1
|
Ullah S, Park TJ, Park JS, Atiq A, Ali J, Kang MH, Ali W, Choe K, Kim MO. Ambroxol attenuates detrimental effect of LPS-induced glia-mediated neuroinflammation, oxidative stress, and cognitive dysfunction in mice brain. Front Immunol 2025; 16:1494114. [PMID: 40114925 PMCID: PMC11923628 DOI: 10.3389/fimmu.2025.1494114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 02/14/2025] [Indexed: 03/22/2025] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD), are multifactorial. Among various factors, lipopolysaccharides (LPSs) from Gram-negative bacteria, such as E. coli, are considered potential causative agents. Despite significant advancements in the field, there is still no cure. In this study, we investigated the neuroprotective effects of ambroxol against LPS-induced neuroinflammation, oxidative stress, neurodegeneration, and the associated cognitive dysfunction. Intraperitoneal injection of LPS (250 µg/kg every alternative day for a total of seven doses over 14 days) triggered glial cell activation, neuroinflammation, oxidative stress, and neurodegeneration in the mouse brain. Ambroxol treatment (30 mg/kg/day for 14 days) significantly reduced neuroinflammation and oxidative stress compared to LPS-treated mice. Immunoblotting and immunofluorescence results showed that ambroxol reduced levels of Toll-like receptor 4 (TLR4) and oxidative stress kinase phospho-c-Jun N-terminal Kinase 1 (p-JNK). It also decreased astrocyte and microglia activation in the cortex and hippocampus of LPS+ Amb-treated mice, as indicated by the downregulation of GFAP and Iba-1. Furthermore, ambroxol-reversed LPS-induced neuroinflammation by inhibiting inflammatory mediators, such as IL-1β and TNF-α, through regulation of the transcription factor p-NFkB. Persistent neuroinflammation disrupted the natural antioxidant mechanisms, leading to oxidative stress. Ambroxol treatment upregulated antioxidant markers, including Nrf-2, HO-1, and SOD, which were downregulated in the LPS-treated group. Additionally, ambroxol-inhibited lipid peroxidation, maintaining malondialdehyde levels in the mouse brain. Ambroxol also improves synaptic integrity by upregulating synaptic biomarkers, including PSD-95 and SNAP-23. Overall, ambroxol demonstrated anti-inflammatory, antioxidant, and neuroprotective effects in LPS-treated mice, highlighting its potential benefits in neurological disorders.
Collapse
Affiliation(s)
- Safi Ullah
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Tae Ju Park
- Haemato-oncology/Systems Medicine Group, Paul O'Gorman Leukaemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences (MVLS), University of Glasgow, Glasgow, United Kingdom
| | - Jun Sung Park
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Abubakar Atiq
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Jawad Ali
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Min Hwa Kang
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Waqar Ali
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Kyonghwan Choe
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Mastricht, Netherlands
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
- Alz-Dementia Korea Co., Jinju, Republic of Korea
| |
Collapse
|
2
|
Sakrajda K, Langwiński W, Stachowiak Z, Ziarniak K, Narożna B, Szczepankiewicz A. Immunomodulatory effect of lithium treatment on in vitro model of neuroinflammation. Neuropharmacology 2025; 265:110238. [PMID: 39586495 DOI: 10.1016/j.neuropharm.2024.110238] [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: 07/21/2024] [Revised: 09/20/2024] [Accepted: 11/21/2024] [Indexed: 11/27/2024]
Abstract
Bipolar disorder (BD) is psychiatric disorder of not fully acknowledged pathophysiology. Studies show the involvement of innate-immune system activation and inflammation in BD course and treatment efficiency. Microglia are crucial players in the inflammatory response possibly responsible for BD innate-immune activity. Lithium is a mood stabilizer used in treatment for 75 years. Immunomodulation was previously described as one of the potential modes of its action. We hypothesized that lithium might modulate the microglia response to innate-immune-associated cytokines (10 ng/mL TNF-α, 50 ng/mL IL-1β, 20 ng/mL IFN-γ). We aimed to investigate whether lithium treatment and pretreatment of microglia modify the expression of genes associated with NLRP3 inflammasome. We also aimed to verify lithium treatment effect on caspase activity and extracellular IL-1β concentration. For the first time, our study used human microglial cell line - HMC3, the cytokine stimuli and lithium in concentration corresponding to that in the brains of patients. To analyze lithium mode of action, we analyzed the short- and long-term treatment and pretreatment. To assess the influence on microglia responding to innate-immune cytokines, we analyzed the expression of genes involved in innate-immune and inflammasome (TSPO, TLR4, NFKB1, CASP1, CASP4, NLRP3, IL-1β, IL-6), caspase activity, extracellular IL-1β concentration, phospho-GSK-3β(Ser9) expression and lactate concentration. We found that lithium treatment significantly reduced NLRP3 inflammasome-related genes expression. We observed that lithium treatment reduces inflammasome activity, which may attenuate the inflammatory state. Interestingly, the lithium pretreatment resulted in significantly elevated inflammasome activity, suggesting that lithium does not impair the immune response to additional stimuli.
Collapse
Affiliation(s)
- Kosma Sakrajda
- Molecular and Cell Biology Unit, Poznan University of Medical Sciences, 60-572, Poznan, Poland; Doctoral School, Poznan University of Medical Sciences, 60-812, Poznan, Poland.
| | - Wojciech Langwiński
- Molecular and Cell Biology Unit, Poznan University of Medical Sciences, 60-572, Poznan, Poland
| | - Zuzanna Stachowiak
- Molecular and Cell Biology Unit, Poznan University of Medical Sciences, 60-572, Poznan, Poland
| | - Kamil Ziarniak
- Molecular and Cell Biology Unit, Poznan University of Medical Sciences, 60-572, Poznan, Poland
| | - Beata Narożna
- Molecular and Cell Biology Unit, Poznan University of Medical Sciences, 60-572, Poznan, Poland
| | | |
Collapse
|
3
|
Bhat K, Helmholz H, Willumeit-Römer R. Application of an in vitro neuroinflammation model to evaluate the efficacy of magnesium-lithium alloys. Front Cell Neurosci 2024; 18:1485427. [PMID: 39539342 PMCID: PMC11558531 DOI: 10.3389/fncel.2024.1485427] [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: 08/23/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Mg-Li alloys can be promising candidates as bioresorbable Li-releasing implants for bipolar disorder and other neurodegenerative disorders. In order to compare the therapeutic efficacy of conventional Li salts and Li delivered through Mg-Li alloy extracts, we tested an in vitro model based on the neuroinflammation hypothesis of mood disorders (peripheral inflammation inducing neuroinflammation) wherein, a coculture of microglia and astrocytes was treated with conditioned medium from pro-inflammatory macrophages. Two alloys, Mg-1.6Li and Mg-9.5Li, were tested in the form of material extracts and well-known outcomes of Li treatment such as GSK3β phosphorylation (indirect flow cytometry) and influence on inflammation-related gene expression (qPCR) were compared against Li salts. This is the first study demonstrating that Li can increase the phosphorylation of GSK3β in glial cells in the presence of excess Mg. Furthermore, Mg-Li alloys were more effective than Li salts in downregulating IL6 and upregulating the neurotrophin GDNF. Mg had no antagonistic effects toward Li-driven downregulation of astrogliosis markers. Overall, the results provide evidence to support further studies employing Mg-Li alloys for neurological applications.
Collapse
|
4
|
Nassar A, Kaplanski J, Azab AN. A Selective Nuclear Factor-κB Inhibitor, JSH-23, Exhibits Antidepressant-like Effects and Reduces Brain Inflammation in Rats. Pharmaceuticals (Basel) 2024; 17:1271. [PMID: 39458912 PMCID: PMC11509963 DOI: 10.3390/ph17101271] [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: 08/29/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Accumulating evidence suggests that nuclear factor (NF)-κB is involved in the pathophysiology of mood disorders. OBJECTIVES AND METHODS We conducted two experimental protocols in rats to investigate the effects of a selective NF-κB inhibitor (JSH-23) on (i) lipopolysaccharide (LPS)-induced inflammation and (ii) on behavioral phenotypes in rat models of depression (sucrose consumption test and forced swim test) and mania (amphetamine-induced hyperactivity test). Additionally, we tested the effects of JSH-23 on levels of inflammatory components (interleukin-6, prostaglandin E2, nuclear phospho-p65, and tumor necrosis factor-α) in the brain. RESULTS Acute treatment with JSH-23 (10 mg/kg, intraperitoneally [ip]) led to potent anti-inflammatory effects in LPS-treated rats, including a diminished hypothermic response to LPS and a reduction in pro-inflammatory mediators' levels in the brain. Chronic treatment with JSH-23 (3 mg/kg, ip, once daily, for 14 days) resulted in robust antidepressant-like effects (increased sucrose consumption and decreased immobility time). The antidepressant-like effects of JSH-23 were mostly accompanied by a reduction in levels of pro-inflammatory mediators in the brain. On the other hand, JSH-23 did not reduce amphetamine-induced hyperactivity. CONCLUSIONS Altogether, these data suggest that NF-κB may be a potential therapeutic target for pharmacological interventions for depression.
Collapse
Affiliation(s)
- Ahmad Nassar
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Jacob Kaplanski
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Abed N. Azab
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Department of Nursing, School for Community Health Professions, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| |
Collapse
|
5
|
Choe K, Park JS, Park HY, Tahir M, Park TJ, Kim MO. Lupeol protect against LPS-induced neuroinflammation and amyloid beta in adult mouse hippocampus. Front Nutr 2024; 11:1414696. [PMID: 39050141 PMCID: PMC11266137 DOI: 10.3389/fnut.2024.1414696] [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: 04/09/2024] [Accepted: 07/01/2024] [Indexed: 07/27/2024] Open
Abstract
Neuroinflammation includes the activation of immune glial cells in the central nervous system, release pro-inflammatory cytokines, which disrupt normal neural function and contribute to various neurological disorders, including Alzheimer's disease (AD), Parkinson's disease, multiple sclerosis, and stroke. AD is characterized by various factors including amyloidogenesis, synaptic dysfunction, memory impairment and neuroinflammation. Lipopolysaccharide (LPS) constitutes a vital element of membrane of the gram-negative bacterial cell, triggering vigorous neuroinflammation and facilitating neurodegeneration. Lupeol, a naturally occurring pentacyclic triterpene, has demonstrated several pharmacological properties, notably its anti-inflammatory activity. In this study, we evaluated the anti-inflammatory and anti-Alzheimer activity of lupeol in lipopolysaccharide (LPS)-injected mice model. LPS (250ug/kg) was administered intraperitoneally to C57BL/6 N male mice for 1 week to induce neuroinflammation and cognitive impairment. For biochemical analysis, acetylcholinesterase (AChE) assay, western blotting and confocal microscopy were performed. AChE, western blot and immunofluorescence results showed that lupeol treatment (50 mg/kg) along with LPS administration significantly inhibited the LPS-induced activation of neuroinflammatory mediators and cytokines like nuclear factor (NF-κB), tumor necrosis factor (TNF-α), cyclooxygenase (COX-2) and interleukin (IL-1β). Furthermore, we found that LPS-induced systemic inflammation lead to Alzheimer's symptoms as LPS treatment enhances level of amyloid beta (Aβ), amyloid precursor protein (APP), Beta-site APP cleaving enzyme (BACE-1) and hyperphosphorylated Tau (p-Tau). Lupeol treatment reversed the LPS-induced elevated level of Aβ, APP, BACE-1 and p-Tau in the hippocampus, showing anti-Alzheimer's properties. It is also determined that lupeol prevented LPS-induced synaptic dysfunction via enhanced expression of pre-and post-synaptic markers like SNAP-23, synaptophysin and PSD-95. Overall, our study shows that lupeol prevents memory impairment and synaptic dysfunction via inhibition of neuroinflammatory processes. Hence, we suggest that lupeol might be a useful therapeutic agent in prevention of neuroinflammation-induced neurological disorders like AD.
