1
|
Sarnyai Z, Ben-Shachar D. Schizophrenia, a disease of impaired dynamic metabolic flexibility: A new mechanistic framework. Psychiatry Res 2024; 342:116220. [PMID: 39369460 DOI: 10.1016/j.psychres.2024.116220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 09/21/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
Schizophrenia is a chronic, neurodevelopmental disorder with unknown aetiology and pathophysiology that emphasises the role of neurotransmitter imbalance and abnormalities in synaptic plasticity. The currently used pharmacological approach, the antipsychotic drugs, which have limited efficacy and an array of side-effects, have been developed based on the neurotransmitter hypothesis. Recent research has uncovered systemic and brain abnormalities in glucose and energy metabolism, focusing on altered glycolysis and mitochondrial oxidative phosphorylation. These findings call for a re-conceptualisation of schizophrenia pathophysiology as a progressing bioenergetics failure. In this review, we provide an overview of the fundamentals of brain bioenergetics and the changes identified in schizophrenia. We then propose a new explanatory framework positing that schizophrenia is a disease of impaired dynamic metabolic flexibility, which also reconciles findings of abnormal glucose and energy metabolism in the periphery and in the brain along the course of the disease. This evidence-based framework and testable hypothesis has the potential to transform the way we conceptualise this debilitating condition and to develop novel treatment approaches.
Collapse
Affiliation(s)
- Zoltán Sarnyai
- Laboratory of Psychobiology, Department of Neuroscience, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Department of Psychiatry, Rambam Health Campus, Haifa, Israel; Laboratory of Psychiatric Neuroscience, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia.
| | - Dorit Ben-Shachar
- Laboratory of Psychobiology, Department of Neuroscience, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Department of Psychiatry, Rambam Health Campus, Haifa, Israel.
| |
Collapse
|
2
|
Katsuma Y, Matsui I, Matsumoto A, Okushima H, Imai A, Sakaguchi Y, Yamamoto T, Mizui M, Uchinomiya S, Kato H, Ojida A, Takashima S, Inoue K, Isaka Y. Endogenous activation of peroxisome proliferator-activated receptor-α in proximal tubule cells in counteracting phosphate toxicity. Am J Physiol Renal Physiol 2024; 327:F208-F223. [PMID: 38870264 DOI: 10.1152/ajprenal.00046.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/17/2024] [Accepted: 05/30/2024] [Indexed: 06/15/2024] Open
Abstract
Increased dietary phosphate consumption intensifies renal phosphate burden. Several mechanisms for phosphate-induced renal tubulointerstitial fibrosis have been reported. Considering the dual nature of phosphate as both a potential renal toxin and an essential nutrient for the body, kidneys may possess inherent protective mechanisms against phosphate overload, rather than succumbing solely to injury. However, there is limited understanding of such mechanisms. To identify these mechanisms, we conducted single-cell RNA sequencing (scRNA-seq) analysis of the kidneys of control and dietary phosphate-loaded (Phos) mice at a time point when the Phos group had not yet developed tubulointerstitial fibrosis. scRNA-seq analysis identified the highest number of differentially expressed genes in the clusters belonging to proximal tubular epithelial cells (PTECs). Based on these differentially expressed genes, in silico analyses suggested that the Phos group activated peroxisome proliferator-activated receptor-α (PPAR-α) and fatty acid β-oxidation (FAO) in the PTECs. This activation was further substantiated through various experiments, including the use of an FAO activity visualization probe. Compared with wild-type mice, Ppara knockout mice exhibited exacerbated tubulointerstitial fibrosis in response to phosphate overload. Experiments conducted with cultured PTECs demonstrated that activation of the PPAR-α/FAO pathway leads to improved cellular viability under high-phosphate conditions. The Phos group mice showed a decreased serum concentration of free fatty acids, which are endogenous PPAR-α agonists. Instead, experiments using cultured PTECs revealed that phosphate directly activates the PPAR-α/FAO pathway. These findings indicate that noncanonical metabolic reprogramming via endogenous activation of the PPAR-α/FAO pathway in PTECs is essential to counteract phosphate toxicity.NEW & NOTEWORTHY This study revealed the activation of peroxisome proliferator-activated receptor-α and fatty acid β-oxidation in proximal tubular epithelial cells as an endogenous mechanism to protect the kidney from phosphate toxicity. These findings highlight noncanonical metabolic reprogramming as a potential target for suppressing phosphate toxicity in the kidneys.
