1
|
de Camargo RW, Joaquim L, Machado RS, de Souza Ramos S, da Rosa LR, de Novais Junior LR, Mathias K, Maximiano L, Strickert YR, Nord R, Gava ML, Scarpari E, Martins HM, Lins EMF, Chaves JS, da Silva LE, de Oliveira MP, da Silva MR, Fernandes BB, Tiscoski ADB, Piacentini N, Santos FP, Inserra A, Bobinski F, Rezin GT, Yonamine M, Petronilho F, de Bitencourt RM. Ayahuasca Pretreatment Prevents Sepsis-Induced Anxiety-Like Behavior, Neuroinflammation, and Oxidative Stress, and Increases Brain-Derived Neurotrophic Factor. Mol Neurobiol 2024:10.1007/s12035-024-04597-4. [PMID: 39613951 DOI: 10.1007/s12035-024-04597-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/28/2024] [Accepted: 10/26/2024] [Indexed: 12/01/2024]
Abstract
The psychoactive decoction Ayahuasca (AYA) used for therapeutic and religious purposes by indigenous groups and peoples from Amazonian regions produces anti-inflammatory and neuroprotective effects. Thus, it may be useful to attenuate the neuroinflammation and related anxiety- and depressive-like symptoms elicited by inflammatory insults such as sepsis. Rats were pretreated for 3 days with different doses of AYA. Twenty-four hours after, cecal ligation and puncture (CLP) was performed. On days 1-4, post-CLP behavioral tests to assess anxiety-like behavior were performed. After 24-h, neuroinflammation, oxidative stress, myeloperoxidase activity, and mitochondrial metabolism were assessed in the prefrontal cortex (PFC), hippocampus (HP), and cortex. AYA pretreatment increased the time spent in the open arms of the elevated plus maze and prevented the sepsis-induced hyper-grooming and -rearing behavior, suggesting an anxiolytic effect. AYA pretreatment increased the levels of the anti-inflammatory interleukin 4, in the PFC and the cortex, and brain-derived neurotrophic factor in the cortex. Moreover, AYA pretreatment increased myeloperoxidase activity in the PFC and the HP and decreased nitrite/nitrate concentration in the PFC, HP, and cortex of septic rats, suggesting enhanced neutrophil activation and decreased nitric oxide signaling. Furthermore, AYA pretreatment prevented lipid peroxidation in the PFC, HP, and cortex of septic rats as measured by decreased levels of thiobarbituric acid reactive substances. Levels of protein carbonyls and activity of superoxide dismutase, citrate synthase, succinate dehydrogenase, and mitochondrial respiratory chain were not affected. Together, AYA represents a promising approach to prevent sepsis-induced neuroinflammatory and oxidative stress and associated anxiety-like symptoms.
Collapse
Affiliation(s)
- Rick Wilhiam de Camargo
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Larissa Joaquim
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Richard Simon Machado
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Suelen de Souza Ramos
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Lara Rodrigues da Rosa
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Linério Ribeiro de Novais Junior
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Khiany Mathias
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Lara Maximiano
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Yasmin Ribeiro Strickert
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Rafael Nord
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Maria Laura Gava
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Eduarda Scarpari
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Helena Mafra Martins
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Palhoça, Santa Catarina, Brazil
| | - Elisa Mitkus Flores Lins
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Palhoça, Santa Catarina, Brazil
| | - Jéssica Schaefer Chaves
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Palhoça, Santa Catarina, Brazil
| | - Larissa Espindola da Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Mariana Pacheco de Oliveira
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Mariella Reinol da Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Bruna Barros Fernandes
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Anita Dal Bó Tiscoski
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Natália Piacentini
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Fabiana Pereira Santos
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Antonio Inserra
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
- Previous Affiliation: Department of Psychiatry, McGill University, Montreal, Canada
| | - Franciane Bobinski
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Palhoça, Santa Catarina, Brazil
| | - Gislaine Tezza Rezin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil
| | - Mauricio Yonamine
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Fabrícia Petronilho
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Rafael Mariano de Bitencourt
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Santa Catarina, Brazil.