Collapse
Affiliation(s)
- Kyonghwan Choe
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju-si, Republic of Korea
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, Netherlands
| | - Jun Sung Park
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju-si, Republic of Korea
| | - Hyun Young Park
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, Netherlands
- Department of Pediatrics, Maastricht University Medical Center (MUMC+), Maastricht, Netherlands
| | - Muhammad Tahir
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju-si, Republic of Korea
| | - Tae Ju Park
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences (MVLS), University of Glasgow, Glasgow, United Kingdom
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju-si, Republic of Korea
- Alz-Dementia Korea Co., Jinju, Republic of Korea
| |
Collapse
|
6
|
Sakrajda K, Bilska K, Czerski PM, Narożna B, Dmitrzak-Węglarz M, Heilmann-Heimbach S, Brockschmidt FF, Herms S, Nöthen MM, Cichon S, Więckowska B, Rybakowski JK, Pawlak J, Szczepankiewicz A. Abelson Helper Integration Site 1 haplotypes and peripheral blood expression associates with lithium response and immunomodulation in bipolar patients. Psychopharmacology (Berl) 2024; 241:727-738. [PMID: 38036661 DOI: 10.1007/s00213-023-06505-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 11/13/2023] [Indexed: 12/02/2023]
Abstract
RATIONALE In bipolar disorder (BD), immunological factors play a role in the pathogenesis and treatment of the illness. Studies showed the potential link between Abelson Helper Integration Site 1 (AHI1) protein, behavioural changes and innate immunity regulation. An immunomodulatory effect was suggested for lithium, a mood stabilizer used in BD treatment. OBJECTIVES We hypothesized that AHI1 may be an important mediator of lithium treatment response. Our study aimed to investigate whether the AHI1 haplotypes and expression associates with lithium treatment response in BD patients. We also examined whether AHI1 expression and lithium treatment correlate with innate inflammatory response genes. RESULTS We genotyped seven AHI1 single nucleotide polymorphisms in 97 euthymic BD patients and found that TG haplotype (rs7739635, rs9494332) was significantly associated with lithium response. We also showed significantly increased AHI1 expression in the blood of lithium responders compared to non-responders and BD patients compared to healthy controls (HC). We analyzed the expression of genes involved in the innate immune response and inflammatory response regulation (TLR4, CASP4, CASP5, NLRP3, IL1A, IL1B, IL6, IL10, IL18) in 21 lithium-treated BD patients, 20 BD patients treated with other mood stabilizer and 19 HC. We found significantly altered expression between BD patients and HC, but not between BD patients treated with different mood stabilizers. CONCLUSIONS Our study suggests the involvement of AHI1 in the lithium mode of action. Moreover, mood-stabilizing treatment associated with the innate immunity-related gene expression in BD patients and only the lithium-treated BD patients showed significantly elevated expression of anti-inflammatory IL10, suggesting lithium's immunomodulatory potential.
Collapse
Affiliation(s)
- Kosma Sakrajda
- Molecular and Cell Biology Unit, Poznan University of Medical Sciences, Poznan, Poland.
- Doctoral School, Poznan University of Medical Sciences, Poznan, Poland.
| | - Karolina Bilska
- Department of Psychiatric Genetics, Poznan University of Medical Sciences, Poznan, Poland
| | - Piotr M Czerski
- Department of Psychiatric Genetics, Poznan University of Medical Sciences, Poznan, Poland
| | - Beata Narożna
- Molecular and Cell Biology Unit, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Stefanie Heilmann-Heimbach
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | | | - Stefan Herms
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - Sven Cichon
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
| | - Barbara Więckowska
- Department of Computer Sciences and Statistics, Poznan University of Medical Sciences, Poznan, Poland
| | - Janusz K Rybakowski
- Department of Adult Psychiatry, Poznan University of Medical Sciences, Poznan, Poland
| | - Joanna Pawlak
- Department of Psychiatric Genetics, Poznan University of Medical Sciences, Poznan, Poland
| | | |
Collapse
|
7
|
Abdelaty AO, Tharwat EK, Abdelrahman AI, Elgohary A, Elsaeed H, El-Feky AS, Ebrahim YM, Habib A, Abd El Latif H, Khadrawy YA, Aboul Ezz HS, Noor NA, Fahmy HM, Mohammed FF, Radwan NM, Ahmed NA. Cerebrolysin potentiates the antidepressant effect of lithium in a rat model of depression. J Psychiatr Res 2024; 172:171-180. [PMID: 38394763 DOI: 10.1016/j.jpsychires.2024.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 01/09/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024]
Abstract
RATIONALE Depression is the most prevalent psychiatric disorder worldwide. Although numerous antidepressant treatments are available, there is a serious clinical concern due to their severe side effects and the fact that some depressed patients are resistant to them. Lithium is the drug of choice for bipolar depression and has been used as adjunct therapy with other groups of antidepressants. OBJECTIVES The present study aims to investigate the effect of lithium augmentation with cerebrolysin on the neurochemical, behavioral and histopathological alterations induced in the reserpine model of depression. METHODS The animals were divided into control and reserpine-induced model of depression. The model animals were further divided into rat model of depression, rat model treated with lithium, rat model treated with cerebrolysin and rat model treated with a combination of lithium and cerebrolysin. RESULTS Treatment with lithium, cerebrolysin, or their combination alleviated most of the changes in behavior, oxidative stress parameters, acetylcholinesterase and monoamines in the cortex and hippocampus of the reserpine-induced model of depression. It also improved the alterations in brain-derived neurotrophic factor (BDNF) and histopathology induced by reserpine. CONCLUSIONS The augmentation of lithium with cerebrolysin showed a clear beneficial effect in the present model of depression suggesting the use of cerebrolysin as an adjuvant in antidepressant treatment.
Collapse
Affiliation(s)
- Ahmed O Abdelaty
- Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Engy K Tharwat
- Bioinformatics Group Center of Informatics Science, Nile University, Giza, Egypt
| | | | - Ayatallah Elgohary
- School of Biotechnology, Badr University in Cairo, Badr City, Cairo, Egypt
| | | | - Amena S El-Feky
- Biophysics Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Yasmina M Ebrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Abdelaziz Habib
- Biophysics Department, Faculty of Science, Cairo University, Cairo, Egypt
| | | | - Yasser A Khadrawy
- Medical Physiology Department, Medical Division, National Research Center, Egypt
| | - Heba S Aboul Ezz
- Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Neveen A Noor
- Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt.
| | - Heba M Fahmy
- Biophysics Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Faten F Mohammed
- Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt; Department of Pathology, College of Veterinary Medicine, King Faisal University, Al Ahsa, 31982, Saudi Arabia
| | - Nasr M Radwan
- Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Nawal A Ahmed
- Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt
| |
Collapse
|
8
|
Dean B, Scarr E. Common changes in rat cortical gene expression after valproate or lithium treatment particularly affect pre- and post-synaptic pathways that regulate four neurotransmitters systems. World J Biol Psychiatry 2024; 25:54-64. [PMID: 37722808 DOI: 10.1080/15622975.2023.2258972] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/11/2023] [Indexed: 09/20/2023]
Abstract
OBJECTIVES We have postulated that common changes in gene expression after treatment with different therapeutic classes of psychotropic drugs contribute to their common therapeutic mechanisms of action. METHODS To test this hypothesis, we measured levels of cortical coding and non-coding RNA using GeneChip® Rat Exon 1.0 ST Array after treatment with vehicle (chow only), chow containing 1.8 g lithium carbonate/kg (n = 10) or chow containing 12 g sodium valproate/kg (n = 10) for 28 days. Differences in levels of RNA were identified using JMP Genomics 13 and the Panther Gene Ontology Classification System was used to identify potential consequences of RNA. RESULTS Compared to vehicle treatment, levels of cortical RNA for 543 and 583 coding and non-coding RNAs were different after treatment with valproate and lithium, respectively. Moreover, levels of 323 coding and non-coding RNAs were altered in a highly correlated way by treatment with valproate and lithium, changes that would impact on cholinergic, glutamatergic, serotonergic and dopaminergic neurotransmission as well as on voltage gated ion channels. CONCLUSIONS Our study suggests that treating with mood stabilisers cause many common changes in levels of RNA which will impact on CNS function, particularly affecting post-synaptic muscarinic receptor functioning and the release of multiple neurotransmitters.
Collapse
Affiliation(s)
- Brian Dean
- The Molecular Psychiatry Laboratory, The Florey Institute for Neuroscience and Mental Health, Parkville, Australia
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Elizabeth Scarr
- The Department of Psychiatry, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
9
|
Mastella MH, Roggia I, Turra BO, de Afonso Bonotto NC, Teixeira CF, Pulcinelli DLF, Meira GM, Azzolin VF, de Morais-Pinto L, Barbisan F, da Cruz IBM. The Protective Effect of Lithium Against Rotenone may be Evolutionarily Conserved: Evidence from Eisenia fetida, a Primitive Animal with a Ganglionic Brain. Neurochem Res 2023; 48:3538-3559. [PMID: 37526866 DOI: 10.1007/s11064-023-04001-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: 02/20/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 08/02/2023]
Abstract
Chronic exposure to stress is a non-adaptive situation that is associated with mitochondrial dysfunction and the accumulation of reactive oxygen species (ROS), especially superoxide anion (SA). This accumulation of ROS produces damage-associated molecular patterns (DAMPs), which activate chronic inflammatory states and behavioral changes found in several mood disorders. In a previous study, we observed that an imbalance of SA triggered by rotenone (Ro) exposure caused evolutionarily conserved oxi-inflammatory disturbances and behavioral changes in Eisenia fetida earthworms. These results supported our hypothesis that SA imbalance triggered by Ro exposure could be attenuated by lithium carbonate (LC), which has anti-inflammatory properties. The initial protocol exposed earthworms to Ro (30 nM) and four different LC concentrations. LC at a concentration of 12.85 mg/L decreased SA and nitric oxide (NO) levels and was chosen to perform complementary assays: (1) neuromuscular damage evaluated by optical and scanning electron microscopy (SEM), (2) innate immune inefficiency by analysis of Eisenia spp. extracellular neutrophil traps (eNETs), and (3) behavioral changes. Gene expression was also evaluated involving mitochondrial (COII, ND1), inflammatory (EaTLR, AMP), and neuronal transmission (nAchR α5). LC attenuated the high melanized deposits in the circular musculature, fiber disarrangement, destruction of secretory glands, immune inefficiency, and impulsive behavior pattern triggered by Ro exposure. However, the effects of LC and Ro on gene expression were more heterogeneous. In summary, SA imbalance, potentially associated with mitochondrial dysfunction, appears to be an evolutionary component triggering oxidative, inflammatory, and behavioral changes observed in psychiatric disorders that are inhibited by LC exposure.
Collapse
Affiliation(s)
- Moisés Henrique Mastella
- Graduate Program of Pharmacology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil.
