1
|
Ali T, Luo Y, Zheng C, Rahman SU, Murtaza I, Feng J, Li S. Pentoxifylline Prevents Neuroinflammation and Modifies PTEN/TrkB Signaling in an LPS-Induced Depression Model. J Neuroimmune Pharmacol 2025; 20:31. [PMID: 40175765 DOI: 10.1007/s11481-025-10193-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/20/2025] [Indexed: 04/04/2025]
Abstract
Neuroinflammation affects patients with major depressive disorder and is linked to severe, treatment-resistant symptoms, making it a promising therapeutic target for improving depressive symptoms. This study highlighted the neuroprotective role of pentoxifylline (PTX) against lipopolysaccharide (LPS)-induced neuroinflammation and associated behavioral deficits. Mice were injected with LPS (1 mg/kg, i.p) to induce neuroinflammation and treated with PTX (10 mg/kg, i.p). Behavioral and biochemical analyses were performed to evaluate depressive-like behaviors and examine hippocampal protein expression associated with neuroinflammation and synaptic plasticity. LPS administration increased proinflammatory cytokine production (IL-1, IL6, and TNF-α), microglial activation (IBA-1/GFAP), and dysregulation of key synaptic proteins, including BDNF and TrkB, in the hippocampus of mice. Concomitantly, LPS reduced Phosphatase and tensin homolog (PTEN) phosphorylation, potentially contributing to increased neuroinflammation. PTX treatment effectively attenuated LPS-induced effects by suppressing inflammatory responses, restoring BDNF/TrkB signaling, and rescuing synaptic impairments. Mechanistically, PTX treatment increased PTEN phosphorylation and was reversed by the TrkB inhibitor K252a, suggesting that PTX upregulates TrkB/BDNF signaling, leading to increased PTEN phosphorylation and subsequent inhibition of PTEN activity. These findings highlight the potential of PTX as a therapeutic agent for neuroinflammatory conditions, possibly exerting its effects by modulating the PTEN/TrkB/BDNF signaling axis and suggest a novel mechanism of action involving the modulation of the PTEN/TrkB/BDNF signaling pathway.
Collapse
Affiliation(s)
- Tahir Ali
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, China.
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, PR China, 518055.
| | - Yanhua Luo
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, PR China, 518055
| | - Chengyou Zheng
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, PR China, 518055
- Department of Neonatology, Shenzhen Children'S Hospital, Shenzhen, China
| | - Shafiq Ur Rahman
- Department of Pharmacy, Shaheed Benazir Bhutto, University, Dir 18000, Sheringal, KP, Pakistan
| | - Iram Murtaza
- Signal Transduction Laboratory, Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Jinxing Feng
- Department of Neonatology, Shenzhen Children'S Hospital, Shenzhen, China.
| | - Shupeng Li
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, China.
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, PR China, 518055.
| |
Collapse
|
2
|
Yu H, Li X, Ning B, Feng L, Ren Y, Li S, Kang Y, Ma J, Zhao M. SIRT1: a potential therapeutic target for coronary heart disease combined with anxiety or depression. J Drug Target 2025; 33:328-340. [PMID: 39470049 DOI: 10.1080/1061186x.2024.2422882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024]
Abstract
Coronary heart disease (CHD) combined with anxiety or depression is increasingly receiving attention in the clinical field of cardiology, and exploring the comorbidity pathological mechanisms of cardiovascular disease combined with psychological disorders is a hot research topic for scholars in this field. Current research suggests that Silent Information Regulatory Factor 1 (SIRT1) may serve as a potential biomarker for the comorbidity mechanism and treatment of CHD with anxiety or depression. SIRT1 is considered a promising therapeutic target for CHD combined with anxiety or depression, with the ability to regulate inflammatory cytokine levels, alleviate oxidative stress damage, activate multiple signalling pathways, reduce platelet hyperresponsiveness, and exert neuroprotective and cardioprotective effects. In this comprehensive review, we deeply studied the structure, function, and mechanism of SIRT1, and discussed its protective effects in the cardiovascular and nervous system. The latest progress in the mechanism of SIRT1's role in CHD combined with anxiety or depression was emphasised, including its specific mechanisms in regulating inflammatory response, alleviating oxidative stress, and mediating various signalling pathways. In addition, this article also summarises the therapeutic potential of SIRT1 as a potential biomarker in patients with CHD combined with anxiety or depression.
Collapse
Affiliation(s)
- Hubin Yu
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Xinping Li
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Bo Ning
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Lanshuan Feng
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yaolong Ren
- Department of Cardiology, Affliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Shilin Li
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yalong Kang
- School of Graduate, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Jing Ma
- Department of Traditional Chinese Medicine, First Affiliated Hospital of Air Force Military Medical University, Xi'an, China
| | - Mingjun Zhao
- Department of Cardiology, Affliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| |
Collapse
|
3
|
Lovis EP, Pereira GC, Viero FT, Arboit F, de Andrade LG, Becker G, Pessano Fialho MF, da Silva Brum E, de Souza Ferreira JE, Zanchet EM, Marques Portela Junior VV, Dos Santos GT, Oliveira SM, Pillat MM, Bochi GV. Lipopolysaccharide preconditioning disrupts the behavioral and molecular response to restraint stress in male mice. Neuroscience 2025; 567:281-293. [PMID: 39798836 DOI: 10.1016/j.neuroscience.2024.12.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/09/2024] [Accepted: 12/16/2024] [Indexed: 01/15/2025]
Abstract
Major depressive disorder (MDD) is a complex neuropsychiatric disorder potentially influenced by factors such as stress and inflammation. Chronic stress can lead to maladaptive brain changes that may trigger immune hyperactivation, contributing to MDD's pathogenesis. While the involvement of inflammation in MDD is well established, the effects of inflammatory preconditioning in animals subsequently exposed to chronic stress remain unclear. This study aimed to investigate the impact of inflammatory preconditioning on behavioral, biochemical, and molecular changes in adult male Swiss mice subjected to chronic restraint stress (CRS). The mice received a single injection of lipopolysaccharide (LPS) 24 h before thefirst CRS and performed 6 h daily for 28 days. Behavioral tests were conducted 24 h after the last CRS, across 4 days, and euthanasia followed 24 h after the final tests. Results indicated that only the LPS + CRS group exhibited depressive- and anxiety-like behaviors, accompanied by demotivation and apathy. Biochemical and molecular analyses revealed anoxidative imbalance in the hippocampus, marked by elevated H2O2 levels and MPO activity. In the prefrontal cortex, theLPS + CRS group demonstrated a central inflammatory imbalance, with reduced IL-10 levels, increased Iba1 gene expression, and decreased Gfap and Bdnf gene expression. A trend toward elevated IL-17 levels was also observed at the peripheral level. These findings indicate that inflammatory preconditioning contributes significantly to behaviors phenotypically associated with MDD. Furthermore, the study suggests that these behavioral changes are linked to a dysfunctional immune response and impaired neuroplasticity.
Collapse
Affiliation(s)
- Elisa Piton Lovis
- Center of Health Sciences, Department of Physiology and Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil; Center of Health Sciences, Postgraduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Gabriele Cheiran Pereira
- Center of Health Sciences, Department of Physiology and Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil; Center of Health Sciences, Postgraduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Fernanda Tibolla Viero
- Center of Health Sciences, Department of Physiology and Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil; Center of Health Sciences, Postgraduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Francini Arboit
- Center of Rural Sciences, Postgraduate Program in Veterinary Medicine, Federal University of Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - Leonardo Guedes de Andrade
- Laboratory of Biotechnology and Animal Reproduction (BioRep), Veterinary Hospital, Federal University of Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - Gabriela Becker
- Center of Natural and Exact Sciences, Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Maria Fernanda Pessano Fialho
- Center of Natural and Exact Sciences, Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Evelyne da Silva Brum
- Center of Natural and Exact Sciences, Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - José Eduardo de Souza Ferreira
- Center of Health Sciences, Department of Physiology and Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Eliane Maria Zanchet
- Center of Health Sciences, Department of Physiology and Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil; Center of Health Sciences, Postgraduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Valerio Valdetar Marques Portela Junior
- Center of Health Sciences, Department of Physiology and Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil; Laboratory of Biotechnology and Animal Reproduction (BioRep), Veterinary Hospital, Federal University of Santa Maria (UFSM), Santa Maria, RS, Brazil
| | - Gabriela Trevisan Dos Santos
- Center of Health Sciences, Department of Physiology and Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil; Center of Health Sciences, Postgraduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Sara Marchesan Oliveira
- Center of Natural and Exact Sciences, Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Micheli Mainardi Pillat
- Center of Health Sciences, Postgraduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil; Center of Health Sciences, Postgraduate program in Pharmaceutical Sciences, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Guilherme Vargas Bochi
- Center of Health Sciences, Department of Physiology and Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil; Center of Health Sciences, Postgraduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil.
| |
Collapse
|
4
|
Farhadi R, Daniali M, Baeeri M, Khorasani R, Haghi-Aminjan H, Gholami M, Rahimifard M, Navaei-Nigjeh M, Abdollahi M. Molecular evidence of the inhibitory potential of melatonin against sodium arsenite toxicity. Heliyon 2025; 11:e42113. [PMID: 39916822 PMCID: PMC11799970 DOI: 10.1016/j.heliyon.2025.e42113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 02/09/2025] Open
Abstract
Introduction Sodium arsenite (SA), NaAsO2, is among the most hazardous toxicants, and wide use and presence of this toxicant leads to a severe environmental threat. Exposure to SA is associated with many health concerns, such as the prevalence of cancer and diabetes mellitus type 2 (DMT2). Many studies suggest that SA induces inflammation and biochemical impairments through different mechanisms, including increasing oxidative stress and altering vital genes such as biochemical and anti-inflammatory. Recent studies on melatonin (MLT), a harmless hormone secreted in the body generally for induction of sleepiness, find many beneficial and positive effects. Mitigating different harms and toxicities through different mechanisms, such as antioxidant properties, anti-inflammatory effects, and critical gene regulation, is essential. Due to these findings, this study aimed to evaluate the hypothesis that MLT may ameliorate pancreatic damage caused by exposure to SA. Methods Forty-eight adult healthy male wistar rats aged 7-8 weeks were divided into eight for this research. Group 1 did not receive any intervention. Group 2 received 10 mg/kg/day MLT through intraperitoneal (IP) injection. Groups 3, 4, and 5 received 1.5 (1/10 LD50), 5 (1/3 LD50), and 7.5 (1/2 LD50) mg/kg SA, respectively. Groups 6, 7, and 8 were given 1.5 (1/10 LD50), 5 (1/3 LD50), and 7.5 (1/2 LD50) mg/kg of SA along with 10 mg/kg/day MLT, respectively, during the last ten days of the experiment. After 28 days of the experiment, the blood and tissue samples of the pancreas were removed for biochemical and pathological examination. Results MLT attenuates SA toxicity by reducing oxidative stress biomarkers and inflammation markers. Moreover, MLT improves SA exposure's biochemical and functional damages by regulating related genes and pathways. Conclusion MLT poses protective and preventive effects on the pancreas against exposure to SA. However, MLT's therapeutic and beneficial impacts have great potential for further investigation.
Collapse
Affiliation(s)
- Ramtin Farhadi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Marzieh Daniali
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Maryam Baeeri
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Reza Khorasani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Hamed Haghi-Aminjan
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mahdi Gholami
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mahban Rahimifard
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mona Navaei-Nigjeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Abdollahi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
5
|
Mukherjee R, Rana R, Mehan S, Khan Z, Das Gupta G, Narula AS, Samant R. Investigating the Interplay Between the Nrf2/Keap1/HO-1/SIRT-1 Pathway and the p75NTR/PI3K/Akt/MAPK Cascade in Neurological Disorders: Mechanistic Insights and Therapeutic Innovations. Mol Neurobiol 2025:10.1007/s12035-025-04725-8. [PMID: 39920438 DOI: 10.1007/s12035-025-04725-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 01/27/2025] [Indexed: 02/09/2025]
Abstract
Neurological illnesses are debilitating diseases that affect brain function and balance. Due to their complicated aetiologies and progressive nature, neurodegenerative and neuropsychiatric illnesses are difficult to treat. These incurable conditions damage brain functions like mobility, cognition, and emotional regulation, but medication, gene therapy, and physical therapy can manage symptoms. Disruptions in cellular signalling pathways, especially those involving oxidative stress response, memory processing, and neurotransmitter modulation, contribute to these illnesses. This review stresses the interplay between key signalling pathways involved in neurological diseases, such as the Nrf2/Keap1/HO-1/SIRT-1 axis and the p75NTR/PI3K/Akt/MAPK cascade. To protect neurons from oxidative damage and death, the Nrf2 transcription factor promotes antioxidant enzyme production. The Keap1 protein releases Nrf2 during oxidative stress for nuclear translocation and gene activation. The review also discusses how neurotrophin signalling through the p75 neurotrophin receptor (p75NTR) determines cell destiny, whether pro-survival or apoptotic. The article highlights emerging treatment approaches targeting these signalling pathways by mapping these connections. Continued research into these molecular pathways may lead to new neurological disease treatments that restore cellular function and neuronal survival. In addition to enhanced delivery technologies, specific modulators and combination therapies should be developed to fine-tune signalling responses. Understanding these crosstalk dynamics is crucial to strengthening neurological illness treatment options and quality of life.
Collapse
Affiliation(s)
- Ritam Mukherjee
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India
| | - Ravi Rana
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India.
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC, 27516, USA
| | - Rajaram Samant
- Chief Scientific Officer, Celagenex Research, Mumbai, India
| |
Collapse
|
6
|
Sethi P, Mehan S, Khan Z, Maurya PK, Kumar N, Kumar A, Tiwari A, Sharma T, Das Gupta G, Narula AS, Kalfin R. The SIRT-1/Nrf2/HO-1 axis: Guardians of neuronal health in neurological disorders. Behav Brain Res 2025; 476:115280. [PMID: 39368713 DOI: 10.1016/j.bbr.2024.115280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/10/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
SIRT1 (Sirtuin 1) is a NAD+-dependent deacetylase that functions through nucleoplasmic transfer and is present in nearly all mammalian tissues. SIRT1 is believed to deacetylate its protein substrates, resulting in neuroprotective actions, including reduced oxidative stress and inflammation, increased autophagy, increased nerve growth factors, and preserved neuronal integrity in aging or neurological disease. Nrf2 is a transcription factor that regulates the genes responsible for oxidative stress response and substance detoxification. The activation of Nrf2 guards cells against oxidative damage, inflammation, and carcinogenic stimuli. Several neurological abnormalities and inflammatory disorders have been associated with variations in Nrf2 activation caused by either pharmacological or genetic factors. Recent evidence indicates that Nrf2 is at the center of a complex cellular regulatory network, establishing it as a transcription factor with genuine pleiotropy. HO-1 is most likely a component of a defense mechanism in cells under stress, as it provides negative feedback for cell activation and mediator synthesis. This mediator is upregulated by Nrf2, nitric oxide (NO), and other factors in various inflammatory states. HO-1 or its metabolites, such as CO, may mitigate inflammation by modulating signal transduction pathways. Neurological diseases may be effectively treated by modulating the activity of HO-1. Multiple studies have demonstrated that SIRT1 and Nrf2 share an important connection. SIRT1 enhances Nrf2, activates HO-1, protects against oxidative injury, and decreases neuronal death. This has been associated with numerous neurodegenerative and neuropsychiatric disorders. Therefore, activating the SIRT1/Nrf2/HO-1 pathway may help treat various neurological disorders. This review focuses on the current understanding of the SIRT1 and Nrf2/HO-1 neuroprotective processes and the potential therapeutic applications of their target activators in neurodegenerative and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Pranshul Sethi
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India.