Collapse
Affiliation(s)
- Yusuke Katsuma
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Isao Matsui
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
- Transdimensional Life Imaging Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
| | - Ayumi Matsumoto
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroki Okushima
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Atsuhiro Imai
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yusuke Sakaguchi
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takeshi Yamamoto
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masayuki Mizui
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shohei Uchinomiya
- Medical Chemistry and Chemical Biology, Department of Medicinal Sciences, Graduate School of Pharmaceutical Science, Kyushu University, Fukuoka, Japan
| | - Hisakazu Kato
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Akio Ojida
- Medical Chemistry and Chemical Biology, Department of Medicinal Sciences, Graduate School of Pharmaceutical Science, Kyushu University, Fukuoka, Japan
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazunori Inoue
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
3
|
Shen HP, Dong X, Li ZB, Wu JZ, Zheng CM, Hu XJ, Qian C, Wang SP, Zhao YL, Li JC. Protein Profiles and Novel Molecular Biomarkers of Schizophrenia Based on 4D-DIA Proteomics. J Proteome Res 2024; 23:2376-2385. [PMID: 38856018 DOI: 10.1021/acs.jproteome.4c00040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Schizophrenia is a severe psychological disorder. The current diagnosis mainly relies on clinical symptoms and lacks laboratory evidence, which makes it very difficult to make an accurate diagnosis especially at an early stage. Plasma protein profiles of schizophrenia patients were obtained and compared with healthy controls using 4D-DIA proteomics technology. Furthermore, 79 DEPs were identified between schizophrenia and healthy controls. GO functional analysis indicated that DEPs were predominantly associated with responses to toxic substances and platelet aggregation, suggesting the presence of metabolic and immune dysregulation in patients with schizophrenia. KEGG pathway enrichment analysis revealed that DEPs were primarily enriched in the chemokine signaling pathway and cytokine receptor interactions. A diagnostic model was ultimately established, comprising three proteins, namely, PFN1, GAPDH and ACTBL2. This model demonstrated an AUC value of 0.972, indicating its effectiveness in accurately identifying schizophrenia. PFN1, GAPDH and ACTBL2 exhibit potential as biomarkers for the early detection of schizophrenia. The findings of our studies provide novel insights into the laboratory-based diagnosis of schizophrenia.
Collapse
Affiliation(s)
- Hui-Ping Shen
- Department of Psychiatry, Shaoxing Seventh People's Hospital, Shaoxing 312000, China
| | - Xiaotao Dong
- Major Disease Biomarker Research Laboratory, School of Basic Medical Science, Henan University, Kaifeng 475004, China
| | - Zhi-Bin Li
- Major Disease Biomarker Research Laboratory, School of Basic Medical Science, Henan University, Kaifeng 475004, China
| | - Jing-Zhu Wu
- Department of Psychiatry, Shaoxing Seventh People's Hospital, Shaoxing 312000, China
| | - Chun-Mei Zheng
- Department of Psychiatry, Shaoxing Seventh People's Hospital, Shaoxing 312000, China
| | - Xie-Jun Hu
- Department of Psychiatry, Shaoxing Seventh People's Hospital, Shaoxing 312000, China
| | - Chao Qian
- Department of Psychiatry, Shaoxing Seventh People's Hospital, Shaoxing 312000, China
| | - Sheng-Pang Wang
- Department of Psychiatry, Shaoxing Seventh People's Hospital, Shaoxing 312000, China
| | - Yu-Long Zhao
- Department of Psychiatry, Shaoxing Seventh People's Hospital, Shaoxing 312000, China
| | - Ji-Cheng Li
- Major Disease Biomarker Research Laboratory, School of Basic Medical Science, Henan University, Kaifeng 475004, China
- Institute of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
4
|
Żulińska S, Strosznajder AK, Strosznajder JB. Current View on PPAR-α and Its Relation to Neurosteroids in Alzheimer's Disease and Other Neuropsychiatric Disorders: Promising Targets in a Therapeutic Strategy. Int J Mol Sci 2024; 25:7106. [PMID: 39000217 PMCID: PMC11241121 DOI: 10.3390/ijms25137106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/19/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) may play an important role in the pathomechanism/pathogenesis of Alzheimer's disease (AD) and several other neurological/neuropsychiatric disorders. AD leads to progressive alterations in the redox state, ion homeostasis, lipids, and protein metabolism. Significant alterations in molecular processes and the functioning of several signaling pathways result in the degeneration and death of synapses and neuronal cells, leading to the most severe dementia. Peroxisome proliferator-activated receptor alpha (PPAR-α) is among the processes affected by AD; it regulates the transcription of genes related to the metabolism of cholesterol, fatty acids, other lipids and neurotransmission, mitochondria biogenesis, and function. PPAR-α is involved in the cholesterol transport to mitochondria, the substrate for neurosteroid biosynthesis. PPAR-α-coding enzymes, such as sulfotransferases, which are responsible for neurosteroid sulfation. The relation between PPAR-α and cholesterol/neurosteroids may have a significant impact on the course and progression of neurodegeneration/neuroprotection processes. Unfortunately, despite many years of intensive studies, the pathogenesis of AD is unknown and therapy for AD and other neurodegenerative diseases is symptomatic, presenting a significant goal and challenge today. This review presents recent achievements in therapeutic approaches for AD, which are targeting PPAR-α and its relation to cholesterol and neurosteroids in AD and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sylwia Żulińska
- Department of Cellular Signaling, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland;
| | - Anna K. Strosznajder
- Department of Psychiatry, Medical University of Warsaw, Nowowiejska St. 27, 00-665 Warsaw, Poland;
| | - Joanna B. Strosznajder
- Department of Cellular Signaling, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego St., 02-106 Warsaw, Poland;
| |
Collapse
|
5
|