- Biogenomics Lab, Health Sciences Center, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Av. Roraima 1000, Building 19, 97105-900, Brazil.
| | - Isabel Roggia
- Biogenomics Lab, Health Sciences Center, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Av. Roraima 1000, Building 19, 97105-900, Brazil
| | - Bárbara Osmarin Turra
- Graduate Program of Pharmacology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
- Biogenomics Lab, Health Sciences Center, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Av. Roraima 1000, Building 19, 97105-900, Brazil
| | - Nathália Cardoso de Afonso Bonotto
- Graduate Program of Pharmacology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
- Biogenomics Lab, Health Sciences Center, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Av. Roraima 1000, Building 19, 97105-900, Brazil
| | - Cibele Ferreira Teixeira
- Graduate Program of Pharmacology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
- Biogenomics Lab, Health Sciences Center, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Av. Roraima 1000, Building 19, 97105-900, Brazil
| | - Débora Luisa Filipetto Pulcinelli
- Biogenomics Lab, Health Sciences Center, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Av. Roraima 1000, Building 19, 97105-900, Brazil
| | - Graziela Moro Meira
- Biogenomics Lab, Health Sciences Center, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Av. Roraima 1000, Building 19, 97105-900, Brazil
| | - Verônica Farina Azzolin
- Center for Research, Teaching and Technological Development (Gerontec/FUnATI), Manaus, Amazonas, Brazil
- Graduate Program of Gerontology, Center for Physical Education and Sports, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Luciano de Morais-Pinto
- Anatomical Design Laboratory, Morphology Department, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Fernanda Barbisan
- Graduate Program of Pharmacology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
- Biogenomics Lab, Health Sciences Center, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Av. Roraima 1000, Building 19, 97105-900, Brazil
- Graduate Program of Gerontology, Center for Physical Education and Sports, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Ivana Beatrice Mânica da Cruz
- Graduate Program of Pharmacology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
- Biogenomics Lab, Health Sciences Center, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Av. Roraima 1000, Building 19, 97105-900, Brazil
- Graduate Program of Gerontology, Center for Physical Education and Sports, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| |
Collapse
|
10
|
Rana AK, Kumar R, Shukla DN, Singh D. Lithium co-administration with rutin improves post-stroke neurological outcomes via suppressing Gsk-3β activity in a rat model. Free Radic Biol Med 2023; 207:107-119. [PMID: 37414348 DOI: 10.1016/j.freeradbiomed.2023.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/24/2023] [Accepted: 07/03/2023] [Indexed: 07/08/2023]
Abstract
Cerebral ischemic stroke is one of the leading causes of adult disability worldwide. Reperfusion is the only therapeutic option with a lot of side effects. In the current study, we investigated the efficacy of rutin and lithium co-treatment in improving post-stroke neurological outcomes in a transient global cerebral ischemia-reperfusion injury rat model. Middle-aged male rats were subjected to transient global cerebral ischemia-reperfusion. NORT and Y-maze were used to assess the cognitive processes. Lipid peroxidation, protein carbonylation, and nitric oxide assays were performed to study oxidative stress. The excitotoxicity index was calculated by HPLC. Real time-PCR and western blotting were performed to study gene and protein expressions. The co-administration of rutin and lithium improved the overall survival, recognition memory, spatial working memory, and neurological score following cerebral ischemia-reperfusion in rats. Further, a marked decrease in malonaldehyde, protein carbonyls, and nitric oxide levels was observed following combined treatment. The mRNA expression of antioxidant (Hmox1 and Nqo1) and pro-inflammatory (Il2, Il6, and Il1β) markers were significantly attenuated in the rutin and lithium co-administrated group. The treatment inhibited the Gsk-3β and maintained a normal pool of the downstream β-catenin and Nrf2 proteins. The results revealed that co-administration of rutin and lithium had a neuroprotective potential, suggesting it to be a viable treatment to overcome post-stroke deaths and neurological complications.
Collapse
Affiliation(s)
- Anil Kumar Rana
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, Himachal Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Rajneesh Kumar
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, Himachal Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Durgesh Nandan Shukla
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, Himachal Pradesh, India
| | - Damanpreet Singh
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, Himachal Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
11
|
Atiq A, Lee HJ, Khan A, Kang MH, Rehman IU, Ahmad R, Tahir M, Ali J, Choe K, Park JS, Kim MO. Vitamin E Analog Trolox Attenuates MPTP-Induced Parkinson's Disease in Mice, Mitigating Oxidative Stress, Neuroinflammation, and Motor Impairment. Int J Mol Sci 2023; 24:9942. [PMID: 37373089 DOI: 10.3390/ijms24129942] [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: 04/28/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Trolox is a potent antioxidant and a water-soluble analog of vitamin E. It has been used in scientific studies to examine oxidative stress and its impact on biological systems. Trolox has been shown to have a neuroprotective effect against ischemia and IL-1β-mediated neurodegeneration. In this study, we investigated the potential protective mechanisms of Trolox against a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinson's disease mouse model. Western blotting, immunofluorescence staining, and ROS/LPO assays were performed to investigate the role of trolox against neuroinflammation, the oxidative stress mediated by MPTP in the Parkinson's disease (PD) mouse model (wild-type mice (C57BL/6N), eight weeks old, average body weight 25-30 g). Our study showed that MPTP increased the expression of α-synuclein, decreased tyrosine hydroxylase (TH) and dopamine transporter (DAT) levels in the striatum and substantia nigra pars compacta (SNpc), and impaired motor function. However, Trolox treatment significantly reversed these PD-like pathologies. Furthermore, Trolox treatment reduced oxidative stress by increasing the expression of nuclear factor erythroid-2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1). Lastly, Trolox treatment inhibited the activated astrocytes (GFAP) and microglia (Iba-1), also reducing phosphorylated nuclear factor-κB, (p-NF-κB) and tumor necrosis factor-alpha (TNF-α) in the PD mouse brain. Overall, our study demonstrated that Trolox may exert neuroprotection on dopaminergic neurons against MPTP-induced oxidative stress, neuroinflammation, motor dysfunction, and neurodegeneration.
Collapse
Affiliation(s)
- Abubakar Atiq
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Hyeon Jin Lee
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Amjad Khan
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Min Hwa Kang
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Inayat Ur Rehman
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Riaz Ahmad
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Muhammad Tahir
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Jawad Ali
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Kyonghwan Choe
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Jun Sung Park
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 FOUR), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea
- Alz-Dementia Korea Co., Jinju 52828, Republic of Korea
| |
Collapse
|
12
|
Tharwat EK, Abdelaty AO, Abdelrahman AI, Elsaeed H, Elgohary A, El-Feky AS, Ebrahim YM, Sakraan A, Ismail HA, Khadrawy YA, Aboul Ezz HS, Noor NA, Fahmy HM, Mohammed HS, Mohammed FF, Radwan NM, Ahmed NA. Evaluation of the therapeutic potential of cerebrolysin and/or lithium in the male Wistar rat model of Parkinson's disease induced by reserpine. Metab Brain Dis 2023; 38:1513-1529. [PMID: 36847968 PMCID: PMC10185619 DOI: 10.1007/s11011-023-01189-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 02/13/2023] [Indexed: 03/01/2023]
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disease worldwide and represents a challenge for clinicians. The present study aims to investigate the effects of cerebrolysin and/or lithium on the behavioral, neurochemical and histopathological alterations induced by reserpine as a model of PD. The rats were divided into control and reserpine-induced PD model groups. The model animals were further divided into four subgroups: rat PD model, rat PD model treated with cerebrolysin, rat PD model treated with lithium and rat PD model treated with a combination of cerebrolysin and lithium. Treatment with cerebrolysin and/or lithium ameliorated most of the alterations in oxidative stress parameters, acetylcholinesterase and monoamines in the striatum and midbrain of reserpine-induced PD model. It also ameliorated the changes in nuclear factor-kappa and improved the histopathological picture induced by reserpine. It could be suggested that cerebrolysin and/or lithium showed promising therapeutic potential against the variations induced in the reserpine model of PD. However, the ameliorating effects of lithium on the neurochemical, histopathological and behavioral alterations induced by reserpine were more prominent than those of cerebrolysin alone or combined with lithium. It can be concluded that the antioxidant and anti-inflammatory effects of both drugs played a significant role in their therapeutic potency.
Collapse
Affiliation(s)
- Engy K Tharwat
- Biotechnology Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Ahmed O Abdelaty
- Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt
| | | | | | - Ayatallah Elgohary
- School of Biotechnology, Badr University in Cairo, Badr City, Cairo, Egypt
| | - Amena S El-Feky
- School of Biotechnology, Badr University in Cairo, Badr City, Cairo, Egypt
| | - Yasmina M Ebrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Alaa Sakraan
- Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Hossam A Ismail
- Biophysics Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Yasser A Khadrawy
- Medical Physiology Department, Medical Division, National Research Center, Dokki, Egypt
| | - Heba S Aboul Ezz
- Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Neveen A Noor
- Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt.
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt.
| | - Heba M Fahmy
- Biophysics Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Haitham S Mohammed
- Biophysics Department, Faculty of Science, Cairo University, Cairo, Egypt
| | | | - Nasr M Radwan
- Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt
| | - Nawal A Ahmed
- Zoology Department, Faculty of Science, Cairo University, Cairo, Egypt
| |
Collapse
|
13
|
Elhabak M, Salama AAA, Salama AH. Nose-to-brain delivery of galantamine loaded nanospray dried polyacrylic acid/taurodeoxycholate mixed matrix as a protective therapy in lipopolysaccharide-induced Alzheimer's in mice model. Int J Pharm 2023; 632:122588. [PMID: 36623740 DOI: 10.1016/j.ijpharm.2023.122588] [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: 08/07/2022] [Revised: 01/02/2023] [Accepted: 01/04/2023] [Indexed: 01/09/2023]
Abstract
One of the promising drug delivery approaches is performed by nanosizing the administered drug product using the nanospray drying technique. In this study, a combination of several formulation factors was integrated and exploited to augment the bioavailability of galantamine hydrobromide (GAL) via the intranasal route. Nanosized polymeric particles were fabricated using the mucoadhesive polymer, polyacrylic acid (PAA), and the permeability booster, sodium taurodeoxycholate (TDC). First, a preliminary study was conducted to adjust the nanospray drying conditions. Then, formulations were prepared on the basis of a mixed factorial experimental design and further analyzed using Design Expert® software. Different responses were investigated: particle size, polydispersity index, spray rate, drying efficiency, and percent yield. The optimized formulation was further assessed for physical morphology using the scanning electron microscope, flowability, in vitro drug release, and in vivo brain cell uptake using confocal laser scanning microscopy. The promising formulation (F6), composed of equal ratio of PAA and TDC and 20 mg GAL, exhibited a particle size of 185.55 ± 4.3 nm, polydispersity index of 0.413 ± 0.02, and yield-value of 69.58 ± 5.82 %. It also displayed good flowability, complete drug release within 2 h, and enhanced in vivo fluorescent dye uptake and penetration in brain cells. The efficacy of the optimized formulation was examined using lipopolysaccharide-induced Alzheimer's in mice. Results revealed the advantageous influence of the optimized formulation (F6) through downregulation of NF-κβ, IL-1β and GFAP as well as upregulating TGF-1β in adult mice.
Collapse
Affiliation(s)
- Mona Elhabak
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ahram Canadian University, 6(th) of October City, Cairo, Egypt.
| | - Abeer A A Salama
- Pharmacology Department, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Alaa H Salama
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ahram Canadian University, 6(th) of October City, Cairo, Egypt; Pharmaceutical Technology Department, National Research Centre, Dokki, Cairo 12622, Egypt
| |
Collapse
|
14
|
Ghanaatfar F, Ghanaatfar A, Isapour P, Farokhi N, Bozorgniahosseini S, Javadi M, Gholami M, Ulloa L, Coleman-Fuller N, Motaghinejad M. Is lithium neuroprotective? An updated mechanistic illustrated review. Fundam Clin Pharmacol 2023; 37:4-30. [PMID: 35996185 DOI: 10.1111/fcp.12826] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 06/17/2022] [Accepted: 08/19/2022] [Indexed: 01/25/2023]
Abstract
Neurodegeneration is a pathological process characterized by progressive neuronal impairment, dysfunction, and loss due to mitochondrial dysfunction, oxidative stress, inflammation, and apoptosis. Many studies have shown that lithium protects against neurodegeneration. Herein, we summarize recent clinical and laboratory studies on the neuroprotective effects of lithium against neurodegeneration and its potential to modulate mitochondrial dysfunction, oxidative stress, inflammation, and apoptosis. Recent findings indicate that lithium regulates critical intracellular pathways such as phosphatidylinositol-3 (PI3)/protein kinase B (Akt)/glycogen synthase kinase-3 (GSK3β) and PI3/Akt/response element-binding protein (CREB)/brain-derived neurotrophic factor (BDNF). We queried PubMed, Web of Science, Scopus, Elsevier, and other related databases using search terms related to lithium and its neuroprotective effect in various neurodegenerative diseases and events from January 2000 to May 2022. We reviewed the major findings and mechanisms proposed for the effects of lithium. Lithium's neuroprotective potential against neural cell degeneration is mediated by inducing anti-inflammatory factors, antioxidant enzymes, and free radical scavengers to prevent mitochondrial dysfunction. Lithium effects are regulated by two essential pathways: PI3/Akt/GSK3β and PI3/Akt/CREB/BDNF. Lithium acts as a neuroprotective agent against neurodegeneration by preventing inflammation, oxidative stress, apoptosis, and mitochondrial dysfunction using PI3/Akt/GSK3β and PI3/Akt/CREB/BDNF signaling pathways.
