1
|
Strekalova T, Svirin E, Gorlova A, Sheveleva E, Burova A, Khairetdinova A, Sitdikova K, Zakharova E, Dudchenko AM, Lyundup A, Morozov S. Resilience and Vulnerability to Stress-Induced Anhedonia: Unveiling Brain Gene Expression and Mitochondrial Dynamics in a Mouse Chronic Stress Depression Model. Biomolecules 2023; 13:1782. [PMID: 38136653 PMCID: PMC10741640 DOI: 10.3390/biom13121782] [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: 11/17/2023] [Revised: 12/07/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
The role of altered brain mitochondrial regulation in psychiatric pathologies, including Major Depressive Disorder (MDD), has attracted increasing attention. Aberrant mitochondrial functions were suggested to underlie distinct inter-individual vulnerability to stress-related MDD syndrome. In this context, insulin receptor sensitizers (IRSs) that regulate brain metabolism have become a focus of recent research, as their use in pre-clinical studies can help to elucidate the role of mitochondrial dynamics in this disorder and contribute to the development of new antidepressant treatment. Here, following 2-week chronic mild stress (CMS) using predation, social defeat, and restraint, MDD-related behaviour and brain molecular markers have been investigated along with the hippocampus-dependent performance and emotionality in mice that received the IRS dicholine succinate (DS). In a sucrose test, mice were studied for the key feature of MDD, a decreased sensitivity to reward, called anhedonia. Based on this test, animals were assigned to anhedonic and resilient-to-stress-induced-anhedonia groups, using a previously established criterion of a decrease in sucrose preference below 65%. Such assignment was based on the fact that none of control, non-stressed animals displayed sucrose preference that would be smaller than this value. DS-treated stressed mice displayed ameliorated behaviours in a battery of assays: sucrose preference, coat state, the Y-maze, the marble test, tail suspension, and nest building. CMS-vulnerable mice exhibited overexpression of the inflammatory markers Il-1β, tnf, and Cox-1, as well as 5-htt and 5-ht2a-R, in various brain regions. The alterations in hippocampal gene expression were the closest to clinical findings and were studied further. DS-treated, stressed mice showed normalised hippocampal expression of the plasticity markers Camk4, Camk2, Pka, Adcy1, Creb-ar, Nmda-2r-ar, and Nmda-2r-s. DS-treated and non-treated stressed mice who were resilient or vulnerable to anhedonia were compared for hippocampal mitochondrial pathway regulation using Illumina profiling. Resilient mice revealed overexpression of the mitochondrial complexes NADH dehydrogenase, succinate dehydrogenase, cytochrome bc1, cytochrome c oxidase, F-type and V-type ATPases, and inorganic pyrophosphatase, which were decreased in anhedonic mice. DS partially normalised the expression of both ATPases. We conclude that hippocampal reduction in ATP synthesis is associated with anhedonia and pro-inflammatory brain changes that are ameliorated by DS.
Collapse
Affiliation(s)
- Tatyana Strekalova
- Division of Molecular Psychiatry, Center of Mental Health, University of Hospital Würzburg, 97080 Wuerzburg, Germany
| | - Evgeniy Svirin
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia (A.G.); (E.S.); (A.B.); (A.K.); (K.S.); (E.Z.); (A.M.D.); (S.M.)
| | - Anna Gorlova
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia (A.G.); (E.S.); (A.B.); (A.K.); (K.S.); (E.Z.); (A.M.D.); (S.M.)
| | - Elizaveta Sheveleva
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia (A.G.); (E.S.); (A.B.); (A.K.); (K.S.); (E.Z.); (A.M.D.); (S.M.)
| | - Alisa Burova
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia (A.G.); (E.S.); (A.B.); (A.K.); (K.S.); (E.Z.); (A.M.D.); (S.M.)
| | - Adel Khairetdinova
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia (A.G.); (E.S.); (A.B.); (A.K.); (K.S.); (E.Z.); (A.M.D.); (S.M.)
| | - Kseniia Sitdikova
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia (A.G.); (E.S.); (A.B.); (A.K.); (K.S.); (E.Z.); (A.M.D.); (S.M.)
| | - Elena Zakharova
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia (A.G.); (E.S.); (A.B.); (A.K.); (K.S.); (E.Z.); (A.M.D.); (S.M.)
| | - Alexander M. Dudchenko
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia (A.G.); (E.S.); (A.B.); (A.K.); (K.S.); (E.Z.); (A.M.D.); (S.M.)
| | - Aleksey Lyundup
- Endocrinology Research Centre, Dmitry Ulyanov St. 19, Moscow 117036, Russia;
- Research and Education Resource Center, Peoples Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya St, Moscow 117198, Russia
| | - Sergey Morozov
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow 125315, Russia (A.G.); (E.S.); (A.B.); (A.K.); (K.S.); (E.Z.); (A.M.D.); (S.M.)
| |
Collapse
|
2
|
Rodrigues T, Bressan GN, Krum BN, Soares FAA, Fachinetto R. Influence of the dose of ketamine used on schizophrenia-like symptoms in mice: A correlation study with TH, GAD 67, and PPAR-γ. Pharmacol Biochem Behav 2023; 233:173658. [PMID: 37804866 DOI: 10.1016/j.pbb.2023.173658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/03/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
Schizophrenia is a chronic, debilitating mental illness that has not yet been completely understood. In this study, we aimed to investigate the effects of different doses of ketamine, a non-competitive NMDA receptor antagonist, on the positive- and negative-like symptoms of schizophrenia. We also explored whether these effects are related to changes in the immunoreactivity of GAD67, TH, and PPAR-γ in brain structures. To conduct the study, male mice received ketamine (20-40 mg/kg) or its vehicle (0.9 % NaCl) intraperitoneally for 14 consecutive days. We quantified stereotyped behavior, the time of immobility in the forced swimming test (FST), and locomotor activity after 7 or 14 days. In addition, we performed ex vivo analysis of the immunoreactivity of GAD, TH, and PPAR-γ, in brain tissues after 14 days. The results showed that ketamine administration for 14 days increased the grooming time in the nose region at all tested doses. It also increased immobility in the FST at 30 mg/kg doses and decreased the number of rearing cycles during stereotyped behavior at 40 mg/kg. These behavioral effects were not associated with changes in locomotor activity. We did not observe any significant alterations regarding the immunoreactivity of brain proteins. However, we found that GAD and TH were positively correlated with the number of rearing during the stereotyped behavior at doses of 20 and 30 mg/kg ketamine, respectively. GAD was positively correlated with the number of rearing in the open field test at a dose of 20 mg/kg. TH was inversely correlated with immobility time in the FST at a dose of 30 mg/kg. PPAR-γ was inversely correlated with the number of bouts of stereotyped behavior at a dose of 40 mg/kg of ketamine. In conclusion, the behavioral alterations induced by ketamine in positive-like symptoms were reproduced with all doses tested and appear to depend on the modulatory effects of TH, GAD, and PPAR-γ. Conversely, negative-like symptoms were associated with a specific dose of ketamine.
Collapse
Affiliation(s)
- Talita Rodrigues
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Getulio Nicola Bressan
- Programa de Pós-Graduação em Ciências Biológicas, Bioquímica Toxicológica, Universidade Federal de Santa Maria, RS, Brazil
| | - Bárbara Nunes Krum
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Félix Alexandre Antunes Soares
- Programa de Pós-Graduação em Ciências Biológicas, Bioquímica Toxicológica, Universidade Federal de Santa Maria, RS, Brazil
| | - Roselei Fachinetto
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil; Programa de Pós-Graduação em Ciências Biológicas, Bioquímica Toxicológica, Universidade Federal de Santa Maria, RS, Brazil.
| |
Collapse
|
3
|
Kumar S, Mehan S, Narula AS. Therapeutic modulation of JAK-STAT, mTOR, and PPAR-γ signaling in neurological dysfunctions. J Mol Med (Berl) 2023; 101:9-49. [PMID: 36478124 DOI: 10.1007/s00109-022-02272-6] [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] [Received: 05/31/2022] [Revised: 10/10/2022] [Accepted: 11/11/2022] [Indexed: 12/12/2022]
Abstract
The cytokine-activated Janus kinase (JAK)-signal transducer and activator of transcription (STAT) cascade is a pleiotropic pathway that involves receptor subunit multimerization. The mammalian target of rapamycin (mTOR) is a ubiquitously expressed serine-threonine kinase that perceives and integrates a variety of intracellular and environmental stimuli to regulate essential activities such as cell development and metabolism. Peroxisome proliferator-activated receptor-gamma (PPARγ) is a prototypical metabolic nuclear receptor involved in neural differentiation and axon polarity. The JAK-STAT, mTOR, and PPARγ signaling pathways serve as a highly conserved signaling hub that coordinates neuronal activity and brain development. Additionally, overactivation of JAK/STAT, mTOR, and inhibition of PPARγ signaling have been linked to various neurocomplications, including neuroinflammation, apoptosis, and oxidative stress. Emerging research suggests that even minor disruptions in these cellular and molecular processes can have significant consequences manifested as neurological and neuropsychiatric diseases. Of interest, target modulators have been proven to alleviate neuronal complications associated with acute and chronic neurological deficits. This research-based review explores the therapeutic role of JAK-STAT, mTOR, and PPARγ signaling modulators in preventing neuronal dysfunctions in preclinical and clinical investigations.
Collapse
Affiliation(s)
- Sumit Kumar
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Punjab, Moga, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Punjab, Moga, India.
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC, 27516, USA
| |
Collapse
|
4
|
Yang Y, Shi GN, Wu X, Xu M, Chen CJ, Zhou Y, Wei YZ, Wu L, Cui FF, Sun L, Zhang TT. Quercetin Impedes Th17 Cell Differentiation to Mitigate Arthritis Involving PPARγ-Driven Transactivation of SOCS3 and Redistribution Corepressor SMRT from PPARγ to STAT3. Mol Nutr Food Res 2022; 66:e2100826. [PMID: 35384292 DOI: 10.1002/mnfr.202100826] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 03/03/2022] [Indexed: 01/08/2023]
Abstract
SCOPE Quercetin (QU) is one of the most abundant flavonoids in plants and has attracted the attention of researchers because of its remarkable antirheumatoid arthritis (RA) effects and extremely low adverse reactions. However, the underlying mechanism needs further study. METHODS AND RESULTS Flow cytometry, immunofluorescence, enzyme linked immunosorbent assay (ELISA), and quantitative real-time polymerase chain reaction (qRT-PCR) reveal the obvious inhibitory effects of QU on Th17 cell differentiation in arthritic mice. More importantly, QU markedly limits the development of Th17 cell polarization, which is virtually compromised by the treatment with peroxisome proliferator activated receptor γ (PPARγ) inhibitor GW9662 and knockdown of PPARγ. Additionally, molecular dynamics simulation and immunofluorescence exhibit QU directly binds to PPARγ and increases PPARγ nuclear translocation. Besides, QU confers its moderation effect on suppressor of cytokine signaling protein (SOCS3)/signal transducer and activator of transcription 3 (STAT3) axis partially depending on PPARγ. Furthermore, coimmunoprecipitation shows QU redistributes the corepressor silencing mediator for retinoid and thyroid-hormone receptors (SMRT) from PPARγ to STAT3. Finally, the inhibition of Th17 response and the antiarthritic effect of QU are nullified by GW9662 treatment in arthritic mice. CONCLUSION QU targets PPARγ and consequently inhibits Th17 cell differentiation by dual inhibitory activity of STAT3 to exert antiarthritic effect. The findings facilitate its development and put forth a stage for uncovering the mechanism of other naturally occurring compounds with chemical structures similar to QU.
Collapse
Affiliation(s)
- Yan Yang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Department of Pharmacy, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Gao-Na Shi
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xin Wu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Min Xu
- Department of Pharmacy, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Cheng-Juan Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yu Zhou
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ya-Zi Wei
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lei Wu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fen-Fang Cui
- Department of Pharmacy, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Lan Sun
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Tian-Tai Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
5
|
Xiao L, Loh YP. Neurotrophic Factor-α1/Carboxypeptidase E Functions in Neuroprotection and Alleviates Depression. Front Mol Neurosci 2022; 15:918852. [PMID: 35711734 PMCID: PMC9197069 DOI: 10.3389/fnmol.2022.918852] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Depression is a major psychiatric disease affecting all ages and is often co-morbid with neurodegeneration in the elderly. Depression and neurodegeneration are associated with decreased neurotrophic factors. In this mini-review the functions and potential therapeutic use of a newly discovered trophic factor, Neurotrophic factor-α1 (NF-α1), also known as Carboxypeptidase E (CPE), in depression and neuroprotection are discussed. NF-α1/CPE expression is enriched in CA3 neurons of the hippocampus. Families carrying null and homozygous non-sense mutations of the NF-α1/CPE gene share common clinical features including childhood onset obesity, type 2 diabetes, impaired intellectual abilities and hypogonadotrophic hypogonadism. Studies in animal models such as CPE knockout (KO) mice and CPEfat/fat mutant mice exhibit similar phenotypes. Analysis of CPE-KO mouse brain revealed that hippocampal CA3 was completely degenerated after weaning stress, along with deficits in hippocampal long-term potentiation. Carbamazepine effectively blocked weaning stress-induced hippocampal CA3 degeneration, suggesting the stress induced epileptic-like neuronal firing led to the degeneration. Analysis of possible mechanisms underlying NF-α1/CPE -mediated neuroprotection revealed that it interacts with the serotonin receptor, 5-HTR1E, and via β arrestin activation, subsequently upregulates ERK1/2 signaling and pro-survival protein, BCL2, levels. Furthermore, the NF-α1/CPE promoter contains a peroxisome proliferator-activated receptor (PPARγ) binding site which can be activated by rosiglitazone, a PPARγ agonist, to up-regulate expression of NF-α1/CPE and neurogenesis, resulting in anti-depression in animal models. Rosiglitazone, an anti-diabetic drug administered to diabetic patients resulted in decline of depression. Thus, NF-α1/CPE is a potential therapeutic agent or drug target for treating depression and neurodegenerative disorders.