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Pankaj Kumar Maurya
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Nitish Kumar
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh 201204, India
| | - Aakash Kumar
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Aarti Tiwari
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Tarun Sharma
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar), Moga, Punjab 144603, India
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| | - Reni Kalfin
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev St., Block 23, Sofia 1113, Bulgaria; Department of Healthcare, South-West University "NeofitRilski", Ivan Mihailov St. 66, Blagoevgrad 2700, Bulgaria
| |
Collapse
|
7
|
Gao M, Li J, Han X, Zhang B, Chen J, Lang J, Zhang Q. Effect of melatonin on gut microbiome and metabolomics in diabetic cognitive impairment. Front Pharmacol 2024; 15:1489834. [PMID: 39640487 PMCID: PMC11619431 DOI: 10.3389/fphar.2024.1489834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/22/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction Diabetic cognitive impairment(DCI) presents as a central nervous complication of diabetes especially among aging population. Melatonin (MEL) is known for its antioxidant and anti-inflammation effects in neuroprotective aspects. Recent evidence has demonstrated that the gut microbiome plays a key role in DCI by modulating cognitive function through the gut-brain crosstalk. MEL has been shown to modulate gut microbiota composition in diabetic model. However, the underlying mechanism through which the gut microbiome contributes to DCI remains unclear. This study aims to investigate the effect and mechanism of MEL in attenuating DCI in relation to regulating the gut microbiome and metabolomics. Methods Cognitive and memory function were assessed by the Morris water maze test, histopathological assessment of brain tissues, and immunoblotting of neuroinflammation and apoptosis. The levels of serum tumor necrosis factor-α (TNF-α) and Interleukin-18 (IL-18) were measured by enzyme-linked immunoassays to reflect the circulatory inflammation level.16S rRNA microbiome sequencing analysis was performed on control mice(db-m group), diabetic mice(db-db group) and MEL-treated diabetic mice(db-dbMEL group). Gut metabolites changes were characterized using liquid chromatography tandem mass spectrometry (LC-MS/MS). Results Our study confirmed that MEL alleviated diabetes-induced cognition and memory dysfunction. MEL protected against neuroinflammation and apoptosis in hippocampus of db-db mice. MEL corrected the increased abundance of Bacteroides and Dorea and the reduced abundance of Prevotella in db-db mice. The vast majority of differential metabolites among the three groups were lipids and lipid-like molecules. MEL significantly restored the reduced levels of pyruvate and lactic acid. Discussion Our results supported the use of MEL as a promising therapeutic agent for DCI, in which the underlying mechanism may be associated with gut microbiome and metabolomics regulation.
Collapse
Affiliation(s)
- Ming Gao
- Department of Endocrinology and Rare Disease, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Rare Disease, Hebei Provincial Department of Science and Technology, Shijiazhuang, Hebei, China
| | - Jie Li
- Department of Endocrinology and Rare Disease, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Rare Disease, Hebei Provincial Department of Science and Technology, Shijiazhuang, Hebei, China
| | - Xu Han
- Department of Endocrinology and Rare Disease, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Beiyao Zhang
- Department of Endocrinology and Rare Disease, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jinting Chen
- Core Facilities and Centers, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jiadong Lang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qiangqiang Zhang
- Department of Endocrinology and Rare Disease, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
8
|
Li W, Luo Y, Ali T, Huang Y, Yu ZJ, Hao L, Li S. Hsp60 deletion in cholinergic neurons: Impact on neuroinflammation and memory. Int Immunopharmacol 2024; 141:113022. [PMID: 39213869 DOI: 10.1016/j.intimp.2024.113022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/25/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Cholinergic circuit defects have been linked to various neurological abnormalities, yet the precise mechanisms underlying the impact of cholinergic signaling on cognitive functions, particularly in the context of neuroinflammation-associated, remain poorly understood. Similarly, while the dopamine receptor (D2R) has been implicated in the pausing of cholinergic interneurons (CIN), its relationship with behavior remains inadequately elucidated. In this study, we aimed to investigate whether D2R plays a role in the regulation of fear and memory in the Hsp60 knockout condition, given the non-canonical involvement of Hsp60 in inflammation. Using a CRE-floxed system, we selectively generated cholinergic neurons specific to Hsp60 knockout mice and subjected them to memory tests. Our results revealed a significant increase in freezing levels during recall and contextual tests in Hsp60-deprived mice. We also observed dysregulation of neurotransmitters and D2R in the hippocampus of Hsp60 knockout mice, along with enhanced impairments in cytokine levels and synaptic protein dysregulations. These changes were accompanied by alterations in PI3K/eIF4E/Jak/ERK/CREB signaling pathways. Notably, D2R agonism via Quinpirole led to a decrease in freezing levels during recall and contextual tests, alongside an increase in IBA-1 expression and improvements in inflammatory response-linked signaling pathways, including JAK/STAT/P38/JNK impairments. Given that these pathways are well-known downstream signaling cascades of D2R, our findings suggest that D2R signaling may contribute to the neuroinflammation induced by Hsp60 deprivation, potentially exacerbating memory impairments.
Collapse
Affiliation(s)
- Weifen Li
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518060, China; Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, The 6th Affiliated Hospital of Shenzhen University Health Science Center. No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China; State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China.
| | - Yanhua Luo
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China.
| | - Tahir Ali
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China.
| | - Yangmei Huang
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China.
| | - Zhi-Jian Yu
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, The 6th Affiliated Hospital of Shenzhen University Health Science Center. No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China.
| | - Liangliang Hao
- Hospital of Chengdu University of Traditional Chinese Medicine, No.39 Shier-Qiao Road, Chengdu, China.
| | - Shupeng Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China; Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
9
|
Luo B. Insights into the advances in therapeutic drugs for neuroinflammation-related diseases. Int J Neurosci 2024; 134:1256-1281. [PMID: 37722706 DOI: 10.1080/00207454.2023.2260088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/05/2023] [Accepted: 09/12/2023] [Indexed: 09/20/2023]
Abstract
Studies have shown that neurodegenerative diseases such as AD and PD are related to neuroinflammation. Neuroinflammation is a common inflammatory condition that can lead to a variety of dysfunction in the body. At present, it is no medications specifically approved to prevent or cure neuroinflammation, so even though many drugs can temporarily control the neurological symptoms of neuroinflammation, but no one can reverse the progress of neuroinflammation, let al.one completely cure neuroinflammation. Therefore, it is urgent to develop new drug development for neuroinflammation treatment. In this review, we highlight the therapeutic advancement in the field of neurodegenerative disorders, by focusing on the impact of neuroinflammation treatment has on these conditions, and the effective drugs for the treatment of neuroinflammation and neurodegenerative diseases and their latest research progress are reviewed according to the related signaling pathway, as well as the prospect of their clinical application is also discussed. The purpose of this review is to enable specialists to better understand the mechanisms underlying neuroinflammation and anti-inflammatory drugs, promote the development of therapeutic drugs for neuroinflammation and neurodegenerative diseases, and further provide therapeutic references for clinical neurologists.
Collapse
Affiliation(s)
- Bozhi Luo
- School of Basic Medicine, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
10
|
Jan A, Shah M, Shah SA, Habib SH, Ehtesham E, Ahmed N. Melatonin rescues pregnant female mice and their juvenile offspring from high fat diet-induced alzheimer disease neuropathy. Heliyon 2024; 10:e36921. [PMID: 39281480 PMCID: PMC11395765 DOI: 10.1016/j.heliyon.2024.e36921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/18/2024] Open
Abstract
High fat diet (HFD) is a prime factor, which contributes to the present epidemic of metabolic syndrome. Prolonged intake of HFD induces oxidative stress (OS) that in turn causes neuroinflammation, neurodegeneration, insulin resistance, amyloid burden, synaptic dysfunction and cognitive impairment hence leading to Alzheimer's disease neuropathy. Melatonin (secreted by the Pineal gland) has the potential to nullify the toxic effects of reactive oxygen species (ROS) and have been shown to ameliorate various complications induced by HFD in rodent models. This study aimed to assess the neurotherapeutic effects of melatonin on HFD-induced neuroinflammation and neurodegeneration mediated by OS in pregnant female mice and their offspring. Western blotting, immunohistochemistry and antioxidant enzyme assays were used for quantification of samples from the hippocampal region of the brain of pregnant albino mice and their offspring. Short- and long-term memory was assessed by Y-maze and Morris Water Maze tests. HFD significantly induced OS leading to AD like neuropathology in the pregnant mice and their offspring while melatonin administration simultaneously with the HFD significantly prevented this neuropathy. This study reports that melatonin exerts these effects through the stimulation of SIRT1/Nrf2/HO-1 pathway that in turn reduces the HFD-induced OS and its downstream signaling. In conclusion melatonin prevents HFD-induced multiple complications that ultimately leads to the memory dysfunction in pregnant female mice and their successive generation via activation of SIRT1/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Amin Jan
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Mohsin Shah
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Shahid Ali Shah
- Department of Biochemistry, Haripur University, Haripur, Pakistan
| | - Syed Hamid Habib
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Ehtesham Ehtesham
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Naseer Ahmed
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| |
Collapse
|
11
|
Gao X, Sun H, Wei Y, Niu J, Hao S, Sun H, Tang G, Qi C, Ge J. Protective effect of melatonin against metabolic disorders and neuropsychiatric injuries in type 2 diabetes mellitus mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 131:155805. [PMID: 38851097 DOI: 10.1016/j.phymed.2024.155805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/11/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is a metabolic disease characterized by hyperglycemia and progressive cognitive dysfunction, and our clinical investigation revealed that the plasma concentration of melatonin (Mlt) decreased and was closely related to cognition in T2DM patients. However, although many studies have suggested that Mlt has a certain protective effect on glucose and lipid metabolism disorders and neuropsychiatric injury, the underlying mechanism of Mlt against T2DM-related metabolic and cognitive impairments remains unclear. PURPOSE The aim of the present study was to investigate the therapeutic effect of Mlt on metabolic disorders and Alzheimer's disease (AD)-like neuropsychiatric injuries in T2DM mice and to explore the possible underlying molecular mechanism involved. METHODS A T2DM mouse model was established by a combination of a high-fat diet (HFD) and streptozotocin (STZ, 100 mg/kg, i.p.), and Mlt (5, 10 or 20 mg/kg) was intragastrically administered for six consecutive weeks. The serum levels of glycolipid metabolism indicators were measured, behavioral performance was tested, and the protein expression of key molecules involved in the regulation of synaptic plasticity, circadian rhythms, and neuroinflammation in the hippocampus was detected. Moreover, the fluorescence intensities of glial fibrillary acidic protein (GFAP), ionized calcium binding adapter molecule 1 (IBA-1), amyloid β-protein (Aβ) and phosphorylated Tau (p-Tau) in the hippocampus were also observed. RESULTS Treatment with Mlt not only improved T2DM-related metabolic disorders, as indicated by increased serum concentrations of fasting blood glucose (FBG), glycosylated hemoglobin (HbAlc), insulin (INS), total cholesterol (TC) and triglyceride (TG), improved glucose tolerance and liver and pancreas function but also alleviated AD-like neuropsychiatric injuries in a HFD/STZ-induced mouse model, as indicated by decreased immobility time in the tail suspension test (TST) and forced swimming test (FST), increased preference indices of novel objects or novel arms in the novel object recognition test (NOR) and Y-maze test (Y-maze), and improved platform positioning capability in the Morris water maze (MWM) test. Moreover, treatment with Mlt also improved the hyperactivation of astrocytes and microglia in the hippocampus of mice, accompanied by reduced expression of interleukin 1β (IL-1β), interleukin 6 (IL-6), tumor necrosis factor (TNF-α), Aβ, and p-Tau and increased expression of brain-derived neurotrophic factor (BDNF), Synapsin I, Synaptotagmin I, melatonin receptor 1B (MT1B), brain muscle arnt-like protein 1 (Bmal1), circadian locomotor output cycles kaput (Clock), period 2 (Per2), and cryptochrome 2 (Cry2). CONCLUSION Mlt alleviated T2DM-related metabolic disorders and AD-like neuropsychiatric injuries in a HFD/STZ-induced mouse model, possibly through a mechanism involving the regulation of glial activation and associated neuroinflammation and the balancing of synaptic plasticity and circadian rhythms in the hippocampus.
Collapse
Affiliation(s)
- Xinran Gao
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, PR China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, PR China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, PR China
| | - Huaizhi Sun
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, PR China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, PR China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, PR China
| | - Yadong Wei
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, PR China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, PR China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, PR China
| | - Jiachun Niu
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, PR China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, PR China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, PR China
| | - Shengwei Hao
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, PR China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, PR China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, PR China
| | - Huimin Sun
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, PR China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, PR China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, PR China
| | - Guozhang Tang
- School of 1st Clinic Medicine, Anhui Medical University, 81 Meishan Road, Hefei 230032, PR China
| | - Congcong Qi
- Department of Laboratory Animal Science, Fudan University, Shanghai, PR China.
| | - Jinfang Ge
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, PR China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, PR China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, PR China.
| |
Collapse
|
12
|
Branković M, Gmizić T, Dukić M, Zdravković M, Daskalović B, Mrda D, Nikolić N, Brajković M, Gojgić M, Lalatović J, Kralj Đ, Pantić I, Vojnović M, Milovanović T, Đurašević S, Todorović Z. Therapeutic Potential of Palmitoylethanolamide in Gastrointestinal Disorders. Antioxidants (Basel) 2024; 13:600. [PMID: 38790705 PMCID: PMC11117950 DOI: 10.3390/antiox13050600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/02/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Palmitoylethanolamide (PEA) is an endocannabinoid-like bioactive lipid mediator belonging to the family of N-acylethanolamines, most abundantly found in peanuts and egg yolk. When the gastrointestinal (GI) effects of PEA are discussed, it must be pointed out that it affects intestinal motility but also modulates gut microbiota. This is due to anti-inflammatory, antioxidant, analgesic, antimicrobial, and immunomodulatory features. Additionally, PEA has shown beneficial effects in several GI diseases, particularly irritable bowel syndrome and inflammatory bowel diseases, as various studies have shown, and it is important to emphasize its relative lack of toxicity, even at high dosages. Unfortunately, there is not enough endogenous PEA to treat disturbed gut homeostasis, even though it is produced in the GI tract in response to inflammatory stimuli, so exogenous intake is mandatory to achieve homeostasis. Intake of PEA could be through animal and/or vegetable food, but bearing in mind that a high dosage is needed to achieve a therapeutic effect, it must be compensated through dietary supplements. There are still open questions pending to be answered, so further studies investigating PEA's effects and mechanisms of action, especially in humans, are crucial to implementing PEA in everyday clinical practice.