Collapse
Affiliation(s)
- Fateme Ghanaatfar
- Student Research Committee, School of Nursing and Midwifery, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Ghanaatfar
- Student Research Committee, Qom University of Medical Sciences, Qom, Iran
| | - Parisa Isapour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Negin Farokhi
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University (IUAPS), Tehran, Iran
| | | | - Mahshid Javadi
- Chronic Respiratory Disease Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mina Gholami
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Luis Ulloa
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University, Durham, North Carolina, USA
| | - Natalie Coleman-Fuller
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, 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
| |
Collapse
|
15
|
GSK3β Inhibition by Phosphorylation at Ser 389 Controls Neuroinflammation. Int J Mol Sci 2022; 24:ijms24010337. [PMID: 36613781 PMCID: PMC9820301 DOI: 10.3390/ijms24010337] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
The inhibition of Glycogen Synthase Kinase 3 β (GSK3β) by Ser9 phosphorylation affects many physiological processes, including the immune response. However, the consequences of GSK3β inhibition by alternative Ser389 phosphorylation remain poorly characterized. Here we have examined neuroinflammation in GSK3β Ser389 knock-in (KI) mice, in which the phosphorylation of Ser389 GSK3β is impaired. The number of activated microglia/infiltrated macrophages, astrocytes, and infiltrated neutrophils was significantly higher in these animals compared to C57BL/6J wild-type (WT) counterparts, which suggests that the failure to inactivate GSK3β by Ser389 phosphorylation results in sustained low-grade neuroinflammation. Moreover, glial cell activation and brain infiltration of immune cells in response to lipopolysaccharide (LPS) failed in GSK3β Ser389 KI mice. Such effects were brain-specific, as peripheral immunity was not similarly affected. Additionally, phosphorylation of the IkB kinase complex (IKK) in response to LPS failed in GSK3β Ser389 KI mice, while STAT3 phosphorylation was fully conserved, suggesting that the NF-κB signaling pathway is specifically affected by this GSK3β regulatory pathway. Overall, our findings indicate that GSK3β inactivation by Ser389 phosphorylation controls the brain inflammatory response, raising the need to evaluate its role in the progression of neuroinflammatory pathologies.
Collapse
|
16
|
Shvartsur R, Agam G, Uzzan S, Azab AN. Low-Dose Aspirin Augments the Anti-Inflammatory Effects of Low-Dose Lithium in Lipopolysaccharide-Treated Rats. Pharmaceutics 2022; 14:pharmaceutics14050901. [PMID: 35631487 PMCID: PMC9143757 DOI: 10.3390/pharmaceutics14050901] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/17/2022] [Accepted: 04/19/2022] [Indexed: 12/23/2022] Open
Abstract
Mounting evidence suggests that immune-system dysfunction and inflammation play a role in the pathophysiology and treatment of mood-disorders in general and of bipolar disorder in particular. The current study examined the effects of chronic low-dose aspirin and low-dose lithium (Li) treatment on plasma and brain interleukin-6 and tumor necrosis factor-α production in lipopolysaccharide (LPS)-treated rats. Rats were fed regular or Li-containing food (0.1%) for six weeks. Low-dose aspirin (1 mg/kg) was administered alone or together with Li. On days 21 and 42 rats were injected with 1 mg/kg LPS or saline. Two h later body temperature was measured and rats were sacrificed. Blood samples, the frontal-cortex, hippocampus, and the hypothalamus were extracted. To assess the therapeutic potential of the combined treatment, rats were administered the same Li + aspirin protocol without LPS. We found that the chronic combined treatment attenuated LPS-induced hypothermia and significantly reduced plasma and brain cytokine level elevation, implicating the potential neuroinflammatory diminution purportedly present among the mentally ill. The combined treatment also significantly decreased immobility time and increased struggling time in the forced swim test, suggestive of an antidepressant-like effect. This preclinical evidence provides a potential approach for treating inflammation-related mental illness.
Collapse
Affiliation(s)
- Rachel Shvartsur
- Department of Nursing, School for Community Health Professions, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel;
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel; (G.A.); (S.U.)
| | - Galila Agam
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel; (G.A.); (S.U.)
| | - Sarit Uzzan
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel; (G.A.); (S.U.)
| | - Abed N. Azab
- Department of Nursing, School for Community Health Professions, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel;
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel; (G.A.); (S.U.)
- Correspondence: ; Tel.: +972-86-479880; Fax: +972-86-477-683
| |
Collapse
|
17
|
Kolmogorova D, Ah-Yen EG, Taylor BC, Vaggas T, Liang J, Davis T, Ismail N. Sex-specific responses of the pubertal neuroimmune axis in CD-1 mice. Brain Behav Immun Health 2021; 13:100229. [PMID: 34589744 PMCID: PMC8474685 DOI: 10.1016/j.bbih.2021.100229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 02/18/2021] [Indexed: 12/30/2022] Open
Abstract
The mechanistic relationship between the sexually dimorphic neuroimmune system and the sex-specific outcomes of a pubertal immune challenge is unclear. Therefore, we examined sex differences in the progression of cytotoxic microglial responses and blood-brain barrier (BBB) disruption to a peripubertal lipopolysaccharide (LPS) treatment in brain regions relevant to stress responses and cognitive function. Six-week-old (i.e., stress-sensitive pubertal period) male and female CD-1 mice were treated with LPS (1.5 mg/kg body weight, ip) or 0.9% saline (LPS-matched volume, ip). Sex and treatment differences in microglial (Iba1+) and apoptotic neuronal (caspase-3+/NeuN+) and non-neuronal (caspase-3+/NeuN−) expression were examined in the hippocampus, medial prefrontal cortex (mPFC), and paraventricular nucleus 24 h (sickness), one week (symptomatic recovery) and four weeks (early adulthood) post-treatment (n = 8/group). Microglial morphology was quantified with fractal analyses. Group differences in BBB permeability to 14C-sucrose were examined 24 h (whole-brain, hippocampus, prefrontal cortex, hypothalamus, and cerebellum) and one week (whole-brain) post-treatment. The acute effects of pubertal LPS were specific to females (i.e., global BBB disruption, altered microglial expression and morphology in the mPFC and hippocampus, increased hippocampal apoptosis). The residual effects of pubertal LPS-induced sickness observed in microglia persisted into adulthood in a sex- and region-specific manner. In addition to highlighting these sex-specific responses of the pubertal neuroimmune system, we report baseline region-specific sex differences in microglia spanning puberty through adulthood. We propose that these sex differences in neuroimmune-neurovascular interactions during the stress-sensitive pubertal period create sex biases in stress-related disorders of brain and behaviour. Pubertal LPS alters baseline sex differences in microglial numbers and morphology. Pubertal CD-1 mice mount sexually dimorphic neuroimmune responses to systemic LPS. Treatment effects on microglial expression and morphology differ by sex and region. The acute LPS-induced effects were specific to females.
Collapse
Affiliation(s)
- Daria Kolmogorova
- NISE Laboratory, School of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | - Emily Grace Ah-Yen
- NISE Laboratory, School of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Tiffany Vaggas
- School of Biosciences, Cardiff University, Cardiff, Wales, United Kingdom
| | - Jacky Liang
- NISE Laboratory, School of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | - Tama Davis
- NISE Laboratory, School of Psychology, University of Ottawa, Ottawa, Ontario, Canada
| | - Nafissa Ismail
- NISE Laboratory, School of Psychology, University of Ottawa, Ottawa, Ontario, Canada.,Brain and Mind Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
18
|
Protective effect of methanol leaf extract of Cnidoscolus aconitifolius against lipopolysaccharides-induced cortico-hippocampal neuroinflammation, oxidative stress and memory impairment. ADVANCES IN TRADITIONAL MEDICINE 2021. [DOI: 10.1007/s13596-021-00578-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
19
|
Ali T, Khan A, Alam SI, Ahmad S, Ikram M, Park JS, Lee HJ, Kim MO. Cadmium, an Environmental Contaminant, Exacerbates Alzheimer's Pathology in the Aged Mice's Brain. Front Aging Neurosci 2021; 13:650930. [PMID: 34248598 PMCID: PMC8263901 DOI: 10.3389/fnagi.2021.650930] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/19/2021] [Indexed: 01/06/2023] Open
Abstract
Cadmium (Cd) is an environmental contaminant, which is a potential risk factor in the progression of aging-associated neurodegenerative diseases. Herein, we have assessed the effects of chronic administration of Cd on cellular oxidative stress and its associated Alzheimer's disease (AD) pathologies in animal models. Two groups of mice were used, one group administered with saline and the other with Cd (1 mg/kg/day; intraperitoneally) for 3 months. After behavioral studies, molecular/biochemical (Immunoblotting, ELISAs, ROS, LPO, and GSH assays) and morphological analyses were performed. We observed an exacerbation of memory and synaptic deficits in chronic Cd-injected mice. Subacute and chronic Cd escalated reactive oxygen species (ROS), suppressed the master antioxidant enzymes, e.g., nuclear factor-erythroid 2-related factor 2 and heme oxygenase-1, and evoked the stress kinase phospho-c-Jun N-terminal kinase 1 signaling pathways, which may escalate AD pathologies possibly associated with amyloidogenic processes. These findings suggest the regulation of oxidative stress/ROS and its associated amyloid beta pathologies for targeting the Cd-exacerbated AD pathogenesis. In addition, these preclinical animal studies represent a paradigm for epidemiological studies of the human population exposed to chronic and subacute administration of Cd, suggesting avoiding environmental contaminants.