Collapse
|
6
|
PPARγ Dysfunction in the Medial Prefrontal Cortex Mediates High-Fat Diet-Induced Depression. Mol Neurobiol 2022; 59:4030-4043. [DOI: 10.1007/s12035-022-02806-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/16/2022] [Indexed: 11/25/2022]
|
7
|
Wang B, Chen D, Jiang R, Ntim M, Lu J, Xia M, Yang X, Wang Y, Kundu S, Guan R, Li S. TIP60 buffers acute stress response and depressive behaviour by controlling PPARγ-mediated transcription. Brain Behav Immun 2022; 101:410-422. [PMID: 35114329 DOI: 10.1016/j.bbi.2022.01.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/29/2021] [Accepted: 01/25/2022] [Indexed: 12/23/2022] Open
Abstract
Tat-interacting protein 60 (TIP60) as nuclear receptors (NRs) coregulator, acts as a tumor suppressor and also has promising therapeutic potential to target Alzheimer's disease. Stress has been implicated in many psychiatric disorders, and these disorders are characterized by impairments in cognitive function. Until now, there are no experimental data available on the regulatory effect of TIP60 in acute stress and depression. There is also no definitive explanation on which specific modulation of target gene expression is achieved by TIP60. Here, we identify TIP60 as a novel positive regulator in response to acute restraint stress (ARS) and a potentially effective target of antidepressants. Firstly, we discovered increased hippocampal TIP60 expressions in the ARS model. Furthermore, using the TIP60 inhibitor, MG149, we proved that TIP60 function correlates with behavioral and synaptic activation in the two-hour ARS. Secondly, the lentivirus vector (LV)-TIP60overexpression (OE) was injected into the hippocampus prior to the chronic restraint stress (CRS) experiments and it was found that over-expressed TIP60 compensates for TIP60 decrease and improves depression index in CRS. Thirdly, through the intervention of TIP60 expression in vitro, we established the genetic regulation of TIP60 on synaptic proteins, confirmed the TIP60 function as a specific coactivator for PPARγ and found that the PPARγ-mediated TIP60 function modulates transcriptional activation of synaptic proteins. Finally, the LV-TIP60OE and PPARγ antagonist, GW9662, were both administered in the CRS model and the data indicated that blocking PPARγ significantly weakened the protective effect of TIP60 against the CRS-induced depression. Conclusively, these findings together support TIP60 as a novel positive factor in response to acute stress and interacts with PPARγ to modulate the pathological mechanism of CRS-induced depression.
Collapse
Affiliation(s)
- Bin Wang
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning, China
| | - Defang Chen
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning, China
| | - Rong Jiang
- Department of Physiology, Binzhou Medical University, Yantai Campus, 346 Guanhai Road, Laishan District, Yantai, Shandong, China
| | - Michael Ntim
- Department of Physiology, School of Medicine and Dentistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Jincheng Lu
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning, China
| | - Min Xia
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning, China
| | - XueWei Yang
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning, China
| | - Ying Wang
- Department of Cardiology, Institute of Heart and Vessel Diseases of Dalian Medical University, the Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Supratik Kundu
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning, China
| | - Rongxiao Guan
- Department of Cardiology, Institute of Heart and Vessel Diseases of Dalian Medical University, the Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Shao Li
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
8
|
Sexton HG, Olszewski NA, Risher ML. The Effects of Rosiglitazone on Task Specific Anxiety-Like Behavior and Novelty Seeking in a Model of Chronic Adolescent Unpredictable Stress. Front Behav Neurosci 2022; 16:830310. [PMID: 35221947 PMCID: PMC8874210 DOI: 10.3389/fnbeh.2022.830310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/20/2022] [Indexed: 12/02/2022] Open
Abstract
Adolescence is characterized as a period of increased social behavior, risk taking, and novelty seeking, partly due to ongoing maturation in critical brain areas and the hypothalamic-pituitary-adrenal (HPA) negative-feedback system. During this period there is heightened vulnerability to stress that can drive neuro-immune-endocrine remodeling, resulting in the emergence of maladaptive behaviors that increase susceptibility to alcohol and substance abuse. Here we used a rat model to investigate the impact of chronic adolescent unpredictable stress on a battery of behavioral measures to assess anxiety, novelty seeking, risk taking, depression, and voluntary ethanol consumption while determining whether the PPARγ agonist rosiglitazone can attenuate these effects. Adolescent female rats that experienced stress showed increased risk taking behavior and novelty seeking behavior with no change in ethanol consumption. The administration of rosiglitazone during stress induction attenuated stress-induced cortisol elevation, normalized risk taking behavior in a model anxiety, and attenuated novelty seeking in a task-specific manner. Depressive-like behavior was not impacted by adolescent unpredictable stress or the administration of rosiglitazone. The results from this study demonstrate that exposure to unpredictable stress during adolescence increases the prevalence of maladaptive behaviors that are known to increase susceptibility to alcohol and substance abuse, and that rosiglitazone may be an effective therapeutic to attenuate the emergence of select risk taking and novelty seeking behaviors in females.
Collapse
Affiliation(s)
- Hannah G. Sexton
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
- Hershel ‘Woody’ Williams Veterans Affairs Medical Center, Huntington, WV, United States
| | - Nathan A. Olszewski
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
| | - Mary-Louise Risher
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States
- Hershel ‘Woody’ Williams Veterans Affairs Medical Center, Huntington, WV, United States
| |
Collapse
|
9
|
Tsyglakova M, Huskey AM, Hurst EH, Telep NM, Wilding MC, Babington ME, Rainville JR, Hodes GE. Sex and region-specific effects of variable stress on microglia morphology. Brain Behav Immun Health 2021; 18:100378. [PMID: 34820640 PMCID: PMC8600001 DOI: 10.1016/j.bbih.2021.100378] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 12/25/2022] Open
Abstract
Major Depressive Disorder (MDD) is a common and debilitating mood disorder that is more prevalent in women than men. In humans, PET imaging of microglia activation is currently being explored as a potential biomarker of MDD and suicidal ideation. Stress is a trigger for many mood disorders, including MDD. Microglial changes in morphology and activation state in response to stress has been reported in various brain regions, but most studies only examined male subjects. Here we report changes in microglia morphology in the nucleus accumbens (NAc) and subregions of the hippocampus (HPC) in both male and female mice following variable stress of 6 or 28 days in duration. Our data demonstrate that after 6 days of stress, microglia in the female NAc and dentate gyrus have a reduction in homeostatic associated morphology and an increase in primed microglia. After 28 days some of these sex specific stress effects were still present in microglia within the NAc but not the dentate gyrus. There were no effects of stress in either sex at either timepoint in CA1. In female mice, anti-inflammatory activation of microglia using rosiglitazone promoted sociability behavior after 6 days of stress. Furthermore, both drug and stress have impact on microglia morphology and activation state in the NAc. These data suggest that microglia morphology and activation state are altered by 6 days of variable stress in a region-specific manner and may contribute to, or potentially compensate for, the onset of stress susceptibility rather than impacting long term exposure to stress.
Collapse
Affiliation(s)
- Mariya Tsyglakova
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
- Graduate Program in Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Alisa M. Huskey
- Department of Psychology, University of Arizona, Tucson, AZ, USA
| | - Emily H. Hurst
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Natalie M. Telep
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Mary C. Wilding
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Meghan E. Babington
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Jennifer R. Rainville
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Georgia E. Hodes
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| |
Collapse
|
10
|
Repurposing Peroxisome Proliferator-Activated Receptor Agonists in Neurological and Psychiatric Disorders. Pharmaceuticals (Basel) 2021; 14:ph14101025. [PMID: 34681249 PMCID: PMC8538250 DOI: 10.3390/ph14101025] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 12/15/2022] Open
Abstract
Common pathophysiological mechanisms have emerged for different neurological and neuropsychiatric conditions. In particular, mechanisms of oxidative stress, immuno-inflammation, and altered metabolic pathways converge and cause neuronal and non-neuronal maladaptative phenomena, which underlie multifaceted brain disorders. The peroxisome proliferator-activated receptors (PPARs) are nuclear receptors modulating, among others, anti-inflammatory and neuroprotective genes in diverse tissues. Both endogenous and synthetic PPAR agonists are approved treatments for metabolic and systemic disorders, such as diabetes, fatty liver disease, and dyslipidemia(s), showing high tolerability and safety profiles. Considering that some PPAR-acting drugs permeate through the blood-brain barrier, the possibility to extend their scope from the periphery to central nervous system has gained interest in recent years. Here, we review preclinical and clinical evidence that PPARs possibly exert a neuroprotective role, thereby providing a rationale for repurposing PPAR-targeting drugs to counteract several diseases affecting the central nervous system.
Collapse
|
11
|
Jones BDM, Farooqui S, Kloiber S, Husain MO, Mulsant BH, Husain MI. Targeting Metabolic Dysfunction for the Treatment of Mood Disorders: Review of the Evidence. Life (Basel) 2021; 11:819. [PMID: 34440563 PMCID: PMC8401631 DOI: 10.3390/life11080819] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 12/28/2022] Open
Abstract
Major depressive disorder (MDD) and bipolar disorder (BD) are often chronic with many patients not responding to available treatments. As these mood disorders are frequently associated with metabolic dysfunction, there has been increased interest in novel treatments that would target metabolic pathways. The objectives of this scoping review were to synthesize evidence on the impact on mood symptoms of lipid lowering agents and anti-diabetics drugs, while also reviewing current knowledge on the association between mood disorders and dyslipidemia or hyperglycemia. We propose that metabolic dysfunction is prevalent in both MDD and BD and it may contribute to the development of these disorders through a variety of pathophysiological processes including inflammation, brain structural changes, hormonal alterations, neurotransmitter disruptions, alteration on brain cholesterol, central insulin resistance, and changes in gut microbiota. Current evidence is conflicting on the use of statins, polyunsaturated fatty acids, thiazolidinediones, glucagon-like peptide agonists, metformin, or insulin for the treatment of MDD and BD. Given the paucity of high-quality randomized controlled trials, additional studies are needed before any of these medications can be repurposed in routine clinical practice. Future trials need to enrich patient recruitment, include evaluations of mechanism of action, and explore differential effects on specific symptom domains such as anhedonia, suicidality, and cognition.
Collapse
Affiliation(s)
- Brett D. M. Jones
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada; (B.D.M.J.); (S.K.); (M.O.H.); (B.H.M.)
- Centre for Addiction and Mental Health, Toronto, ON M6J 1H4, Canada
| | - Salman Farooqui
- Department of Biology, University of Ottawa, Ottawa, ON K1N 6N5, Canada;
| | - Stefan Kloiber
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada; (B.D.M.J.); (S.K.); (M.O.H.); (B.H.M.)
- Centre for Addiction and Mental Health, Toronto, ON M6J 1H4, Canada
| | - Muhammad Omair Husain
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada; (B.D.M.J.); (S.K.); (M.O.H.); (B.H.M.)
- Centre for Addiction and Mental Health, Toronto, ON M6J 1H4, Canada
| | - Benoit H. Mulsant
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada; (B.D.M.J.); (S.K.); (M.O.H.); (B.H.M.)
- Centre for Addiction and Mental Health, Toronto, ON M6J 1H4, Canada
| | - Muhammad Ishrat Husain
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada; (B.D.M.J.); (S.K.); (M.O.H.); (B.H.M.)
- Centre for Addiction and Mental Health, Toronto, ON M6J 1H4, Canada
| |
Collapse
|
12
|
Cosgrove KT, Kuplicki R, Savitz J, Burrows K, Simmons WK, Khalsa SS, Teague TK, Aupperle RL, Paulus MP. Impact of ibuprofen and peroxisome proliferator-activated receptor gamma on emotion-related neural activation: A randomized, placebo-controlled trial. Brain Behav Immun 2021; 96:135-142. [PMID: 34052365 PMCID: PMC8319138 DOI: 10.1016/j.bbi.2021.05.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 05/08/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) such as ibuprofen have shown initial promise in producing antidepressant effects. This is perhaps due to these drugs being peroxisome proliferator-activated receptor gamma (PPARγ) agonists, in addition to their inhibition of cyclooxygenase enzymes. Some, albeit mixed, evidence suggests that PPARγ agonists have antidepressant effects in humans and animals. This double-blind, placebo-controlled, pharmacologic functional magnetic resonance imaging (ph-fMRI) study aimed to elucidate the impact of ibuprofen on emotion-related neural activity and determine whether observed effects were due to changes in PPARγ gene expression. Twenty healthy volunteers completed an emotional face matching task during three fMRI sessions, conducted one week apart. Placebo, 200 mg, or 600 mg ibuprofen was administered 1 h prior to each scan in a pseudo-randomized order. Peripheral blood mononuclear cells were collected at each session to isolate RNA for PPARγ gene expression. At the doses used, ibuprofen did not significantly change PPARγ gene expression. Ibuprofen dose was associated with decreased blood oxygen level-dependent (BOLD) activation in the dorsolateral prefrontal cortex and fusiform gyrus during emotional face processing (faces-shapes). Additionally, PPARγ gene expression was associated with increased BOLD activation in the insula and transverse and superior temporal gyri (faces-shapes). No interaction effects between ibuprofen dose and PPARγ gene expression on BOLD activation were observed. Thus, results suggest that ibuprofen and PPARγ may have independent effects on emotional neurocircuitry. Future studies are needed to further delineate the roles of ibuprofen and PPARγ in exerting antidepressant effects in healthy as well as clinical populations.