Peng Y, Li Z, Zhang Z, Chen Y, Wang R, Xu N, Cao Y, Jiang C, Chen Z, Lin H. Bromocriptine protects perilesional spinal cord neurons from lipotoxicity after spinal cord injury. Neural Regen Res 2024; 19:1142-1149. [PMID: 37862220 PMCID: PMC10749608 DOI: 10.4103/1673-5374.385308] [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: 03/25/2023] [Revised: 06/28/2023] [Accepted: 07/13/2023] [Indexed: 10/22/2023] Open
Abstract
Recent studies have revealed that lipid droplets accumulate in neurons after brain injury and evoke lipotoxicity, damaging the neurons. However, how lipids are metabolized by spinal cord neurons after spinal cord injury remains unclear. Herein, we investigated lipid metabolism by spinal cord neurons after spinal cord injury and identified lipid-lowering compounds to treat spinal cord injury. We found that lipid droplets accumulated in perilesional spinal cord neurons after spinal cord injury in mice. Lipid droplet accumulation could be induced by myelin debris in HT22 cells. Myelin debris degradation by phospholipase led to massive free fatty acid production, which increased lipid droplet synthesis, β-oxidation, and oxidative phosphorylation. Excessive oxidative phosphorylation increased reactive oxygen species generation, which led to increased lipid peroxidation and HT22 cell apoptosis. Bromocriptine was identified as a lipid-lowering compound that inhibited phosphorylation of cytosolic phospholipase A2 by reducing the phosphorylation of extracellular signal-regulated kinases 1/2 in the mitogen-activated protein kinase pathway, thereby inhibiting myelin debris degradation by cytosolic phospholipase A2 and alleviating lipid droplet accumulation in myelin debris-treated HT22 cells. Motor function, lipid droplet accumulation in spinal cord neurons and neuronal survival were all improved in bromocriptine-treated mice after spinal cord injury. The results suggest that bromocriptine can protect neurons from lipotoxic damage after spinal cord injury via the extracellular signal-regulated kinases 1/2-cytosolic phospholipase A2 pathway.
Collapse
Affiliation(s)
- Ying Peng
- Trauma Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhuoxuan Li
- Trauma Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyang Zhang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yinglun Chen
- Department of Rehabilitation Medicine, Shanghai Geriatric Medical Center, Shanghai, China
| | - Renyuan Wang
- Trauma Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nixi Xu
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuanwu Cao
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chang Jiang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zixian Chen
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haodong Lin
- Trauma Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Maekawa M. [The nuclear receptor PPARα (peroxisome proliferator-activated receptor α) as a novel therapeutic target for schizophrenia]. Nihon Yakurigaku Zasshi 2023; 158:238-241. [PMID: 36990793 DOI: 10.1254/fpj.22142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Our previous study has suggested that peroxisome proliferator-activated receptor α (PPARα) plays a crucial role in the pathophysiology of schizophrenia. In the current study, we screened and identified rare variants in the PPARA gene (encoding PPARα) of schizophrenia subjects. In vitro study showed that those variants decreased activities of PPARα as a transcription factor. Ppara KO mice exhibited a deficit in the sensorimotor gating function and schizophrenia-related histological abnormalities. RNA-seq analysis revealed that PPARα regulates the expression of synaptogenesis signaling pathway-related genes in the brain. Remarkably, treatment of mice with the PPARα agonist fenofibrate alleviated an NMDA receptor antagonist, phencyclidine (PCP)-induced spine pathology and reduced sensitivity to MK-801, another NMDA receptor antagonist. In conclusion, the current study further supports the idea that perturbation in the PPARα-regulated transcriptional machinery leads to a predisposition to schizophrenia, probably by affecting synapse physiology. This study also demonstrates that PPARα can serve as a novel therapeutic target for schizophrenia.
Collapse
Affiliation(s)
- Motoko Maekawa
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine
| |
Collapse
|
7
|
Gómez-Cañas M, Rodríguez-Cueto C, Satta V, Hernández-Fisac I, Navarro E, Fernández-Ruiz J. Endocannabinoid-Binding Receptors as Drug Targets. Methods Mol Biol 2023; 2576:67-94. [PMID: 36152178 DOI: 10.1007/978-1-0716-2728-0_6] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Cannabis plant has been used from ancient times with therapeutic purposes for treating human pathologies, but the identification of the cellular and molecular mechanisms underlying the therapeutic properties of the phytocannabinoids, the active compounds in this plant, occurred in the last years of the past century. In the late 1980s and early 1990s, seminal studies demonstrated the existence of cannabinoid receptors and other elements of the so-called endocannabinoid system. These G protein-coupled receptors (GPCRs) are a key element in the functions assigned to endocannabinoids and appear to serve as promising pharmacological targets. They include CB1, CB2, and GPR55, but also non-GPCRs can be activated by endocannabinoids, like ionotropic receptor TRPV1 and even nuclear receptors of the PPAR family. Their activation, inhibition, or simply modulation have been associated with numerous physiological effects at both central and peripheral levels, which may have therapeutic value in different human pathologies, then providing a solid experimental explanation for both the ancient medicinal uses of Cannabis plant and the recent advances in the development of cannabinoid-based specific therapies. This chapter will review the scientific knowledge generated in the last years around the research on the different endocannabinoid-binding receptors and their signaling mechanisms. Our intention is that this knowledge may help readers to understand the relevance of these receptors in health and disease conditions, as well as it may serve as the theoretical basis for the different experimental protocols to investigate these receptors and their signaling mechanisms that will be described in the following chapters.