Collapse
Affiliation(s)
- Marija Branković
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia; (T.G.); (M.D.); (M.Z.); (D.M.); (N.N.); (M.B.); (J.L.); (Z.T.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Tijana Gmizić
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia; (T.G.); (M.D.); (M.Z.); (D.M.); (N.N.); (M.B.); (J.L.); (Z.T.)
| | - Marija Dukić
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia; (T.G.); (M.D.); (M.Z.); (D.M.); (N.N.); (M.B.); (J.L.); (Z.T.)
| | - Marija Zdravković
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia; (T.G.); (M.D.); (M.Z.); (D.M.); (N.N.); (M.B.); (J.L.); (Z.T.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | | | - Davor Mrda
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia; (T.G.); (M.D.); (M.Z.); (D.M.); (N.N.); (M.B.); (J.L.); (Z.T.)
| | - Novica Nikolić
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia; (T.G.); (M.D.); (M.Z.); (D.M.); (N.N.); (M.B.); (J.L.); (Z.T.)
| | - Milica Brajković
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia; (T.G.); (M.D.); (M.Z.); (D.M.); (N.N.); (M.B.); (J.L.); (Z.T.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Milan Gojgić
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia; (T.G.); (M.D.); (M.Z.); (D.M.); (N.N.); (M.B.); (J.L.); (Z.T.)
| | - Jovana Lalatović
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia; (T.G.); (M.D.); (M.Z.); (D.M.); (N.N.); (M.B.); (J.L.); (Z.T.)
| | - Đorđe Kralj
- University Hospital Medical Center Zvezdara, 11000 Belgrade, Serbia;
| | - Ivana Pantić
- Clinic of Gastroenterology and Hepatology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (I.P.); (M.V.)
| | - Marko Vojnović
- Clinic of Gastroenterology and Hepatology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (I.P.); (M.V.)
| | - Tamara Milovanović
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
- Clinic of Gastroenterology and Hepatology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (I.P.); (M.V.)
| | - Siniša Đurašević
- Department for Comparative Physiology and Ecophysiology, Institute for Physiology and Biochemistry Ivan Đaja, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia;
| | - Zoran Todorović
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia; (T.G.); (M.D.); (M.Z.); (D.M.); (N.N.); (M.B.); (J.L.); (Z.T.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| |
Collapse
|
13
|
Zemniaçak ÂB, Ribeiro RT, Pinheiro CV, de Azevedo Cunha S, Tavares TQ, Castro ET, Leipnitz G, Wajner M, Amaral AU. In Vivo Intracerebral Administration of α-Ketoisocaproic Acid to Neonate Rats Disrupts Brain Redox Homeostasis and Promotes Neuronal Death, Glial Reactivity, and Myelination Injury. Mol Neurobiol 2024; 61:2496-2513. [PMID: 37910283 DOI: 10.1007/s12035-023-03718-9] [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: 08/27/2023] [Accepted: 10/17/2023] [Indexed: 11/03/2023]
Abstract
Maple syrup urine disease (MSUD) is caused by severe deficiency of branched-chain α-keto acid dehydrogenase complex activity, resulting in tissue accumulation of branched-chain α-keto acids and amino acids, particularly α-ketoisocaproic acid (KIC) and leucine. Affected patients regularly manifest with acute episodes of encephalopathy including seizures, coma, and potentially fatal brain edema during the newborn period. The present work investigated the ex vivo effects of a single intracerebroventricular injection of KIC to neonate rats on redox homeostasis and neurochemical markers of neuronal viability (neuronal nuclear protein (NeuN)), astrogliosis (glial fibrillary acidic protein (GFAP)), and myelination (myelin basic protein (MBP) and 2',3'-cyclic-nucleotide 3'-phosphodiesterase (CNPase)) in the cerebral cortex and striatum. KIC significantly disturbed redox homeostasis in these brain structures 6 h after injection, as observed by increased 2',7'-dichlorofluorescein oxidation (reactive oxygen species generation), malondialdehyde levels (lipid oxidative damage), and carbonyl formation (protein oxidative damage), besides impairing the antioxidant defenses (diminished levels of reduced glutathione and altered glutathione peroxidase, glutathione reductase, and superoxide dismutase activities) in both cerebral structures. Noteworthy, the antioxidants N-acetylcysteine and melatonin attenuated or normalized most of the KIC-induced effects on redox homeostasis. Furthermore, a reduction of NeuN, MBP, and CNPase, and an increase of GFAP levels were observed at postnatal day 15, suggesting neuronal loss, myelination injury, and astrocyte reactivity, respectively. Our data indicate that disruption of redox homeostasis, associated with neural damage caused by acute intracerebral accumulation of KIC in the neonatal period may contribute to the neuropathology characteristic of MSUD patients.
Collapse
Affiliation(s)
- Ângela Beatris Zemniaçak
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Rafael Teixeira Ribeiro
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Camila Vieira Pinheiro
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Sâmela de Azevedo Cunha
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Tailine Quevedo Tavares
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Ediandra Tissot Castro
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Guilhian Leipnitz
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Moacir Wajner
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Alexandre Umpierrez Amaral
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Programa de Pós-Graduação em Atenção Integral à Saúde, Universidade Regional Integrada do Alto Uruguai e das Missões, Avenida Sete de Setembro, 1621, Erechim, RS, 99709-910, Brazil.
| |
Collapse
|
14
|
Li W, Ali T, He K, Zheng C, Li N, Yu Z, Li S. ApoE4 dysregulation incites depressive symptoms and mitochondrial impairments in mice. J Cell Mol Med 2024; 28:e18160. [PMID: 38506067 PMCID: PMC10951871 DOI: 10.1111/jcmm.18160] [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: 07/10/2023] [Revised: 01/12/2024] [Accepted: 01/18/2024] [Indexed: 03/21/2024] Open
Abstract
Apolipoprotein E4 (ApoE4) is involved in the stress-response processes and is hypothesized to be a risk factor for depression by means of mitochondrial dysfunction. However, their exact roles and underlying mechanisms are largely unknown. ApoE4 transgenic mice (B6. Cg-ApoEtm1Unc Cdh18Tg( GFAP-APOE i4)1Hol /J) were subjected to stress (lipopolysaccharides, LPS) to elucidate the aetiology of ApoE4-induced depression. LPS treatment significantly aggravated depression-like behaviours, concurrent with neuroinflammation and impaired mitochondrial changes, and melatonin/Urolithin A (UA) + 5-aminoimidazole-4-carboxamide 1-β-D-ribofuranoside (AICAR) reversed these effects in ApoE4 mice. Concurrently, ApoE4 mice exhibited mitophagy deficits, which could be further exacerbated by LPS stimulation, as demonstrated by reduced Atg5, Beclin-1 and Parkin levels, while PINK1 levels were increased. However, these changes were reversed by melatonin treatment. Additionally, proteomic profiling suggested mitochondria-related signalling and network changes in ApoE4 mice, which may underlie the exaggerated response to LPS. Furthermore, HEK 293T cells transfected with ApoE4 showed mitochondria-associated protein and mitophagy defects, including PGC-1α, TFAM, p-AMPKα, PINK1 and LC3B impairments. Additionally, it aggravates mitochondrial impairment (particularly mitophagy), which can be attenuated by triggering autophagy. Collectively, ApoE4 dysregulation enhanced depressive behaviour upon LPS stimulation.
Collapse
Affiliation(s)
- Weifen Li
- Department of Infectious Diseases, Huazhong University of Science and Technology Union Shenzhen HospitalShenzhen University School of MedicineShenzhenChina
- State Key Laboratory of Oncogenomics, School of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| | - Tahir Ali
- State Key Laboratory of Oncogenomics, School of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
- Shenzhen Bay LaboratoryShenzhenChina
| | - Kaiwu He
- State Key Laboratory of Oncogenomics, School of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| | - Chengyou Zheng
- State Key Laboratory of Oncogenomics, School of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| | - Ningning Li
- Tomas Lindahl Nobel Laureate Laboratory, Precision Medicine Research CentreThe Seventh Affiliated Hospital of Sun Yat‐sen UniversityShenzhenChina
| | - Zhi‐Jian Yu
- Department of Infectious Diseases, Huazhong University of Science and Technology Union Shenzhen HospitalShenzhen University School of MedicineShenzhenChina
| | - Shupeng Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
- Shenzhen Bay LaboratoryShenzhenChina
- Campbell Research Institute, Centre for Addiction and Mental HealthTorontoOntarioCanada
- Department of PsychiatryUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
15
|
Pan HC, Yang CN, Lee WJ, Sheehan J, Wu SM, Chen HS, Lin MH, Shen LW, Lee SH, Shen CC, Pan LY, Liu SH, Sheu ML. Melatonin Enhanced Microglia M2 Polarization in Rat Model of Neuro-inflammation Via Regulating ER Stress/PPARδ/SIRT1 Signaling Axis. J Neuroimmune Pharmacol 2024; 19:11. [PMID: 38530514 DOI: 10.1007/s11481-024-10108-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 02/15/2024] [Indexed: 03/28/2024]
Abstract
Neuro-inflammation involves distinct alterations of microglial phenotypes, containing nocuous pro-inflammatory M1-phenotype and neuroprotective anti-inflammatory M-phenotype. Currently, there is no effective treatment for modulating such alterations. M1/M2 marker of primary microglia influenced by Melatonin were detected via qPCR. Functional activities were explored by western blotting, luciferase activity, EMSA, and ChIP assay. Structure interaction was assessed by molecular docking and LIGPLOT analysis. ER-stress detection was examined by ultrastructure TEM, calapin activity, and ERSE assay. The functional neurobehavioral evaluations were used for investigation of Melatonin on the neuroinflammation in vivo. Melatonin had targeted on Peroxisome Proliferator Activated Receptor Delta (PPARδ) activity, boosted LPS-stimulated alterations in polarization from the M1 to the M2 phenotype, and thereby inhibited NFκB-IKKβ activation in primary microglia. The PPARδ agonist L-165,041 or over-expression of PPARδ plasmid (ov-PPARδ) showed similar results. Molecular docking screening, dynamic simulation approaches, and biological studies of Melatonin showed that the activated site was located at PPARδ (phospho-Thr256-PPARδ). Activated microglia had lowered PPARδ activity as well as the downstream SIRT1 formation via enhancing ER-stress. Melatonin, PPARδ agonist and ov-PPARδ all effectively reversed the above-mentioned effects. Melatonin blocked ER-stress by regulating calapin activity and expression in LPS-activated microglia. Additionally, Melatonin or L-165,041 ameliorated the neurobehavioral deficits in LPS-aggravated neuroinflammatory mice through blocking microglia activities, and also promoted phenotype changes to M2-predominant microglia. Melatonin suppressed neuro-inflammation in vitro and in vivo by tuning microglial activation through the ER-stress-dependent PPARδ/SIRT1 signaling cascade. This treatment strategy is an encouraging pharmacological approach for the remedy of neuro-inflammation associated disorders.
Collapse
Affiliation(s)
- Hung-Chuan Pan
- Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Ph.D. Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- Doctoral Program in Biotechnology Industrial Management and Innovation, National Chung Hsing University, Taichung, Taiwan
- College of Medicine and Life Science, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Cheng-Ning Yang
- Department of Dentistry, School of Dentistry, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wen-Jane Lee
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Jason Sheehan
- Department of Neurosurgery, University of Virginia, Charlottesville, VA, USA
| | - Sheng-Mao Wu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Hong-Shiu Chen
- Department of Neurosurgery, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Mao-Hsun Lin
- Division of Neurology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi, Taiwan
| | - Li-Wei Shen
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Shu-Hua Lee
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Chin-Chang Shen
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Liang-Yi Pan
- School of Medicine, Kaohsiung Medical University, Taichung, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Meei-Ling Sheu
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan.
- Ph.D. Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan.
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan.
- Institute of Biomedical Sciences, College of Life Sciences, National Chung Hsing University, 250, Kuo Kuang Road, Taichung, 402, Taiwan.
| |
Collapse
|
16
|
Lan ZF, Yao W, Xie YC, Chen W, Zhu YY, Chen JQ, Zhou XY, Huang JQ, Wu MS, Chen JX. Oral Troxerutin Alleviates Depression Symptoms in Mice by Modulating Gut Microbiota and Microbial Metabolism. Mol Nutr Food Res 2024; 68:e2300603. [PMID: 38072646 DOI: 10.1002/mnfr.202300603] [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] [Indexed: 02/10/2024]
Abstract
SCOPE A growing body of evidence suggests that the harmful gut microbiota in depression patients can play a role in the progression of depression. There is limited research on troxerutin's impact on the central nervous system (CNS), especially in depression. The study finds that troxerutin effectively alleviates depression and anxiety-like behavior in mice by increasing the abundance of beneficial bacteria like Lactobacillus and Firmicutes while decreasing the abundance of harmful bacteria like Proteobacteria, Bacteroides, and Actinobacteria in the gut. Furthermore, the research reveals that troxerutin regulates various metabolic pathways in mice, including nucleotide metabolism, caffeine metabolism, purine metabolism, arginine biosynthesis, histidine metabolism, 2-oxocarboxylic acid metabolism, biosynthesis of amino acids, glycine, serine and threonine metabolism, and Arginine and proline metabolism. CONCLUSIONS In conclusion, the study provides compelling evidence for the antidepressant efficacy of troxerutin. Through the investigation of the role of intestinal microorganisms and metabolites, the study identifies these factors as key players in troxerutin's ability to prevent depression. Troxerutin achieves its neuroprotective effects and effectively prevents depression and anxiety by modulating the abundance of gut microbiota, including Proteobacteria, Bacteroides, and Actinobacteria, as well as regulating metabolites such as creatine.
Collapse
Affiliation(s)
- Zhi-Fang Lan
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Wei Yao
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Yi-Ci Xie
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Wushisi Chen
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Yin-Ying Zhu
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Jia-Qi Chen
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Xing-Yi Zhou
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Jun-Qing Huang
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Man-Si Wu
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
- School of Food and Biotechnology, Guangdong Industry Polytechnic, Guangzhou, 510300, China
| | - Jia-Xu Chen
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
17
|
Guo J, Fang M, Xiong Z, Zhou K, Zeng P. Mechanistic insights into the anti-depressant effect of curcumin based on network pharmacology and experimental validation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:583-598. [PMID: 37490124 DOI: 10.1007/s00210-023-02628-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 07/12/2023] [Indexed: 07/26/2023]
Abstract
Curcumin (CUR) exhibits a definite curative effect in the treatment of depression. To identify potential antidepressant targets and mechanisms of action of CUR. This study used network pharmacology to explore the signaling pathways and CUR-related targets in depression. C57BL/6 J mice (male,12-14 weeks old) were randomly divided into four groups (n = 8): saline-treated (control mice), lipopolysaccharide (LPS, 2 mg/kg/day, intraperitoneally), LPS + CUR (50 mg/kg/day, intragastrically), and LPS + CUR + LY294002 (7.5 mg/kg/day, intraperitoneally). After 1 week, behavioral tests were performed. Then, neuronal damage in the prefrontal cortex of mice was evaluated by hematoxylin-eosin (HE) staining. We uncovered the main active mechanism of CUR against depression using Western blotting and enzyme-linked immunosorbent assay (ELISA). Gene set enrichment analysis (GSEA) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways showed that the most significantly enriched pathway in CUR against depression was the PI3K-Akt pathway. Moreover, 52 targets were significantly correlated with the PI3K-Akt signaling pathway and CUR-related targets. In addition, among the top 50 targets ranked by degree in the protein-protein interaction (PPI) network, there were 23 targets involved in the 52 intersection targets. Administration of LPS alone extended immobility time in the open field test (OFT) and tail suspension test (TST) and decreased sucrose consumption in the sucrose preference test (SPT). Pretreatment with CUR relieved LPS-induced changes in the behavioral tests, activity of the PI3K-Akt signaling pathway, neuronal damage in the prefrontal cortex (PFC), and inflammatory response. Moreover, inhibition of the PI3K-Akt signaling pathway by LY294002 blocked the therapeutic effects of CUR. Our study indicates that CUR may be an effective antidepressant agent in an LPS-induced mouse model, partly because of its anti-inflammatory action through the PI3K-Akt signaling pathway.