| |
Collapse
|
2
|
Low JJL, Tan BJW, Yi LX, Zhou ZD, Tan EK. Genetic susceptibility to caffeine intake and metabolism: a systematic review. J Transl Med 2024; 22:961. [PMID: 39438936 PMCID: PMC11515775 DOI: 10.1186/s12967-024-05737-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/21/2024] [Accepted: 10/06/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Coffee and tea consumption account for most caffeine intake and 2-3 billion cups are taken daily around the world. Caffeine dependence is a widespread but under recognized problem. OBJECTIVES To conduct a systematic review on the genetic susceptibility factors affecting caffeine metabolism and caffeine reward and their association with caffeine intake. METHODOLOGY We conducted PubMed and Embase searches using the terms "caffeine", "reward", "gene", "polymorphism", "addiction", "dependence" and "habit" from inception till 2024. The demographics, genetic and clinical data from included studies were extracted and analyzed. Only case-control studies on habitual caffeine drinkers with at least 100 in each arm were included. RESULTS A total of 2552 studies were screened and 26 studies involving 1,851,428 individuals were included. Several genes that were involved with caffeine metabolism such as CYP1A2, ADORA2A, AHR, POR, ABCG2, CYP2A6, PDSS2 and HECTD4 rs2074356 (A allele specific to East Asians and monomorphic in Europeans, Africans and Americans) were associated with habitual caffeine consumption with effect size difference of 3% to 32% in number of cups of caffeinated drink per day per effect allele. In addition, ALDH2 was linked to the Japanese population. Genes associated with caffeine reward included BDNF, SLC6A4, GCKR, MLXIPL and dopaminergic genes such as DRD2 and DAT1 which had around 2-5% effect size difference in number of cups of caffeinated drink for each allele per day. CONCLUSION Several genes that were involved in caffeine metabolism and reward were associated with up to 30% effect size difference in number of cups of caffeinated drink per day, and some associations were specific to certain ethnicities. Identification of at-risk caffeine dependence individuals can lead to early diagnosis and stratification of at-risk vulnerable individuals such as pregnant women and children, and can potentially lead to development of drug targets for dependence to caffeine.
Collapse
Affiliation(s)
- Jazreel Ju-Li Low
- Department of Neurology, Singapore General Hospital Campus, National Neuroscience Institute, Singapore, Singapore
- Neuroscience and Behavioural Disorders, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Brendan Jen-Wei Tan
- Department of Neurology, Singapore General Hospital Campus, National Neuroscience Institute, Singapore, Singapore
| | - Ling-Xiao Yi
- Neuroscience and Behavioural Disorders, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Zhi-Dong Zhou
- Department of Neurology, Singapore General Hospital Campus, National Neuroscience Institute, Singapore, Singapore
- Neuroscience and Behavioural Disorders, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Eng-King Tan
- Department of Neurology, Singapore General Hospital Campus, National Neuroscience Institute, Singapore, Singapore.
- Neuroscience and Behavioural Disorders, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore.
| |
Collapse
|
3
|
Chen Z, Lu N, Li X, Liu Q, Li Y, Li X, Yu X, Zhao H, Liu C, Tang X, Wang X, Huang W. The Effect of a Caffeine and Nicotine Combination on Nicotine Withdrawal Syndrome in Mice. Nutrients 2024; 16:3048. [PMID: 39339647 PMCID: PMC11435009 DOI: 10.3390/nu16183048] [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] [Academic Contribution Register] [Received: 08/06/2024] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
Nicotine dependence is an important cause of excessive exposure to tobacco combustion compounds in most smokers. Nicotine replacement therapy is the main method to treat nicotine dependence, but it still has its shortcomings, such as the inability to mitigate withdrawal effects and limited applicability. It has been hypothesized that a combination of low-dose nicotine and caffeine could achieve the same psychological stimulation effect as a high dose of nicotine without causing nicotine withdrawal effects. To establish a model of nicotine dependence, male C57BL/6J mice were subcutaneously injected four times a day with nicotine (2 mg/kg) for 15 days and fed with water containing nicotine at the same time. They were randomly divided into four groups. After 24 h of withdrawal, different groups were injected with saline, nicotine (0.25 mg/kg or 0.1 mg/kg), or nicotine (0.1 mg/kg) and caffeine (20 mg/kg). Behavioral and physiological changes were evaluated by an assessment of physical signs, open field tests, elevated plus maze experiments, forced swimming tests, hot plate tests, and new-object-recognition tests. The changes in dopamine release in the prefrontal cortex (PFC) and ventral tegmental area (VTA) in the midbrain were analyzed using ELISA. The results showed that a combination of caffeine and nicotine could effectively relieve nicotine withdrawal syndrome, increase movement ability and pain thresholds, reduce anxiety and depression, enhance memory and cognitive ability, and increase the level of dopamine release in the PFC and VTA. Thus, caffeine combined with nicotine has potential as a stable and effective treatment option to help humans with smoking cessation.
