerosis progression and regression, respectively. This review will look at the interface of phospholipase activity, immune cell function, and atherosclerosis.
Collapse
|