Collapse
Affiliation(s)
- María Gómez-Cañas
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Carmen Rodríguez-Cueto
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Valentina Satta
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Inés Hernández-Fisac
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Elisa Navarro
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Javier Fernández-Ruiz
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.
| |
Collapse
|
8
|
Matrisciano F. Functional Nutrition as Integrated Intervention for In- and Outpatient with Schizophrenia. Curr Neuropharmacol 2023; 21:2409-2423. [PMID: 36946488 PMCID: PMC10616917 DOI: 10.2174/1570159x21666230322160259] [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: 07/05/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 03/23/2023] Open
Abstract
Schizophrenia is a chronic and progressive disorder characterized by cognitive, emotional, and behavioral abnormalities associated with neuronal development and synaptic plasticity alterations. Genetic and epigenetic abnormalities in cortical parvalbumin-positive GABAergic interneurons and consequent alterations in glutamate-mediated excitatory neurotransmission during early neurodevelopment underlie schizophrenia manifestation and progression. Also, epigenetic alterations during pregnancy or early phases of postnatal life are associated with schizophrenia vulnerability and inflammatory processes, which are at the basis of brain pathology and a higher risk of comorbidities, including cardiovascular diseases and metabolic syndrome. In addition, schizophrenia patients adopt an unhealthy lifestyle and poor nutrition, leading to premature death. Here, I explored the role of functional nutrition as an integrated intervention for the long-term management of patients with schizophrenia. Several natural bioactive compounds in plant-based whole foods, including flavonoids, phytonutrients, vitamins, fatty acids, and minerals, modulate brain functioning by targeting neuroinflammation and improving cognitive decline. Although further clinical studies are needed, a functional diet rich in natural bioactive compounds might be effective in synergism with standard treatments to improve schizophrenia symptoms and reduce the risk of comorbidities.
Collapse
Affiliation(s)
- Francesco Matrisciano
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois Chicago (UIC), Chicago, IL, USA
| |
Collapse
|
9
|
Virendra SA, Kumar A, Chawla PA, Mamidi N. Development of Heterocyclic PPAR Ligands for Potential Therapeutic Applications. Pharmaceutics 2022; 14:2139. [PMID: 36297575 PMCID: PMC9611956 DOI: 10.3390/pharmaceutics14102139] [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: 06/20/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022] Open
Abstract
The family of nuclear peroxisome proliferator-activated receptors (PPARα, PPARβ/δ, and PPARγ) is a set of ligand-activated transcription factors that regulate different functions in the body. Whereas activation of PPARα is known to reduce the levels of circulating triglycerides and regulate energy homeostasis, the activation of PPARγ brings about insulin sensitization and increases the metabolism of glucose. On the other hand, PPARβ when activated increases the metabolism of fatty acids. Further, these PPARs have been claimed to be utilized in various metabolic, neurological, and inflammatory diseases, neurodegenerative disorders, fertility or reproduction, pain, and obesity. A series of different heterocyclic scaffolds have been synthesized and evaluated for their ability to act as PPAR agonists. This review is a compilation of efforts on the part of medicinal chemists around the world to find novel compounds that may act as PPAR ligands along with patents in regards to PPAR ligands. The structure-activity relationship, as well as docking studies, have been documented to better understand the mechanistic investigations of various compounds, which will eventually aid in the design and development of new PPAR ligands. From the results of the structural activity relationship through the pharmacological and in silico evaluation the potency of heterocycles as PPAR ligands can be described in terms of their hydrogen bonding, hydrophobic interactions, and other interactions with PPAR.
Collapse
Affiliation(s)
- Sharma Arvind Virendra
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga 142001, Punjab, India
| | - Ankur Kumar
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga 142001, Punjab, India
| | - Pooja A. Chawla
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga 142001, Punjab, India
| | - Narsimha Mamidi
- Department of Chemistry and Nanotechnology, School of Engineering and Sciences, Tecnologico de Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey 64849, Nuevo Leon, Mexico
| |
Collapse
|
10
|
Zhou H, Wang D, Cao B, Zhang X. Association of reduced cortical thickness and psychopathological symptoms in patients with first-episode drug-naïve schizophrenia. Int J Psychiatry Clin Pract 2022; 27:42-50. [PMID: 36193901 DOI: 10.1080/13651501.2022.2129067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/10/2022]
Abstract
OBJECTIVE There is growing evidence that reduced cortical thickness has been considered to be a central abnormality in schizophrenia. Brain imaging studies have demonstrated that the cerebral cortex becomes thinner in patients with first-episode schizophrenia. This study aimed to examine whether cortical thickness is altered in drug-naïve schizophrenia in a Chinese Han population and the relationship between cortical thickness and clinical symptoms. METHODS We compared cortical thickness in 41 schizophrenia patients and 30 healthy controls. Psychopathology of patients with schizophrenia was assessed using the Positive and Negative Syndrome Scale (PANSS). RESULTS The cortical thickness of left banks of superior temporal sulcus, left lateral occipital gyrus, left rostral middle frontal gyrus, right inferior parietal lobule and right lateral occipital gyrus in schizophrenia patients was generally thinner compared with healthy controls. Correlation analysis revealed a negative correlation between cortical thickness of the left banks of superior temporal sulcus and general psychopathology of PANSS. CONCLUSIONS Our results suggest that cortical thickness abnormalities are already present early in the onset of schizophrenia and are associated with psychopathological symptoms, suggesting that it plays an important role in the pathogenesis and symptomatology of schizophrenia.Key points(1) The first-episode drug-naïve schizophrenia had reduced cortical thickness than the controls.(2) Cortical thickness was associated with psychopathological symptoms in patients with schizophrenia.