Collapse
Affiliation(s)
- Zhe Chen
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Naiyan Lu
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Sports and Health Research Institute, Jiangnan University, Wuxi 214122, China
| | - Xu Li
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Qingrun Liu
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yujie Li
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Xiyue Li
- Department of Pulmonary and Critical Care Medicine, Jiangnan University Medical Center, Jiangnan University, Wuxi 214126, China
| | - Ximiao Yu
- Department of Pulmonary and Critical Care Medicine, Jiangnan University Medical Center, Jiangnan University, Wuxi 214126, China
| | - Haotian Zhao
- Sports and Health Research Institute, Jiangnan University, Wuxi 214122, China
| | - Chang Liu
- School of Sport Science, Beijing Sport University, Beijing 100084, China
| | - Xue Tang
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xun Wang
- Department of Pulmonary and Critical Care Medicine, Jiangnan University Medical Center, Jiangnan University, Wuxi 214126, China
| | - Weisun Huang
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, China
| |
Collapse
|
4
|
Hammad AM, Alzaghari LF, Alfaraj M, Al-Qerem W, Talib WH, Alasmari F, Amawi H, Hall FS. Acetylsalicylic acid reduces cigarette smoke withdrawal-induced anxiety in rats via modulating the expression of NFĸB, GLT-1, and xCT. Front Pharmacol 2023; 13:1047236. [PMID: 36699078 PMCID: PMC9868824 DOI: 10.3389/fphar.2022.1047236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/17/2022] [Accepted: 12/16/2022] [Indexed: 01/11/2023] Open
Abstract
Background: Chronic exposure to cigarette smoke produces neuroinflammation and long-term changes in neurotransmitter systems, especially glutamatergic systems. Objective: We examined the effects of cigarette smoke on astroglial glutamate transporters as well as NF-κB expression in mesocorticolimbic brain regions, prefrontal cortex (PFC) and nucleus accumbens (NAc). The behavioral consequences of cigarette smoke exposure were assessed using open field (OF) and light/dark (LD) tests to assess withdrawal-induced anxiety-like behavior. Methods: Sprague-Dawley rats were randomly assigned to five experimental groups: a control group exposed only to standard room air, a cigarette smoke exposed group treated with saline vehicle, two cigarette smoke exposed groups treated with acetylsalicylic acid (ASA) (15 mg/kg and 30 mg/kg, respectively), and a group treated only with ASA (30 mg/kg). Cigarette smoke exposure was performed for 2 h/day, 5 days/week, for 31 days. Behavioral tests were conducted weekly, 24 h after cigarette smoke exposure, during acute withdrawal. At the end of week 4, rats were given either saline or ASA 45 min before cigarette exposure for 11 days. Results: Cigarette smoke increased withdrawal-induced anxiety, and 30 mg/kg ASA attenuated this effect. Cigarette smoke exposure increased the relative mRNA and protein expression of nuclear factor ĸB (NFĸB) in PFC and NAc, and ASA treatment reversed this effect. Also, cigarette smoke decreased the relative mRNA and protein expression of glutamate transporter1 (GLT-1) and the cystine-glutamate transporter (xCT) in the PFC and the NAc, while ASA treatment normalized their expression. Conclusion: Cigarette smoke caused neuroinflammation, alterations in glutamate transporter expression, and increased anxiety-like behavior, and these effects were attenuated by acetylsalicylic acid treatment.