Collapse
Affiliation(s)
- Jing Guo
- School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Meng Fang
- School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Zhe Xiong
- School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Ke Zhou
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical College, University of South China, Hengyang, 421001, China
| | - Peng Zeng
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical College, University of South China, Hengyang, 421001, China.
| |
Collapse
|
18
|
Tong T, Hao C, Shen J, Liu S, Yan S, Aslam MS, Chen Y, Chen W, Li J, Li Y, Zeng J, Li M, You Z, Gulizhaerkezi T, Wei S, Zhu A, Meng X. Electroacupuncture ameliorates chronic unpredictable mild stress-induced depression-like behavior and cognitive impairment through suppressing oxidative stress and neuroinflammation in rats. Brain Res Bull 2024; 206:110838. [PMID: 38123022 DOI: 10.1016/j.brainresbull.2023.110838] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 11/01/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Depression is associated with lowered mood, anxiety, anhedonia, cognitive impairments, and even suicidal tendencies in severe cases. Yet few studies have directed acupuncture's mechanism toward enhancing axonal repair correlated with synaptic plasticity and anti-inflammatory effects related to oxidative stress in the hippocampus. METHODS Male Sprague-Dawley (SD) rats were randomly divided into control group (CON), chronic unpredictable mild stress (CUMS) group, CUMS + electroacupuncture group (EA), and CUMS + fluoxetine group (FLX) (n = 10/group). Rats were given a 28-day treatment at the Shangxing (GV23) and Fengfu (GV16) acupoints with electroacupuncture or fluoxetine (2.1 mg/kg). RESULTS Rats exposed to CUMS induced depression-like behaviors and spatial learning-memory impairment, changed the ionized calcium binding adaptor molecule 1 (IBA-1), Vglut1, myelin basic protein (MBP), and postsynaptic density protein 95 (PSD95) level of hippocampal, increased the Nod-like receptor protein 3 (NLRP3), atypical squamous cell (ASC), Caspase level and hippocampal reactive oxygen species (ROS), and prompted the activation of Epha4-mediated signaling and an inflammatory response. Conversely, electroacupuncture administration reduced these changes and prevented depression-like behaviors and cognitive impairment. Electroacupuncture also promoted hippocampal expression of Sirtuin1(SIRT1), Nuclear factor erythroid 2-like (Nrf2), Heme oxygenase-1 (HO-1); reduced the expression of interleukin-1β (IL-1β), interleukin-18 (IL-18), and tumor necrosis factor-alpha (TNF-α); and prevented neural damage, particularly the synaptic myelin sheath, and neuroinflammation by regulating Eph receptor A4 (EphA4) in the hippocampal. CONCLUSION These results indicate that electroacupuncture prevents depression-like behaviors with cognitive impairment and synaptic and neuronal damage, probably by reducing EphA4, which mediates ROS hyperfunction and the inflammatory response.
Collapse
Affiliation(s)
- Tao Tong
- Department of Traditional Chinese Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Second Clinical College, Shanxi University of Traditional Chinese Medicine, Taiyuan, Shanxi, PR China.
| | - Chongyao Hao
- Second Clinical College, Shanxi University of Traditional Chinese Medicine, Taiyuan, Shanxi, PR China.
| | - Junliang Shen
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, PR China.
| | - Siyu Liu
- Longyan Hospital of Traditional Chinese Medicine of Xiamen University, Longyan, Fujian, PR China.
| | - Simin Yan
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, PR China.
| | | | - Yiping Chen
- Second Clinical College, Shanxi University of Traditional Chinese Medicine, Taiyuan, Shanxi, PR China.
| | - Wenjie Chen
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, PR China.
| | - Jianguo Li
- Second Clinical College, Shanxi University of Traditional Chinese Medicine, Taiyuan, Shanxi, PR China.
| | - Yuhan Li
- The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, PR China.
| | - Jingyu Zeng
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, PR China.
| | - Meng Li
- Second Clinical College, Shanxi University of Traditional Chinese Medicine, Taiyuan, Shanxi, PR China.
| | - Zhuoran You
- Second Clinical College, Shanxi University of Traditional Chinese Medicine, Taiyuan, Shanxi, PR China.
| | - Tuergong Gulizhaerkezi
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, PR China.
| | - Simiao Wei
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, PR China.
| | - Anning Zhu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xianjun Meng
- Department of Traditional Chinese Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, PR China.
| |
Collapse
|
19
|
Qiu F, Zeng C, Liu Y, Pan H, Ke C. J147 ameliorates sepsis-induced depressive-like behaviors in mice by attenuating neuroinflammation through regulating the TLR4/NF-κB signaling pathway. J Mol Histol 2023; 54:725-738. [PMID: 37676534 PMCID: PMC10635911 DOI: 10.1007/s10735-023-10147-4] [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: 04/28/2023] [Accepted: 08/18/2023] [Indexed: 09/08/2023]
Abstract
Neuroinflammation is associated with the pathophysiology of depression. The molecular mechanism of depressive-like behavior caused by sepsis-associated encephalopathy (SAE) is incompletely understood. J147 (an analog of curcumin) has been reported to improve memory and has neuroprotective activity, but its biological function in the depressive-like behavior observed in SAE is not known. We investigated the effects of J147 on lipopolysaccharide (LPS)-induced neuroinflammatory, depressive-like behaviors, and the toll-like receptor 4 (TLR4)/nuclear factor-κB (NF-κB) signal pathway in the mouse hippocampus and microglia (BV2 cells). The forced-swimming test (FST) and tail-suspension test (TST) were undertaken for assessment of depressive-like behaviors. Expression of the proinflammatory genes interleukin (IL)-6, IL-1β, and tumor necrosis factor (TNF)-α were measured using RT-qPCR and ELISA. Microglia activation was detected using immunofluorescence staining. The TLR4/NF-κB signaling pathway was studied using western blotting and immunofluorescence staining. J147 pretreatment markedly downregulated expression of IL-6, IL-1β, and TNF-α, and the mean fluorescence intensity of ionized calcium-binding adapter protein-1 in microglia. J147 restrained LPS-induced nuclear translocation of nuclear factor-kappa B (NF-κB), inhibitor of nuclear factor kappa B (IκB) degradation, and TLR4 activation in microglia. J147 administration inhibited bodyweight loss, mortality, microglia activation, and depressive-like behaviors in LPS-treated mice. In conclusion, J147 ameliorated the sepsis-induced depressive-like behaviors induced by neuroinflammation through attenuating the TLR4/NF-κB signaling pathway in microglia.
Collapse
Affiliation(s)
- Fang Qiu
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, Guangdong, China
- Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China
| | - Changchun Zeng
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong, China
| | - Yuqiang Liu
- Department of Anesthesiology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518025, Guangdong, China.
| | - Haobo Pan
- Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China.
| | - Changneng Ke
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, Guangdong, China.
| |
Collapse
|
20
|
Li W, Ali T, Mou S, Gong Q, Li N, Hao L, Yu ZJ, Li S. D1R-5-HT2AR Uncoupling Reduces Depressive Behaviours via HDAC Signalling. Neurotherapeutics 2023; 20:1875-1892. [PMID: 37782408 PMCID: PMC10684469 DOI: 10.1007/s13311-023-01436-7] [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] [Accepted: 09/03/2023] [Indexed: 10/03/2023] Open
Abstract
Dopamine and serotonin signalling are associated with major depressive disorder, which is a prevalent life-threatening illness worldwide. Numerous FDA-approved dopamine/serotonin signalling-modifying drugs are available but are associated with concurrent side effects and limited efficacy. Thus, identifying and targeting their signalling pathway is crucial for improving depression treatment. Here, we determined that serotonin receptor 2A (5-HT2AR) abundantly forms a protein complex with dopamine receptor 1 (D1R) in high abundance via its carboxy-terminus in the brains of mice subjected to various chronic stress paradigms. Furthermore, the D1R/5-HT2AR interaction elicited CREB/ERK/AKT modulation during synaptic regulation. An interfering peptide (TAT-5-HT2AR-SV) agitated the D1R/5-HT2AR interaction and attenuated depressive symptoms accompanied by CREB/ERK molecule costimulation. Interestingly, HDAC antagonism but not TrkB antagonism reversed the antidepressant effect of competitive peptides. These findings revealed a novel D1R/5-HT2AR heteroreceptor complex mechanism in the pathophysiology of depression, and their uncoupling ameliorates depressive-like behaviours through HDAC-, and not BDNF-, dependent mechanisms.
Collapse
Affiliation(s)
- Weifen Li
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, the 6th Affiliated Hospital of Shenzhen University Health Science Center, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
| | - Tahir Ali
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Shengnan Mou
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Qichao Gong
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Ningning Li
- Department of Neurology, School of Medicine, Affiliated ZhongDa Hospital, Southeast University, Nanjing, China
- Precision Medicine Research Centre, Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Liangliang Hao
- Hospital of Chengdu, University of Traditional Chinese Medicine, No.39 Shi-er-qiao Road, Chengdu, People's Republic of China
| | - Zhi-Jian Yu
- Department of Infectious Diseases and Shenzhen Key Laboratory for Endogenous Infections, the 6th Affiliated Hospital of Shenzhen University Health Science Center, No 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China.
| | - Shupeng Li
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
- Campbell Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
21
|
Sethi P, Mehan S, Khan Z, Chhabra S. Acetyl-11-keto-beta boswellic acid(AKBA) modulates CSTC-pathway by activating SIRT-1/Nrf2-HO-1 signalling in experimental rat model of obsessive-compulsive disorder: Evidenced by CSF, blood plasma and histopathological alterations. Neurotoxicology 2023; 98:61-85. [PMID: 37549874 DOI: 10.1016/j.neuro.2023.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 07/23/2023] [Accepted: 08/03/2023] [Indexed: 08/09/2023]
Abstract
Obsessive-Compulsive disorder (OCD) is a long-term and persistent mental illness characterised by obsessive thoughts and compulsive behaviours. Numerous factors can contribute to the development or progression of OCD. These factors may result from the dysregulation of multiple intrinsic cellular pathways, including SIRT-1, Nrf2, and HO-1. Inhibitors of selective serotonin reuptake (SSRIs) are effective first-line treatments for OCD. In our ongoing research, we have investigated the role of SIRT-1, Nrf2, and HO-1, as well as the neuroprotective potential of Acetyl-11-keto-beta boswellic acid (AKBA) against behavioural and neurochemical changes in rodents treated with 8-OH-DPAT. In addition, the effects of AKBA were compared to those of fluvoxamine (FLX), a standard OCD medication. Injections of 8-OH-DPAT into the intra-dorso raphe nuclei (IDRN) of rats for seven days induced repetitive and compulsive behaviour accompanied by elevated oxidative stress, inflammatory processes, apoptosis, and neurotransmitter imbalances in CSF, blood plasma, and brain samples. Chronic administration of AKBA at 50 mg/kg and 100 mg/kg p.o. restored histopathological alterations in the cortico-striatal-thalamo-cortical (CSTC) pathway, including the cerebral cortex, striatum, and hippocampal regions. Our investigation revealed that when AKBA and fluvoxamine were administered together, the alterations were restored to a greater degree than when administered separately. These findings demonstrate that the neuroprotective effect of AKBA can serve as an effective basis for developing a novel OCD treatment.
Collapse
Affiliation(s)
- Pranshul Sethi
- Division of Neuroscience, Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Division of Neuroscience, Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India.
| | - Zuber Khan
- Division of Neuroscience, Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Swesha Chhabra
- Division of Neuroscience, Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
22
|
Sani G, Margoni S, Brugnami A, Ferrara OM, Bernardi E, Simonetti A, Monti L, Mazza M, Janiri D, Moccia L, Kotzalidis GD, Chieffo DPR, Janiri L. The Nrf2 Pathway in Depressive Disorders: A Systematic Review of Animal and Human Studies. Antioxidants (Basel) 2023; 12:817. [PMID: 37107192 PMCID: PMC10135298 DOI: 10.3390/antiox12040817] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/12/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
There is increasing interest in the involvement of antioxidative systems in protecting from depression. Among these, Nrf2 occupies a central place. We aimed to review the role of Nrf2 in depression. For this reason, we conducted a PubMed search using as search strategy (psychiatr*[ti] OR schizo*[ti] OR psychot*[ti] OR psychos*[ti] OR depress*[ti] OR MDD[ti] OR BD[ti] OR bipolar[ti] OR Anxiety[ti] OR antidepress*[ti] OR panic[ti] OR obsess*[ti] OR compulsio*[ti] OR "mood disord*"[ti] OR phobi*[ti] OR agoraphob*[ti] OR anorex*[ti] OR anorect*[ti] OR bulimi*[ti] OR "eating disorder*"[ti] OR neurodevelopm*[ti] OR retardation[ti] OR autism[ti] OR autistic[ti] OR ASM[ti] OR adhd[ti] OR "attention-deficit"[ti]) AND nrf2, which on the 9th of March produced 208 results of which 89 were eligible for our purposes. Eligible articles were studies reporting data of Nrf2 manipulations or content by any treatment in human patients or animals with any animal model of depression. Most studies were on mice only (N = 58), 20 on rats only, and three on both rats and mice. There were two studies on cell lines (in vitro) and one each on nematodes and fish. Only four studies were conducted in humans, one of which was post mortem. Most studies were conducted on male animals; however, human studies were carried out on both men and women. The results indicate that Nrf2 is lower in depression and that antidepressant methods (drugs or other methods) increase it. Antioxidant systems and plasticity-promoting molecules, such as those in the Nrf2-HO-1, BDNF-TrkB, and cyclic AMP-CREB pathways, could protect from depression, while glycogen synthase kinase-3β and nuclear factor κB oppose these actions, thus increasing depressive-like behaviours. Since Nrf2 is also endowed with tumorigenic and atherogenic potential, the balance between benefits and harms must be taken into account in designing novel drugs aiming at increasing the intracellular content of Nrf2.
Collapse
Affiliation(s)
- Gabriele Sani
- Institute of Psychiatry, Department of Neuroscience, Catholic University of the Sacred Hearth, Rome, Largo Francesco Vito 1, 00168 Rome, Italy
- Department of Psychiatry, Department of Neuroscience, Head, Neck and Thorax, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 1, 00168 Rome, Italy
| | - Stella Margoni
- Institute of Psychiatry, Department of Neuroscience, Catholic University of the Sacred Hearth, Rome, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Andrea Brugnami
- Institute of Psychiatry, Department of Neuroscience, Catholic University of the Sacred Hearth, Rome, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Ottavia Marianna Ferrara
- Institute of Psychiatry, Department of Neuroscience, Catholic University of the Sacred Hearth, Rome, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Evelina Bernardi
- Institute of Psychiatry, Department of Neuroscience, Catholic University of the Sacred Hearth, Rome, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Alessio Simonetti
- Institute of Psychiatry, Department of Neuroscience, Catholic University of the Sacred Hearth, Rome, Largo Francesco Vito 1, 00168 Rome, Italy
- Department of Psychiatry, Department of Neuroscience, Head, Neck and Thorax, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 1, 00168 Rome, Italy
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
- Centro Lucio Bini, Via Crescenzio 42, 00193 Rome, Italy
| | - Laura Monti
- UOS Clinical Psychology, Clinical Government, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 1, 00168 Rome, Italy
| | - Marianna Mazza
- Institute of Psychiatry, Department of Neuroscience, Catholic University of the Sacred Hearth, Rome, Largo Francesco Vito 1, 00168 Rome, Italy
- Department of Psychiatry, Department of Neuroscience, Head, Neck and Thorax, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 1, 00168 Rome, Italy
| | - Delfina Janiri
- Institute of Psychiatry, Department of Neuroscience, Catholic University of the Sacred Hearth, Rome, Largo Francesco Vito 1, 00168 Rome, Italy
- Department of Psychiatry, Department of Neuroscience, Head, Neck and Thorax, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 1, 00168 Rome, Italy
| | - Lorenzo Moccia
- Institute of Psychiatry, Department of Neuroscience, Catholic University of the Sacred Hearth, Rome, Largo Francesco Vito 1, 00168 Rome, Italy
- Department of Psychiatry, Department of Neuroscience, Head, Neck and Thorax, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 1, 00168 Rome, Italy
| | - Georgios D. Kotzalidis
- Institute of Psychiatry, Department of Neuroscience, Catholic University of the Sacred Hearth, Rome, Largo Francesco Vito 1, 00168 Rome, Italy
- NESMOS Department, Faculty of Medicine and Psychology, Sant’Andrea University Hospital, University of Rome La Sapienza, Via di Grottarossa, 1035-1039, 00189 Rome, Italy
| | - Daniela Pia Rosaria Chieffo
- UOS Clinical Psychology, Clinical Government, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 1, 00168 Rome, Italy
| | - Luigi Janiri
- Institute of Psychiatry, Department of Neuroscience, Catholic University of the Sacred Hearth, Rome, Largo Francesco Vito 1, 00168 Rome, Italy
- Department of Psychiatry, Department of Neuroscience, Head, Neck and Thorax, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 1, 00168 Rome, Italy
| |
Collapse
|
23
|
Zhang W, Wang X, Tang Y, Huang C. Melatonin alleviates doxorubicin-induced cardiotoxicity via inhibiting oxidative stress, pyroptosis and apoptosis by activating Sirt1/Nrf2 pathway. Biomed Pharmacother 2023; 162:114591. [PMID: 36965257 DOI: 10.1016/j.biopha.2023.114591] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/13/2023] [Accepted: 03/22/2023] [Indexed: 03/27/2023] Open
Abstract
Melatonin confers cardioprotective effects on multiple cardiovascular diseases, including doxorubicin-induced cardiomyopathy. The effectiveness of melatonin in mitigating myocardial injuries caused by Doxorubicin through enhancement of mitochondrial function is already established, however, the role of melatonin in regulating the Sirtuin-1 (Sirt1)/Nuclear factor E2-associated factor 2 (Nrf2) pathway in lessening the onset of Doxorubicin-induced cardiomyopathy is yet to be elucidated. To address this, H9C2 cardiomyocytes and C57BL/6 mice were employed to construct in vitro and in vivo models of Dox-induced myocardial impairments, respectively. Results showed that Dox markedly evoked oxidative stress, pyroptosis and apoptosis both in vitro and in vivo, which were significantly alleviated by melatonin administration. Mechanistically, melatonin attenuated Dox-induced downregulation of Sirt1 and Nrf2, and both inhibition of Sirt1 and Nrf2 significantly reversed the cardioprotective effects of melatonin. In conclusion, our studies suggest that the activation of the Sirt1/Nrf2 pathway is the underlying mechanism behind melatonin's ability to curtail oxidative stress, pyroptosis, and apoptosis in Dox-induced cardiomyopathy. These promising results demonstrated the potential application of melatonin as a treatment for doxorubicin-induced cardiac injury.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China.