Collapse
Affiliation(s)
- Huixia Zhou
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, PR China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, PR China
| | - Dongmei Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, PR China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, PR China
| | - Bo Cao
- Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada.,Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xiangyang Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, PR China.,Department of Psychology, University of Chinese Academy of Sciences, Beijing, PR China
| |
Collapse
|
11
|
Witchey SK, Doyle MG, Fredenburg JD, St Armour G, Horman B, Odenkirk MT, Aylor DL, Baker ES, Patisaul HB. Impacts of Gestational FireMaster 550 Exposure on the Neonatal Cortex Are Sex Specific and Largely Attributable to the Organophosphate Esters. Neuroendocrinology 2022; 113:1262-1282. [PMID: 36075192 PMCID: PMC9992460 DOI: 10.1159/000526959] [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: 05/04/2022] [Accepted: 08/24/2022] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Flame retardants (FRs) are common bodily and environmental pollutants, creating concern about their potential toxicity. We and others have found that the commercial mixture FireMaster® 550 (FM 550) or its individual brominated (BFR) and organophosphate ester (OPFR) components are potential developmental neurotoxicants. Using Wistar rats, we previously reported that developmental exposure to FM 550 or its component classes produced sex- and compound-specific effects on adult socioemotional behaviors. The underlying mechanisms driving the behavioral phenotypes are unknown. METHODS To further mechanistic understanding, here we conducted transcriptomics in parallel with a novel lipidomics approach using cortical tissues from newborn siblings of the rats in the published behavioral study. Inclusion of lipid composition is significant because it is rarely examined in developmental neurotoxicity studies. Pups were gestationally exposed via oral dosing to the dam to FM 550 or the BFR or OPFR components at environmentally relevant doses. RESULTS The neonatal cortex was highly sexually dimorphic in lipid and transcriptome composition, and males were more significantly impacted by FR exposure. Multiple adverse modes of action for the BFRs and OPFRs on neurodevelopment were identified, with the OPFRs being more disruptive than the BFRs via multiple mechanisms including dysregulation of mitochondrial function and disruption of cholinergic and glutamatergic systems. Disrupted mitochondrial function by environmental factors has been linked to a higher risk of autism spectrum disorders and neurodegenerative disorders. Impacted lipid classes included ceramides, sphingomyelins, and triacylglycerides. Robust ceramide upregulation in the OPFR females could suggest a heightened risk of brain metabolic disease. CONCLUSIONS This study reveals multiple mechanisms by which the components of a common FR mixture are developmentally neurotoxic and that the OPFRs may be the compounds of greatest concern.
Collapse
Affiliation(s)
- Shannah K Witchey
- Department of Biological Sciences, NC State University, Raleigh, North Carolina, USA
| | - Michael G Doyle
- Department of Chemistry, NC State University, Raleigh, North Carolina, USA
| | - Jacob D Fredenburg
- Department of Biological Sciences, NC State University, Raleigh, North Carolina, USA
| | - Genevieve St Armour
- Department of Biological Sciences, NC State University, Raleigh, North Carolina, USA
| | - Brian Horman
- Department of Biological Sciences, NC State University, Raleigh, North Carolina, USA
| | - Melanie T Odenkirk
- Department of Chemistry, NC State University, Raleigh, North Carolina, USA
| | - David L Aylor
- Department of Biological Sciences, NC State University, Raleigh, North Carolina, USA
- Center for Human Health and the Environment, NC State University, Raleigh, North Carolina, USA
| | - Erin S Baker
- Department of Chemistry, NC State University, Raleigh, North Carolina, USA
| | - Heather B Patisaul
- Center for Human Health and the Environment, NC State University, Raleigh, North Carolina, USA
| |
Collapse
|
12
|
PPARα Signaling: A Candidate Target in Psychiatric Disorder Management. Biomolecules 2022; 12:biom12050723. [PMID: 35625650 PMCID: PMC9138493 DOI: 10.3390/biom12050723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/07/2022] [Accepted: 05/18/2022] [Indexed: 02/01/2023] Open
Abstract
Peroxisome proliferator-activator receptors (PPARs) regulate lipid and glucose metabolism, control inflammatory processes, and modulate several brain functions. Three PPAR isoforms have been identified, PPARα, PPARβ/δ, and PPARγ, which are expressed in different tissues and cell types. Hereinafter, we focus on PPARα involvement in the pathophysiology of neuropsychiatric and neurodegenerative disorders, which is underscored by PPARα localization in neuronal circuits involved in emotion modulation and stress response, and its role in neurodevelopment and neuroinflammation. A multiplicity of downstream pathways modulated by PPARα activation, including glutamatergic neurotransmission, upregulation of brain-derived neurotrophic factor, and neurosteroidogenic effects, encompass mechanisms underlying behavioral regulation. Modulation of dopamine neuronal firing in the ventral tegmental area likely contributes to PPARα effects in depression, anhedonia, and autism spectrum disorder (ASD). Based on robust preclinical evidence and the initial results of clinical studies, future clinical trials should assess the efficacy of PPARα agonists in the treatment of mood and neurodevelopmental disorders, such as depression, schizophrenia, and ASD.