Collapse
Affiliation(s)
- Alaa M. Hammad
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | - Lujain F. Alzaghari
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | - Malek Alfaraj
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | - Walid Al-Qerem
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | - Wamidh H. Talib
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman, Jordan
| | - Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Haneen Amawi
- Department of Pharmacy Practice, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - F. Scott Hall
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, United States
| |
Collapse
|
5
|
Bayassi-Jakowicka M, Lietzau G, Czuba E, Patrone C, Kowiański P. More than Addiction—The Nucleus Accumbens Contribution to Development of Mental Disorders and Neurodegenerative Diseases. Int J Mol Sci 2022; 23:ijms23052618. [PMID: 35269761 PMCID: PMC8910774 DOI: 10.3390/ijms23052618] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/04/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 01/09/2023] Open
Abstract
Stress and negative emotions evoked by social relationships and working conditions, frequently accompanied by the consumption of addictive substances, and metabolic and/or genetic predispositions, negatively affect brain function. One of the affected structures is nucleus accumbens (NAc). Although its function is commonly known to be associated with brain reword responses and addiction, a growing body of evidence also suggests its role in some mental disorders, such as depression and schizophrenia, as well as neurodegenerative diseases, such as Alzheimer’s, Huntington’s, and Parkinson’s. This may result from disintegration of the extensive connections based on numerous neurotransmitter systems, as well as impairment of some neuroplasticity mechanisms in the NAc. The consequences of NAc lesions are both morphological and functional. They include changes in the NAc’s volume, cell number, modifications of the neuronal dendritic tree and dendritic spines, and changes in the number of synapses. Alterations in the synaptic plasticity affect the efficiency of synaptic transmission. Modification of the number and structure of the receptors affects signaling pathways, the content of neuromodulators (e.g., BDNF) and transcription factors (e.g., pCREB, DeltaFosB, NFκB), and gene expression. Interestingly, changes in the NAc often have a different character and intensity compared to the changes observed in the other parts of the basal ganglia, in particular the dorsal striatum. In this review, we highlight the role of the NAc in various pathological processes in the context of its structural and functional damage, impaired connections with the other brain areas cooperating within functional systems, and progression of the pathological processes.
Collapse
Affiliation(s)
- Martyna Bayassi-Jakowicka
- Division of Anatomy and Neurobiology, Faculty of Medicine, Medical University of Gdańsk, Dębinki 1, 80-211 Gdansk, Poland; (M.B.-J.); (E.C.)
| | - Grazyna Lietzau
- Division of Anatomy and Neurobiology, Faculty of Medicine, Medical University of Gdańsk, Dębinki 1, 80-211 Gdansk, Poland; (M.B.-J.); (E.C.)
- Correspondence: (G.L.); (P.K.); Tel.: +48-58-349-14-01 (G.L. & P.K.)
| | - Ewelina Czuba
- Division of Anatomy and Neurobiology, Faculty of Medicine, Medical University of Gdańsk, Dębinki 1, 80-211 Gdansk, Poland; (M.B.-J.); (E.C.)
| | - Cesare Patrone
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Sjukhusbacken 17, 11883 Stockholm, Sweden;
| | - Przemysław Kowiański
- Division of Anatomy and Neurobiology, Faculty of Medicine, Medical University of Gdańsk, Dębinki 1, 80-211 Gdansk, Poland; (M.B.-J.); (E.C.)
- Correspondence: (G.L.); (P.K.); Tel.: +48-58-349-14-01 (G.L. & P.K.)
| |
Collapse
|
6
|
Abstract
Neuroepigenetics, a new branch of epigenetics, plays an important role in the regulation of gene expression. Neuroepigenetics is associated with holistic neuronal function and helps in formation and maintenance of memory and learning processes. This includes neurodevelopment and neurodegenerative defects in which histone modification enzymes appear to play a crucial role. These modifications, carried out by acetyltransferases and deacetylases, regulate biologic and cellular processes such as apoptosis and autophagy, inflammatory response, mitochondrial dysfunction, cell-cycle progression and oxidative stress. Alterations in acetylation status of histone as well as non-histone substrates lead to transcriptional deregulation. Histone deacetylase decreases acetylation status and causes transcriptional repression of regulatory genes involved in neural plasticity, synaptogenesis, synaptic and neural plasticity, cognition and memory, and neural differentiation. Transcriptional deactivation in the brain results in development of neurodevelopmental and neurodegenerative disorders. Mounting evidence implicates histone deacetylase inhibitors as potential therapeutic targets to combat neurologic disorders. Recent studies have targeted naturally-occurring biomolecules and micro-RNAs to improve cognitive defects and memory. Multi-target drug ligands targeting HDAC have been developed and used in cell-culture and animal-models of neurologic disorders to ameliorate synaptic and cognitive dysfunction. Herein, we focus on the implications of histone deacetylase enzymes in neuropathology, their regulation of brain function and plausible involvement in the pathogenesis of neurologic defects.