| |
Collapse
|
24
|
Li A, Liu Z, Ali T, Gao R, Luo Y, Gong Q, Zheng C, Li W, Guo H, Liu X, Li S, Li T. Roxadustat (FG-4592) abated lipopolysaccharides-induced depressive-like symptoms via PI3K signaling. Front Mol Neurosci 2023; 16:1048985. [PMID: 37008780 PMCID: PMC10056220 DOI: 10.3389/fnmol.2023.1048985] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 02/07/2023] [Indexed: 03/17/2023] Open
Abstract
BackgroundDespite its role in inflammation and the redox system under hypoxia, the effects and molecular mechanisms of hypoxia-inducible factor (HIF) in neuroinflammation-associated depression are poorly explored. Furthermore, Prolyl hydroxylase domain-containing proteins (PHDs) regulate HIF-1; however, whether and how PHDs regulate depressive-like behaviors under Lipopolysaccharides (LPS)-induced stress conditions remain covered.MethodsTo highlight the roles and underlying mechanisms of PHDs-HIF-1 in depression, we employed behavioral, pharmacological, and biochemical analyses using the LPS-induced depression model.ResultsLipopolysaccharides treatment induced depressive-like behaviors, as we found, increased immobility and decreased sucrose preference in the mice. Concurrently, we examined increased cytokine levels, HIF-1 expression, mRNA levels of PHD1/PHD2, and neuroinflammation upon LPS administration, which Roxadustat reduced. Furthermore, the PI3K inhibitor wortmannin reversed Roxadustat-induced changes. Additionally, Roxadustat treatment attenuated LPS-induced synaptic impairment and improved spine numbers, ameliorated by wortmannin.ConclusionLipopolysaccharides-dysregulates HIF-PHDs signaling may contribute to neuroinflammation-coincides depression via PI3K signaling.
Collapse
Affiliation(s)
- Axiang Li
- College of Forensic Medicine, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
- Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Zizhen Liu
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Tahir Ali
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
- Shenzhen Bay Laboratory, Institute of Chemical Biology, Shenzhen, China
| | - Ruyan Gao
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Yanhua Luo
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Qichao Gong
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Chenyou Zheng
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Weifen Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Hongling Guo
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Xinshe Liu
- College of Forensic Medicine, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
- Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- NHC Key Laboratory of Forensic Science, College of Forensic Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Shupeng Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
- Shenzhen Bay Laboratory, Institute of Chemical Biology, Shenzhen, China
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- *Correspondence: Shupeng Li, ; Tao Li,
| | - Tao Li
- College of Forensic Medicine, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
- Institute of Forensic Injury, Institute of Forensic Bio-Evidence, Western China Science and Technology Innovation Harbor, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- NHC Key Laboratory of Forensic Science, College of Forensic Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- *Correspondence: Shupeng Li, ; Tao Li,
| |
Collapse
|
25
|
Exogenous melatonin alleviates neuropathic pain-induced affective disorders by suppressing NF-κB/ NLRP3 pathway and apoptosis. Sci Rep 2023; 13:2111. [PMID: 36747075 PMCID: PMC9902529 DOI: 10.1038/s41598-023-28418-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 01/18/2023] [Indexed: 02/08/2023] Open
Abstract
In this study, we aimed to evaluate the anti-inflammatory and anti-apoptotic effects of melatonin (MLT) on neuropathic pain (NP)-induced anxiety and depression in a rat model. Adult male rats were separated into four groups, i.e., Sham-VEH: healthy animals received a vehicle, Sham-MLT (10 mg/kg), and chronic constrictive injury (CCI)-VEH: nerve ligation received the vehicle, and CCI-MLT. Next, we used behavioral tests to evaluate pain severity, anxiety, and depression. Finally, rats were sacrificed for molecular and histopathological studies. Behavioral tests showed that NP could induce depressive- and anxiety-like behaviors. NP activated NF-κB/NLRP3 inflammasome pathways by upregulating NF-κB, NLRP3, ASC, active Caspase-1, also enhancing the concentrations of cytokines (IL-1β and IL-18) in the prefrontal cortex (PFC) and hippocampus (HC). NP upregulated Bax, downregulated Bcl2, and increased cell apoptosis in the HC and PFC. The rats treated with MLT eliminated the effects of NP, as the reduced pain severity, improved anxiety- and depressive-like behaviors, ameliorated NF-κB/NLRP3 inflammasome pathways, and modulated levels of cytokines in the HC and PFC. MLT could promote cell survival from apoptosis by modulating Bax and Bcl2. Therefore, it might be inferred that its anti-inflammatory and anti-apoptotic properties mediate the beneficial effects of MLT in NP-induced affective disorders.
Collapse
|
26
|
Luo X, Xiao P, Li L, Duan Y, Sk CA, Xie J. Research progress in circadian rhythms in the application of psychological rehabilitation of cancer patients. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2022; 47:1740-1747. [PMID: 36748386 PMCID: PMC10930277 DOI: 10.11817/j.issn.1672-7347.2022.220245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Indexed: 02/08/2023]
Abstract
The psychological distress of cancer patients seriously affects their therapeutic effects. Effective psychological rehabilitation of cancer patients significantly improves their survival chance and quality of life. Circadian rhythm results from adaptation to the environment during the organism's evolution. When the endogenous clock system is disrupted or the external environment is changed, the body and the environment are out of synchronization, and the circadian rhythm will be disrupted. Circadian rhythm disorder is a common phenomenon in cancer patients, and the changes of circadian rhythm are closely related to their psychological distress. Many studies believe that the circadian rhythm disorder of cancer patients may directly or indirectly affect their psychology through various mechanisms, and targeted intervention by regulating the circadian rhythm of patients may be an essential means to promote the psychological rehabilitation.
Collapse
Affiliation(s)
- Xiaofei Luo
- Department of Nursing, Third Xiangya Hospital, Central South University, Changsha 410013.
- Xiangya Nursing School, Central South University, Changsha 410013.
| | - Panpan Xiao
- Xiangya Nursing School, Central South University, Changsha 410013
| | - Lijun Li
- Xiangya Nursing School, Central South University, Changsha 410013
| | - Yinglong Duan
- Department of Nursing, Third Xiangya Hospital, Central South University, Changsha 410013
| | - Cheng Andy Sk
- Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Jianfei Xie
- Department of Nursing, Third Xiangya Hospital, Central South University, Changsha 410013.
| |
Collapse
|
27
|
Sobhani S, Tehrani AA, Sobhani G, Fatima S, Ulloa L, Motaghinejad M, Atif A. Melatonin Protects Against Titanium Oxide-Induced Neurotoxicity: Neurochemical, Neurobehavioral, and Histopathological Evidences. Biol Trace Elem Res 2022:10.1007/s12011-022-03464-4. [PMID: 36378265 DOI: 10.1007/s12011-022-03464-4] [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: 04/01/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022]
Abstract
titania (titanium dioxide, TiO2) is known to induce neurotoxicity and CNS dysfunctions. Numerous studies have explored the neuroprotective effects of melatonin against neurotoxicity. This study evaluates the potential of melatonin to protect against titania-induced neurotoxicity and the role of the Keap1/Nrf2/ARE signaling pathway. One group of animals were treated with Titania (0.045 and 0.075 g/rat) alone while the other with added melatonin (1 mg/kg and 3 mg/kg) and behavioral alterations were assessed using OFT (open field test). Neurochemical and histopathological changes were also studied in the hippocampus by analyzing kelch ECH associating protein 1 (Keap1), nuclear factor erythroid 2-related factor 2 (Nrf2), and antioxidant response element (ARE). It was seen that the animals with added Melatonin had improved behavioral scores in the OFT, like anxiety and motor dysfunction triggered by TiO2. Melatonin also reduced lipid peroxidation, ROS, GSSG, IL1β, TNFα, Bax, and Keap1 levels, but boosted GSH, GPx, GR, SOD,IL10,IL4, Bcl2, Nrf2, and ARE levels and improved quadruple mitochondrial enzyme complex activity in titania-treated animals. Histopathological examination showed melatonin induced cytoprotection against vacuolization and necrosis in granular cells of DG and pyramidal cells of CA1 area of the hippocampus. In our study, pretreatment with melatonin reduced titania-induced neurotoxicity in the hippocampus through a mechanism potentially mediated by the Keap-1/Nrf2/ARE pathway.
Collapse
Affiliation(s)
- Sarvenaz Sobhani
- Department of Pathobiology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Ali-Asghar Tehrani
- Department of Pathobiology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran.
| | - Golnar Sobhani
- Department of Pathobiology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Sulail Fatima
- Department of Physiology, Jinnah Medical & Dental College, Sohail University, Karachi, Pakistan
| | - Luis Ulloa
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University, Durham, NC, USA
| | - Majid Motaghinejad
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Masih Daneshvari Hospital, Darabad Avenue, Shahid Bahonar roundabout, Tehran, Iran.
| | - Alina Atif
- Department of Physiology, Jinnah Medical & Dental College, Sohail University, Karachi, Pakistan
| |
Collapse
|
28
|
Bian H, Xiao L, Liang L, Xie Y, Wang H, Slevin M, Tu WJ, Wang G. Polydatin Prevents Neuroinflammation and Relieves Depression via Regulating Sirt1/HMGB1/NF-κB Signaling in Mice. Neurotox Res 2022; 40:1393-1404. [PMID: 35986876 DOI: 10.1007/s12640-022-00553-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/22/2022] [Accepted: 07/29/2022] [Indexed: 12/21/2022]
Abstract
Depression is a prevalent psychiatric disorder with a significant health impact and economic burden worldwide. Unfortunately, the exact pathogenesis of depression is not well understood. Neuroinflammation and microglial activation play an essential role in the pathogenesis of depression. Previous studies have shown that polydatin has anti-inflammatory and antioxidant properties. However, the link between polydatin and depression remains unclear. Therefore, the objective of this study was to investigate the antidepressant effect of polydatin in lipopolysaccharide (LPS)-induced depression in mice and its possible mechanism. Adult male C57BL/6 J mice were used in this study. The polydatin and LPS were injected intraperitoneally daily for 5 days. In addition, the EX527, an inhibitor of Sirt1, was injected intraperitoneally daily and 1 h before the polydatin injection. The behavior tests were performed to elucidate the depression-like behaviors. The Sirt1/HMGB1/NF-κB pathway expression was detected by western blot, ELISA, and immunofluorescence staining. Polydatin can significantly improve LPS-induced depression-like behavior in mice. Treatment with polydatin increased the expression of the Sirt1 but decreased the expression of the HMGB1, p-NF-κB, IL-1b, and TNF-α in the LPS-induced depression mice. In addition, the EX527 abolished the anti-depressive effects of the polydatin and the levels of Sirt1 protein. These findings suggested that the polydatin reversed the depressive effects through the Sirt1/HMGB1/NF-κB signaling in the LPS-induced depression mice. Therefore, polydatin can be used in the treatment of depression.
Collapse
Affiliation(s)
- Hetao Bian
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, 430060, Hubei, People's Republic of China
| | - Ling Xiao
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, 430060, Hubei, People's Republic of China
| | - Liang Liang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, 430060, Hubei, People's Republic of China
| | - Yinping Xie
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, 430060, Hubei, People's Republic of China
| | - Huiling Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, 430060, Hubei, People's Republic of China
| | - Mark Slevin
- School of Healthcare Science, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, M156GX, UK.
| | - Wen-Jun Tu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 238, Baiti Road, Tianjin, 300192, People's Republic of China.
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China.
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, 430060, Hubei, People's Republic of China.
| |
Collapse
|
29
|
Masanetz RK, Winkler J, Winner B, Günther C, Süß P. The Gut-Immune-Brain Axis: An Important Route for Neuropsychiatric Morbidity in Inflammatory Bowel Disease. Int J Mol Sci 2022; 23:11111. [PMID: 36232412 PMCID: PMC9570400 DOI: 10.3390/ijms231911111] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Inflammatory bowel disease (IBD) comprises Crohn's disease (CD) and ulcerative colitis (UC) and is associated with neuropsychiatric symptoms like anxiety and depression. Both conditions strongly worsen IBD disease burden. In the present review, we summarize the current understanding of the pathogenesis of depression and anxiety in IBD. We present a stepwise cascade along a gut-immune-brain axis initiated by evasion of chronic intestinal inflammation to pass the epithelial and vascular barrier in the gut and cause systemic inflammation. We then summarize different anatomical transmission routes of gut-derived peripheral inflammation into the central nervous system (CNS) and highlight the current knowledge on neuroinflammatory changes in the CNS of preclinical IBD mouse models with a focus on microglia, the brain-resident macrophages. Subsequently, we discuss how neuroinflammation in IBD can alter neuronal circuitry to trigger symptoms like depression and anxiety. Finally, the role of intestinal microbiota in the gut-immune-brain axis in IBD will be reviewed. A more comprehensive understanding of the interaction between the gastrointestinal tract, the immune system and the CNS accounting for the similarities and differences between UC and CD will pave the path for improved prediction and treatment of neuropsychiatric comorbidities in IBD and other inflammatory diseases.
Collapse
Affiliation(s)
- Rebecca Katharina Masanetz
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Jürgen Winkler
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Beate Winner
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Center of Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Claudia Günther
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Department of Internal Medicine 1, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Patrick Süß
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| |
Collapse
|
30
|
Sundram S, Malviya R, Awasthi R. Genetic Causes of Alzheimer's Disease and the Neuroprotective Role of Melatonin in its Management. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 22:CNSNDDT-EPUB-126085. [PMID: 36056839 DOI: 10.2174/1871527321666220901125730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/15/2022] [Accepted: 07/16/2022] [Indexed: 06/15/2023]
Abstract
Dementia is a global health concern owing to its complexity, which also poses a great challenge to pharmaceutical scientists and neuroscientists. The global dementia prevalence is approximately 47 million, which may increase by three times by 2050. Alzheimer's disease (AD) is the most common cause of dementia. AD is a severe age-related neurodegenerative disorder characterized by short-term memory loss, aphasia, mood imbalance, and executive function. The etiology of AD is still unknown, and the exact origin of the disease is still under investigation. Aggregation of Amyloid β (Aβ) plaques or neurotoxic Aβo oligomers outside the neuron is the most common cause of AD development. Amyloid precursor protein (APP) processing by β secretase and γ secretase produces abnormal Aβ monomers. This aggregation of Aβ and NFT is promoted by various genes like BACE1, ADAM10, PIN1, GSK-3, APOE, PPARα, etc. Identification of these genes can discover several therapeutic targets that can be useful in studying pathogenesis and underlying treatments. Melatonin modulates the activities of these genes, thereby reducing Aβ production and increasing its clearance. Melatonin also reduces the expression of APP by attenuating cAMP, thereby enhancing the non-amyloidogenic process. Present communication explored and discussed the neuroprotective role of melatonin against Aβ-dependent AD pathogenesis. The manuscript also discussed potential molecular and genetic mechanisms of melatonin in the production and clearance of Aβ that could ameliorate neurotoxicity.