Collapse
Affiliation(s)
- Kelly T. Cosgrove
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136,Department of Psychology, University of Tulsa, 800 S. Tucker Dr., Tulsa, OK, 74104
| | - Rayus Kuplicki
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK 74136 USA.
| | - Jonathan Savitz
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK 74136 USA.
| | - Kaiping Burrows
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK 74136 USA.
| | - W. Kyle Simmons
- Center for Health Sciences, Oklahoma State University, 1013 E 66th Pl, Tulsa, OK 74136
| | - Sahib S. Khalsa
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136,School of Community Medicine, University of Tulsa, 800 S. Tucker Dr., Tulsa, OK, 74104
| | - T. Kent Teague
- School of Community Medicine, University of Oklahoma, 4502 E. 41st St., Tulsa, OK, 74135
| | - Robin L. Aupperle
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136,School of Community Medicine, University of Tulsa, 800 S. Tucker Dr., Tulsa, OK, 74104
| | - Martin P. Paulus
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136,School of Community Medicine, University of Tulsa, 800 S. Tucker Dr., Tulsa, OK, 74104
| |
Collapse
|
13
|
Peroxisome proliferator-activated receptor gamma: a novel therapeutic target for cognitive impairment and mood disorders that functions via the regulation of adult neurogenesis. Arch Pharm Res 2021; 44:553-563. [PMID: 34138417 DOI: 10.1007/s12272-021-01333-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022]
Abstract
The proliferation, differentiation, and migration of neural precursor cells occur not only during embryonic development but also within distinct regions of the adult brain through the process of adult neurogenesis. As neurogenesis can potentially regulate brain cognition and neuronal plasticity, the factors that enhance neurogenesis can be attractive therapeutic targets for improving cognitive function and regulating neurodegenerative and neuropsychiatric disorders, including affective and mood disorders. Peroxisome proliferator-activated receptors (PPARs) are a class of ligand-activated transcription factors belonging to the nuclear receptor superfamily. PPARγ is a target for insulin sensitizers and plays an essential role in regulating various metabolic processes, including adipogenesis and glucose homeostasis. Interestingly, evidence demonstrates the role of PPARγ activation in regulating neurogenesis. The pharmacological activation of PPARγ using specific ligands increases the proliferation and differentiation of neural stem cells in specific brain regions, including the hippocampus, and prevents neurodegeneration and improves cognition and anxiety/depression-like behaviors in animal models. We summarize here recent reports on the role of PPARγ in adult neurogenesis, as well as the mechanisms involved, and suggest that PPARγ can serve as a potential therapeutic target for neurological and/or neurodegenerative diseases.
Collapse
|
14
|
Rawlinson S, Andrews ZB. Hypothalamic insulin signalling as a nexus regulating mood and metabolism. J Neuroendocrinol 2021; 33:e12939. [PMID: 33634518 DOI: 10.1111/jne.12939] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/05/2021] [Accepted: 01/11/2021] [Indexed: 01/23/2023]
Abstract
Insulin has long been known as a metabolic hormone critical in the treatment of diabetes for its peripheral effects on blood glucose. However, in the last 50 years, insulin has entered the realm of neuroendocrinology and many studies have described its function on insulin receptors in the brain in relation to both metabolic and mood disorders. Indeed, rodent models of impaired insulin signalling show signs of dysregulated energy and glucose homeostasis, as well as anxiety-like and depressive behaviours. Importantly, many metabolic diseases such as obesity and diabetes increase the risk of developing mood disorders; however, the brain mechanisms underlying the connection between metabolism and mood remain unresolved. We present the current literature on the importance of the insulin receptor with respect to regulating glucose and energy homeostasis and mood-related behaviours. Specifically, we hypothesise that the insulin receptor in the hypothalamus, classically known as the homeostatic centre of the brain, plays a causal role in linking metabolic and behavioural effects of insulin signalling. In this review, we discuss insulin signalling in the hypothalamus as a critical point of neural integration controlling metabolism and mood.
Collapse
Affiliation(s)
- Sasha Rawlinson
- Department of Physiology, Monash Biomedicine Discovery Institute Monash University, Clayton, VIC, Australia
| | - Zane B Andrews
- Department of Physiology, Monash Biomedicine Discovery Institute Monash University, Clayton, VIC, Australia
| |
Collapse
|
15
|
Drug repositioning for treatment-resistant depression: Hypotheses from a pharmacogenomic study. Prog Neuropsychopharmacol Biol Psychiatry 2021; 104:110050. [PMID: 32738352 DOI: 10.1016/j.pnpbp.2020.110050] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/20/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023]
Abstract
About 20-30% of patients with major depressive disorder (MDD) develop treatment-resistant depression (TRD) and finding new effective treatments for TRD has been a challenge. This study aimed to identify new possible pharmacological options for TRD. Genes in pathways included in predictive models of TRD in a previous whole exome sequence study were compared with those coding for targets of drugs in any phase of development, nutraceuticals, proteins and peptides from Drug repurposing Hub, Drug-Gene Interaction database and DrugBank database. We tested if known gene targets were enriched in TRD-associated genes by a hypergeometric test. Compounds enriched in TRD-associated genes after false-discovery rate (FDR) correction were annotated and compared with those showing enrichment in genes associated with MDD in the last Psychiatric Genomics Consortium genome-wide association study. Among a total of 15,475 compounds, 542 were enriched in TRD-associated genes (FDR p < .05). Significant results included drugs which are currently used in TRD (e.g. lithium and ketamine), confirming the rationale of this approach. Interesting molecules included modulators of inflammation, renin-angiotensin system, proliferator-activated receptor agonists, glycogen synthase kinase 3 beta inhibitors and the rho associated kinase inhibitor fasudil. Nutraceuticals, mostly antioxidant polyphenols, were also identified. Drugs showing enrichment for TRD-associated genes had a higher probability of enrichment for MDD-associated genes compared to those having no TRD-genes enrichment (p = 6.21e-55). This study suggested new potential treatments for TRD using a in silico approach. These analyses are exploratory only but can contribute to the identification of drugs to study in future clinical trials.
Collapse
|
16
|
Estrada-Camarena EM, López-Rubalcava C, Ramírez-Rodríguez GB, Pulido D, Cervantes-Anaya N, Azpilcueta-Morales G, Granados-Juárez A, Vega-Rivera NM, Islas-Preciado D, Treviño S, de Gortari P, González-Trujano ME, García-Viguera C. Aqueous extract of pomegranate enriched in ellagitannins prevents anxiety-like behavior and metabolic changes induced by cafeteria diet in an animal model of menopause. Neurochem Int 2020; 141:104876. [PMID: 33049337 DOI: 10.1016/j.neuint.2020.104876] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 09/29/2020] [Accepted: 10/06/2020] [Indexed: 01/15/2023]
Abstract
Women around menopause are vulnerable to present psychiatric and metabolic disorders; thus, therapies that contribute to treat both pathologies are required. Previous reports showed that an aqueous extract of pomegranate (Punica granatum), enriched in ellagitannins, exerts an antidepressant-like effect in ovariectomized rats. We analyze whether this aqueous extract of P. granatum (AE-PG) prevents the anxiety-like behavior induced by a cafeteria diet (CAF) in middle-aged ovariectomized rats at the same time that it prevents an increase in body weight, glucose, lipids, and the changes on mRNA expression of the peroxisome proliferator-activated receptor-gamma (PPAR-γ) in the liver. Also, the effects of AE-PG on the protein levels of PPAR-γphospho-PPAR-γ, extracellular signal-regulated protein kinase (ERK1/2) and phospho-ERK1/2 were measured in the hippocampus and amygdala. CAF induced anxiety-like behavior, augmented lipids and glucose blood levels, body weight, visceral fat, insulin resistance, and decreased mRNA expression of PPAR-γ in the liver. In rats fed with the CAF, AE-PG prevented the anxiety-like behavior, reduced body weight, lowered lipid levels, reduced insulin resistance, and increased PPAR-γ mRNA expression in the liver. In the hippocampus, ERK1/2 but not PPAR-γ protein levels were decreased by CAF, while AE-PG prevented these effects. In the amygdala, CAF increased the phosphorylation of PPARγ, and AE-PG prevented it. In contrast, AE-PG rescued the decreased ERK1/2 protein level in the hippocampus caused by CAF. In conclusion, AE-PG treatment prevented anxiogenic and metabolic effects induced by CAF, and its effects appear to be mediated by ERK1/2 and PPARγ depending on the brain area studied.
Collapse
Affiliation(s)
- E M Estrada-Camarena
- Laboratorio de Neuropsicofarmacología. Dirección de Neurociencias. Instituto Nacional de Psiquiatría "Ramón de la Fuente", Mexico City, Mexico.
| | - C López-Rubalcava
- Departamento de Farmacobiología, Centro de Investigación y Estudios Avanzados (Sede Sur-Coapa), Mexico City, Mexico
| | - G B Ramírez-Rodríguez
- Laboratorio de Neurogénesis. Subdirección de Investigaciones Clínicas. Dirección de Neurociencias. Instituto Nacional de Psiquiatría "Ramón de la Fuente". Mexico City, Mexico
| | - D Pulido
- Laboratorio de Neuropsicofarmacología. Dirección de Neurociencias. Instituto Nacional de Psiquiatría "Ramón de la Fuente", Mexico City, Mexico
| | - N Cervantes-Anaya
- Laboratorio de Neuropsicofarmacología. Dirección de Neurociencias. Instituto Nacional de Psiquiatría "Ramón de la Fuente", Mexico City, Mexico
| | - G Azpilcueta-Morales
- Laboratorio de Neuropsicofarmacología. Dirección de Neurociencias. Instituto Nacional de Psiquiatría "Ramón de la Fuente", Mexico City, Mexico
| | - A Granados-Juárez
- Laboratorio de Neurogénesis. Subdirección de Investigaciones Clínicas. Dirección de Neurociencias. Instituto Nacional de Psiquiatría "Ramón de la Fuente". Mexico City, Mexico
| | - N M Vega-Rivera
- Laboratorio de Neuropsicofarmacología. Dirección de Neurociencias. Instituto Nacional de Psiquiatría "Ramón de la Fuente", Mexico City, Mexico
| | - D Islas-Preciado
- Laboratorio de Neuropsicofarmacología. Dirección de Neurociencias. Instituto Nacional de Psiquiatría "Ramón de la Fuente", Mexico City, Mexico
| | - S Treviño
- Facultad de Ciencias Químicas. Benemérita Universidad de Puebla. Puebla, Mexico
| | - P de Gortari
- Laboratorio de Neurofisiología Molecular. Dirección de Neurociencias. Dirección de Neurociencias. Instituto Nacional de Psiquiatría "Ramón de la Fuente", Mexico City, Mexico
| | - M E González-Trujano
- Laboratorio de Neurofarmacología de Productos Naturales. Dirección de Neurociencias. Instituto Nacional de Psiquiatría "Ramón de la Fuente". Mexico City, Mexico
| | - C García-Viguera
- Department of Food Science and Technology, CEBAS-CSIC, Murcia, Spain
| |
Collapse
|
17
|
Tufano M, Pinna G. Is There a Future for PPARs in the Treatment of Neuropsychiatric Disorders? Molecules 2020; 25:molecules25051062. [PMID: 32120979 PMCID: PMC7179196 DOI: 10.3390/molecules25051062] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 02/07/2023] Open
Abstract
Recently, peroxisome proliferator-activated receptor (PPAR)-α and γ isoforms have been gaining consistent interest in neuropathology and treatment of neuropsychiatric disorders. Several studies have provided evidence that either the receptor expression or the levels of their endogenously-produced modulators are downregulated in several neurological and psychiatric disorders and in their respective animal models. Remarkably, administration of these endogenous or synthetic ligands improves mood and cognition, suggesting that PPARs may offer a significant pharmacological target to improve several neuropathologies. Furthermore, various neurological and psychiatric disorders reflect sustained levels of systemic inflammation. Hence, the strategy of targeting PPARs for their anti-inflammatory role to improve these disorders is attracting attention. Traditionally, classical antidepressants fail to be effective, specifically in patients with inflammation. Non-steroidal anti-inflammatory drugs exert potent antidepressant effects by acting along with PPARs, thereby strongly substantiating the involvement of these receptors in the mechanisms that lead to development of several neuropathologies. We reviewed running findings in support of a role for PPARs in the treatment of neurological diseases, including Alzheimer's disease or psychiatric disorders, such as major depression. We discuss the opportunity of targeting PPARs as a future pharmacological approach to decrease neuropsychiatric symptoms at the same time that PPAR ligands resolve neuroinflammatory processes.