Collapse
|
7
|
Brown RW, Bhide PG, Gill WD, Peeters LD. The adenosine A(2A) receptor agonist CGS 21680 alleviates auditory sensorimotor gating deficits and increases in accumbal CREB in rats neonatally treated with quinpirole. Psychopharmacology (Berl) 2020; 237:3519-3527. [PMID: 32772144 PMCID: PMC7686116 DOI: 10.1007/s00213-020-05631-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 01/24/2020] [Accepted: 07/28/2020] [Indexed: 12/21/2022]
Abstract
RATIONALE AND OBJECTIVE The adenosine A(2A) receptor forms a mutually inhibitory heteromer with the dopamine D2 receptor, and A(2A) agonists decrease D2 signaling. This study analyzed whether an adenosine A(2A) agonist would alleviate deficits in sensorimotor gating and increases in cyclic-AMP response element binding protein (CREB) in the nucleus accumbens (NAc) in the neonatal quinpirole model of schizophrenia (SZ). METHODS Male and female Sprague-Dawley rats were neonatally treated with saline (NS) or quinpirole HCl (NQ; 1 mg/kg) from postnatal days (P) 1-21. Animals were raised to P44 and behaviorally tested on auditory sensorimotor gating as measured through prepulse inhibition (PPI) from P44 to P48. Approximately 15 min before each session, animals were given an ip administration of saline or the adenosine A(2A) agonist CGS 21680 (0.03 or 0.09 mg/kg). One day after PPI was complete on P49, animals were administered a locomotor activity test in the open field after saline or CGS 21680 treatment, respectively. On P50, the nucleus accumbens (NAc) was evaluated for CREB protein. RESULTS NQ-treated rats demonstrated a deficit in PPI that was alleviated to control levels by either dose of CGS 21680. The 0.03 mg/kg dose of CGS 21680 increased startle amplitude in males. The 0.09 mg/kg dose of CGS 21680 resulted in an overall decrease in locomotor activity. NQ treatment significantly increased NAc CREB that was attenuated to control levels by either dose of CGS 21680. CONCLUSIONS This study revealed that an adenosine A(2A) receptor agonist was effective to alleviate PPI deficits in the NQ model of SZ in both male and female rats.
Collapse
Affiliation(s)
- Russell W. Brown
- Department of Biomedical Sciences, James H. Quillen College of Medicine East Tennessee State University Johnson City, TN 37614
| | - Pradeep G. Bhide
- Department of Biomedical Sciences and Neuroscience, Florida State University College of Medicine, Tallahassee, FL 32306
| | - W. Drew Gill
- Department of Biomedical Sciences, James H. Quillen College of Medicine East Tennessee State University Johnson City, TN 37614
| | - Loren D. Peeters
- Department of Biomedical Sciences, James H. Quillen College of Medicine East Tennessee State University Johnson City, TN 37614
| |
Collapse
|
8
|
Duan S, Ma Y, Xie L, Zheng L, Huang J, Guo R, Sun Z, Xie Y, Lv J, Lin Z, Ma S. Effects of Chronic Ephedrine Toxicity on Functional Connections, Cell Apoptosis, and CREB-Related Proteins in the Prefrontal Cortex of Rhesus Monkeys. Neurotox Res 2020; 37:602-615. [PMID: 31858422 DOI: 10.1007/s12640-019-00146-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/17/2019] [Revised: 11/20/2019] [Accepted: 11/29/2019] [Indexed: 02/05/2023]
Abstract
Ephedrine abuse has spread in many parts of the world, severely threatening human health. The mechanism of ephedrine toxicity is still unclear. To explore the possible neural mechanisms of ephedrine toxicity, this study established a non-human primate model of ephedrine exposure, analyzed the functional connectivity changes in its prefrontal cortex through resting state BOLD-fMRI, and then inspected the pathophysiological changes as well as the expression of the cyclic adenosine monophosphate response element-binding protein (CREB), phosphorylated CREB (P-CREB), and CREB target proteins (c-fos and fosB) in the prefrontal cortex. After ephedrine toxicity, we found that the prefrontal cortex of monkeys strengthened its functional connectivity with the brain regions that perform motivation, drive, reward, and learning and memory functions and weakened its functional connectivity with the brain regions that perform cognitive control. These results suggest that ephedrine toxicity causes abnormal neural circuits that lead to the amplification and enhancement of drug-related cues and the weakening and damage of cognitive control function. Histology showed that the neurocytotoxicity of ephedrine can cause neuronal degeneration and apoptosis. Real-time PCR and Western blot showed increased expression of CREB mRNA and CREB/P-CREB/c-fos/fosB protein in the prefrontal cortex after ephedrine toxicity. Collectively, the present study indicates that the enhancement of drug-related cues and the weakening of cognitive control caused by abnormal neural circuits after drug exposure may be a major mechanism of brain function changes caused by ephedrine. These histological and molecular changes may be the pathophysiological basis of brain function changes caused by ephedrine.