Collapse
Affiliation(s)
- Sonali Sundram
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, U.P., India
- Amity Institute of Pharmacy, Amity University Uttar Pradesh, Sector-125, Noida 201313, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, U.P., India
| | - Rajendra Awasthi
- Department of Pharmaceutical Sciences, School of Health Science and Technology, University of Petroleum and Energy Studies (UPES), Energy Acres, Bidholi, Via-Prem Nagar, Dehradun - 248 007, Uttarakhand, India
| |
Collapse
|
31
|
Kholghi G, Eskandari M, Shokouhi Qare Saadlou MS, Zarrindast MR, Vaseghi S. Night shift hormone: How does melatonin affect depression? Physiol Behav 2022; 252:113835. [PMID: 35504318 DOI: 10.1016/j.physbeh.2022.113835] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/19/2022] [Accepted: 04/28/2022] [Indexed: 12/23/2022]
Abstract
Melatonin is the main hormone secreted by the pineal gland that modulates the circadian rhythm and mood. Previous studies have shown the therapeutic effects of melatonin, or its important analogue, agomelatine, on depression. In this review study, we aimed to discuss the potential mechanisms of melatonin involved in the treatment of depression. It was noted that disrupted circadian rhythm can lead to depressive state, and melatonin via regulating circadian rhythm shows a therapeutic effect. It was also noted that melatonin induces antidepressant effects via promoting antioxidant system and neurogenesis, and suppressing oxidative stress, neuroinflammation, and apoptosis. The interaction effect between melatonin or agomelatine and serotonergic signaling has a significant effect on depression. It was noted that the psychotropic effects of agomelatine are induced by the synergistic interaction between melatonin and 5-HT2C receptors. Agomelatine also interacts with glutamatergic signaling in brain regions involved in regulating mood and circadian rhythm. Interestingly, it was concluded that melatonin exerts both pro- and anti-inflammatory effects, depending on the grade of inflammation. It was suggested that synergistic interaction between melatonin and 5-HT2C receptors may be able to induce therapeutic effects on other psychiatric disorders. Furthermore, dualistic role of melatonin in regulating inflammation is an important point that can be examined at different levels of inflammation in animal models of depression.
Collapse
Affiliation(s)
- Gita Kholghi
- Department of Psychology, Faculty of Human Sciences, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Maliheh Eskandari
- Faculty of Basic Sciences, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | | | - Mohammad-Reza Zarrindast
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Salar Vaseghi
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran.
| |
Collapse
|
32
|
Yan C, Mao J, Yao C, Liu Y, Yan H, Jin W. Neuroprotective effects of mild hypothermia against traumatic brain injury by the involvement of the Nrf2/ARE pathway. Brain Behav 2022; 12:e2686. [PMID: 35803901 PMCID: PMC9392531 DOI: 10.1002/brb3.2686] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/01/2022] [Accepted: 06/14/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is the leading cause of death and disability worldwide. Mild hypothermia (32-35°C) has been found to show neuroprotective effects against TBI. However, the specific mechanism is still elusive. In the current study, we explored the relationship between oxidative damage after TBI and treatment with mild hypothermia as well as the underlying molecular mechanisms. METHODS We used the closed cortex injury model to perform the brain injury and a temperature monitoring and control system to regulate the body temperature of mice after injury. Adult male C57BL/6 mice were adopted in this study and divided into four experimental groups. Tissue samples were harvested 24 h after injury. RESULTS First, our results showed that treatment with mild hypothermia significantly improved neurobehavioral dysfunction and alleviated brain edema after TBI. Moreover, treatment with mild hypothermia enhanced the activity of the antioxidant enzymes superoxide dismutase and glutathione peroxidase and reduced the accumulation of lipid peroxidation malondialdehyde. Importantly, the expression and activation of the nuclear factor erythroid 2-related factor 2-antioxidant response element (Nrf2-ARE) pathway were upregulated by mild hypothermia after TBI. Finally, treatment with hypothermia significantly decreased the cell apoptosis induced by TBI. CONCLUSION Our results showed that the protective effects of mild hypothermia after TBI may be achieved by the upregulation of the Nrf2-ARE pathway and revealed Nrf2 as a potentially important target to improve the prognosis of TBI.
Collapse
Affiliation(s)
- Chaolong Yan
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China.,Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.,Department of Neurosurgery, Zhongshan Hospital, The Affiliated Hospital of Fudan University, Shanghai, China
| | - Jiannan Mao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Chenbei Yao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yang Liu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Huiying Yan
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Wei Jin
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China.,Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
33
|
Jia S, Guo X, Chen Z, Li S, Liu XA. The roles of the circadian hormone melatonin in drug addiction. Pharmacol Res 2022; 183:106371. [PMID: 35907435 DOI: 10.1016/j.phrs.2022.106371] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/19/2022] [Accepted: 07/26/2022] [Indexed: 10/16/2022]
Abstract
Given the devastating social and health consequences of drug addiction and the limitations of current treatments, a new strategy is needed. Circadian system disruptions are frequently associated with drug addiction. Correcting abnormal circadian rhythms and improving sleep quality may thus be beneficial in the treatment of patients with drug addiction. Melatonin, an essential circadian hormone that modulates the biological clock, has anti-inflammatory, analgesic, anti-depressive, and neuroprotective effects via gut microbiota regulation and epigenetic modifications. It has attracted scientists' attention as a potential solution to drug abuse. This review summarized scientific evidence on the roles of melatonin in substance use disorders at the cellular, circuitry, and system levels, and discussed its potential applications as an intervention strategy for drug addiction.
Collapse
Affiliation(s)
- Shuhui Jia
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China; University of Chinese Academy of Sciences, Beijing, China
| | - Xuantong Guo
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China; University of Chinese Academy of Sciences, Beijing, China
| | - Zuxin Chen
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China; University of Chinese Academy of Sciences, Beijing, China.
| | - Shupeng Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Xin-An Liu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
34
|
Xie LL, Rui C, Li ZZ, Li SS, Fan YJ, Qi MM. Melatonin mitigates traumatic brain injury-induced depression-like behaviors through HO-1/CREB signal in rats. Neurosci Lett 2022; 784:136754. [PMID: 35753614 DOI: 10.1016/j.neulet.2022.136754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 06/14/2022] [Accepted: 06/21/2022] [Indexed: 02/01/2023]
Abstract
In addition to significant antioxidant properties, melatonin exhibits neuroprotective effects against various neurological diseases including traumatic brain injury (TBI) and ischemic stroke. Several potential mechanisms have been reported in the neuroprotection of melatonin among patients with TBI. Notably, the heme oxygenase-1 (HO-1)/cAMP response element-binding protein (CREB) signaling pathway is implicated in the development of a depressive state. Moreover, the activity of CREB in the nucleus accumbens (NAc) participates in reward and motivation, further contributing to depression induced by TBI. This study aims to explore whether melatonin could mitigate TBI-induced depression by activating of HO-1/CREB signal in a rodent model of weight-drop. As a consequence, melatonin (10 mg/kg) attenuated TBI-induced elevated immobility time in the force swim test, decreased time spent sniffing the novel rat in 3-chambered social test, and downregulated phosphorylated CERB in the NAc. However, a special inhibitor of HO-1 (SnPP) via intracerebroventricular injection partially reversed the neuroprotective effects of melatonin. Furthermore, melatonin decreased the number of summarized intersects in the astrocyte, A1-type astrocytes, IL-6-positive astrocytes in the NAc after TBI exposure, nevertheless, these changes could partially be restored by SnPP. Therefore, our findings demonstrate a novel neuroprotective mechanism for melatonin against TBI which can be a potential neuroprotective agent for the treatment of TBI-induced depression.
Collapse
Affiliation(s)
- Ling-Ling Xie
- Department of Pharmacy, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China.
| | - Chen Rui
- Department of Pharmacy, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China.
| | - Zhuang-Zhuang Li
- Department of Pharmacy, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China.
| | - Shan-Shan Li
- Clinical Lab, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China.
| | - Yong-Jian Fan
- Department of Ultrasonography, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China.
| | - Man-Man Qi
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China.
| |
Collapse
|
35
|
Melatonergic Receptors (Mt1/Mt2) as a Potential Additional Target of Novel Drugs for Depression. Neurochem Res 2022; 47:2909-2924. [PMID: 35689787 PMCID: PMC9187850 DOI: 10.1007/s11064-022-03646-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 05/21/2022] [Accepted: 05/25/2022] [Indexed: 11/06/2022]
Abstract
A complex pathogenesis involving several physiological systems is theorized to underline the development of depressive disorders. Depression is accompanied by circadian regulation disruption and interaction with the functioning of both central and peripheral oscillators. Many aspects of melatonin function unite these systems. The use of drugs for circadian rhythm disorders could inspire a potential treatment strategy for depression. Melatonin plays an essential role in the regulation of circadian rhythms. It exerts effect by activating two types of melatonin receptors, type 1A (MT1) and 1B (MT2). These are G-protein-coupled receptors, predominantly located in the central nervous system. MT1/MT2 agonists could be a useful treatment approach according to all three prevalent theories of the pathogenesis of depression involving either monoamines, synaptic remodeling, or immune/inflammatory events. MT1/MT2 receptors can be a potential target for novel antidepressants with impact on concentrations of neurotrophins or neurotransmitters, and reducing levels of pro-inflammatory cytokines. There is an interesting cross-talk mediated via the physical association of melatonin and serotonin receptors into functional heteromers. The antidepressive and neurogenetic effects of MT1/MT2 agonists can also be caused by the inhibition of the acid sphingomyelinase, leading to reduced ceramide, or increasing monoamine oxidase A levels in the hippocampus. Compounds targeting MT1 and MT2 receptors could have potential for new anti-depressants that may improve the quality of therapeutic interventions in treating depression and relieving symptoms. In particular, a combined effect on MT1 and/or MT2 receptors and neurotransmitter systems may be useful, since the normalization of the circadian rhythm through the melatonergic system will probably contribute to improved treatment. In this review, we discuss melatonergic receptors as a potential additional target for novel drugs for depression.
Collapse
|
36
|
Zhang J, Fang Y, Tang D, Xu X, Zhu X, Wu S, Yu H, Cheng H, Luo T, Shen Q, Gao Y, Ma C, Liu Y, Wei Z, Chen X, Tao F, He X, Cao Y. Activation of MT1/MT2 to Protect Testes and Leydig Cells against Cisplatin-Induced Oxidative Stress through the SIRT1/Nrf2 Signaling Pathway. Cells 2022; 11:cells11101690. [PMID: 35626727 PMCID: PMC9139217 DOI: 10.3390/cells11101690] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/05/2022] [Accepted: 05/17/2022] [Indexed: 02/06/2023] Open
Abstract
There is growing concern that chemotherapy drugs can damage Leydig cells and inhibit the production of testosterone. Increasing evidence shows that melatonin benefits the reproductive process. This study mainly explores the protective effect and possible molecular mechanism of melatonin regarding cisplatin-induced oxidative stress in testicular tissue and Leydig cells. We found that there were only Leydig and Sertoli cells in the testes of gastrointestinal tumor patients with azoospermia caused by platinum chemotherapeutic drugs. Melatonin (Mel) receptor 1/melatonin receptor 2 (MT1/MT2) was mainly expressed in human and mouse Leydig cells of the testes. We also observed that the melatonin level in the peripheral blood decreased and oxidative stress occurred in mice treated with cisplatin or gastrointestinal tumor patients treated with platinum-based chemotherapeutic drugs. iTRAQ proteomics showed that SIRT1/Nrf2 signaling and MT1 proteins were downregulated in cisplatin-treated mouse testes. The STRING database predicted that MT1 might be able to regulate the SIRT1/Nrf2 signaling pathway. Melatonin reduced oxidative stress and upregulated SIRT1/Nrf2 signaling in cisplatin-treated mouse testes and Leydig cells. Most importantly, after inhibiting MT1/MT2, melatonin could not upregulate SIRT1/Nrf2 signaling in cisplatin-treated Leydig cells. The MT1/MT2 inhibitor aggravated the cisplatin-induced downregulation of SIRT1/Nrf2 signaling and increased the apoptosis of Leydig cells. We believe that melatonin stimulates SIRT1/Nrf2 signaling by activating MT1/MT2 to prevent the cisplatin-induced apoptosis of Leydig cells.
Collapse
Affiliation(s)
- Junqiang Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; (J.Z.); (D.T.); (X.Z.); (H.Y.); (H.C.); (Q.S.); (Y.G.); (C.M.); (Y.L.); (Z.W.); (F.T.)
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People’s Republic of China, Hefei 230032, China
| | - Yuan Fang
- Department of Blood Transfusion, Anhui NO. 2 Provincial People’s Hospital, Hefei 230041, China;
| | - Dongdong Tang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; (J.Z.); (D.T.); (X.Z.); (H.Y.); (H.C.); (Q.S.); (Y.G.); (C.M.); (Y.L.); (Z.W.); (F.T.)
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People’s Republic of China, Hefei 230032, China
| | - Xingyu Xu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China;
| | - Xiaoqian Zhu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; (J.Z.); (D.T.); (X.Z.); (H.Y.); (H.C.); (Q.S.); (Y.G.); (C.M.); (Y.L.); (Z.W.); (F.T.)
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People’s Republic of China, Hefei 230032, China
| | - Shusheng Wu
- Department of Medical Oncology, The First Affiliated Hospital of University of Science and Technology of China, Hefei 230031, China;
| | - Hui Yu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; (J.Z.); (D.T.); (X.Z.); (H.Y.); (H.C.); (Q.S.); (Y.G.); (C.M.); (Y.L.); (Z.W.); (F.T.)
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, China
- Department of Obstetrics and Gynecology, Fuyang Hospital of Anhui Medical University, Fuyang 236000, China
| | - Huiru Cheng
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; (J.Z.); (D.T.); (X.Z.); (H.Y.); (H.C.); (Q.S.); (Y.G.); (C.M.); (Y.L.); (Z.W.); (F.T.)
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People’s Republic of China, Hefei 230032, China
| | - Ting Luo
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei 230032, China;
| | - Qunshan Shen
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; (J.Z.); (D.T.); (X.Z.); (H.Y.); (H.C.); (Q.S.); (Y.G.); (C.M.); (Y.L.); (Z.W.); (F.T.)
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei 230032, China;
| | - Yang Gao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; (J.Z.); (D.T.); (X.Z.); (H.Y.); (H.C.); (Q.S.); (Y.G.); (C.M.); (Y.L.); (Z.W.); (F.T.)
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei 230032, China;
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei 230032, China
| | - Cong Ma
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; (J.Z.); (D.T.); (X.Z.); (H.Y.); (H.C.); (Q.S.); (Y.G.); (C.M.); (Y.L.); (Z.W.); (F.T.)
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei 230032, China;
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei 230032, China
| | - Yajing Liu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; (J.Z.); (D.T.); (X.Z.); (H.Y.); (H.C.); (Q.S.); (Y.G.); (C.M.); (Y.L.); (Z.W.); (F.T.)
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei 230032, China
| | - Zhaolian Wei
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; (J.Z.); (D.T.); (X.Z.); (H.Y.); (H.C.); (Q.S.); (Y.G.); (C.M.); (Y.L.); (Z.W.); (F.T.)