Collapse
Affiliation(s)
| | - Graziano Pinna
- Correspondence: or ; Tel.: +1-312-355-1464; Fax: +1-312-413-4569
| |
Collapse
|
18
|
Aguiniga LM, Searl TJ, Rahman-Enyart A, Yaggie RE, Yang W, Schaeffer AJ, Klumpp DJ. Acyloxyacyl hydrolase regulates voiding activity. Am J Physiol Renal Physiol 2020; 318:F1006-F1016. [PMID: 32003596 DOI: 10.1152/ajprenal.00442.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Corticotropin-releasing factor (CRF) regulates diverse physiological functions, including bladder control. We recently reported that Crf expression is under genetic control of Aoah, the locus encoding acyloxyacyl hydrolase (AOAH), suggesting that AOAH may also modulate voiding. Here, we examined the role of AOAH in bladder function. AOAH-deficient mice exhibited enlarged bladders relative to wild-type mice and had decreased voiding frequency and increased void volumes. AOAH-deficient mice had increased nonvoiding contractions and increased peak voiding pressure in awake cystometry. AOAH-deficient mice also exhibited increased bladder permeability and higher neuronal firing rates of bladder afferents in response to stretch. In wild-type mice, AOAH was expressed in bladder projecting neurons and colocalized in CRF-expressing neurons in Barrington's nucleus, an important brain area for voiding behavior, and Crf was elevated in Barrington's nucleus of AOAH-deficient mice. We had previously identified aryl hydrocarbon receptor (AhR) and peroxisome proliferator-activated receptor-γ as transcriptional regulators of Crf, and conditional knockout of AhR or peroxisome proliferator-activated receptor-γ in Crf-expressing cells restored normal voiding in AOAH-deficient mice. Finally, an AhR antagonist improved voiding in AOAH-deficient mice. Together, these data demonstrate that AOAH regulates bladder function and that the AOAH-Crf axis is a therapeutic target for treating voiding dysfunction.
Collapse
Affiliation(s)
- Lizath M Aguiniga
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Timothy J Searl
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Afrida Rahman-Enyart
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Ryan E Yaggie
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Wenbin Yang
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Anthony J Schaeffer
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - David J Klumpp
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
19
|
Chávez-Castillo M, Nava M, Ortega Á, Rojas M, Núñez V, Salazar J, Bermúdez V, Rojas-Quintero J. Depression as an Immunometabolic Disorder: Exploring Shared Pharmacotherapeutics with Cardiovascular Disease. Curr Neuropharmacol 2020; 18:1138-1153. [PMID: 32282306 PMCID: PMC7709154 DOI: 10.2174/1570159x18666200413144401] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/04/2020] [Accepted: 04/16/2020] [Indexed: 12/12/2022] Open
Abstract
Modern times have seen depression and cardiovascular disease (CVD) become notorious public health concerns, corresponding to alarming proportions of morbidity, mortality, decreased quality of life, and economic costs. Expanding comprehension of the pathogenesis of depression as an immunometabolic disorder has identified numerous pathophysiologic phenomena in common with CVD, including chronic inflammation, insulin resistance, and oxidative stress. These shared components could be exploited to offer improved alternatives in the joint management of these conditions. Abundant preclinical and clinical data on the impact of established treatments for CVD in the management of depression have allowed for potential candidates to be proposed for the joint management of depression and CVD as immunometabolic disorders. However, a large proportion of the clinical investigation currently available exhibits marked methodological flaws which preclude the formulation of concrete recommendations in many cases. This situation may be a reflection of pervasive problems present in clinical research in psychiatry, especially pertaining to study homogeneity. Therefore, further high-quality research is essential in the future in this regard.
Collapse
Affiliation(s)
| | | | | | | | | | - Juan Salazar
- Address correspondence to this author at the Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 20th Avenue 4004, Venezuela; Tel/Fax: ++582617597279; E-mail:
| | | | | |
Collapse
|
20
|
Fakhfouri G, Rahimian R, Dyhrfjeld-Johnsen J, Zirak MR, Beaulieu JM. 5-HT 3 Receptor Antagonists in Neurologic and Neuropsychiatric Disorders: The Iceberg Still Lies beneath the Surface. Pharmacol Rev 2019; 71:383-412. [PMID: 31243157 DOI: 10.1124/pr.118.015487] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
5-HT3 receptor antagonists, first introduced to the market in the mid-1980s, are proven efficient agents to counteract chemotherapy-induced emesis. Nonetheless, recent investigations have shed light on unappreciated dimensions of this class of compounds in conditions with an immunoinflammatory component as well as in neurologic and psychiatric disorders. The promising findings from multiple studies have unveiled several beneficial effects of these compounds in multiple sclerosis, stroke, Alzheimer disease, and Parkinson disease. Reports continue to uncover important roles for 5-HT3 receptors in the physiopathology of neuropsychiatric disorders, including depression, anxiety, drug abuse, and schizophrenia. This review addresses the potential of 5-HT3 receptor antagonists in neurology- and neuropsychiatry-related disorders. The broad therapeutic window and high compliance observed with these agents position them as suitable prototypes for the development of novel pharmacotherapeutics with higher efficacy and fewer adverse effects.
Collapse
Affiliation(s)
- Gohar Fakhfouri
- Department of Psychiatry and Neuroscience, Faculty of Medicine, CERVO Brain Research Centre, Laval University, Quebec, Quebec, Canada (G.F., R.R.); Sensorion SA, Montpellier, France (J.D.-J.); Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran (M.R.Z.); and Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (J.-M.B.)
| | - Reza Rahimian
- Department of Psychiatry and Neuroscience, Faculty of Medicine, CERVO Brain Research Centre, Laval University, Quebec, Quebec, Canada (G.F., R.R.); Sensorion SA, Montpellier, France (J.D.-J.); Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran (M.R.Z.); and Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (J.-M.B.)
| | - Jonas Dyhrfjeld-Johnsen
- Department of Psychiatry and Neuroscience, Faculty of Medicine, CERVO Brain Research Centre, Laval University, Quebec, Quebec, Canada (G.F., R.R.); Sensorion SA, Montpellier, France (J.D.-J.); Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran (M.R.Z.); and Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (J.-M.B.)
| | - Mohammad Reza Zirak
- Department of Psychiatry and Neuroscience, Faculty of Medicine, CERVO Brain Research Centre, Laval University, Quebec, Quebec, Canada (G.F., R.R.); Sensorion SA, Montpellier, France (J.D.-J.); Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran (M.R.Z.); and Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (J.-M.B.)
| | - Jean-Martin Beaulieu
- Department of Psychiatry and Neuroscience, Faculty of Medicine, CERVO Brain Research Centre, Laval University, Quebec, Quebec, Canada (G.F., R.R.); Sensorion SA, Montpellier, France (J.D.-J.); Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran (M.R.Z.); and Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada (J.-M.B.)
| |
Collapse
|
21
|
Ambrée O, Ruland C, Zwanzger P, Klotz L, Baune BT, Arolt V, Scheu S, Alferink J. Social Defeat Modulates T Helper Cell Percentages in Stress Susceptible and Resilient Mice. Int J Mol Sci 2019; 20:ijms20143512. [PMID: 31319604 PMCID: PMC6678569 DOI: 10.3390/ijms20143512] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/13/2019] [Accepted: 07/16/2019] [Indexed: 12/21/2022] Open
Abstract
Altered adaptive immunity involving T lymphocytes has been found in depressed patients and in stress-induced depression-like behavior in animal models. Peripheral T cells play important roles in homeostasis and function of the central nervous system and thus modulate behavior. However, the T cell phenotype and function associated with susceptibility and resilience to depression remain largely unknown. Here, we characterized splenic T cells in susceptible and resilient mice after 10 days of social defeat stress (SDS). We found equally decreased T cell frequencies and comparably altered expression levels of genes associated with T helper (Th) cell function in resilient and susceptible mice. Interleukin (IL)-17 producing CD4+ and CD8+ T cell numbers in the spleen were significantly increased in susceptible mice. These animals further exhibited significantly reduced numbers of regulatory T cells (Treg) and decreased gene expression levels of TGF-β. Mice with enhanced Th17 differentiation induced by conditional deletion of PPARγ in CD4+ cells (CD4-PPARγKO), an inhibitor of Th17 development, were equally susceptible to SDS when compared to CD4-PPARγWT controls. These data indicate that enhanced Th17 differentiation alone does not alter stress vulnerability. Thus, SDS promotes Th17 cell and suppresses Treg cell differentiation predominantly in susceptible mice with yet unknown effects in immune responses after stress exposure.
Collapse
Affiliation(s)
- Oliver Ambrée
- Department of Psychiatry, University of Münster, 48149 Münster, Germany.
- Department of Behavioural Biology, University of Osnabrück, 49076 Osnabrück, Germany.
| | - Christina Ruland
- Department of Psychiatry, University of Münster, 48149 Münster, Germany
| | - Peter Zwanzger
- kbo-Inn-Salzach-Klinikum, 83512 Wasserburg am Inn, Germany
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
| | - Luisa Klotz
- Department of Neurology, University of Münster, 49149 Münster, Germany
| | - Bernhard T Baune
- Department of Psychiatry, University of Münster, 48149 Münster, Germany
- Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Parkville, VIC 3010, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Volker Arolt
- Department of Psychiatry, University of Münster, 48149 Münster, Germany
| | - Stefanie Scheu
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Judith Alferink
- Department of Psychiatry, University of Münster, 48149 Münster, Germany.
- Cluster of Excellence EXC 1003, Cells in Motion, University of Münster, 48149 Münster, Germany.
| |
Collapse
|
22
|
Pomytkin I, Costa‐Nunes JP, Kasatkin V, Veniaminova E, Demchenko A, Lyundup A, Lesch K, Ponomarev ED, Strekalova T. Insulin receptor in the brain: Mechanisms of activation and the role in the CNS pathology and treatment. CNS Neurosci Ther 2018; 24:763-774. [PMID: 29691988 PMCID: PMC6489906 DOI: 10.1111/cns.12866] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/28/2018] [Accepted: 03/30/2018] [Indexed: 12/16/2022] Open
Abstract
While the insulin receptor (IR) was found in the CNS decades ago, the brain was long considered to be an insulin-insensitive organ. This view is currently revisited, given emerging evidence of critical roles of IR-mediated signaling in development, neuroprotection, metabolism, and plasticity in the brain. These diverse cellular and physiological IR activities are distinct from metabolic IR functions in peripheral tissues, thus highlighting region specificity of IR properties. This particularly concerns the fact that two IR isoforms, A and B, are predominantly expressed in either the brain or peripheral tissues, respectively, and neurons express exclusively IR-A. Intriguingly, in comparison with IR-B, IR-A displays high binding affinity and is also activated by low concentrations of insulin-like growth factor-2 (IGF-2), a regulator of neuronal plasticity, whose dysregulation is associated with neuropathologic processes. Deficiencies in IR activation, insulin availability, and downstream IR-related mechanisms may result in aberrant IR-mediated functions and, subsequently, a broad range of brain disorders, including neurodevelopmental syndromes, neoplasms, neurodegenerative conditions, and depression. Here, we discuss findings on the brain-specific features of IR-mediated signaling with focus on mechanisms of primary receptor activation and their roles in the neuropathology. We aimed to uncover the remaining gaps in current knowledge on IR physiology and highlight new therapies targeting IR, such as IR sensitizers.
Collapse
Affiliation(s)
- Igor Pomytkin
- Department of Advanced Cell TechnologiesInstitute of Regenerative MedicineSechenov First Moscow State Medical UniversityMoscowRussia
| | - João P. Costa‐Nunes
- Department of Normal PhysiologyLaboratory of Psychiatric NeurobiologyInstitute of Molecular MedicineSechenov First Moscow State Medical UniversityMoscowRussia
- Faculdade de Medicina de LisboaInstituto de Medicina MolecularUniversidade de LisboaLisboaPortugal
| | - Vladimir Kasatkin
- Dmitry Rogachev National Research Center of Pediatric Hematology, Oncology and ImmunologyMoscowRussia
| | - Ekaterina Veniaminova
- Department of Normal PhysiologyLaboratory of Psychiatric NeurobiologyInstitute of Molecular MedicineSechenov First Moscow State Medical UniversityMoscowRussia
- Laboratory of Cognitive DysfunctionsInstitute of General Pathology and PathophysiologyMoscowRussia
- Department of NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
| | - Anna Demchenko
- Department of Advanced Cell TechnologiesInstitute of Regenerative MedicineSechenov First Moscow State Medical UniversityMoscowRussia
| | - Alexey Lyundup
- Department of Advanced Cell TechnologiesInstitute of Regenerative MedicineSechenov First Moscow State Medical UniversityMoscowRussia
| | - Klaus‐Peter Lesch
- Department of Normal PhysiologyLaboratory of Psychiatric NeurobiologyInstitute of Molecular MedicineSechenov First Moscow State Medical UniversityMoscowRussia
- Department of NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
- Division of Molecular PsychiatryCenter of Mental HealthClinical Research Unit on Disorders of Neurodevelopment and CognitionUniversity of WürzburgWürzburgGermany
| | - Eugene D. Ponomarev
- Faculty of MedicineSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongHong Kong
| | - Tatyana Strekalova
- Department of Normal PhysiologyLaboratory of Psychiatric NeurobiologyInstitute of Molecular MedicineSechenov First Moscow State Medical UniversityMoscowRussia
- Laboratory of Cognitive DysfunctionsInstitute of General Pathology and PathophysiologyMoscowRussia
- Department of NeuroscienceMaastricht UniversityMaastrichtThe Netherlands
| |
Collapse
|
23
|
Song L, Wang H, Wang YJ, Wang JL, Zhu Q, Wu F, Zhang W, Jiang B. Hippocampal PPARα is a novel therapeutic target for depression and mediates the antidepressant actions of fluoxetine in mice. Br J Pharmacol 2018; 175:2968-2987. [PMID: 29722018 DOI: 10.1111/bph.14346] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 03/30/2018] [Accepted: 04/04/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND AND PURPOSE Developing novel pharmacological targets beyond the monoaminergic system is now a popular strategy for treating depression. PPARα is a nuclear receptor protein that functions as a transcription factor,-regulating gene expression. We have previously reported that both WY14643 and fenofibrate, two pharmacological agonists of PPARα, have antidepressant-like effects in mice, implying that PPARα is a potential antidepressant target. EXPERIMENTAL APPROACH We first used various biotechnological methods to evaluate the effects of chronic stress and fluoxetine on hippocampal PPARα. The viral-mediated genetic approach was then employed to explore whether hippocampal PPARα was an antidepressant target. PPARα inhibitors, PPARα-knockout (KO) mice and PPARα-knockdown (KD) mice were further used to determine the role of PPARα in the antidepressant effects of fluoxetine. KEY RESULTS Chronic stress significantly decreased mRNA and protein levels of PPARα in the hippocampus, but not other regions, and also fully reduced the recruitment of hippocampal PPARα to the cAMP response element-binding (CREB) promoter. Genetic overexpression of hippocampal PPARα induced significant antidepressant-like actions in mice by promoting CREB-mediated biosynthesis of brain-derived neurotrophic factor. Moreover, fluoxetine notably restored the stress-induced negative effects on hippocampal PPARα. Using PPARα antagonists fully blocked the antidepressant effects of fluoxetine in mice, and similarly, both PPARα-KO and PPARα-KD abolished the effects of fluoxetine. Besides, PPARα-KO and PPARα-KD aggravated depression in mice. CONCLUSIONS AND IMPLICATIONS Hippocampal PPARα is a potential novel antidepressant target that mediates the antidepressant actions of fluoxetine in mice.