Collapse
Affiliation(s)
- Tahir Ali
- Division of Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju, South Korea
- Calgary Prion Research Unit, Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, Hotchkiss Brain Institute Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Amjad Khan
- Division of Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju, South Korea
| | - Sayed Ibrar Alam
- Division of Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju, South Korea
| | - Sareer Ahmad
- Division of Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju, South Korea
| | - Muhammad Ikram
- Division of Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju, South Korea
| | - Jun Sung Park
- Division of Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju, South Korea
| | - Hyeon Jin Lee
- Division of Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju, South Korea
| | - Myeong Ok Kim
- Division of Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju, South Korea
| |
Collapse
|
20
|
Rehman IU, Ahmad R, Khan I, Lee HJ, Park J, Ullah R, Choi MJ, Kang HY, Kim MO. Nicotinamide Ameliorates Amyloid Beta-Induced Oxidative Stress-Mediated Neuroinflammation and Neurodegeneration in Adult Mouse Brain. Biomedicines 2021; 9:biomedicines9040408. [PMID: 33920212 PMCID: PMC8070416 DOI: 10.3390/biomedicines9040408] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/22/2021] [Accepted: 04/06/2021] [Indexed: 01/02/2023] Open
Abstract
Alzheimer’s disease (AD) is the most predominant age-related neurodegenerative disease, pathologically characterized by the accumulation of aggregates of amyloid beta Aβ1–42 and tau hyperphosphorylation in the brain. It is considered to be the primary cause of cognitive dysfunction. The aggregation of Aβ1–42 leads to neuronal inflammation and apoptosis. Since vitamins are basic dietary nutrients that organisms need for their growth, survival, and other metabolic functions, in this study, the underlying neuroprotective mechanism of nicotinamide (NAM) Vitamin B3 against Aβ1–42 -induced neurotoxicity was investigated in mouse brains. Intracerebroventricular (i.c.v.) Aβ1–42 injection elicited neuronal dysfunctions that led to memory impairment and neurodegeneration in mouse brains. After 24 h after Aβ1–42 injection, the mice were treated with NAM (250 mg/kg intraperitoneally) for 1 week. For biochemical and Western blot studies, the mice were directly sacrificed, while for confocal and “immunohistochemical staining”, mice were perfused transcardially with 4% paraformaldehyde. Our biochemical, immunofluorescence, and immunohistochemical results showed that NAM can ameliorate neuronal inflammation and apoptosis by reducing oxidative stress through lowering malondialdehyde and 2,7-dichlorofluorescein levels in an Aβ1–42-injected mouse brains, where the regulation of p-JNK further regulated inflammatory marker proteins (TNF-α, IL-1β, transcription factor NF-kB) and apoptotic marker proteins (Bax, caspase 3, PARP1). Furthermore, NAM + Aβ treatment for 1 week increased the amount of survival neurons and reduced neuronal cell death in Nissl staining. We also analyzed memory dysfunction via behavioral studies and the analysis showed that NAM could prevent Aβ1–42 -induced memory deficits. Collectively, the results of this study suggest that NAM may be a potential preventive and therapeutic candidate for Aβ1–42 -induced reactive oxygen species (ROS)-mediated neuroinflammation, neurodegeneration, and neurotoxicity in an adult mouse model.
Collapse
Affiliation(s)
- Inayat Ur Rehman
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (I.U.R.); (R.A.); (I.K.); (H.J.L.); (J.P.); (R.U.)
| | - Riaz Ahmad
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (I.U.R.); (R.A.); (I.K.); (H.J.L.); (J.P.); (R.U.)
| | - Ibrahim Khan
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (I.U.R.); (R.A.); (I.K.); (H.J.L.); (J.P.); (R.U.)
| | - Hyeon Jin Lee
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (I.U.R.); (R.A.); (I.K.); (H.J.L.); (J.P.); (R.U.)
| | - Jungsung Park
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (I.U.R.); (R.A.); (I.K.); (H.J.L.); (J.P.); (R.U.)
| | - Rahat Ullah
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (I.U.R.); (R.A.); (I.K.); (H.J.L.); (J.P.); (R.U.)
| | - Myeong Jun Choi
- Research and Development Center, Axceso Bio-pharma co, Anyang 14056, Korea;
| | - Hee Young Kang
- Department of Neurology, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju 52828, Korea;
| | - Myeong Ok Kim
- Division of Life Sciences and Applied Life Science (BK 21 Four), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (I.U.R.); (R.A.); (I.K.); (H.J.L.); (J.P.); (R.U.)
- Correspondence: ; Tel.: +82-55-772-1345; Fax: +82-55-772-2656
| |
Collapse
|
21
|
Giménez-Palomo A, Dodd S, Anmella G, Carvalho AF, Scaini G, Quevedo J, Pacchiarotti I, Vieta E, Berk M. The Role of Mitochondria in Mood Disorders: From Physiology to Pathophysiology and to Treatment. Front Psychiatry 2021; 12:546801. [PMID: 34295268 PMCID: PMC8291901 DOI: 10.3389/fpsyt.2021.546801] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 05/24/2021] [Indexed: 12/30/2022] Open
Abstract
Mitochondria are cellular organelles involved in several biological processes, especially in energy production. Several studies have found a relationship between mitochondrial dysfunction and mood disorders, such as major depressive disorder and bipolar disorder. Impairments in energy production are found in these disorders together with higher levels of oxidative stress. Recently, many agents capable of enhancing antioxidant defenses or mitochondrial functioning have been studied for the treatment of mood disorders as adjuvant therapy to current pharmacological treatments. A better knowledge of mitochondrial physiology and pathophysiology might allow the identification of new therapeutic targets and the development and study of novel effective therapies to treat these specific mitochondrial impairments. This could be especially beneficial for treatment-resistant patients. In this article, we provide a focused narrative review of the currently available evidence supporting the involvement of mitochondrial dysfunction in mood disorders, the effects of current therapies on mitochondrial functions, and novel targeted therapies acting on mitochondrial pathways that might be useful for the treatment of mood disorders.
Collapse
Affiliation(s)
- Anna Giménez-Palomo
- Bipolar and Depressives Disorders Unit, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Mental Health Research Networking Center (CIBERSAM), Madrid, Spain
| | - Seetal Dodd
- Deakin University, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, VIC, Australia.,Department of Psychiatry, Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Gerard Anmella
- Bipolar and Depressives Disorders Unit, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Mental Health Research Networking Center (CIBERSAM), Madrid, Spain
| | - Andre F Carvalho
- Centre for Addiction and Mental Health, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Giselli Scaini
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Joao Quevedo
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.,Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States.,Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, Brazil.,Center of Excellence in Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Isabella Pacchiarotti
- Bipolar and Depressives Disorders Unit, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Mental Health Research Networking Center (CIBERSAM), Madrid, Spain
| | - Eduard Vieta
- Bipolar and Depressives Disorders Unit, Hospital Clínic, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Mental Health Research Networking Center (CIBERSAM), Madrid, Spain
| | - Michael Berk
- School of Medicine, The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Barwon Health, Geelong, VIC, Australia.,Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, VIC, Australia.,Centre for Youth Mental Health, Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
22
|
Ullah R, Ikram M, Park TJ, Ahmad R, Saeed K, Alam SI, Rehman IU, Khan A, Khan I, Jo MG, Kim MO. Vanillic Acid, a Bioactive Phenolic Compound, Counteracts LPS-Induced Neurotoxicity by Regulating c-Jun N-Terminal Kinase in Mouse Brain. Int J Mol Sci 2020; 22:ijms22010361. [PMID: 33396372 PMCID: PMC7795830 DOI: 10.3390/ijms22010361] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/24/2020] [Accepted: 12/24/2020] [Indexed: 12/15/2022] Open
Abstract
The receptor for advanced glycation end products (RAGE), a pattern recognition receptor signaling event, has been associated with several human illnesses, including neurodegenerative diseases, particularly in Alzheimer’s disease (AD). Vanillic acid (V.A), a flavoring agent, is a benzoic acid derivative having a broad range of biological activities, including antioxidant, anti-inflammatory, and neuroprotective effects. However, the underlying molecular mechanisms of V.A in exerting neuroprotection are not well investigated. The present study aims to explore the neuroprotective effects of V.A against lipopolysaccharides (LPS)-induced neuroinflammation, amyloidogenesis, synaptic/memory dysfunction, and neurodegeneration in mice brain. Behavioral tests and biochemical and immunofluorescence assays were applied. Our results indicated increased expression of RAGE and its downstream phospho-c-Jun n-terminal kinase (p-JNK) in the LPS-alone treated group, which was significantly reduced in the V.A + LPS co-treated group. We also found that systemic administration of LPS-injection induced glial cells (microglia and astrocytes) activation and significantly increased expression level of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-KB) and secretion of proinflammatory cytokines including tumor necrosis factor alpha (TNF-α), interleukin-1 β (IL1-β), and cyclooxygenase (COX-2). However, V.A + LPS co-treatment significantly inhibited the LPS-induced activation of glial cells and neuroinflammatory mediators. Moreover, we also noted that V.A treatment significantly attenuated LPS-induced increases in the expression of AD markers, such as β-site amyloid precursor protein (APP)–cleaving enzyme 1 (BACE1) and amyloid-β (Aβ). Furthermore, V.A treatment significantly reversed LPS-induced synaptic loss via enhancing the expression level of pre- and post-synaptic markers (PSD-95 and SYP), and improved memory performance in LPS-alone treated group. Taken together; we suggest that neuroprotective effects of V.A against LPS-induced neurotoxicity might be via inhibition of LPS/RAGE mediated JNK signaling pathway; and encourage future studies that V.A would be a potential neuroprotective and neurotherapeutic candidate in various neurological disorders.
Collapse
Affiliation(s)
- Rahat Ullah
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Muhammad Ikram
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Tae Ju Park
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences (MVLS), University of Glasgow, Glasgow G12OZD, UK;
| | - Riaz Ahmad
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Kamran Saeed
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Sayed Ibrar Alam
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Inayat Ur Rehman
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Amjad Khan
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Ibrahim Khan
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Min Gi Jo
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Myeong Ok Kim
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
- Correspondence: ; Tel.: +82-55-772-1345; Fax: +82-55-772-2656
| |
Collapse
|
23
|
Shishkina GT, Bannova AV, Komysheva NP, Dygalo NN. Anxiogenic-like effect of chronic lipopolysaccharide is associated with increased expression of matrix metalloproteinase 9 in the rat amygdala. Stress 2020; 23:708-714. [PMID: 32748675 DOI: 10.1080/10253890.2020.1793943] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pathways by which inflammatory stimuli influence behaviors can involve changes in neuronal plasticity, however, the evidence for this is still insufficient. This study aimed to evaluate the effects of chronic lipopolysaccharide (LPS) injected alone or together with tetracycline antibiotic doxycycline (Dox) on the levels of Iba-1, BDNF, Bcl-xL and MMP-9 in brain regions in relation to stress-induced behaviors in the elevated plus-maze (EPM). LPS injected to adult rats every 2 days for a total of 7 injections reduced body weight gain, increased spleen and adrenal weights, decreased locomotor activity, and increased anxiety-like behavior. These effects were associated with increased expression of Iba-1, a well-known marker for activated microglia, in most brain regions investigated. Co-treatment of LPS with Dox attenuated LPS-induced microglial activation and behavioral changes, supporting their relation to the neuroinflammation. LPS administration also produced pro-apoptotic changes in the brain. In the hypothalamus and striatum, the levels of anti-apoptotic protein Bcl-xL were decreased, whereas in the amygdala, a significant increase in MMP-9 protein levels was observed. The levels of Iba-1 as well as MMP-9 in the amygdala positively correlated with the numbers of defecation. The data suggest that mechanisms of anxiety associated with neuroinflammation may involve the increase in MMP-9 levels in the amygdala.