Collapse
|
13
|
Parksepp M, Haring L, Kilk K, Koch K, Uppin K, Kangro R, Zilmer M, Vasar E. The Expanded Endocannabinoid System Contributes to Metabolic and Body Mass Shifts in First-Episode Schizophrenia: A 5-Year Follow-Up Study. Biomedicines 2022; 10:biomedicines10020243. [PMID: 35203453 PMCID: PMC8869544 DOI: 10.3390/biomedicines10020243] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 02/07/2023] Open
Abstract
Alterations in the expanded endocannabinoid system (eECS) and cell membrane composition have been implicated in the pathophysiology of schizophrenia spectrum disorders. We enrolled 54 antipsychotic (AP)-naïve first-episode psychosis (FEP) patients and 58 controls and applied a targeted metabolomics approach followed by multivariate data analysis to investigate the profile changes in the serum levels of endocannabinoids: 2-arachidonoylglycerol (2-AG) and anandamide, endocannabinoids-like N-acylethanolamines (NAEs: linoleoylethanolamide, oleoylethanolamide, and palmitoylethanolamide), and their dominating lipid precursor’s phosphatidylcholines. Biomolecule profiles were measured at the onset of first-episode psychosis (FEP) and 0.6 years and 5.1 years after the initiation of AP treatment. The results indicated that FEP might be characterized by elevated concentrations of NAEs and by decreased 2-AG levels. At this stage of the disease, the NAE-mediated upregulation of peroxisome proliferator-activated receptors (PPARs) manifested themselves in energy expenditure. A 5-year disease progression and AP treatment adverse effects led to a robust increase in 2-AG levels, which contributed to strengthened cannabinoid (CB1) receptor-mediated effects, which manifested in obesity. Dynamic 2-AG, NAEs, and their precursors in terms of phosphatidylcholines are relevant to the description of the metabolic shifts resulting from the altered eECS function during and after FEP.
Collapse
Affiliation(s)
- Madis Parksepp
- Institute of Clinical Medicine, University of Tartu, 50406 Tartu, Estonia;
- Psychiatry Clinic of Viljandi Hospital, 71024 Viljandi, Estonia
| | - Liina Haring
- Institute of Clinical Medicine, University of Tartu, 50406 Tartu, Estonia;
- Psychiatry Clinic of Tartu University Hospital, 50406 Tartu, Estonia; (K.K.); (K.U.)
- Centre of Excellence for Genomics and Translational Medicine, Institute of Biomedicine and Translational Medicine, Univesignallingrsity of Tartu, 50090 Tartu, Estonia; (K.K.); (M.Z.); (E.V.)
- Correspondence: ; Tel.: +372-7318-767
| | - Kalle Kilk
- Centre of Excellence for Genomics and Translational Medicine, Institute of Biomedicine and Translational Medicine, Univesignallingrsity of Tartu, 50090 Tartu, Estonia; (K.K.); (M.Z.); (E.V.)
| | - Kadri Koch
- Psychiatry Clinic of Tartu University Hospital, 50406 Tartu, Estonia; (K.K.); (K.U.)
| | - Kärt Uppin
- Psychiatry Clinic of Tartu University Hospital, 50406 Tartu, Estonia; (K.K.); (K.U.)
| | - Raul Kangro
- Institute of Mathematics and Statistics, University of Tartu, 50090 Tartu, Estonia;
| | - Mihkel Zilmer
- Centre of Excellence for Genomics and Translational Medicine, Institute of Biomedicine and Translational Medicine, Univesignallingrsity of Tartu, 50090 Tartu, Estonia; (K.K.); (M.Z.); (E.V.)
| | - Eero Vasar
- Centre of Excellence for Genomics and Translational Medicine, Institute of Biomedicine and Translational Medicine, Univesignallingrsity of Tartu, 50090 Tartu, Estonia; (K.K.); (M.Z.); (E.V.)
| |
Collapse
|
14
|
Yang X, Sun W, Wu Q, Lin H, Lu Z, Shen X, Chen Y, Zhou Y, Huang L, Wu F, Liu F, Chu D. Excess Folic Acid Supplementation before and during Pregnancy and Lactation Alters Behaviors and Brain Gene Expression in Female Mouse Offspring. Nutrients 2021; 14:nu14010066. [PMID: 35010941 PMCID: PMC8746785 DOI: 10.3390/nu14010066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/17/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022] Open
Abstract
Use of folic acid (FA) during early pregnancy protects against birth defects. However, excess FA has shown gender-specific neurodevelopmental toxicity. Previously, we fed the mice with 2.5 times the recommended amount of FA one week prior to mating and during the pregnancy and lactation periods, and detected the activated expression of Fos and related genes in the brains of weaning male offspring, as well as behavioral abnormalities in the adults. Here, we studied whether female offspring were affected by the same dosage of FA. An open field test, three-chamber social approach and social novelty test, an elevated plus-maze, rotarod test and the Morris water maze task were used to evaluate their behaviors. RNA sequencing was performed to identify differentially expressed genes in the brains. Quantitative real time-PCR (qRT-PCR) and Western blots were applied to verify the changes in gene expression. We found increased anxiety and impaired exploratory behavior, motor coordination and spatial memory in FA-exposed females. The brain transcriptome revealed 36 up-regulated and 79 down-regulated genes in their brains at weaning. The increase of Tlr1; Sult1a1; Tph2; Acacb; Etnppl; Angptl4 and Apold1, as well as a decrease of Ppara mRNA were confirmed by qRT-PCR. Among these genes; the mRNA levels of Etnppl; Angptl4andApold1 were increased in the both FA-exposed female and male brains. The elevation of Sult1a1 protein was confirmed by Western blots. Our data suggest that excess FA alteres brain gene expression and behaviors in female offspring, of which certain genes show apparent gender specificity.