Collapse
Affiliation(s)
- Lu Song
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China.,Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu, China
| | - Hao Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China.,Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu, China
| | - Ying-Jie Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China.,Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu, China
| | - Jin-Liang Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China.,Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu, China
| | - Qing Zhu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China.,Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu, China
| | - Feng Wu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China.,Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu, China
| | - Wei Zhang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China.,Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu, China
| | - Bo Jiang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China.,Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu, China
| |
Collapse
|
24
|
Zong J, Liao X, Ren B, Wang Z. The antidepressant effects of rosiglitazone on rats with depression induced by neuropathic pain. Life Sci 2018; 203:315-322. [DOI: 10.1016/j.lfs.2018.04.057] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 04/23/2018] [Accepted: 04/30/2018] [Indexed: 12/22/2022]
|
25
|
Adzic M, Brkic Z, Mitic M, Francija E, Jovicic MJ, Radulovic J, Maric NP. Therapeutic Strategies for Treatment of Inflammation-related Depression. Curr Neuropharmacol 2018; 16:176-209. [PMID: 28847294 PMCID: PMC5883379 DOI: 10.2174/1570159x15666170828163048] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 08/17/2017] [Accepted: 08/24/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Mounting evidence demonstrates enhanced systemic levels of inflammatory mediators in depression, indicating that inflammation may play a role in the etiology and course of mood disorders. Indeed, proinflammatory cytokines induce a behavioral state of conservation- withdrawal resembling human depression, characterized by negative mood, fatigue, anhedonia, psychomotor retardation, loss of appetite, and cognitive deficits. Neuroinflammation also contributes to non-responsiveness to current antidepressant (AD) therapies. Namely, response to conventional AD medications is associated with a decrease in inflammatory biomarkers, whereas resistance to treatment is accompanied by increased inflammation. METHODS In this review, we will discuss the utility and shortcomings of pharmacologic AD treatment strategies focused on inflammatory pathways, applied alone or as an adjuvant component to current AD therapies. RESULTS Mechanisms of cytokine actions on behavior involve activation of inflammatory pathways in the brain, resulting in changes of neurotransmitter metabolism, neuroendocrine function, and neuronal plasticity. Selective serotonin reuptake inhibitors exhibit the most beneficial effects in restraining the inflammation markers in depression. Different anti-inflammatory agents exhibit AD effects via modulating neurotransmitter systems, neuroplasticity markers and glucocorticoid receptor signaling. Anti-inflammatory add-on therapy in depression highlights such treatment as a candidate for enhancement strategy in patients with moderate-to-severe depression. CONCLUSION The interactions between the immune system and CNS are not only involved in shaping behavior, but also in responding to therapeutics. Even though, substantial evidence from animal and human research support a beneficial effect of anti-inflammatory add-on therapy in depression, further research with special attention on safety, particularly during prolonged periods of antiinflammatory co-treatments, is required.
Collapse
Affiliation(s)
- Miroslav Adzic
- Department of Molecular Biology and Endocrinology, VINCA Institute of Nuclear Sciences, University of Belgrade, Serbia
| | - Zeljka Brkic
- Department of Molecular Biology and Endocrinology, VINCA Institute of Nuclear Sciences, University of Belgrade, Serbia
| | - Milos Mitic
- Department of Molecular Biology and Endocrinology, VINCA Institute of Nuclear Sciences, University of Belgrade, Serbia
| | - Ester Francija
- Department of Molecular Biology and Endocrinology, VINCA Institute of Nuclear Sciences, University of Belgrade, Serbia
| | - Milica J. Jovicic
- Clinic for Psychiatry, Clinical Centre of Serbia, Pasterova 2, 11000, Belgrade, Serbia
| | - Jelena Radulovic
- Department of Psychiatry and Behavioral Sciences, The Asher Center of Study and Treatment of Depressive Disorders, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Nadja P. Maric
- Clinic for Psychiatry, Clinical Centre of Serbia, Pasterova 2, 11000, Belgrade, Serbia
- School of Medicine, University of Belgrade, Dr Subotica 8, 11000, Belgrade, Serbia
| |
Collapse
|
26
|
Wu Y, Wang L, Hu K, Yu C, Zhu Y, Zhang S, Shao A. Mechanisms and Therapeutic Targets of Depression After Intracerebral Hemorrhage. Front Psychiatry 2018; 9:682. [PMID: 30618863 PMCID: PMC6304443 DOI: 10.3389/fpsyt.2018.00682] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 11/23/2018] [Indexed: 12/17/2022] Open
Abstract
The relationship between depression and intracerebral hemorrhage (ICH) is complicated. One of the most common neuropsychiatric comorbidities of hemorrhagic stroke is Post-ICH depression. Depression, as a neuropsychiatric symptom, also negatively impacts the outcome of ICH by enhancing morbidity, disability, and mortality. However, the ICH outcome can be improved by antidepressants such as the frequently-used selective serotonin reuptake inhibitors. This review therefore presents the mechanisms of post-ICH depression, we grouped the mechanisms according to inflammation, oxidative stress (OS), apoptosis and autophagy, and explained them through their several associated signaling pathways. Inflammation is mainly related to Toll-like receptors (TLRs), the NF-kB mediated signal pathway, the PPAR-γ-dependent pathway, as well as other signaling pathways. OS is associated to nuclear factor erythroid-2 related factor 2 (Nrf2), the PI3K/Akt pathway and the MAPK/P38 pathway. Moreover, autophagy is associated with the mTOR signaling cascade and the NF-kB mediated signal pathway, while apoptosis is correlated with the death receptor-mediated apoptosis pathway, mitochondrial apoptosis pathway, caspase-independent pathways and others. Furthermore, we found that neuroinflammation, oxidative stress, autophagy, and apoptosis experience interactions with one another. Additionally, it may provide several potential therapeutic targets for patients that might suffer from depression after ICH.
Collapse
Affiliation(s)
- Yinan Wu
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Liangliang Wang
- Interdisciplinary Institute of Neuroscience and Technology, Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, China
| | - Kaimin Hu
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chengcheng Yu
- Department of Orthopedics, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuanhan Zhu
- Department of Neurosurgery, Rongjun Hospital, Jiaxing, China
| | - Suzhan Zhang
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
27
|
Insulin resistance, an unmasked culprit in depressive disorders: Promises for interventions. Neuropharmacology 2017; 136:327-334. [PMID: 29180223 DOI: 10.1016/j.neuropharm.2017.11.038] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 11/22/2017] [Accepted: 11/23/2017] [Indexed: 12/12/2022]
Abstract
Depressive disorders constitute a set of debilitating diseases with psychological, societal, economic and humanitarian consequences for millions of people worldwide. Scientists are beginning to understand the reciprocal communication between the brain and the rest of the body in the etiology of these diseases. In particular, scientists have noted a connection between depressive disorders, which are primarily seen as brain-based, and, insulin resistance (IR), a modifiable metabolic inflammatory state that is typically seen as peripheral. We highlight evidence showing how treating IR, with drugs or behavioral interventions, may ameliorate or possibly prevent, depressive disorders and their long-term consequences at various stages of the life course. This article is part of the Special Issue entitled 'Metabolic Impairment as Risk Factors for Neurodegenerative Disorders.'
Collapse
|
28
|
Liao L, Zhang X, Li J, Zhang Z, Yang C, Rao C, Zhou C, Zeng L, Zhao L, Fang L, Yang D, Xie P. Pioglitazone attenuates lipopolysaccharide-induced depression-like behaviors, modulates NF-κB/IL-6/STAT3, CREB/BDNF pathways and central serotonergic neurotransmission in mice. Int Immunopharmacol 2017; 49:178-186. [DOI: 10.1016/j.intimp.2017.05.036] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 05/21/2017] [Accepted: 05/31/2017] [Indexed: 11/16/2022]
|
29
|
Role of the adipose PPARγ-adiponectin axis in susceptibility to stress and depression/anxiety-related behaviors. Mol Psychiatry 2017; 22:1056-1068. [PMID: 27956741 PMCID: PMC5468488 DOI: 10.1038/mp.2016.225] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 08/31/2016] [Accepted: 10/27/2016] [Indexed: 02/06/2023]
Abstract
Adaptive responses to stressful stimuli involving behavioral, emotional and metabolic changes are orchestrated by the nervous and endocrine systems. Adipose tissue has been recognized as a highly active metabolic and endocrine organ, secreting adipokines that operate as hormones to mediate the crosstalk with other organs including the brain. The role of adipose tissue in sensing and responding to emotional stress and in behavioral regulation, however, remains largely unknown. The nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) is a key transcriptional factor controlling adipokine gene expression. Here we show that chronic social defeat stress decreases messenger RNA and protein levels of PPARγ in adipose tissue of susceptible but not resilient mice, which was correlated with social avoidance behavior. A corresponding reduction in adipose adiponectin production was observed in susceptible mice. Rosiglitazone, a blood-brain barrier-impermeant PPARγ-selective agonist, elicited antidepressant- and anxiolytic-like behavioral effects in wild-type mice, with a concurrent increase in plasma adiponectin levels. These effects of rosiglitazone were absent in mice lacking adiponectin but having normal PPARγ expression in adipose tissue and brain. Moreover, pretreatment with the PPARγ-selective antagonist GW9662 blocked rosiglitazone-induced adiponectin expression and antidepressant/anxiolytic-like effects. Together, these results suggest that the behavioral responses to rosiglitazone are mediated through PPARγ-dependent induction of adiponectin. Our findings support an important role for the adipose PPARγ-adiponectin axis in susceptibility to stress and negative emotion-related behaviors. Selectively targeting PPARγ in adipose tissue may offer novel strategies for combating depression and anxiety.
Collapse
|
30
|
The Role of Nuclear Hormone Receptors in Cannabinoid Function. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 80:291-328. [PMID: 28826538 DOI: 10.1016/bs.apha.2017.03.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Since the early 2000s, evidence has been accumulating that most cannabinoid compounds interact with the nuclear hormone family peroxisome proliferator-activated receptors (PPARs). This can be through direct binding of these compounds to PPARs, metabolism of cannabinoid to other PPAR-activating chemicals, or indirect activation of PPAR through cell signaling pathways. Delivery of cannabinoids to the nucleus may be facilitated by fatty acid-binding proteins and carrier proteins. All PPAR isoforms appear to be activated by cannabinoids, but the majority of evidence is for PPARα and γ. To date, little is known about the potential interaction of cannabinoids with other nuclear hormones. At least some (but not all) of the well-known biological actions of cannabinoids including neuroprotection, antiinflammatory action, and analgesic effects are partly mediated by PPAR-activation, often in combination with activation of the more traditional target sites of action. This has been best investigated for the endocannabinoid-like compounds palmitoylethanolamide and oleoylethanolamine acting at PPARα, and for phytocannabinoids or their derivatives activation acting at PPARγ. However, there are still many aspects of cannabinoid activation of PPAR and the role it plays in the biological and therapeutic effects of cannabinoids that remain to be investigated.
Collapse
|
31
|
SayuriYamagata A, Brietzke E, Rosenblat JD, Kakar R, McIntyre RS. Medical comorbidity in bipolar disorder: The link with metabolic-inflammatory systems. J Affect Disord 2017; 211:99-106. [PMID: 28107669 DOI: 10.1016/j.jad.2016.12.059] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 12/23/2016] [Accepted: 12/31/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND Bipolar disorder (BD) is associated with chronic low-grade inflammation, several medical comorbidities and a decreased life expectancy. Metabolic-inflammatory changes have been postulated as one of the main links between BD and medical comorbidity, although there are few studies exploring possible mechanisms underlying this relationship. Therefore, the aims of the current narrative review were 1) synthesize the evidence for metabolic-inflammatory changes that may facilitate the link between medical comorbidity and BD and 2) discuss therapeutic and preventive implications of these pathways. METHODS The PubMed and Google Scholar databases were searched for relevant studies. RESULTS Identified studies suggested that there is an increased risk of medical comorbidities, such as autoimmune disorders, obesity, diabetes and cardiovascular disease in patients with BD. The association between BD and general medical comorbidities seems to be bidirectional and potentially mediated by immune dysfunction. Targeting the metabolic-inflammatory-mood pathway may potential yield improved outcomes in BD; however, further study is needed to determine which specific interventions may be beneficial. LIMITATIONS The majority of identified studies had cross-sectional designs, small sample sizes and limited measurements of inflammation. CONCLUSIONS Treatment and prevention of general medical comorbidities in mood disorders should include preferential prescribing of metabolically neutral agents and adjunctive lifestyle modifications including increased physical activity, improved diet and decreased substance abuse. In addition, the use of anti-inflammatory agents could be a relevant therapeutic target in future research.