Collapse
Affiliation(s)
- Galina T Shishkina
- Laboratory of Functional Neurogenomics, Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Science, Novosibirsk, Russia
| | - Anita V Bannova
- Laboratory of Functional Neurogenomics, Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Science, Novosibirsk, Russia
| | - Natalya P Komysheva
- Laboratory of Functional Neurogenomics, Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Science, Novosibirsk, Russia
| | - Nikolay N Dygalo
- Laboratory of Functional Neurogenomics, Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Science, Novosibirsk, Russia
| |
Collapse
|
24
|
Zhang Y, Liu H, Chen Z, Yu M, Li J, Dong H, Li N, Ding X, Ge Y, Liu C, Ma T, Gui B. TLR4-mediated hippocampal MMP/TIMP imbalance contributes to the aggravation of perioperative neurocognitive disorder in db/db mice. Neurochem Int 2020; 140:104818. [PMID: 32758588 DOI: 10.1016/j.neuint.2020.104818] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 07/07/2020] [Accepted: 07/19/2020] [Indexed: 01/21/2023]
Abstract
Although type 2 diabetes is an important predictor of perioperative neurocognitive disorder (PND), little is currently known about its mechanism of action. Adult male db/db and db/m mice were subjected to four different treatments, including either sham or tibial fracture surgery as well as intraperitoneal injection of vehicle or TAK-242 (the selective inhibitor of TLR4) at 1, 24, and 48 h after surgery. The fear conditioning test was performed to detect cognitive impairment on post-operative day (POD) 3. The hippocampus was collected on POD 1 for western-blots and on POD 3 for western-blots, transmission electron microscopy, and electrophysiological experiments. Toll-like receptor 4 (TLR4) inhibition reversed more profound decline in the freezing behavior of db/db mice on POD 3. The surgery reduced the slope of hippocampal field excitatory postsynaptic potentials, and induced blood-brain barrier (BBB) damage in db/db mice on POD 3. The surgery also increased protein levels of TLR4, tumor necrosis factor (TNF)-α, interleukin (IL)-1β, albumin, matrix metalloproteinase (MMP)-2, and MMP-9, and decreased protein levels of claudin-5, occludin, tissue inhibitor of matrix metalloproteinase (TIMP)-1, and TIMP-2 in the hippocampus of db/db and db/m mice. These changes were all reversed by TAK-242 treatment. At last, compared with those in post-operative db/m mice, the surgery increased protein levels of TLR4, TNF-α, and IL-1β, decreased protein levels of claudin-5 and occludin, and sustained the MMP/TIMP imbalance in the hippocampus of db/db mice on POD 3. Our results suggest that TLR4-mediated aggravated hippocampal MMP/TIMP imbalance, BBB disruption, sustained inflammatory cytokine release, and impairment of long-term potentiation play a key role in tibial fracture surgery-induced persistent PND in db/db mice.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Anesthesiology and Perioperative Medicine, 1st Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Hailin Liu
- Department of Anesthesiology, Huai'an First People's Hospital, Huai'an, Jiangsu, 223300, China
| | - Zixuan Chen
- Department of Anesthesiology and Perioperative Medicine, 1st Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Min Yu
- Department of Anesthesiology and Perioperative Medicine, 1st Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Jiaxin Li
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Hongquan Dong
- Department of Anesthesiology and Perioperative Medicine, 1st Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Nana Li
- Department of Anesthesiology and Perioperative Medicine, 1st Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Xiahao Ding
- Department of Anesthesiology and Perioperative Medicine, 1st Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Yahe Ge
- Department of Anesthesiology and Perioperative Medicine, 1st Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Cunming Liu
- Department of Anesthesiology and Perioperative Medicine, 1st Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Tengfei Ma
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Bo Gui
- Department of Anesthesiology and Perioperative Medicine, 1st Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| |
Collapse
|
25
|
Fritz M, Klawonn AM, Zhao Q, Sullivan EV, Zahr NM, Pfefferbaum A. Structural and biochemical imaging reveals systemic LPS-induced changes in the rat brain. J Neuroimmunol 2020; 348:577367. [PMID: 32866714 DOI: 10.1016/j.jneuroim.2020.577367] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/11/2022]
Abstract
Despite mounting evidence for the role of inflammation in Major Depressive Disorder (MDD), in vivo preclinical investigations of inflammation-induced negative affect using whole brain imaging modalities are scarce, precluding a valid model within which to evaluate pharmacological interventions. Here we used an E. coli lipopolysaccharide (LPS)-based model of inflammation-induced depressive signs in rats to explore brain changes using multimodal neuroimaging methods. During the acute phase of the LPS response (2 h post injection), prior to the emergence of a task-quantifiable depressive phenotype, striatal glutamine levels and splenial, retrosplenial, and peri-callosal hippocampal cortex volumes were greater than at baseline. LPS-induced depressive behaviors observed at 24 h, however, occurred concurrently with lower than control levels of striatal glutamine and a reversibility of volume expansion (i.e., shrinkage of splenial, retrosplenial, and peri-callosal hippocampal cortex to baseline volumes). In both striatum and hippocampus at 24 h, mRNA expression in LPS relative to control animals demonstrated alterations in enzymes and transporters regulating glutamine homeostasis. Collectively, the observed behavioral, in vivo structural and metabolic, and mRNA expression alterations suggest a critical role for astrocytic regulation of inflammation-induced depressive behaviors.
Collapse
Affiliation(s)
- Michael Fritz
- Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford University, Stanford, CA 94304, United States of America
| | - Anna M Klawonn
- Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford University, Stanford, CA 94304, United States of America
| | - Qingyu Zhao
- Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford University, Stanford, CA 94304, United States of America
| | - Edith V Sullivan
- Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford University, Stanford, CA 94304, United States of America; Neuroscience Program, SRI International, Menlo Park, CA 94025, United States of America
| | - Natalie M Zahr
- Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford University, Stanford, CA 94304, United States of America; Neuroscience Program, SRI International, Menlo Park, CA 94025, United States of America.
| | - Adolf Pfefferbaum
- Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford University, Stanford, CA 94304, United States of America; Neuroscience Program, SRI International, Menlo Park, CA 94025, United States of America
| |
Collapse
|
26
|
da Costa R, Passos GF, Quintão NL, Fernandes ES, Maia JRL, Campos MM, Calixto JB. Taxane-induced neurotoxicity: Pathophysiology and therapeutic perspectives. Br J Pharmacol 2020; 177:3127-3146. [PMID: 32352155 PMCID: PMC7312267 DOI: 10.1111/bph.15086] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/17/2020] [Accepted: 04/25/2020] [Indexed: 12/28/2022] Open
Abstract
Taxane-derived drugs are antineoplastic agents used for the treatment of highly common malignancies. Paclitaxel and docetaxel are the most commonly used taxanes; however, other drugs and formulations have been used, such as cabazitaxel and nab-paclitaxel. Taxane treatment is associated with neurotoxicity, a well-known and relevant side effect, very prevalent amongst patients undergoing chemotherapy. Painful peripheral neuropathy is the most dose-limiting side effect of taxanes, affecting up to 97% of paclitaxel-treated patients. Central neurotoxicity is an emerging side effect of taxanes and it is characterized by cognitive impairment and encephalopathy. Besides impairing compliance to chemotherapy treatment, taxane-induced neurotoxicity (TIN) can adversely affect the patient's life quality on a long-term basis. Despite the clinical relevance, not many reviews have comprehensively addressed taxane-induced neurotoxicity when they are used therapeutically. This article provides an up-to-date review on the pathophysiology of TIN and the novel potential therapies to prevent or treat this side effect.
Collapse
Affiliation(s)
- Robson da Costa
- Faculdade de FarmáciaUniversidade Federal do Rio de JaneiroRio de JaneiroRJBrazil
| | - Giselle F. Passos
- Faculdade de FarmáciaUniversidade Federal do Rio de JaneiroRio de JaneiroRJBrazil
| | - Nara L.M. Quintão
- Programa de Pós‐graduação em Ciências FarmacêuticasUniversidade do Vale do ItajaíItajaíSCBrazil
| | - Elizabeth S. Fernandes
- Instituto Pelé Pequeno PríncipeCuritibaPRBrazil
- Programa de Pós‐graduação em Biotecnologia Aplicada à Saúde da Criança e do AdolescenteFaculdades Pequeno PríncipeCuritibaPRBrazil
| | | | - Maria Martha Campos
- Escola de Ciências da Saúde e da VidaPontifícia Universidade Católica do Rio Grande do SulPorto AlegreRSBrazil
| | - João B. Calixto
- Centro de Inovação e Ensaios Pré‐clínicos ‐ CIEnPFlorianópolisSCBrazil
| |
Collapse
|
27
|
Kongsui R, Sriraksa N, Thongrong S. The Neuroprotective Effect of Zingiber cassumunar Roxb. Extract on LPS-Induced Neuronal Cell Loss and Astroglial Activation within the Hippocampus. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4259316. [PMID: 32596307 PMCID: PMC7273477 DOI: 10.1155/2020/4259316] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 05/10/2020] [Accepted: 05/15/2020] [Indexed: 12/23/2022]
Abstract
The systemic administration of lipopolysaccharide (LPS) has been recognized to induce neuroinflammation which plays a significant role in the pathogenesis of neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. In this study, we aimed to determine the protective effect of Zingiber cassumunar (Z. cassumunar) or Phlai (in Thai) against LPS-induced neuronal cell loss and the upregulation of glial fibrillary acidic protein (GFAP) of astrocytes in the hippocampus. Adult male Wistar rats were orally administered with Z. cassumunar extract at various doses (50, 100, and 200 mg/kg body weight) for 14 days before a single injection of LPS (250 μg/kg/i.p.). The results indicated that LPS-treated animals exhibited neuronal cell loss and the activation of astrocytes and also increased proinflammatory cytokine interleukin- (IL-) 1β in the hippocampus. Pretreatment with Z. cassumunar markedly reduced neuronal cell loss in the hippocampus. In addition, Z. cassumunar extract at a dose of 200 mg/kg BW significantly suppressed the inflammatory response by reducing the expression of GFAP and IL-1ß in the hippocampus. Therefore, the results suggested that Z. cassumunar extract might be valuable as a neuroprotective agent in neuroinflammation-induced brain damage. However, further investigations are essential to validate the possible active ingredients and mechanisms of its neuroprotective effect.
Collapse
Affiliation(s)
- Ratchaniporn Kongsui
- Division of Physiology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Napatr Sriraksa
- Division of Physiology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Sitthisak Thongrong
- Division of Anatomy, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
| |
Collapse
|
28
|
Gao Y, Cui M, Zhong S, Feng C, Nwobodo AK, Chen B, Song Y, Wang Y. Dihydroartemisinin ameliorates LPS-induced neuroinflammation by inhibiting the PI3K/AKT pathway. Metab Brain Dis 2020; 35:661-672. [PMID: 32152798 DOI: 10.1007/s11011-020-00533-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 01/17/2020] [Indexed: 11/30/2022]
Abstract
Neuroinflammation can cause multiple neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD). Recent studies have shown that the artemisinin derivative dihydroartemisinin (DHA) can be used as an immunomodulatory, anti-inflammatory and anti-tumor agent. The anti-neuroinflammatory effects of DHA were evaluated in our study, and the underlying mechanisms were explored using the Morris water maze test (MWMT), Open-field test (OFT) and Closed-field test (CFT), Elevated plus maze test (EPMT), Nissl Staining, Immunofluorescence analysis, RT-PCR, and Western Blot. Our results show that DHA significantly inhibits LPS-induced inflammation and attenuates LPS-induced behavioral and memory disorders. 1. Behavioral test results: 1) in the water maze test, the mice in the LPS group showed increased escape latency and length of the movement path on the third day; they also had a decreased number of crossings of the target quadrant after the platform was removed on the 5th day and remained in the target quadrant for less time; 2) in the open- and closed-field experiment, the number of activities and activities in the open-field were significantly reduced; 3) in the elevated cross maze experiment, LPS-treated mice exhibited a significant reduction in the number of times and the time to enter the open arm; the above behavior was reversed after DHA treatment. 2. Nissl staining results: compared with the Control group, the LPS group showed significant damage, and the number of damaged cells in the hippocampal CA1, CA2, CA3 and DG regions was increased; DHA treatment reduced cell damage. 3. RT-PCR results: compared with the Control group, the LPS group showed increased expression of IL-1β and IL-6 but decreased expression after DHA treatment. 4. GFAP fluorescent staining: compared with the control group, the corresponding reactivity of positive cells in the LPS-induced group was increased in the CA1-CA3 and DG regions of the hippocampus; compared with the LPS-induced mice, cells in the LPS + DHA group showed significantly reduced reactivity (GFAP). 5. Western blot results: compared with the Control group, the LPS group showed increased expression of P-PI3K/PI3K, P-AKT/AKT, IL-6 and TNFα and a decreased expression of P-PI3K/PI3K, IL-6, TNF and P-AKT/AKT after DHA treatment. Our findings provide direct evidence for the potential use of DHA in the treatment of neuroinflammatory diseases.