Collapse
Affiliation(s)
- Shouxing Duan
- Department of Pediatric Surgery, The First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
- Shantou University Medical College, No. 22 Xinling Road, Shantou, Guangdong, 515041, People's Republic of China
- Guangdong Key Laboratory of Medical Molecular Imaging, No. 57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Ye Ma
- Department of Linguistics & Languages, Michigan State University, East Lansing, MI, 48824, USA
| | - Lei Xie
- Shantou University Medical College, No. 22 Xinling Road, Shantou, Guangdong, 515041, People's Republic of China
- Guangdong Key Laboratory of Medical Molecular Imaging, No. 57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Lian Zheng
- Department of Pediatric Surgery, The First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Jinzhuang Huang
- Shantou University Medical College, No. 22 Xinling Road, Shantou, Guangdong, 515041, People's Republic of China
- Guangdong Key Laboratory of Medical Molecular Imaging, No. 57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Ruiwei Guo
- Shantou University Medical College, No. 22 Xinling Road, Shantou, Guangdong, 515041, People's Republic of China
- Guangdong Key Laboratory of Medical Molecular Imaging, No. 57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Zongbo Sun
- Shantou University Medical College, No. 22 Xinling Road, Shantou, Guangdong, 515041, People's Republic of China
- Guangdong Key Laboratory of Medical Molecular Imaging, No. 57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Yao Xie
- Shantou University Medical College, No. 22 Xinling Road, Shantou, Guangdong, 515041, People's Republic of China
- Guangdong Key Laboratory of Medical Molecular Imaging, No. 57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Junyao Lv
- Department of Forensic Medicine, Shantou University Medical College, No. 22 Xinling Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Zhirong Lin
- Shantou University Medical College, No. 22 Xinling Road, Shantou, Guangdong, 515041, People's Republic of China
- Guangdong Key Laboratory of Medical Molecular Imaging, No. 57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Shuhua Ma
- Shantou University Medical College, No. 22 Xinling Road, Shantou, Guangdong, 515041, People's Republic of China.
- Guangdong Key Laboratory of Medical Molecular Imaging, No. 57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China.
- Department of Radiology, The First Affiliated Hospital of Shantou University Medical College, No. 57 Changping Road, Shantou, Guangdong, 515041, People's Republic of China.
| |
Collapse
|
9
|
Pharmacological intervention of histone deacetylase enzymes in the neurodegenerative disorders. Life Sci 2020; 243:117278. [PMID: 31926248 DOI: 10.1016/j.lfs.2020.117278] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/06/2019] [Revised: 12/31/2019] [Accepted: 01/01/2020] [Indexed: 02/06/2023]
Abstract
Reversal of aging symptoms and related disorders are the challenging task where epigenetic is a crucial player that includes DNA methylation, histone modification; chromatin remodeling and regulation that are linked to the progression of various neurodegenerative disorders (NDDs). Overexpression of various histone deacetylase (HDACs) can activate Glycogen synthase kinase 3 which promotes the hyperphosphorylation of tau and inhibits its degradation. While HDAC is important for maintaining the neuronal morphology and brain homeostasis, at the same time, these enzymes are promoting neurodegeneration, if it is deregulated. Different experimental models have also confirmed the neuroprotective effects caused by HDAC enzymes through the regulation of neuronal apoptosis, inflammatory response, DNA damage, cell cycle regulation, and metabolic dysfunction. Apart from transcriptional regulation, protein-protein interaction, histone post-translational modifications, deacetylation mechanism of non-histone protein and direct association with disease proteins have been linked to neuronal imbalance. Histone deacetylases inhibitors (HDACi) can be able to alter gene expression and shown its efficacy on experimental models, and in clinical trials for NDD's and found to be a very promising therapeutic agent with certain limitation, for instance, non-specific target effect, isoform-selectivity, specificity, and limited number of predicted biomarkers. Herein, we discussed (i) the catalytic mechanism of the deacetylation process of various HDAC's in in vivo and in vitro experimental models, (ii) how HDACs are participating in neuroprotection as well as in neurodegeneration, (iii) a comprehensive role of HDACi in maintaining neuronal homeostasis and (iv) therapeutic role of biomolecules to modulate HDACs.
Collapse
|