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, China
- Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei 230032, China;
| | - Xiaoyu Chen
- Department of Histology and Embryology, Anhui Medical University, Hefei 230032, China;
| | - Fangbiao Tao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; (J.Z.); (D.T.); (X.Z.); (H.Y.); (H.C.); (Q.S.); (Y.G.); (C.M.); (Y.L.); (Z.W.); (F.T.)
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People’s Republic of China, Hefei 230032, China
| | - Xiaojin He
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; (J.Z.); (D.T.); (X.Z.); (H.Y.); (H.C.); (Q.S.); (Y.G.); (C.M.); (Y.L.); (Z.W.); (F.T.)
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People’s Republic of China, Hefei 230032, China
- Correspondence: (X.H.); (Y.C.)
| | - Yunxia Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; (J.Z.); (D.T.); (X.Z.); (H.Y.); (H.C.); (Q.S.); (Y.G.); (C.M.); (Y.L.); (Z.W.); (F.T.)
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People’s Republic of China, Hefei 230032, China
- Correspondence: (X.H.); (Y.C.)
| |
Collapse
|
37
|
Jiang N, Zhang Y, Yao C, Huang H, Wang Q, Huang S, He Q, Liu X. Ginsenosides Rb1 Attenuates Chronic Social Defeat Stress-Induced Depressive Behavior via Regulation of SIRT1-NLRP3/Nrf2 Pathways. Front Nutr 2022; 9:868833. [PMID: 35634375 PMCID: PMC9133844 DOI: 10.3389/fnut.2022.868833] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/29/2022] [Indexed: 11/13/2022] Open
Abstract
Ginsenoside Rb1, a diol-type ginseng saponin, has various positive effects on the central nervous system. This study aimed to evaluate the antidepressant effects of Rb1 on chronic social defeat stress (CSDS) induced behavioral deficits and the exact neural cascades linked with inflammatory processes. The results of behavioral tests such as social interaction, tail suspension, and forced swimming revealed that oral treatment of Rb1 (35 and 70 mg/kg) alleviates depression-like behavior. Rb1 treatment increased antioxidant enzyme activity (SOD and CAT) and reduced lipid peroxidation (LPO) content in the hippocampus. Rb1 also suppressed the production of inflammatory cytokines (TNF-α, IL-18, and IL-1β) as well as microglial activation (Iba1) in response to CSDS. Moreover, Rb1 administration considerably reduced the protein expression of NLRP3 (inflammasome) and promoted the protein expressions of Nrf2, HO-1 and Sirtuin1(SIRT1) activation in the hippocampus. Our findings showed that Rb1 effectively restores the depressive-like behavior in CSDS-induced model mice, mediated in part by the normalization of oxidative stress levels. The suppression of neuroinflammation is mediated by the regulation of SIRT1-NLRP3/Nrf2 pathways. Our results asserted that the Rb1 is a novel therapeutic candidate for treating depression.
Collapse
Affiliation(s)
- Ning Jiang
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Hunan University of Chinese Medicine, College of Traditional Chinese Medicine, Changsha, China
| | - Yiwen Zhang
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Caihong Yao
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Hong Huang
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Qiong Wang
- Hunan University of Chinese Medicine, College of Traditional Chinese Medicine, Changsha, China
- Affiliated TCM Hospital, School of Pharmacy, Sino-Portugal TCM International Cooperation Center, Southwest Medical University, Luzhou, China
| | - Shuangxue Huang
- Hunan University of Chinese Medicine, College of Traditional Chinese Medicine, Changsha, China
| | - Qinghu He
- Hunan University of Chinese Medicine, College of Traditional Chinese Medicine, Changsha, China
| | - Xinmin Liu
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Hunan University of Chinese Medicine, College of Traditional Chinese Medicine, Changsha, China
- *Correspondence: Xinmin Liu
| |
Collapse
|
38
|
Targeting NRF2 in Type 2 diabetes mellitus and depression: Efficacy of natural and synthetic compounds. Eur J Pharmacol 2022; 925:174993. [PMID: 35513015 DOI: 10.1016/j.ejphar.2022.174993] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/31/2022] [Accepted: 04/28/2022] [Indexed: 12/18/2022]
Abstract
Evidence supports a strong bidirectional association between depression and Type 2 diabetes mellitus (T2DM). The harmful impact of oxidative stress and chronic inflammation on the development of both disorders is widely accepted. Nuclear factor erythroid 2-related factor 2 (NRF2) is a pertinent target in disease management owing to its reputation as the master regulator of antioxidant responses. NRF2 influences the expression of various cytoprotective phase 2 antioxidant genes, which is hampered in both depression and T2DM. Through interaction and crosstalk with several signaling pathways, NRF2 endeavors to contain the widespread oxidative damage and persistent inflammation involved in the pathophysiology of depression and T2DM. NRF2 promotes the neuroprotective and insulin-sensitizing properties of its upstream and downstream targets, thereby interrupting and preventing disease advancement. Standard antidepressant and antidiabetic drugs may be powerful against these disorders, but unfortunately, they come bearing distressing side effects. Therefore, exploiting the therapeutic potential of NRF2 activators presents an exciting opportunity to manage such bidirectional and comorbid conditions.
Collapse
|
39
|
Yang X, Zhang F, Du Y, Cui W, Dou Y, Lin Y, Zhao Z, Ma X. Effect of tetrahedral DNA nanostructures on LPS‐induced neuroinflammation in mice. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.10.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
40
|
Hu C, Zhang S, Chen Q, Wang R. Ovatodiolide protects ischemia-reperfusion-induced neuronal injury via microglial neuroinflammation via mediating SIRT1/NF-κB pathway. Brain Res Bull 2022; 180:97-107. [PMID: 34968641 DOI: 10.1016/j.brainresbull.2021.12.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 11/30/2021] [Accepted: 12/22/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Ovatodiolide (OVA), a bioactive substance extracted from the bioactive component of Anisomeles indica, is reported to be endowed with anti-inflammatory properties. Nonetheless, its function in ischemia-reperfusion (I/R)-induced neurological deficits and microglial inflammation remains unclear. METHOD A middle cerebral artery occlusion (MCAO) model was set up in SD rats, which were then dealt with varying doses of OVA. The rats' neurological functions were estimated at diverse periods postoperatively. The dry and wet method, triphenyl tetrazolium chloride (TTC) staining, and Nissl's staining were conducted to measure brain edema, cerebral infarction area and neuronal damage, respectively. Immunohistochemistry (IHC) was performed to detect neuronal apoptosis and microglial activation, and the profiles of inflammatory factors in the cerebral tissues were estimated by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). In-vitro assays were implemented on HT22 neuronal cells and BV2 microglia to elaborate the effect of OVA against oxygen-glucose deprivation (OGD)-mediated effects. RESULTS OVA relieved HT22 cell apoptosis and eased inflammation in BV2 microglia, which were induced by OGD. OVA mitigated NF-κB phosphorylation in BV2 cells, whereas boosted SIRT1 expression. However, inhibiting SIRT1 abolished the anti-inflammatory effects of OVA in BV2 microglia under OGD stimulation. The condition medium (CM) of OGD-treated BV2 cells enhanced HT22 cell apoptosis and damage. OVA treatment in BV2 cells relieved BV2-mediated injury on HT22 cells, which was reversed by SIRT1 inhibitor. In-vivo results revealed that OVA dose-dependently attenuated I/R rats' neurological deficits, reduced brain edema, cerebral infarction area, neuronal apoptosis and microglial overactivation. Additionally, OVA inactivated the NF-κB pathway and up-regulated SIRT1 in the I/R rat model. CONCLUSION OVA prevented rats from brain I/R damage by hampering neuronal apoptosis and microglial inflammation via the SIRT1-NF-κB pathway. DATA AVAILABILITY The data sets used and analyzed during the current study are available from the corresponding author on reasonable request.
Collapse
Affiliation(s)
- Chaojun Hu
- Department of Emergency, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Shan Zhang
- Department of Neurology, The People's Hospital of Leshan, Leshan 614000, Sichuan, China
| | - Qian Chen
- Department of Neurology, The First People's Hospital of Ziyang, Ziyang 641300, Sichuan, China
| | - Rong Wang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China.
| |
Collapse
|
41
|
Bakaeva Z, Lizunova N, Tarzhanov I, Boyarkin D, Petrichuk S, Pinelis V, Fisenko A, Tuzikov A, Sharipov R, Surin A. Lipopolysaccharide From E. coli Increases Glutamate-Induced Disturbances of Calcium Homeostasis, the Functional State of Mitochondria, and the Death of Cultured Cortical Neurons. Front Mol Neurosci 2022; 14:811171. [PMID: 35069113 PMCID: PMC8767065 DOI: 10.3389/fnmol.2021.811171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
Lipopolysaccharide (LPS), a fragment of the bacterial cell wall, specifically interacting with protein complexes on the cell surface, can induce the production of pro-inflammatory and apoptotic signaling molecules, leading to the damage and death of brain cells. Similar effects have been noted in stroke and traumatic brain injury, when the leading factor of death is glutamate (Glu) excitotoxicity too. But being an amphiphilic molecule with a significant hydrophobic moiety and a large hydrophilic region, LPS can also non-specifically bind to the plasma membrane, altering its properties. In the present work, we studied the effect of LPS from Escherichia coli alone and in combination with the hyperstimulation of Glu-receptors on the functional state of mitochondria and Ca2+ homeostasis, oxygen consumption and the cell survival in primary cultures from the rats brain cerebellum and cortex. In both types of cultures, LPS (0.1–10 μg/ml) did not change the intracellular free Ca2+ concentration ([Ca2+]i) in resting neurons but slowed down the median of the decrease in [Ca2+]i on 14% and recovery of the mitochondrial potential (ΔΨm) after Glu removal. LPS did not affect the basal oxygen consumption rate (OCR) of cortical neurons; however, it did decrease the acute OCR during Glu and LPS coapplication. Evaluation of the cell culture survival using vital dyes and the MTT assay showed that LPS (10 μg/ml) and Glu (33 μM) reduced jointly and separately the proportion of live cortical neurons, but there was no synergism or additive action. LPS-effects was dependent on the type of culture, that may be related to both the properties of neurons and the different ratio between neurons and glial cells in cultures. The rapid manifestation of these effects may be the consequence of the direct effect of LPS on the rheological properties of the cell membrane.
Collapse
Affiliation(s)
- Zanda Bakaeva
- Laboratory of Neurobiology, “National Medical Research Center of Children’s Health”, Russian Ministry of Health, Moscow, Russia
- Department of General Biology and Physiology, Kalmyk State University named after B.B. Gorodovikov, Elista, Russia
- *Correspondence: Zanda Bakaeva, ,
| | - Natalia Lizunova
- Laboratory of Neurobiology, “National Medical Research Center of Children’s Health”, Russian Ministry of Health, Moscow, Russia
- Department of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Ivan Tarzhanov
- Laboratory of Neurobiology, “National Medical Research Center of Children’s Health”, Russian Ministry of Health, Moscow, Russia
- Institute of Pharmacy, The Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Dmitrii Boyarkin
- Laboratory of Neurobiology, “National Medical Research Center of Children’s Health”, Russian Ministry of Health, Moscow, Russia
| | - Svetlana Petrichuk
- Laboratory of Neurobiology, “National Medical Research Center of Children’s Health”, Russian Ministry of Health, Moscow, Russia
| | - Vsevolod Pinelis
- Laboratory of Neurobiology, “National Medical Research Center of Children’s Health”, Russian Ministry of Health, Moscow, Russia
| | - Andrey Fisenko
- Laboratory of Neurobiology, “National Medical Research Center of Children’s Health”, Russian Ministry of Health, Moscow, Russia
| | - Alexander Tuzikov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Rinat Sharipov
- Laboratory of Fundamental and Applied Problems of Pain, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Alexander Surin
- Laboratory of Neurobiology, “National Medical Research Center of Children’s Health”, Russian Ministry of Health, Moscow, Russia
- Laboratory of Fundamental and Applied Problems of Pain, Institute of General Pathology and Pathophysiology, Moscow, Russia
| |
Collapse
|
42
|
Wang YQ, Jiang YJ, Zou MS, Liu J, Zhao HQ, Wang YH. Antidepressant actions of melatonin and melatonin receptor agonist: Focus on pathophysiology and treatment. Behav Brain Res 2021; 420:113724. [PMID: 34929236 DOI: 10.1016/j.bbr.2021.113724] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/15/2021] [Accepted: 12/15/2021] [Indexed: 12/27/2022]
Abstract
Depression has become one of the most commonly prevalent neuropsychiatric disorders, and the main characteristics of depression are sleep disorders and melatonin secretion disorders caused by circadian rhythm disorders. Abnormal endogenous melatonin alterations can contribute to the occurrence and development of depression. However, molecular mechanisms underlying this abnormality remain ambiguous. The present review summarizes the mechanisms underlying the antidepressant effects of melatonin, which is related to its functions in the regulation of the hypothalamic-pituitary-adrenal axis, inhibition of neuroinflammation, inhibition of oxidative stress, alleviation of autophagy, and upregulation of neurotrophic, promotion of neuroplasticity and upregulation of the levels of neurotransmitters, etc. Also, melatonin receptor agonists, such as agomelatine, ramelteon, piromelatine, tasimelteon, and GW117, have received considerable critical attention and are highly implicated in treating depression and comorbid disorders. This review focuses on melatonin and various melatonin receptor agonists in the pathophysiology and treatment of depression, aiming to provide further insight into the pathogenesis of depression and explore potential targets for novel agent development.
Collapse
Affiliation(s)
- Ye-Qing Wang
- Institute of Innovation and Applied Research, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Ya-Jie Jiang
- Institute of Innovation and Applied Research, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Man-Shu Zou
- Institute of Innovation and Applied Research, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Jian Liu
- The First Hospital, Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Hong-Qing Zhao
- Institute of Innovation and Applied Research, Hunan University of Chinese Medicine, Changsha, Hunan Province, China.
| | - Yu-Hong Wang
- Institute of Innovation and Applied Research, Hunan University of Chinese Medicine, Changsha, Hunan Province, China.
| |
Collapse
|
43
|
Siwicka-Gieroba D, Dabrowski W. Credibility of the Neutrophil-to-Lymphocyte Count Ratio in Severe Traumatic Brain Injury. Life (Basel) 2021; 11:life11121352. [PMID: 34947883 PMCID: PMC8706648 DOI: 10.3390/life11121352] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 02/07/2023] Open
Abstract
Traumatic brain injury (TBI) is one of the leading causes of morbidity and mortality worldwide. The consequences of a TBI generate the activation and accumulation of inflammatory cells. The peak number of neutrophils entering into an injured brain is observed after 24 h; however, cells infiltrate within 5 min of closed brain injury. Neutrophils release toxic molecules including free radicals, proinflammatory cytokines, and proteases that advance secondary damage. Regulatory T cells impair T cell infiltration into the central nervous system and elevate reactive astrogliosis and interferon-γ gene expression, probably inducing the process of healing. Therefore, the neutrophil-to-lymphocyte ratio (NLR) may be a low-cost, objective, and available predictor of inflammation as well as a marker of secondary injury associated with neutrophil activation. Recent studies have documented that an NLR value on admission might be effective for predicting outcome and mortality in severe brain injury patients.