Collapse
Affiliation(s)
- Ana SayuriYamagata
- University of São Paulo (USP), São Paulo, Brazil; Research Group in Molecular and Behavioral Neuroscience of Bipolar Disorder, Federal University of São Paulo (Unifesp), São Paulo, Brazil
| | - Elisa Brietzke
- Research Group in Molecular and Behavioral Neuroscience of Bipolar Disorder, Federal University of São Paulo (Unifesp), São Paulo, Brazil
| | - Joshua D Rosenblat
- Mood Disorders Psychopharmacology Unit (MDPU), University Health Network (UHN), University of Toronto, Toronto, Canada
| | - Ron Kakar
- Mood Disorders Psychopharmacology Unit (MDPU), University Health Network (UHN), University of Toronto, Toronto, Canada
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit (MDPU), University Health Network (UHN), University of Toronto, Toronto, Canada.
| |
Collapse
|
32
|
Colle R, de Larminat D, Rotenberg S, Hozer F, Hardy P, Verstuyft C, Fève B, Corruble E. Pioglitazone could induce remission in major depression: a meta-analysis. Neuropsychiatr Dis Treat 2017; 13:9-16. [PMID: 28031713 PMCID: PMC5182046 DOI: 10.2147/ndt.s121149] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Pioglitazone, a selective agonist of the nuclear transcription factor peroxisome proliferator-activated receptor-gamma (PPAR-γ), prescribed for the treatment of type 2 diabetes, could have antidepressant properties. However, its potential to induce remission of major depressive episodes, the optimal clinical target for an antidepressant drug, is a matter of concern. Indeed, only one out of four double-blind randomized controlled trials show higher remission rates with pioglitazone than with control treatments. Hence, the main aim of this study was to perform a meta-analysis of the efficacy of pioglitazone for the treatment of MDE, focusing on remission rates. METHODS Four double-blind randomized controlled trials, comprising 161 patients with an MDE, were included in this meta-analysis. Pioglitazone was studied either alone (one study) or as add-on therapy to conventional treatments (antidepressant drugs or lithium salts). It was compared either to placebo (three studies) or to metformin (one study). Remission was defined by a Hamilton Depression Rating Scale score <8 after treatment. RESULTS Pioglitazone could induce higher remission rates than control treatments (27% versus 10%, I2=17.3%, fixed-effect model: odds ratio [OR] =3.3, 95% confidence interval [95% CI; 1.4; 7.8], P=0.008). The OR was even higher in the subgroup of patients with major depressive disorder (n=80; 23% versus 8%, I2=0.0%; fixed-effect model: OR =5.9, 95% CI [1.6; 22.4], P=0.009) and in the subgroup of patients without metabolic comorbidities (n=84; 33% versus 10%, I2=0.0%; fixed-effect model: OR =5.1, 95% CI [1.5; 17.9], P=0.01). As compared to control treatments, results suggest six patients would need to be treated with pioglitazone in order to achieve the possibility of one more remission. CONCLUSION Pioglitazone, either alone or as add-on therapy to conventional treatments, could induce remission of MDE, suggesting that drugs with PPAR-γ agonist properties may be true and clinically relevant antidepressants, even in patients without metabolic comorbidities.
Collapse
Affiliation(s)
- Romain Colle
- Psychiatry Department, Hôpital Bicêtre, INSERM, UMR S1178, University Paris-Sud, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, France
| | - Delphine de Larminat
- Psychiatry Department, Hôpital Bicêtre, INSERM, UMR S1178, University Paris-Sud, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, France
| | - Samuel Rotenberg
- Psychiatry Department, Hôpital Bicêtre, INSERM, UMR S1178, University Paris-Sud, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, France
| | - Franz Hozer
- Psychiatry Department, Hôpital Bicêtre, INSERM, UMR S1178, University Paris-Sud, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, France
| | - Patrick Hardy
- Psychiatry Department, Hôpital Bicêtre, INSERM, UMR S1178, University Paris-Sud, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, France
| | - Céline Verstuyft
- Molecular Genetic, Pharmacogenetics and Hormonology Department, Hôpital Bicêtre, INSERM UMR_S1184, Centre IMVA, University Paris-Sud, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, France
| | - Bruno Fève
- Endocrinology Department, INSERM UMR_S938, Hôpital Saint-Antoine, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire ICAN, Sorbonne Universités, Université Pierre et Marie Curie, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Emmanuelle Corruble
- Psychiatry Department, Hôpital Bicêtre, INSERM, UMR S1178, University Paris-Sud, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, France
| |
Collapse
|
33
|
Diniz BS, Lin CW, Sibille E, Tseng G, Lotrich F, Aizenstein HJ, Reynolds CF, Butters MA. Circulating biosignatures of late-life depression (LLD): Towards a comprehensive, data-driven approach to understanding LLD pathophysiology. J Psychiatr Res 2016; 82:1-7. [PMID: 27447786 PMCID: PMC9344393 DOI: 10.1016/j.jpsychires.2016.07.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 06/28/2016] [Accepted: 07/05/2016] [Indexed: 11/30/2022]
Abstract
There is scarce information about the pathophysiological processes underlying Late-Life Depression (LLD). We aimed to determine the neurobiological abnormalities related to LLD through a multi-modal biomarker approach combining a large, unbiased peripheral proteomic panel and structural brain imaging. We examined data from 44 LLD and 31 control participants. Plasma proteomic analysis was performed using a multiplex immunoassay. We evaluated the differential protein expression between groups with random intercept models. We carried out enrichment pathway analyses (EPA) to uncover biological pathways and processes related to LLD. Machine learning analysis was applied to the combined dataset to determine the accuracy with which specific proteins could correctly discriminate LLD versus control participants. Sixty-one proteins were differentially expressed in LLD (p < 0.05 and FDR < 0.01). EPA showed that these proteins were related to abnormal immune-inflammatory control, cell survival and proliferation, proteostasis control, lipid metabolism, intracellular signaling. Machine learning analysis showed that a panel of three proteins (C-peptide, FABP-liver, ApoA-IV) discriminated LLD and control participants with 100% accuracy. The plasma proteomic profile in LLD revealed dysregulation in biological processes essential to the maintenance of homeostasis at cellular and systemic levels. These abnormalities increase brain and systemic allostatic load leading to the downstream negative outcomes of LLD, including increased risk of medical comorbidities and dementia. The peripheral biosignature of LLD has predictive power and may suggest novel putative therapeutic targets for prevention, treatment, and neuroprotection in LLD.
Collapse
Affiliation(s)
- Breno Satler Diniz
- Department of Psychiatry & Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chien-Wei Lin
- Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute of CAMH, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - George Tseng
- Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - Francis Lotrich
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Howard J. Aizenstein
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Charles F. Reynolds
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Meryl A. Butters
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA,Corresponding author. 3811 O’Hara Street, Pittsburgh, PA, 15213, USA. (M.A. Butters)
| |
Collapse
|
34
|
Huang CW, Hong TW, Wang YJ, Chen KC, Pei JC, Chuang TY, Lai WS, Tsai SH, Chu R, Chen WC, Sheen LY, Takahashi S, Ding ST, Shen TL. Ophiocordyceps formosana improves hyperglycemia and depression-like behavior in an STZ-induced diabetic mouse model. Altern Ther Health Med 2016; 16:310. [PMID: 27553852 PMCID: PMC4995616 DOI: 10.1186/s12906-016-1278-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 08/11/2016] [Indexed: 12/27/2022]
Abstract
Background A newly defined Cordyceps species, Ophiocordyceps formosana (O. formosana) has been implicated in multitudinous bioactivities, including lowering glucose and cholesterol levels and modulating the immune system. However, few literatures demonstrate sufficient evidence to support these proposed functions. Although the use of Cordyceps spp. has been previously addressed to improve insulin insensitivity and improve the detrimental symptoms of depression; its mechanistic nature remains unsettled. Herein, we reveal the effects of O. formosana in ameliorating hyperglycemia accompanied with depression. Methods Diabetes was induced in mice by employing streptozotocin(STZ), a chemical that is toxic to insulin-producing β cells of the pancreas. These streptozotocin (STZ)-induced diabetic mice showed combined symptoms of hyperglycemia and depressive behaviors. Twenty-four STZ-induced mice were randomly divided into 3 groups subjected to oral gavage with 100 μL solution of either PBS or 25 mg/mL Ophiocordyceps formosana extract (OFE) or 2 mg/mL rosiglitazone (Rosi, positive control group). Treatments were administered once per day for 28 days. An additional 6 mice without STZ induction were treated with PBS to serve as the control group. Insulin sensitivity was measured by a glucose tolerance test and levels of adiponectin in plasma and adipose tissue were also quantified. Behavioral tests were conducted and levels of monoamines in various brain regions relating to depression were evaluated. Results HPLC analysis uncovered three major constituents, adenosine, D-mannitol and cordycepin, within O. formosana similar to other prestigious medicinal Cordyceps spp.. STZ-induced diabetic mice demonstrated decreased body weight and subcutaneous adipose tissue, while these symptoms were recovered in mice receiving OFE treatment. Moreover, the OFE group displayed improved insulin sensitivity and elevated adiponectin within the plasma and adipose tissue. The anti-depressive effect of OFE was observed in various depression-related behavior tests. Concurrently, neurotransmitters, like 5-HT and dopamine in the frontal cortex, striatum and hippocampus were found to be up-regulated in OFE-treated mice. Conclusions Our findings illustrated, for the first time, the medicinal merits of O. formosana on Type I diabetes and hyperglycemia-induced depression. OFE were found to promote the expression of adiponectin, which is an adipokine involved in insulin sensitivity and hold anti-depressive effects. In addition, OFE administration also displayed altered levels of neurotransmitters in certain brain regions that may have contributed to its anti-depressive effect. Collectively, this current study provided insights to the potential therapeutic effects of O. formosana extracts in regards to hyperglycemia and its depressive complications.
Collapse
|
35
|
Wincewicz D, Juchniewicz A, Waszkiewicz N, Braszko JJ. Angiotensin II type 1 receptor blockade by telmisartan prevents stress-induced impairment of memory via HPA axis deactivation and up-regulation of brain-derived neurotrophic factor gene expression. Pharmacol Biochem Behav 2016; 148:108-18. [PMID: 27375198 DOI: 10.1016/j.pbb.2016.06.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 06/10/2016] [Accepted: 06/29/2016] [Indexed: 01/19/2023]
Abstract
Physical and psychological aspects of chronic stress continue to be a persistent clinical problem for which new pharmacological treatment strategies are aggressively sought. By the results of our previous work it has been demonstrated that telmisartan (TLM), an angiotensin type 1 receptor (AT1) blocker (ARB) and partial agonist of peroxisome proliferator-activated receptor gamma (PPARγ), alleviates stress-induced cognitive decline. Understanding of mechanistic background of this phenomenon is hampered by both dual binding sites of TLM and limited data on the consequences of central AT1 blockade and PPARγ activation. Therefore, a critical need exists for progress in the characterization of this target for pro-cognitive drug discovery. An unusual ability of novel ARBs to exert various PPARγ binding activities is commonly being viewed as predominant over angiotensin blockade in terms of neuroprotection. Here we aimed to verify this hypothesis using an animal model of chronic psychological stress (Wistar rats restrained 2.5h daily for 21days) with simultaneous oral administration of TLM (1mg/kg), GW9662 - PPARγ receptor antagonist (0.5mg/kg), or both in combination, followed by a battery of behavioral tests (open field, elevated plus maze, inhibitory avoidance - IA, object recognition - OR), quantitative determination of serum corticosterone (CORT) and evaluation of brain-derived neurotrophic factor (BDNF) gene expression in the medial prefrontal cortex (mPFC) and hippocampus (HIP). Stressed animals displayed decreased recall of the IA behavior (p<0.001), decreased OR (p<0.001), substantial CORT increase (p<0.001) and significantly downregulated expression of BDNF in the mPFC (p<0.001), which were attenuated in rats receiving TLM and TLM+GW9662. These data indicate that procognitive effect of ARBs in stressed subjects do not result from PPAR-γ activation, but AT1 blockade and subsequent hypothalamus-pituitary-adrenal axis deactivation associated with changes in primarily cortical gene expression. This study confirms the dual activities of TLM that controls hypertension and cognition through AT1 blockade.