Collapse
Affiliation(s)
- Xingyue Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (X.Y.); (Q.W.); (X.S.); (L.H.)
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China; (H.L.); (F.W.)
| | - Wenyan Sun
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong 226001, China; (W.S.); (Z.L.); (Y.C.); (Y.Z.)
| | - Qian Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (X.Y.); (Q.W.); (X.S.); (L.H.)
| | - Hongyan Lin
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China; (H.L.); (F.W.)
| | - Zhixing Lu
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong 226001, China; (W.S.); (Z.L.); (Y.C.); (Y.Z.)
| | - Xin Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (X.Y.); (Q.W.); (X.S.); (L.H.)
| | - Yongqi Chen
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong 226001, China; (W.S.); (Z.L.); (Y.C.); (Y.Z.)
| | - Yan Zhou
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong 226001, China; (W.S.); (Z.L.); (Y.C.); (Y.Z.)
| | - Li Huang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (X.Y.); (Q.W.); (X.S.); (L.H.)
| | - Feng Wu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China; (H.L.); (F.W.)
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
- Correspondence: (F.L.); (D.C.)
| | - Dandan Chu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (X.Y.); (Q.W.); (X.S.); (L.H.)
- Correspondence: (F.L.); (D.C.)
| |
Collapse
|
15
|
A loss-of-function variant in SUV39H2 identified in autism-spectrum disorder causes altered H3K9 trimethylation and dysregulation of protocadherin β-cluster genes in the developing brain. Mol Psychiatry 2021; 26:7550-7559. [PMID: 34262135 DOI: 10.1038/s41380-021-01199-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/21/2021] [Accepted: 06/10/2021] [Indexed: 02/06/2023]
Abstract
Recent evidence has documented the potential roles of histone-modifying enzymes in autism-spectrum disorder (ASD). Aberrant histone H3 lysine 9 (H3K9) dimethylation resulting from genetic variants in histone methyltransferases is known for neurodevelopmental and behavioral anomalies. However, a systematic examination of H3K9 methylation dynamics in ASD is lacking. Here we resequenced nine genes for histone methyltransferases and demethylases involved in H3K9 methylation in individuals with ASD and healthy controls using targeted next-generation sequencing. We identified a novel rare variant (A211S) in the SUV39H2, which was predicted to be deleterious. The variant showed strongly reduced histone methyltransferase activity in vitro. In silico analysis showed that the variant destabilizes the hydrophobic core and allosterically affects the enzyme activity. The Suv39h2-KO mice displayed hyperactivity and reduced behavioral flexibility in learning the tasks that required complex behavioral adaptation, which is relevant for ASD. The Suv39h2 deficit evoked an elevated expression of a subset of protocadherin β (Pcdhb) cluster genes in the embryonic brain, which is attributable to the loss of H3K9 trimethylation (me3) at the gene promoters. Reduced H3K9me3 persisted in the cerebellum of Suv39h2-deficient mice to an adult stage. Congruently, reduced expression of SUV39H1 and SUV39H2 in the postmortem brain samples of ASD individuals was observed, underscoring the role of H3K9me3 deficiency in ASD etiology. The present study provides direct evidence for the role of SUV39H2 in ASD and suggests a molecular cascade of SUV39H2 dysfunction leading to H3K9me3 deficiency followed by an untimely, elevated expression of Pcdhb cluster genes during early neurodevelopment.
Collapse
|
16
|
Lee D, Tomita Y, Allen W, Tsubota K, Negishi K, Kurihara T. PPARα Modulation-Based Therapy in Central Nervous System Diseases. Life (Basel) 2021; 11:life11111168. [PMID: 34833044 PMCID: PMC8622664 DOI: 10.3390/life11111168] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 10/29/2021] [Accepted: 10/30/2021] [Indexed: 12/11/2022] Open
Abstract
The burden of neurodegenerative diseases in the central nervous system (CNS) is increasing globally. There are various risk factors for the development and progression of CNS diseases, such as inflammatory responses and metabolic derangements. Thus, curing CNS diseases requires the modulation of damaging signaling pathways through a multitude of mechanisms. Peroxisome proliferator-activated receptors (PPARs) are a family of nuclear hormone receptors (PPARα, PPARβ/δ, and PPARγ), and they work as master sensors and modulators of cellular metabolism. In this regard, PPARs have recently been suggested as promising therapeutic targets for suppressing the development of CNS diseases and their progressions. While the therapeutic role of PPARγ modulation in CNS diseases has been well reviewed, the role of PPARα modulation in these diseases has not been comprehensively summarized. The current review focuses on the therapeutic roles of PPARα modulation in CNS diseases, including those affecting the brain, spinal cord, and eye, with recent advances. Our review will enable more comprehensive therapeutic approaches to modulate PPARα for the prevention of and protection from various CNS diseases.