Collapse
Affiliation(s)
- Yuting Gao
- Medical Technology School, Xuzhou Medical University, Xuzhou, 221004, China
| | - Miaomiao Cui
- Department of Genetics, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Sijin Zhong
- Department of Clinical, Xuzhou Medical University, Xuzhou, China
| | - Chenyao Feng
- Department of Clinical, Xuzhou Medical University, Xuzhou, China
| | - Alexander Kenechukwu Nwobodo
- Department of Genetics, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Bin Chen
- Department of Rehabilitation, The Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, 350004, Fujian Province, China
| | - Yuanjian Song
- Department of Genetics, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, People's Republic of China.
| | - Yulan Wang
- Department of Human Anatomy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, People's Republic of China.
| |
Collapse
|
29
|
Song Y, Qin L, Yang R, Yang F, Kenechukwu NA, Zhao X, Zhou X, Wen X, Li L. Inhibition of HDAC6 alleviating lipopolysaccharide-induced p38MAPK phosphorylation and neuroinflammation in mice. PHARMACEUTICAL BIOLOGY 2019; 57:263-268. [PMID: 31124385 PMCID: PMC8871618 DOI: 10.1080/13880209.2018.1563620] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Context: Researchers in a variety of fields have extensively focused on histone deacetylase 6 (HDAC6) due to its aggravation of inflammatory reaction. However, relevant studies examining whether HDAC6 could exacerbate lipopolysaccharide (LPS)-induced inflammation are still lacking. Objective: We assessed the role of HDAC6 in LPS-induced brain inflammation and used the HDAC6-selective inhibitor Tubastatin A (TBSA) to investigate the potential mechanisms further. Materials and methods: Brain inflammation was induced in Kunming (KM) mice via intraperitoneal (I.P.), injection of Lipopolysaccharide (LPS) (1 mg/kg), the TBSA (0.5 mg/kg) was delivered via intraperitoneal. The phosphorylated p38 (p-p38) Mitogen-activated protein kinases (MAPK) and expression of typical inflammatory mediators, including tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in both the hippocampus and cortex, were examined by immunoblotting. Nissl staining was used to detect the neuronal damage in the hippocampus and the cortex. Results: About 1 mg/kg LPS via daily intraperitoneal (I.P.) injections for 12 days significantly increased p38 MAPK phosphorylation, TNF-α and IL-6 expression, and neuronal loss. However, 0.5 mg/kg TBSA (three days before LPS treatment) by I.P. injections for 15 days could reverse the above results. Conclusions: This present study provided evidence that TBSA significantly suppressed LPS-induced neuroinflammation and the expression of p-p38. Results derived from our study might help reveal the effective targeting strategies of LPS-induced brain inflammation through inhibiting HDAC6.
Collapse
Affiliation(s)
- Yuanjian Song
- Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu, PR China
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu, PR China
- Department of Geriatrics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Li Qin
- Department of Geriatrics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Rongli Yang
- Department of Geriatrics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Fan Yang
- Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu, PR China
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Nwobodo Alexander Kenechukwu
- Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu, PR China
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Xiaofang Zhao
- Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Xiaoyan Zhou
- Laboratory of Morphology, Xuzhou Medical University, Xuzhou, Jiangsu, PR China
- CONTACT Xiaoyan Zhou Xuzhou Medical University Xuzhou, 209 Tongshan Road, Jiangsu, 221004, PR China
| | - Xiangru Wen
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu, PR China
- Xiangru Wen Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu221004, PR China
| | - Lei Li
- Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu, PR China
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, Jiangsu, PR China
- Department of Geriatrics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
- Lei Li Department of Genetics; Department of Geriatrics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| |
Collapse
|
30
|
Samad N, Imran I, Zulfiqar I, Bilal K. Ameliorative effect of lithium chloride against d-galactose induced behavioral and memory impairment, oxidative stress and alteration in serotonin function in rats. Pharmacol Rep 2019; 71:909-916. [PMID: 31426009 DOI: 10.1016/j.pharep.2019.04.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 04/13/2019] [Accepted: 04/29/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Aging is a phenomenon that all living organisms surely face. d-galactose (D-gal) has been used to develop an aging model of brain. Lithium (Li) has been proposed to have neuroprotective properties in relation to several neurological disorders. The goal of the current studyis to evaluate the effect of Lithium Chloride (LiCl) on D-gal induced neurological disorders and oxidative stress. METHODS Rats were treated with D-gal at a dose of 300 mg/ml/kg and various doses of LiCl (20, 40 and 80 mg/ml/kg) for 14 days. After that behavioral analysis (Elevated plus maze (EPM); Light dark box test (LDT); Morris water maze (MWM); Forced swim test (FST)) were performed. Animals were decapitated after behavioral tests and brain samples were collected for biochemical (malondialdehyde (MDA); superoxide dismutase (SOD); catalase (CAT); glutathione peroxidase (GPx); acetylcholiesterase (AChE)) and neurochemical analysis (5-hydroxytryptamine (5-HT) and 5-hydroxyindoleacetic acid (5-HIAA)). RESULTS The results showed that administration of LiCl at all doses ameliorates D-gal induced, decreased time spent in the open arm and light box in EPM and LDT respectively, increased immobility in FST, increased latency escape in MWM, increased MDA levels, decreased antioxidant enzyme, increased AChE activity and decreased 5-HT metabolism. CONCLUSIONS In conclusion, the present study indicated that D-gal induced anxiety/depression like symptoms and memory impairment were ameliorated by LiCl (at all doses) possibly via its antioxidant effects and normalizing 5-HT function.
Collapse
Affiliation(s)
- Noreen Samad
- Department of Biochemistry, Faculty of Science, Bahauddin Zakariya University, Multan, Pakistan.
| | - Imran Imran
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Iqra Zulfiqar
- Department of Biochemistry, Faculty of Science, Bahauddin Zakariya University, Multan, Pakistan
| | - Kainat Bilal
- Department of Biochemistry, Faculty of Science, Bahauddin Zakariya University, Multan, Pakistan
| |
Collapse
|
31
|
Robertson OD, Coronado NG, Sethi R, Berk M, Dodd S. Putative neuroprotective pharmacotherapies to target the staged progression of mental illness. Early Interv Psychiatry 2019; 13:1032-1049. [PMID: 30690898 DOI: 10.1111/eip.12775] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 12/26/2018] [Indexed: 12/22/2022]
Abstract
AIM Neuropsychiatric disorders including depression, bipolar and schizophrenia frequently exhibit a neuroprogressive course from prodrome to chronicity. There are a range of agents exhibiting capacity to attenuate biological mechanisms associated with neuroprogression. This review will update the evidence for putative neuroprotective agents including clinical efficacy, mechanisms of action and limitations in current assessment tools, and identify novel agents with neuroprotective potential. METHOD Data for this review were sourced from online databases PUBMED, Embase and Web of Science. Only data published since 2012 were included in this review, no data were excluded based on language or publication origin. RESULTS Each of the agents reviewed inhibit one or multiple pathways of neuroprogression including: inflammatory gene expression and cytokine release, oxidative and nitrosative stress, mitochondrial dysfunction, neurotrophin dysregulation and apoptotic signalling. Some demonstrate clinical efficacy in preventing neural damage or loss, relapse or cognitive/functional decline. Agents include: the psychotropic medications lithium, second generation antipsychotics and antidepressants; other pharmacological agents such as minocycline, aspirin, cyclooxygenase-2 inhibitors, statins, ketamine and alpha-2-delta ligands; and others such as erythropoietin, oestrogen, leptin, N-acetylcysteine, curcumin, melatonin and ebselen. CONCLUSIONS Signals of evidence of clinical neuroprotection are evident for a number of candidate agents. Adjunctive use of multiple agents may present a viable avenue to clinical realization of neuroprotection. Definitive prospective studies of neuroprotection with multimodal assessment tools are required.
Collapse
Affiliation(s)
- Oliver D Robertson
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Victoria, Australia.,Mental Health, Drugs and Alcohol Services, University Hospital Geelong, Barwon Health, Geelong, Victoria, Australia
| | - Nieves G Coronado
- Unidad de Gestión Clinica Salud Mental, Hospital Universitario Virgen del Rocio, Sevilla, Spain
| | - Rickinder Sethi
- Department of Psychiatry, Western University, London, Ontario, Canada
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Victoria, Australia.,Mental Health, Drugs and Alcohol Services, University Hospital Geelong, Barwon Health, Geelong, Victoria, Australia.,Department of Psychiatry, The University of Melbourne, Parkville, Victoria, Australia.,Mood Disorders Research Program, Orygen, the National Centre of Excellence in Youth Mental Health, Parkville, Victoria, Australia.,Department of Psychiatry, Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Seetal Dodd
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Victoria, Australia.,Mental Health, Drugs and Alcohol Services, University Hospital Geelong, Barwon Health, Geelong, Victoria, Australia.,Department of Psychiatry, The University of Melbourne, Parkville, Victoria, Australia.,Mood Disorders Research Program, Orygen, the National Centre of Excellence in Youth Mental Health, Parkville, Victoria, Australia
| |
Collapse
|
32
|
Yang C, Wang W, Zhu K, Liu W, Luo Y, Yuan X, Wang J, Cheng T, Zhang X. Lithium chloride with immunomodulatory function for regulating titanium nanoparticle-stimulated inflammatory response and accelerating osteogenesis through suppression of MAPK signaling pathway. Int J Nanomedicine 2019; 14:7475-7488. [PMID: 31571859 PMCID: PMC6750619 DOI: 10.2147/ijn.s210834] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 08/30/2019] [Indexed: 12/26/2022] Open
Abstract
Background Wear particle-induced inflammatory osteolysis and the consequent aseptic loosening constitute the leading reasons for prosthesis failure and revision surgery. Several studies have demonstrated that the macrophage polarization state and immune response play critical roles in periprosthetic osteolysis and tissue repair, but the immunomodulatory role of lithium chloride (LiCl), which has a protective effect on wear particle-induced osteolysis by suppressing osteoclasts and attenuating inflammatory responses, has never been investigated. Methods In this work, the immunomodulatory capability of LiCl on titanium (Ti) nanoparticle-stimulated transformation of macrophage phenotypes and the subsequent effect on osteogenic differentiation were investigated. We first speculated that LiCl attenuated Ti nanoparticle-stimulated inflammation responses by driving macrophage polarization and generating an immune micro-environment to improve osteogenesis. Furthermore, a metal nanoparticle-stimulated murine air pouch inflammatory model was applied to confirm this protective effect in vivo. Results The results revealed that metal nanoparticles significantly activate M1 phenotype (proinflammatory macrophage) expression and increase proinflammatory cytokines secretions in vitro and in vivo, whereas LiCl drives macrophages to the M2 phenotype (anti-inflammatory macrophage) and increases the release of anti-inflammatory and bone-related cytokines. This improved the osteogenic differentiation capability of rat bone marrow mesenchymal stem cells (rBMSCs). In addition, we also provided evidence that LiCl inhibits the phosphorylation of the p38 mitogen-activated protein kinase (p38) and extracellular signal-regulated kinase (ERK) pathways in wear particle-treated macrophages. Conclusion LiCl has the immunomodulatory effects to alleviate Ti nanoparticle-mediated inflammatory reactions and enhance the osteogenic differentiation of rBMSCs by driving macrophage polarization. Thus, LiCl may be an effective therapeutic alternative for preventing and treating wear debris-induced inflammatory osteolysis.