Collapse
Affiliation(s)
- D Wincewicz
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15a, 15274 Bialystok, Poland; Department of Psychiatry, Medical University of Bialystok, Poland.
| | - A Juchniewicz
- Department of Clinical Molecular Biology, Medical University of Bialystok, Poland
| | - N Waszkiewicz
- Department of Psychiatry, Medical University of Bialystok, Poland
| | - J J Braszko
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15a, 15274 Bialystok, Poland
| |
Collapse
|
36
|
Kurhe Y, Mahesh R. Pioglitazone, a PPARγ agonist rescues depression associated with obesity using chronic unpredictable mild stress model in experimental mice. Neurobiol Stress 2016; 3:114-121. [PMID: 27981184 PMCID: PMC5146196 DOI: 10.1016/j.ynstr.2016.05.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 03/19/2016] [Accepted: 05/16/2016] [Indexed: 12/13/2022] Open
Abstract
Pioglitazone, a peroxisome proliferator activated receptor gamma (PPARγ) agonist belonging to thiazolidinedione class, is mainly used in diabetes mellitus. Obese subjects are twice likely to become depressed than non-obese individuals. The biological mechanisms linking depression with obesity still remain poorly understood and there is immense need for better therapeutic intervention against such co-morbid disorders. The present study investigates the effect of pioglitazone on the chronic unpredictable mild stress (CUMS) induced depression in obese mice by using behavioral tests and biochemical estimations. Mice were fed with high fat diet (HFD) for 14 weeks and were further subjected to different stress procedures for 28 days to induce depressive behavior. Animals were administered orally with pioglitazone (30 mg/kg p.o.)/escitalopram (10 mg/kg p.o.)/vehicle (10 ml/kg p.o.) daily from day 15-28. Various behavioral paradigms such as sucrose preference test, forced swim test (FST), tail suspension test (TST) and elevated plus maze (EPM) were performed. Biochemical estimations including plasma glucose, total cholesterol, triglycerides, and total proteins were performed. The data obtained from behavioral assays and biochemical assessments indicated that obese animals exhibited severe depressive-like behavior compared to non-obese animals. Furthermore, obese animals subjected to CUMS worsen the depressive behavior compared to obese control animals. Repetitive treatment with pioglitazone reversed the CUMS induced behavioral and biochemical alterations in HFD fed obese mice which atleast in part may be mediated through improving altered plasma glucose. The study suggests that pioglitazone needs further attention with respect to molecular mechanisms that could provide a better therapeutic strategy against depression associated with obesity.
Collapse
Affiliation(s)
- Yeshwant Kurhe
- Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Rajasthan, 333031, India
| | | |
Collapse
|
37
|
Strekalova T, Costa-Nunes JP, Veniaminova E, Kubatiev A, Lesch KP, Chekhonin VP, Evans MC, Steinbusch HWM. Insulin receptor sensitizer, dicholine succinate, prevents both Toll-like receptor 4 (TLR4) upregulation and affective changes induced by a high-cholesterol diet in mice. J Affect Disord 2016; 196:109-16. [PMID: 26921863 DOI: 10.1016/j.jad.2016.02.045] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 02/03/2016] [Accepted: 02/16/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND High cholesterol intake in mice induces hepatic lipid dystrophy and inflammation, signs of non-alcoholic fatty liver disease (NAFLD), depressive- and anxiety-like behaviors, and the up-regulation of brain and liver Toll-like receptor 4 (Tlr4). Here, we investigated whether dicholine succinate (DS), an insulin receptor sensitizer and mitochondrial complex II substrate would interact with these effects. METHODS C57BL/6J mice were given a 0.2%-cholesterol diet for 3 weeks, alone or along with oral DS administration, or a control feed. Outcomes included behavioral measures of anxiety/depression, and Tlr4 and peroxisome-proliferator-activated-receptor-gamma coactivator-1b (PPARGC1b) expression. RESULTS 50mg/kg DS treatment for 3 weeks partially ameliorated the cholesterol-induced anxiety- and depressive-like changes. Mice were next treated at the higher dose (180mg/kg), either for the 3-week period of dietary intervention, or for the last two weeks. Three-week DS administration normalized behaviors in the forced swim and O-maze tests and abolished the Tlr4 up-regulation in the brain and liver. The delayed, 2-week DS treatment had similar effects on Tlr4 expression and largely rescued the above-mentioned behaviors. Suppression of PPARGC1b, a master regulator of mitochondrial biogenesis, by the high cholesterol diet, was prevented with the 3-week administration, and markedly diminished by the a 2-week administration of DS. None of treatments prevented hepatic dystrophy and triglyceride accumulation. LIMITATIONS Other conditions have to be tested to define possible limitations of reported effects of DS. CONCLUSIONS DS treatment did not alter the patho-morphological substrates of NAFLD syndrome in mice, but ameliorated its molecular and behavioral consequences, likely by activating mitochondrial functions and anti-inflammatory mechanisms.
Collapse
Affiliation(s)
- Tatyana Strekalova
- Department of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, 6200 MD Maastricht, The Netherlands.
| | - João P Costa-Nunes
- Department of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, 6200 MD Maastricht, The Netherlands; CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, FCM, Universidade Nova de Lisboa, Campo Mártires da Pátria, 130, 1169-056 Lisboa, Portugal
| | - Ekaterina Veniaminova
- Department of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, 6200 MD Maastricht, The Netherlands; Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Baltiyskaya 8, Moscow 125315, Russia
| | - Aslan Kubatiev
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Baltiyskaya 8, Moscow 125315, Russia
| | - Klaus-Peter Lesch
- Department of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, 6200 MD Maastricht, The Netherlands; Division of Molecular Psychiatry, Laboratory of Translational Neuroscience, Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Fuechsleinstr. 15, 97080 Wuerzburg, Germany
| | - Vladimir P Chekhonin
- Serbsky National Research Center for Social and Forensic Psychiatry, Department of Fundamental and Applied Neurobiology, per. Kropotkin 23, Moscow 119034, Russian Federation
| | - Matthew C Evans
- Department of Pharmacology, Oxford University, Mansfield Road, OX1 3QT Oxford, UK
| | - Harry W M Steinbusch
- Department of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
38
|
Kamble M, Gupta R, Rehan HS, Gupta LK. Neurobehavioral effects of liraglutide and sitagliptin in experimental models. Eur J Pharmacol 2016; 774:64-70. [DOI: 10.1016/j.ejphar.2016.02.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Revised: 01/27/2016] [Accepted: 02/01/2016] [Indexed: 01/14/2023]
|
39
|
Apigenin ameliorates chronic mild stress-induced depressive behavior by inhibiting interleukin-1β production and NLRP3 inflammasome activation in the rat brain. Behav Brain Res 2016; 296:318-325. [DOI: 10.1016/j.bbr.2015.09.031] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 09/16/2015] [Accepted: 09/23/2015] [Indexed: 12/12/2022]
|
40
|
Lin KW, Wroolie TE, Robakis T, Rasgon NL. Adjuvant pioglitazone for unremitted depression: Clinical correlates of treatment response. Psychiatry Res 2015; 230:846-52. [PMID: 26602230 PMCID: PMC4978223 DOI: 10.1016/j.psychres.2015.10.013] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 09/23/2015] [Accepted: 10/09/2015] [Indexed: 10/22/2022]
Abstract
Previous studies suggest that insulin-sensitizing agents could play a significant role in the treatment of major depression, particularly depression in patients with documented insulin resistance or those who are resistant to standard psychopharmacological approaches. This study aimed to assess the effects on depressive symptoms with adjuvant treatment with the PPARγ-agonist pioglitazone. Patients (N=37) with non-psychotic, non-remitting depression receiving standard psychiatric regimens for depression were randomized across an insulin sensitivity spectrum in a 12-week double blind, randomized controlled trial of pioglitazone or placebo. Improvement in depression was associated with improvement in glucose metabolism but only in patients with insulin resistance. An age effect was also shown in that response to pioglitazone was more beneficial in younger aged patients. Study findings suggest differential improvement in depression severity according to both glucose metabolic status and level of depression at baseline. A greater understanding of the reciprocal links between depression and IR may lead to a dramatic shift in the way in which depression is conceptualized and treated, with a greater focus on treating and/or preventing metabolic dysfunction.
Collapse
Affiliation(s)
| | | | | | - Natalie L. Rasgon
- Corresponding author: Natalie Rasgon M.D Ph.D., Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 401 Quarry Road, Stanford, CA 94305-5723, Phone: (650) 724-6689, Fax: (650) 724-3144,
| |
Collapse
|
41
|
Noland RC. Exercise and Regulation of Lipid Metabolism. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 135:39-74. [PMID: 26477910 DOI: 10.1016/bs.pmbts.2015.06.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The increased prevalence of hyperlipidemia, hypertriglyceridemia, hypercholesterolemia, and fatty liver disease has provided increasingly negative connotations toward lipids. However, it is important to remember that lipids are essential components supporting life. Lipids are a class of molecules defined by their inherent insolubility in water. In biological systems, lipids are either hydrophobic (containing only polar groups) or amphipathic (possess polar and nonpolar groups). These characteristics lend lipids to be highly diverse with a multitude of functions including hormone and membrane synthesis, involvement in numerous signaling cascades, as well as serving as a source of metabolic fuel supporting energy production. Exercise can induce changes in the lipid composition of membranes that effect fluidity and cellular function, as well as modify the cellular and circulating environment of lipids that regulate signaling cascades. The purpose of this chapter is to focus on lipid utilization as metabolic fuel in response to acute and chronic exercise training. Lipids utilized as an energy source during exercise include circulating fatty acids bound to albumin, triglycerides stored in very-low-density lipoprotein, and intramuscular triglyceride stores. Dynamic changes in these lipid pools during and after exercise are discussed, as well as key factors that may be responsible for regulating changes in fat oxidation in response to varying exercise conditions.
Collapse
Affiliation(s)
- Robert C Noland
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA.
| |
Collapse
|
42
|
Thouennon E, Cheng Y, Falahatian V, Cawley NX, Loh YP. Rosiglitazone-activated PPARγ induces neurotrophic factor-α1 transcription contributing to neuroprotection. J Neurochem 2015; 134:463-70. [PMID: 25940785 DOI: 10.1111/jnc.13152] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 03/04/2015] [Accepted: 04/02/2015] [Indexed: 12/20/2022]
Abstract
Brain peroxisome proliferator-activated receptor gamma (PPARγ), a member of the nuclear receptor superfamily of ligand-dependent transcription factors, is involved in neuroprotection. It is activated by the drug rosiglitazone, which then can increase the pro-survival protein B-cell lymphoma 2 (BCL-2), to mediate neuroprotection. However, the mechanism underlying this molecular cascade remains unknown. Here, we show that the neuroprotective protein neurotrophic factor-α1 (NF-α1), which also induces the expression of BCL-2, has a promoter that contains PPARγ-binding sites that are activated by rosiglitazone. Treatment of Neuro2a cells and primary hippocampal neurons with rosiglitazone increased endogenous NF-α1 expression and prevented H2 O2 -induced cytotoxicity. Concomitant with the increase in NF-α1, BCL-2 was also increased in these cells. When siRNA against NF-α1 was used, the induction of BCL-2 by rosiglitazone was prevented, and the neuroprotective effect of rosiglitazone was reduced. These results demonstrate that rosiglitazone-activated PPARγ directly induces the transcription of NF-α1, contributing to neuroprotection in neurons. We proposed the following cascade for neuroprotection against oxidative stress by rosiglitazone: Rosiglitazone enters the neuron and binds to peroxisome proliferator-activated receptor gamma (PPARγ) in the nucleus. The PPARγ-rosiglitazone complex binds to the neurotrophic factor-α1 (NF-α1) promoter and activates the transcription of NF-α1 mRNA which is then translated to the protein. NF-α1 is the secreted, binds to a cognate receptor and activates the extracellular signal-regulated kinases (ERK) pathway. This in turn enhances the expression of the pro-survival protein, B-cell lymphoma 2 (BCL-2) and inhibition of caspase 3 (Csp-3) to mediate neuroprotection under oxidative stress. Akt, protein kinase B (PKB).
Collapse
Affiliation(s)
- Erwan Thouennon
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Yong Cheng
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Vida Falahatian
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Niamh X Cawley
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Yoke Peng Loh
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
43
|
Cheng Y, Rodriguiz RM, Murthy SRK, Senatorov V, Thouennon E, Cawley NX, Aryal DK, Ahn S, Lecka-Czernik B, Wetsel WC, Loh YP. Neurotrophic factor-α1 prevents stress-induced depression through enhancement of neurogenesis and is activated by rosiglitazone. Mol Psychiatry 2015; 20:744-54. [PMID: 25330741 PMCID: PMC4405386 DOI: 10.1038/mp.2014.136] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 08/14/2014] [Accepted: 09/04/2014] [Indexed: 12/24/2022]
Abstract
Major depressive disorder is often linked to stress. Although short-term stress is without effect in mice, prolonged stress leads to depressive-like behavior, indicating that an allostatic mechanism exists in this difference. Here we demonstrate that mice after short-term (1 h per day for 7 days) chronic restraint stress (CRS), do not display depressive-like behavior. Analysis of the hippocampus of these mice showed increased levels of neurotrophic factor-α1 (NF-α1; also known as carboxypeptidase E, CPE), concomitant with enhanced fibroblast growth factor 2 (FGF2) expression, and an increase in neurogenesis in the dentate gyrus. In contrast, after prolonged (6 h per day for 21 days) CRS, mice show decreased hippocampal NF-α1 and FGF2 levels and depressive-like responses. In NF-α1-knockout mice, hippocampal FGF2 levels and neurogenesis are reduced. These mice exhibit depressive-like behavior that is reversed by FGF2 administration. Indeed, studies in cultured hippocampal neurons reveal that NF-α1 treatment directly upregulates FGF2 expression through extracellular signal-regulated kinase-Sp1 signaling. Thus, during short-term CRS, hippocampal NF-α1 expression is upregulated and has a key role in preventing the onset of depressive-like behavior through enhanced FGF2-mediated neurogenesis. To evaluate the therapeutic potential of this pathway, we examined, rosiglitazone (Rosi), a PPARγ agonist, which has been shown to have antidepressant activity in rodents and humans. Rosi upregulates FGF2 expression in a NF-α1-dependent manner in hippocampal neurons. Mice fed Rosi show increased hippocampal NF-α1 levels and neurogenesis compared with controls, thereby indicating the antidepressant action of this drug. Development of drugs that activate the NF-α1/FGF2/neurogenesis pathway can offer a new approach to depression therapy.