Collapse
|
44
|
Song L, Wu X, Wang J, Guan Y, Zhang Y, Gong M, Wang Y, Li B. Antidepressant effect of catalpol on corticosterone-induced depressive-like behavior involves the inhibition of HPA axis hyperactivity, central inflammation and oxidative damage probably via dual regulation of NF-κB and Nrf2. Brain Res Bull 2021; 177:81-91. [PMID: 34500039 DOI: 10.1016/j.brainresbull.2021.09.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 12/24/2022]
Abstract
This study aimed to investigate the antidepressant effect and mechanism of catalpol on corticosterone (CORT)-induced depressive-like behavior in mice for the first time. As a result, CORT injection induced depressive-like behaviors of mice in behavioral tests, aggravated the serum CORT, adrenocorticotropic hormone, and corticotropin-releasing hormone levels, and conspicuously elevated the phosphorylations of nuclear factor kappa-B (NF-κB) in the hippocampus and frontal cortex, and down-regulated the expression levels of nuclear factor erythroid-2-related factor 2 (Nrf2). Furthermore, CORT exposure dramatically augmented the levels of inflammatory factors (interleukin-1β, tumor necrosis factor-α, nitric oxide synthase, and nitric oxide) and lipid peroxidation product malondialdehyde, and attenuated the levels of antioxidants including reduced glutathione, glutathione S-transferase, total superoxide dismutase, and heme oxygenase-1 in the mouse hippocampus and frontal cortex. On the contrary, catalpol administration markedly suppressed the abnormalities of the above indicators. From the overall results, this study displayed that catalpol exerted a beneficial effect on CORT-induced depressive-like behavior in mice possibly via the inhibition of hypothalamus-pituitary-adrenal (HPA) axis hyperactivity, central inflammation and oxidative damage at least partially through dual regulation of NF-κB and Nrf2.
Collapse
Affiliation(s)
- Lingling Song
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Xiaohui Wu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Junming Wang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| | - Yuechen Guan
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Yueyue Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Mingzhu Gong
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Yanmei Wang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Bingyin Li
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| |
Collapse
|
45
|
Nrf2 Alleviates Cognitive Dysfunction and Brain Inflammatory Injury via Mediating Wfs1 in Rats with Depression-Like Behaviors. Inflammation 2021; 45:399-413. [PMID: 34495404 DOI: 10.1007/s10753-021-01554-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/26/2021] [Accepted: 08/23/2021] [Indexed: 12/26/2022]
Abstract
Depression is a major threat to global mental health and demands targeted therapeutic regimens. The current study set out to evaluate the regulatory mechanism of nuclear factor erythroid-2 related factor 2 (Nrf2) in depression-induced cognitive dysfunction and inflammatory injury. First, depressive rat models were established via chronic unpredicted mild stress (CUMS) treatment. Cognitive function of rats was assessed by a series of behavioral tests. Rats were further stereotactically injected with Nrf2 overexpression vector, with expression patterns of Nrf2, miR-17-5p, and wolfram syndrome 1 (Wfs1) detected using qRT-PCR and Western blot assay. In addition, pathological changes of murine hippocampus were analyzed using hematoxylin-eosin staining. In vitro cell models were additionally established using lipopolysaccharide. Cell viability was detected via the CCK-8 method. Moreover, levels of TNF-α, IL-1β, and IL-10 were detected via ELISA. Furthermore, the binding relationships between Nrf2 and the miR-17-5p promoter, miR-17-5p, and Wfs1 were verified. It was found that Nrf2 was weakly expressed in CUMS-treated rats, whereas Nrf2 upregulation alleviated cognitive dysfunction and brain inflammatory injury. Meanwhile, Nrf2 inhibited miR-17-5p expression via binding to the miR-17-5p promoter. miR-17-5p was also found to limit Wfs1 transcription. miR-17-5p overexpression or Wfs1 downregulation partly reversed the role of Nrf2 in reliving inflammatory injury of murine hippocampal neurons. Overall, our findings indicated that Nrf2 inhibited miR-17-5p expression and promoted Wfs1 transcription, thereby alleviating cognitive dysfunction and inflammatory injury in rats with depression-like behaviors.
Collapse
|
46
|
Zhang Y, Cong P, Tong C, Jin H, Liu Y, Hou M. Melatonin pretreatment alleviates blast-induced oxidative stress in the hypothalamic-pituitary-gonadal axis by activating the Nrf2/HO-1 signaling pathway. Life Sci 2021; 280:119722. [PMID: 34153300 DOI: 10.1016/j.lfs.2021.119722] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/02/2021] [Accepted: 06/09/2021] [Indexed: 01/14/2023]
Abstract
Although melatonin has been demonstrated to exert a potent antioxidant effect, the ability of melatonin to alleviate blast-induced oxidative stress in the hypothalamic-pituitary-gonadal (HPG) axis remains unclear. This study aimed to elucidate the effects and underlying mechanism of melatonin pretreatment on the HPG axis disrupted by blast injury. Sixty C57BL/6 mice were randomly divided into control, blast, and blast + melatonin groups for behavioral experiments. The elevated maze experiment, open field experiment, and Morris Water Maze experiment were carried out on the 7th, 14th and 28th day after the blast injury. Fifty Sprague Dawley rats were randomly divided into control, blast, blast + melatonin, and blast + melatonin + luzindole groups for hormone assays and molecular and pathological experiments. Blood samples were used for HPG axis hormone detection and ELISA assays, and tissue samples were used to detect oxidative stress, inflammation, apoptosis, and stress-related protein levels. The results showed that melatonin pretreatment alleviated blast-induced behavioral abnormalities in mice and maintained the HPG axis hormone homeostasis in rats. Additionally, melatonin significantly reduced MDA5 expression and increased the expression of Nrf2/HO-1. Moreover, melatonin significantly inhibited NF-κB expression and upregulated IL-10 expression, and it reversed the blast-induced high expression of caspase-3 and Bax and the low expression of Bcl-2. Furthermore, luzindole counteracted melatonin inhibition of NF-κB and upregulated Nrf2/HO-1. Melatonin significantly alleviated blast-induced HPG axis hormone dyshomeostasis, behavioral abnormalities, oxidative stress, inflammation, and apoptosis, which may be achieved by upregulating the Nrf2/HO-1 signaling pathway. Our study suggested that melatonin pretreatment is a potential treatment for blast-induced HPG axis hormonal and behavioral abnormalities.
Collapse
Affiliation(s)
- Yin Zhang
- Graduate School, Dalian Medical University, Dalian, Liaoning Province 116044, China
| | - Peifang Cong
- Department of Emergency Medicine, The General Hospital of Northern Theater Command, Laboratory of Rescue Center of Severe Trauma PLA, Shenyang, Liaoning Province 116044, China
| | - Changci Tong
- Department of Emergency Medicine, The General Hospital of Northern Theater Command, Laboratory of Rescue Center of Severe Trauma PLA, Shenyang, Liaoning Province 116044, China
| | - Hongxu Jin
- Department of Emergency Medicine, The General Hospital of Northern Theater Command, Laboratory of Rescue Center of Severe Trauma PLA, Shenyang, Liaoning Province 116044, China
| | - Yunen Liu
- The Second Affiliated Hospital of Shenyang Medical College, The Veterans General Hospital of Liaoning Province, No. 20 Beijiu Road, Heping District, Shenyang 110001, China; Shenyang Medical College, No. 146, Huanghe North Street, Shenyang 110034, China.
| | - Mingxiao Hou
- Graduate School, Dalian Medical University, Dalian, Liaoning Province 116044, China; Department of Emergency Medicine, The General Hospital of Northern Theater Command, Laboratory of Rescue Center of Severe Trauma PLA, Shenyang, Liaoning Province 116044, China.
| |
Collapse
|
47
|
Hu D, Zhang L, Jiang R, Liao C, Xu J, Jiang S, Yang Y, Lin L, Huang J, Shen Y, Tang L, Li L. Nicotinic Acid against Acetaminophen-Induced Hepatotoxicity via Sirt1/Nrf2 Antioxidative Pathway in Mice. J Nutr Sci Vitaminol (Tokyo) 2021; 67:145-152. [PMID: 34193673 DOI: 10.3177/jnsv.67.145] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Acetaminophen (N-acetyl-p-aminophenol, APAP) overdose causes hepatotoxicity, even liver failure, and oxidative stress plays pivotal role in its pathogenesis. Nicotinic acid (NA) is one form of vitamin B3, which has been used to treat a series of diseases in clinic for decades. To date, several studies have evidenced that NA has anti-oxidative property. Therefore, NA may have the hepatoprotective potential against APAP-induced toxicity. Here, our aim was to investigate the beneficial effect of NA against hepatotoxicity induced by APAP and its mechanism in vivo. BALB/c mice were intraperitoneally injected with NA (100 mg/kg) 3 times at 24, 12 and 1 h before APAP (600 mg/kg or 400 mg/kg) challenge. The results showed that pretreatment of NA markedly improved the survival rate, alleviated serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels and mitigated the histopathological injuries compared to APAP-exposed mice. Furthermore, NA significantly elevated the activities of superoxide dismutase (SOD), catalase (CAT) and glutathione (GSH) content, while reduced malondialdehyde (MDA) level. Finally, the signaling pathway was probed. The western blot revealed that NA up-regulated Sirtuin1 (Sirt1), nuclear factor erythroid 2-related factor 2 (Nrf2) and NAD(P)H quinone dehydrogenase-1 (NQO-1) expression and down-regulated Kelch-like ECH-associated protein 1 (Keap1) level in liver followed APAP exposure, implying Sirt1/Nrf2 axis exerted an essential role in the protective mechanism of NA on APAP toxicity. In brief, pretreatment of NA effectively protects liver against hepatotoxicity due to overdose of APAP through an antioxidant dependent manner modulated by Sirt1/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Da Hu
- Department of Pathophysiology, Chongqing Medical University
| | - Li Zhang
- Department of Pathophysiology, Chongqing Medical University
| | - Rong Jiang
- Department of Histology and Embryology, Chongqing Medical University
| | - Cuiting Liao
- Department of Pathophysiology, Chongqing Medical University
| | - Juanjuan Xu
- Department of Pathophysiology, Chongqing Medical University
| | - Shifang Jiang
- Department of Pathophysiology, Chongqing Medical University
| | - Yongqiang Yang
- Department of Pathophysiology, Chongqing Medical University
| | - Ling Lin
- Department of Pathophysiology, Chongqing Medical University
| | - Jiayi Huang
- Department of Pathophysiology, Chongqing Medical University
| | - Yi Shen
- Department of Pathophysiology, Chongqing Medical University
| | - Li Tang
- Department of Pathophysiology, Chongqing Medical University
| | - Longjiang Li
- Department of Pathophysiology, Chongqing Medical University
| |
Collapse
|
48
|
Xanthohumol Attenuates Lipopolysaccharide-Induced Depressive Like Behavior in Mice: Involvement of NF-κB/Nrf2 Signaling Pathways. Neurochem Res 2021; 46:3135-3148. [PMID: 34398408 DOI: 10.1007/s11064-021-03396-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 07/04/2021] [Accepted: 07/05/2021] [Indexed: 02/08/2023]
Abstract
Depression is the most common psychiatric disorder associated with brain and immune system abnormalities. In recent years, xanthohumol (Xn) a bioactive prenylated flavonoid has received ample attention for its polypharmacological effects, therefore, here we aimed to explore the protective effects of Xn against the LPS-induced depressive-like symptoms mediated by inflammation and oxidative stress. We tested the effect of Xn against LPS-induced behavioural changes in mice by means of forced swimming test (FST), tail suspention test (TST), sucrose preference test (SPT) and open field test (OPT). Examined the neuroinflammation and oxido-nitrosative stress (O&NS) markers and analyze Nrf2 and NF-κB signalling pathways in the hippocampus. Our results indicated that peripheral repeated administration of lipopolysaccharides (LPS) (1 mg/kg, intra peritoneally) induced depressive-like behavior, neuroinflammation and O&NS in mice. Pretreatment with Xn (10 and 20 mg/kg, intra gastrically) reverse the behavioural impairments prophylactically as obvious in the FST and TST without effecting locomotion, however only 20 mg dose improve anhedonic behavior as observed in SPT. Similarly, Xn pretreatment in dose-dependent manner prevented the LPS induced neuro-inflammation and O&NS. Immunofluorescence analysis showed that Xn reduced activated gliosis via attenuation of Iba-1 and GFAP in hippocampus. In addition, Xn considerably reduced the expression of phospho-NF-κB and cleaved caspase-3 while enhanced Nrf2 and HO-1 expression in the hippocampus. To the best of our knowledge, this is the first study to examine the underlying beneficial prophylactic effects of the Xn in neuroinflammation and O&NS mediating depressive-like behaviors.
Collapse
|
49
|
Businaro R, Vauzour D, Sarris J, Münch G, Gyengesi E, Brogelli L, Zuzarte P. Therapeutic Opportunities for Food Supplements in Neurodegenerative Disease and Depression. Front Nutr 2021; 8:669846. [PMID: 34055858 PMCID: PMC8160227 DOI: 10.3389/fnut.2021.669846] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/19/2021] [Indexed: 12/16/2022] Open
Abstract
Emerging evidence is showing nutrition as a crucial factor in the high prevalence and incidence of neurodegenerative mental disorders. Preventive interventions on neuroinflammation seem to be able to interfere with neurodegeneration. Supplementation of essential nutrients, such as long-chain-polyunsaturated fatty acids, vitamin E and mineral elements, may minimize inflammation, enhancing antioxidative defense, and lowering the risk and incidence of age-related diseases, such as cardiovascular diseases and neurodegenerative diseases. This manuscript reviews the current evidence on the role of neuroinflammation in the pathophysiology of neurodegenerative and mental disorders, and preventive strategies for food supplementation in these neuropsychiatric diseases. Dietary supplementation-based strategies have been demonstrated to be effective in subjects with mild cognitive impairment, while weaker results have been obtained in patients with advance neurodegenerative disease. Adjunctive supplementation has also been demonstrated to improve depression, this being of marked benefit considering the comorbidity between cognitive impairment/dementia and depression. Further research is needed to improve the prescriptive precision of supplementation in patients, and to better understand potential interactions with clinical and pharmacokinetic factors.
Collapse
Affiliation(s)
- Rita Businaro
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - David Vauzour
- Faculty of Medicine and Health Sciences, Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Jerome Sarris
- NICM Health Research Institute, Western Sydney University, Westmead, NSW, Australia.,Professorial Unit, The Melbourne Clinic, Department of Psychiatry, Melbourne University, Melbourne, VIC, Australia
| | - Gerald Münch
- Pharmacology Unit, School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Erika Gyengesi
- Pharmacology Unit, School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | | | - Pedro Zuzarte
- Psychiatric Clinic, Faculty of Medicine, University of Lisbon, Lisbon, Portugal.,Neuropsychiatry Research Department, GNR Clinical Center, Lisbon, Portugal
| |
Collapse
|
50
|
Ma H, Kang J, Fan W, He H, Huang F. ROR: Nuclear Receptor for Melatonin or Not? Molecules 2021; 26:molecules26092693. [PMID: 34064466 PMCID: PMC8124216 DOI: 10.3390/molecules26092693] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/28/2021] [Accepted: 05/03/2021] [Indexed: 12/14/2022] Open
Abstract
Whether the retinoic acid-related orphan receptor (ROR) is a nuclear receptor of melatonin remains controversial. ROR is inextricably linked to melatonin in terms of its expression, function, and mechanism of action. Additionally, studies have illustrated that melatonin functions analogous to ROR ligands, thereby modulating the transcriptional activity of ROR. However, studies supporting these interactions have since been withdrawn. Furthermore, recent crystallographic evidence does not support the view that ROR is a nuclear receptor of melatonin. Some other studies have proposed that melatonin indirectly regulates ROR activity rather than directly binding to ROR. This review aims to delve into the complex relationship of the ROR receptor with melatonin in terms of its structure, expression, function, and mechanism. Thus, we provide the latest evidence and views on direct binding as well as indirect regulation of ROR by melatonin, dissecting both viewpoints in-depth to provide a more comprehensive perspective on this issue.
Collapse
Affiliation(s)
- Haozhen Ma
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; (H.M.); (J.K.); (W.F.)
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, China
| | - Jun Kang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; (H.M.); (J.K.); (W.F.)
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, China
| | - Wenguo Fan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; (H.M.); (J.K.); (W.F.)
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, China
| | - Hongwen He
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510080, China
- Correspondence: (H.H.); (F.H.); Tel.: +86-20-8733-0570 (H.H. & F.H.)
| | - Fang Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; (H.M.); (J.K.); (W.F.)
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510080, China
- Correspondence: (H.H.); (F.H.); Tel.: +86-20-8733-0570 (H.H. & F.H.)
| |
Collapse
|