Collapse
Affiliation(s)
- Deokho Lee
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo 160-8582, Japan;
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan;
| | - Yohei Tomita
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo 160-8582, Japan;
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan;
- Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
- Correspondence: (Y.T.); (T.K.); Tel.: +1-617-919-2533 (Y.T.); +81-3-5636-3204 (T.K.)
| | - William Allen
- Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | | | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan;
| | - Toshihide Kurihara
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo 160-8582, Japan;
- Department of Ophthalmology, Keio University School of Medicine, Tokyo 160-8582, Japan;
- Correspondence: (Y.T.); (T.K.); Tel.: +1-617-919-2533 (Y.T.); +81-3-5636-3204 (T.K.)
| |
Collapse
|
17
|
Matrisciano F, Pinna G. PPAR-α Hypermethylation in the Hippocampus of Mice Exposed to Social Isolation Stress Is Associated with Enhanced Neuroinflammation and Aggressive Behavior. Int J Mol Sci 2021; 22:ijms221910678. [PMID: 34639019 PMCID: PMC8509148 DOI: 10.3390/ijms221910678] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/24/2021] [Accepted: 08/28/2021] [Indexed: 12/18/2022] Open
Abstract
Social behavioral changes, including social isolation or loneliness, increase the risk for stress-related disorders, such as major depressive disorder, posttraumatic stress disorder (PTSD), and suicide, which share a strong neuroinflammatory etiopathogenetic component. The peroxisome-proliferator activated receptor (PPAR)-α, a newly discovered target involved in emotional behavior regulation, is a ligand-activated nuclear receptor and a transcription factor that, following stimulation by endogenous or synthetic ligands, may induce neuroprotective effects by modulating neuroinflammation, and improve anxiety and depression-like behaviors by enhancing neurosteroid biosynthesis. How stress affects epigenetic mechanisms with downstream effects on inflammation and emotional behavior remains poorly understood. We studied the effects of 4-week social isolation, using a mouse model of PTSD/suicide-like behavior, on hippocampal PPAR-α epigenetic modification. Decreased PPAR-α expression in the hippocampus of socially isolated mice was associated with increased levels of methylated cytosines of PPAR-α gene CpG-rich fragments and deficient neurosteroid biosynthesis. This effect was associated with increased histone deacetylases (HDAC)1, methyl-cytosine binding protein (MeCP)2 and decreased ten-eleven translocator (TET)2 expression, which favor hypermethylation. These alterations were associated with increased TLR-4 and pro-inflammatory markers (e.g., TNF-α,), mediated by NF-κB signaling in the hippocampus of aggressive mice. This study contributes the first evidence of stress-induced brain PPAR-α epigenetic regulation. Social isolation stress may constitute a risk factor for inflammatory-based psychiatric disorders associated with neurosteroid deficits, and targeting epigenetic marks linked to PPAR-α downregulation may offer a valid therapeutic approach.
Collapse
|
18
|
Ohnishi T, Kiyama Y, Arima‐Yoshida F, Kadota M, Ichikawa T, Yamada K, Watanabe A, Ohba H, Tanaka K, Nakaya A, Horiuchi Y, Iwayama Y, Toyoshima M, Ogawa I, Shimamoto‐Mitsuyama C, Maekawa M, Balan S, Arai M, Miyashita M, Toriumi K, Nozaki Y, Kurokawa R, Suzuki K, Yoshikawa A, Toyota T, Hosoya T, Okuno H, Bito H, Itokawa M, Kuraku S, Manabe T, Yoshikawa T. Cooperation of LIM domain-binding 2 (LDB2) with EGR in the pathogenesis of schizophrenia. EMBO Mol Med 2021; 13:e12574. [PMID: 33656268 PMCID: PMC8033514 DOI: 10.15252/emmm.202012574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 01/21/2021] [Accepted: 01/25/2021] [Indexed: 01/15/2023] Open
Abstract
Genomic defects with large effect size can help elucidate unknown pathologic architecture of mental disorders. We previously reported on a patient with schizophrenia and a balanced translocation between chromosomes 4 and 13 and found that the breakpoint within chromosome 4 is located near the LDB2 gene. We show here that Ldb2 knockout (KO) mice displayed multiple deficits relevant to mental disorders. In particular, Ldb2 KO mice exhibited deficits in the fear-conditioning paradigm. Analysis of the amygdala suggested that dysregulation of synaptic activities controlled by the immediate early gene Arc is involved in the phenotypes. We show that LDB2 forms protein complexes with known transcription factors. Consistently, ChIP-seq analyses indicated that LDB2 binds to > 10,000 genomic sites in human neurospheres. We found that many of those sites, including the promoter region of ARC, are occupied by EGR transcription factors. Our previous study showed an association of the EGR family genes with schizophrenia. Collectively, the findings suggest that dysregulation in the gene expression controlled by the LDB2-EGR axis underlies a pathogenesis of subset of mental disorders.
Collapse
|