Collapse
Affiliation(s)
- Yong Cheng
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ramona M. Rodriguiz
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA
| | - Saravana R. K. Murthy
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Vladimir Senatorov
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Erwan Thouennon
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Niamh X. Cawley
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dipendra K. Aryal
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA
| | - Sohyun Ahn
- Program in Genomics Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Beata Lecka-Czernik
- Departments of Orthopaedic Surgery and Physiology and Pharmacology, Center for Diabetes and Endocrine Research, University of Toledo Health Sciences Campus, Toledo, Ohio 43614, USA
| | - William C. Wetsel
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA,Departments of Neurobiology and Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Y. Peng Loh
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA,Correspondence should be addressed to: Dr. Y. Peng Loh, 49 Convent Drive, Bldg. 49, Room 6C80, National Institutes of Health, Bethesda, MD 20892, USA. , Tel: 301-496-3239, Fax: 301-496-9938
| |
Collapse
|
44
|
Cline BH, Costa-Nunes JP, Cespuglio R, Markova N, Santos AI, Bukhman YV, Kubatiev A, Steinbusch HWM, Lesch KP, Strekalova T. Dicholine succinate, the neuronal insulin sensitizer, normalizes behavior, REM sleep, hippocampal pGSK3 beta and mRNAs of NMDA receptor subunits in mouse models of depression. Front Behav Neurosci 2015; 9:37. [PMID: 25767439 PMCID: PMC4341562 DOI: 10.3389/fnbeh.2015.00037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 02/01/2015] [Indexed: 11/13/2022] Open
Abstract
Central insulin receptor-mediated signaling is attracting the growing attention of researchers because of rapidly accumulating evidence implicating it in the mechanisms of plasticity, stress response, and neuropsychiatric disorders including depression. Dicholine succinate (DS), a mitochondrial complex II substrate, was shown to enhance insulin-receptor mediated signaling in neurons and is regarded as a sensitizer of the neuronal insulin receptor. Compounds enhancing neuronal insulin receptor-mediated transmission exert an antidepressant-like effect in several pre-clinical paradigms of depression; similarly, such properties for DS were found with a stress-induced anhedonia model. Here, we additionally studied the effects of DS on several variables which were ameliorated by other insulin receptor sensitizers in mice. Pre-treatment with DS of chronically stressed C57BL6 mice rescued normal contextual fear conditioning, hippocampal gene expression of NMDA receptor subunit NR2A, the NR2A/NR2B ratio and increased REM sleep rebound after acute predation. In 18-month-old C57BL6 mice, a model of elderly depression, DS restored normal sucrose preference and activated the expression of neural plasticity factors in the hippocampus as shown by Illumina microarray. Finally, young naïve DS-treated C57BL6 mice had reduced depressive- and anxiety-like behaviors and, similarly to imipramine-treated mice, preserved hippocampal levels of the phosphorylated (inactive) form of GSK3 beta that was lowered by forced swimming in pharmacologically naïve animals. Thus, DS can ameliorate behavioral and molecular outcomes under a variety of stress- and depression-related conditions. This further highlights neuronal insulin signaling as a new factor of pathogenesis and a potential pharmacotherapy of affective pathologies.
Collapse
Affiliation(s)
- Brandon H Cline
- Faculté de Médecine, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg Strasbourg, France
| | - Joao P Costa-Nunes
- Department of Neuroscience, Maastricht University Maastricht, Netherlands ; Group of Behavioural Neuroscience and Pharmacology, Institute for Hygiene and Tropical Medicine, New University of Lisbon Lisbon, Portugal
| | - Raymond Cespuglio
- Faculty of Medicine, Neuroscience Research Center of Lyon, INSERM U1028, C. Bernard University Lyon, France
| | - Natalyia Markova
- Laboratory of Biomolecular Screening, Institute of Physiologically Active Compounds, Russian Academy of Sciences Moscow, Russia ; Laboratory of Cognitive Dysfunctions, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences Moscow, Russia
| | - Ana I Santos
- Faculdade de Ciências Médicas, NOVA Medical School, Universidade Nova de Lisboa Lisboa, Portugal
| | - Yury V Bukhman
- Great Lakes Bioenergy Research Center, Computational Biology, Wisconsin Energy Institute, University of Wisconsin Madison, WI, USA
| | - Aslan Kubatiev
- Laboratory of Cognitive Dysfunctions, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences Moscow, Russia
| | | | - Klaus-Peter Lesch
- Department of Neuroscience, Maastricht University Maastricht, Netherlands ; Laboratory of Translational Neuroscience, Division of Molecular Psychiatry, Centre of Mental Health, University of Wuerzburg Wuerzburg, Germany
| | - Tatyana Strekalova
- Department of Neuroscience, Maastricht University Maastricht, Netherlands ; Group of Behavioural Neuroscience and Pharmacology, Institute for Hygiene and Tropical Medicine, New University of Lisbon Lisbon, Portugal ; Laboratory of Biomolecular Screening, Institute of Physiologically Active Compounds, Russian Academy of Sciences Moscow, Russia
| |
Collapse
|
45
|
Brand SJ, Moller M, Harvey BH. A Review of Biomarkers in Mood and Psychotic Disorders: A Dissection of Clinical vs. Preclinical Correlates. Curr Neuropharmacol 2015; 13:324-68. [PMID: 26411964 PMCID: PMC4812797 DOI: 10.2174/1570159x13666150307004545] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 02/04/2015] [Accepted: 03/06/2015] [Indexed: 11/23/2022] Open
Abstract
Despite significant research efforts aimed at understanding the neurobiological underpinnings of mood (depression, bipolar disorder) and psychotic disorders, the diagnosis and evaluation of treatment of these disorders are still based solely on relatively subjective assessment of symptoms as well as psychometric evaluations. Therefore, biological markers aimed at improving the current classification of psychotic and mood-related disorders, and that will enable patients to be stratified on a biological basis into more homogeneous clinically distinct subgroups, are urgently needed. The attainment of this goal can be facilitated by identifying biomarkers that accurately reflect pathophysiologic processes in these disorders. This review postulates that the field of psychotic and mood disorder research has advanced sufficiently to develop biochemical hypotheses of the etiopathology of the particular illness and to target the same for more effective disease modifying therapy. This implies that a "one-size fits all" paradigm in the treatment of psychotic and mood disorders is not a viable approach, but that a customized regime based on individual biological abnormalities would pave the way forward to more effective treatment. In reviewing the clinical and preclinical literature, this paper discusses the most highly regarded pathophysiologic processes in mood and psychotic disorders, thereby providing a scaffold for the selection of suitable biomarkers for future studies in this field, to develope biomarker panels, as well as to improve diagnosis and to customize treatment regimens for better therapeutic outcomes.
Collapse
Affiliation(s)
| | | | - Brian H Harvey
- Division of Pharmacology and Center of Excellence for Pharmaceutical Sciences, School of Pharmacy, North-West University, Potchefstroom, South Africa.
| |
Collapse
|
46
|
Bishop-Bailey D. Mechanisms governing the health and performance benefits of exercise. Br J Pharmacol 2014; 170:1153-66. [PMID: 24033098 DOI: 10.1111/bph.12399] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 07/18/2013] [Accepted: 07/23/2013] [Indexed: 12/18/2022] Open
Abstract
Humans are considered among the greatest if not the greatest endurance land animals. Over the last 50 years, as the population has become more sedentary, rates of cardiovascular disease and its associated risk factors such as obesity, type 2 diabetes and hypertension have all increased. Aerobic fitness is considered protective for all-cause mortality, cardiovascular disease, a variety of cancers, joint disease and depression. Here, I will review the emerging mechanisms that underlie the response to exercise, focusing on the major target organ the skeletal muscle system. Understanding the mechanisms of action of exercise will allow us to develop new therapies that mimic the protective actions of exercise.
Collapse
Affiliation(s)
- D Bishop-Bailey
- Comparative Biomedical Sciences, The Royal Veterinary College, London, UK
| |
Collapse
|
47
|
Barbiero JK, Santiago RM, Persike DS, da Silva Fernandes MJ, Tonin FS, da Cunha C, Lucio Boschen S, Lima MM, Vital MA. Neuroprotective effects of peroxisome proliferator-activated receptor alpha and gamma agonists in model of parkinsonism induced by intranigral 1-methyl-4-phenyl-1,2,3,6-tetrahyropyridine. Behav Brain Res 2014; 274:390-9. [DOI: 10.1016/j.bbr.2014.08.014] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 08/01/2014] [Accepted: 08/05/2014] [Indexed: 12/20/2022]
|
48
|
Physical exercise-induced hippocampal neurogenesis and antidepressant effects are mediated by the adipocyte hormone adiponectin. Proc Natl Acad Sci U S A 2014; 111:15810-5. [PMID: 25331877 DOI: 10.1073/pnas.1415219111] [Citation(s) in RCA: 225] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Adiponectin (ADN) is an adipocyte-secreted protein with insulin-sensitizing, antidiabetic, antiinflammatory, and antiatherogenic properties. Evidence is also accumulating that ADN has neuroprotective activities, yet the underlying mechanism remains elusive. Here we show that ADN could pass through the blood-brain barrier, and elevating its levels in the brain increased cell proliferation and decreased depression-like behaviors. ADN deficiency did not reduce the basal hippocampal neurogenesis or neuronal differentiation but diminished the effectiveness of exercise in increasing hippocampal neurogenesis. Furthermore, exercise-induced reduction in depression-like behaviors was abrogated in ADN-deficient mice, and this impairment in ADN-deficient mice was accompanied by defective running-induced phosphorylation of AMP-activated protein kinase (AMPK) in the hippocampal tissue. In vitro analyses indicated that ADN itself could increase cell proliferation of both hippocampal progenitor cells and Neuro2a neuroblastoma cells. The neurogenic effects of ADN were mediated by the ADN receptor 1 (ADNR1), because siRNA targeting ADNR1, but not ADNR2, inhibited the capacity of ADN to enhance cell proliferation. These data suggest that adiponectin may play a significant role in mediating the effects of exercise on hippocampal neurogenesis and depression, possibly by activation of the ADNR1/AMPK signaling pathways, and also raise the possibility that adiponectin and its agonists may represent a promising therapeutic treatment for depression.
Collapse
|
49
|
Abstract
Food is a potent natural reward and food intake is a complex process. Reward and gratification associated with food consumption leads to dopamine (DA) production, which in turn activates reward and pleasure centers in the brain. An individual will repeatedly eat a particular food to experience this positive feeling of gratification. This type of repetitive behavior of food intake leads to the activation of brain reward pathways that eventually overrides other signals of satiety and hunger. Thus, a gratification habit through a favorable food leads to overeating and morbid obesity. Overeating and obesity stems from many biological factors engaging both central and peripheral systems in a bi-directional manner involving mood and emotions. Emotional eating and altered mood can also lead to altered food choice and intake leading to overeating and obesity. Research findings from human and animal studies support a two-way link between three concepts, mood, food, and obesity. The focus of this article is to provide an overview of complex nature of food intake where various biological factors link mood, food intake, and brain signaling that engages both peripheral and central nervous system signaling pathways in a bi-directional manner in obesity.
Collapse
Affiliation(s)
- Minati Singh
- Department of Pediatrics, University of Iowa Iowa City, IA, USA ; Department of Pediatrics, HHMI, University of Iowa Iowa City, IA, USA
| |
Collapse
|
50
|
Roohafza H, Shokouh P, Sadeghi M, Alikhassy Z, Sarrafzadegan N. A Possible Role for Pioglitazone in the Management of Depressive Symptoms in Metabolic Syndrome Patients (EPICAMP Study): A Double Blind, Randomized Clinical Trial. INTERNATIONAL SCHOLARLY RESEARCH NOTICES 2014; 2014:697617. [PMID: 27433505 PMCID: PMC4897538 DOI: 10.1155/2014/697617] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 06/16/2014] [Indexed: 01/04/2023]
Abstract
The present trial aimed to evaluate the effects of pioglitazone on the mental status of nondiabetic metabolic syndrome patients. From 145 patients screened, 104 eligible volunteers (57% female; age 20-70 years) were enrolled and randomly assigned to receive either pioglitazone (uptitrated to 30 mg/day; P = 53) or matching placebo (P = 51) for 24 weeks. Depression and anxiety were quantified using the hospital anxiety and depression scale and stress level using the general health questionnaire 12 at baseline, week 12, and endpoint. Homeostasis model assessment was used to estimate insulin resistance. At week 24, pioglitazone was superior in mitigating depression score (P = 0.011). In trend analysis, the effect of time (P < 0.001) and group (P = 0.023) as well as the time by group interaction (P = 0.032) on the mean depression score was considerable. In contrast, significant decrements in anxiety and stress levels (P < 0.001 and P = 0.012, resp.) were comparable between two groups. With respect to our findings, alterations in depression severity were not correlated with changes in insulin resistance level (P = 0.145). In conclusion, our findings suggest that pioglitazone might be able to improve mood in nondiabetic insulin resistant patients. (Registered at Australian New Zealand Clinical Trials Registry; ACTRN12611000351910.).
Collapse
Affiliation(s)
- Hamidreza Roohafza
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, P.O. Box 81465-1148, Iran
| | - Pedram Shokouh
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, P.O. Box 81465-1148, Iran
| | - Masoumeh Sadeghi
- Isfahan Cardiac Rehabilitation Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, P.O. Box 81465-1148, Iran
| | - Zahra Alikhassy
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, P.O. Box 81465-1148, Iran
| | - Nizal Sarrafzadegan
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, P.O. Box 81465-1148, Iran
| |
Collapse
|