Collapse
Affiliation(s)
- Chao Yang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Wei Wang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Kechao Zhu
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Wei Liu
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Yao Luo
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Xiangwei Yuan
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Jiaxing Wang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Tao Cheng
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Xianlong Zhang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| |
Collapse
|
33
|
Rivera AD, Butt AM. Astrocytes are direct cellular targets of lithium treatment: novel roles for lysyl oxidase and peroxisome-proliferator activated receptor-γ as astroglial targets of lithium. Transl Psychiatry 2019; 9:211. [PMID: 31477687 PMCID: PMC6718419 DOI: 10.1038/s41398-019-0542-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/11/2019] [Accepted: 07/07/2019] [Indexed: 12/26/2022] Open
Abstract
Astrocytes are multifunctional glial cells that play essential roles in supporting synaptic signalling and white matter-associated connectivity. There is increasing evidence that astrocyte dysfunction is involved in several brain disorders, including bipolar disorder (BD), depression and schizophrenia. The mood stabiliser lithium is a frontline treatment for BD, but the mechanisms of action remain unclear. Here, we demonstrate that astrocytes are direct targets of lithium and identify unique astroglial transcriptional networks that regulate specific molecular changes in astrocytes associated with BD and schizophrenia, together with Alzheimer's disease (AD). Using pharmacogenomic analyses, we identified novel roles for the extracellular matrix (ECM) regulatory enzyme lysyl oxidase (LOX) and peroxisome proliferator-activated receptor gamma (PPAR-γ) as profound regulators of astrocyte morphogenesis. This study unravels new pathophysiological mechanisms in astrocytes that have potential as novel biomarkers and potential therapeutic targets for regulating astroglial responses in diverse neurological disorders.
Collapse
Affiliation(s)
- Andrea D. Rivera
- 0000 0001 0728 6636grid.4701.2Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Science, University of Portsmouth, St Michael’s Building, White Swan Road, Portsmouth, PO1 2DT UK
| | - Arthur M. Butt
- 0000 0001 0728 6636grid.4701.2Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Science, University of Portsmouth, St Michael’s Building, White Swan Road, Portsmouth, PO1 2DT UK
| |
Collapse
|
34
|
Ahmad A, Ali T, Rehman SU, Kim MO. Phytomedicine-Based Potent Antioxidant, Fisetin Protects CNS-Insult LPS-Induced Oxidative Stress-Mediated Neurodegeneration and Memory Impairment. J Clin Med 2019; 8:E850. [PMID: 31207963 PMCID: PMC6616651 DOI: 10.3390/jcm8060850] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/05/2019] [Accepted: 06/11/2019] [Indexed: 12/14/2022] Open
Abstract
Phytomedicine based natural flavonoids have potent antioxidant, anti-inflammatory, and neuroprotective activities against neurodegenerative diseases. The aim of the present study is to investigate the potent neuroprotective and antioxidant potential effects of fisetin (natural flavonoid) against central nervous system (CNS)-insult, lipopolysaccharide (LPS)-induced reactive oxygen species (ROS), neuroinflammation, neurodegeneration, and synaptic/memory deficits in adult mice. The mice were injected intraperitoneally (i.p.) with LPS (250 μg/kg/day for 1 week) and a fisetin dosage regimen (20 mg/kg/day i.p. for 2 weeks, 1 week pre-treated to LPS and 1 week co-treated with LPS). Behavioral tests, and biochemical and immunofluorescence assays were applied. Our results revealed that fisetin markedly abrogated the LPS-induced elevated ROS/oxidative stress and activated phosphorylated c-JUN N-terminal Kinase (p-JNK) in the adult mouse hippocampus. Fisetin significantly alleviated LPS-induced activated gliosis. Moreover, fisetin treatment inhibited LPS-induced activation of the inflammatory Toll-like Receptors (TLR4)/cluster of differentiation 14 (CD14)/phospho-nuclear factor kappa (NF-κB) signaling and attenuated other inflammatory mediators (tumor necrosis factor-α (TNF-α), interleukin-1 β (IL1-β), and cyclooxygenase (COX-2). Furthermore, immunoblotting and immunohistochemical results revealed that fisetin significantly reversed LPS-induced apoptotic neurodegeneration. Fisetin improved the hippocampal-dependent synaptic and memory functions in LPS-treated adult mice. In summary, our results strongly recommend that fisetin, a natural potent antioxidant, and neuroprotective phytomedicine, represents a promising, valuable, and therapeutic candidate for the prevention and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Ashfaq Ahmad
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea.
| | - Tahir Ali
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea.
| | - Shafiq Ur Rehman
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea.
| | - Myeong Ok Kim
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea.
| |
Collapse
|
35
|
Caffeine Modulates Cadmium-Induced Oxidative Stress, Neuroinflammation, and Cognitive Impairments by Regulating Nrf-2/HO-1 In Vivo and In Vitro. J Clin Med 2019; 8:jcm8050680. [PMID: 31091792 PMCID: PMC6572702 DOI: 10.3390/jcm8050680] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/10/2019] [Accepted: 05/12/2019] [Indexed: 02/06/2023] Open
Abstract
Cadmium (Cd), a nonbiodegradable heavy metal and one of the most neurotoxic environmental and industrial pollutants, promotes disturbances in major organs and tissues following both acute and chronic exposure. In this study, we assessed the neuroprotective potential of caffeine (30 mg/kg) against Cd (5 mg/kg)-induced oxidative stress-mediated neuroinflammation, neuronal apoptosis, and cognitive deficits in male C57BL/6N mice in vivo and in HT-22 and BV-2 cell lines in vitro. Interestingly, our findings indicate that caffeine markedly reduced reactive oxygen species (ROS) and lipid peroxidation (LPO) levels and enhanced the expression of nuclear factor-2 erythroid-2 (Nrf-2) and hemeoxygenase-1 (HO-1), which act as endogenous antioxidant regulators. Also, 8-dihydro-8-oxoguanine (8-OXO-G) expression was considerably reduced in the caffeine-treated group as compared to the Cd-treated group. Similarly, caffeine ameliorated Cd-mediated glial activation by reducing the expression of glial fibrillary acidic protein (GFAP), ionized calcium-binding adapter molecule 1 (Iba-1), and other inflammatory mediators in the cortical and hippocampal regions of the mouse brain. Moreover, caffeine markedly attenuated Cd-induced neuronal loss, synaptic dysfunction, and learning and cognitive deficits. Of note, nuclear factor-2 erythroid-2 (Nrf-2) gene silencing and nuclear factor-κB (NF-κB) inhibition studies revealed that caffeine exerted neuroprotection via regulation of Nrf-2- and NF-κB-dependent mechanisms in the HT-22 and BV-2 cell lines, respectively. On the whole, these findings reveal that caffeine rescues Cd-induced oxidative stress-mediated neuroinflammation, neurodegeneration, and memory impairment. The present study suggests that caffeine might be a potential antioxidant and neuroprotective agent against Cd-induced neurodegeneration.
Collapse
|
36
|
Mohammadi G, Dargahi L, Naserpour T, Mirzanejad Y, Alizadeh SA, Peymani A, Nassiri-Asl M. Probiotic mixture of Lactobacillus helveticus R0052 and Bifidobacterium longum R0175 attenuates hippocampal apoptosis induced by lipopolysaccharide in rats. Int Microbiol 2018; 22:317-323. [PMID: 30810993 DOI: 10.1007/s10123-018-00051-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 11/11/2018] [Accepted: 11/29/2018] [Indexed: 02/07/2023]
Abstract
In recent years, the beneficial impact of targeted gut microbiota manipulation in various neurological disorders has become more evident. Therefore, probiotics have been considered as a promising approach to modulate brain gene expression and neuronal pathways even in some neurodegenerative diseases. The purpose of this study was to determine the effect of probiotic biotherapy with combination of Lactobacillus helveticus R0052 and Bifidobacterium longum R0175 on the expression levels of proteins critical to neuronal apoptosis in hippocampus of lipopolysaccharide (LPS)-exposed rats. Four groups of animals (Control, LPS, Probiotic + LPS, and Probiotic) were treated with maltodextrin (placebo) or probiotic (109 CFU/ml/rat) for 2 weeks by gavage. On the 15th day, a single intraperitoneal dose of saline or LPS (1 mg/kg) was injected and 4 h later, protein assessment was performed by western blotting in hippocampal tissues. LPS significantly increased the Bax, Bax/Bcl-2 ratio, and cleaved caspase-3 expression along with decreased the Bcl-2 and procaspase-3 protein levels. However, probiotic pretreatment (L. helveticus R0052 + B. longum R0175) significantly downregulated the Bax and Bax/Bcl-2 ratio accompanied with upregulated Bcl-2 expression. Prophylactic treatment with these bacteria also attenuated LPS-induced caspase-3 activation by remarkably increasing the expression of procaspase-3 while reducing the level of cleaved caspase-3 in target tissues. Our data indicate that probiotic formulation (L. helveticus R0052 + B. longum R0175) alleviated hippocampal apoptosis induced by LPS in rats via the gut-brain axis and suggest that this probiotic could play a beneficial role in some neurodegenerative conditions.
Collapse
Affiliation(s)
- Ghazaleh Mohammadi
- Cellular and Molecular Research Center, Department of Molecular Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Leila Dargahi
- NeuroBiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Taghi Naserpour
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Yazdan Mirzanejad
- Division of Infectious Diseases, University of British Columbia, Vancouver, Canada
| | - Safar Ali Alizadeh
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Amir Peymani
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran.
| | - Marjan Nassiri-Asl
- Cellular and Molecular Research Center, Department of Pharmacology, Qazvin University of Medical Sciences, P.O. Box 341197-5981, Qazvin, Iran.
| |
Collapse
|
37
|
Khan A, Ali T, Rehman SU, Khan MS, Alam SI, Ikram M, Muhammad T, Saeed K, Badshah H, Kim MO. Neuroprotective Effect of Quercetin Against the Detrimental Effects of LPS in the Adult Mouse Brain. Front Pharmacol 2018; 9:1383. [PMID: 30618732 PMCID: PMC6297180 DOI: 10.3389/fphar.2018.01383] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 11/09/2018] [Indexed: 12/19/2022] Open
Abstract
Chronic neuroinflammation is responsible for multiple neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. Lipopolysaccharide (LPS) is an essential component of the gram-negative bacterial cell wall and acts as a potent stimulator of neuroinflammation that mediates neurodegeneration. Quercetin is a natural flavonoid that is abundantly found in fruits and vegetables and has been shown to possess multiple forms of desirable biological activity including anti-inflammatory and antioxidant properties. This study aimed to evaluate the neuroprotective effect of quercetin against the detrimental effects of LPS, such as neuroinflammation-mediated neurodegeneration and synaptic/memory dysfunction, in adult mice. LPS [0.25 mg/kg/day, intraperitoneally (I.P.) injections for 1 week]-induced glial activation causes the secretion of cytokines/chemokines and other inflammatory mediators, which further activate the mitochondrial apoptotic pathway and neuronal degeneration. Compared to LPS alone, quercetin (30 mg/kg/day, I.P.) for 2 weeks (1 week prior to the LPS and 1 week cotreated with LPS) significantly reduced activated gliosis and various inflammatory markers and prevented neuroinflammation in the cortex and hippocampus of adult mice. Furthermore, quercetin rescued the mitochondrial apoptotic pathway and neuronal degeneration by regulating Bax/Bcl2, and decreasing activated cytochrome c, caspase-3 activity and cleaving PARP-1 in the cortical and hippocampal regions of the mouse brain. The quercetin treatment significantly reversed the LPS-induced synaptic loss in the cortex and hippocampus of the adult mouse brain and improved the memory performance of the LPS-treated mice. In summary, our results demonstrate that natural flavonoids such as quercetin can be beneficial against LPS-induced neurotoxicity in adult mice.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Myeong Ok Kim
- Division of Applied Life Science (BK 21), College of Natural Science, Gyeongsang National University, Jinju, South Korea
| |
Collapse
|
38
|
Melatonin Rescue Oxidative Stress-Mediated Neuroinflammation/ Neurodegeneration and Memory Impairment in Scopolamine-Induced Amnesia Mice Model. J Neuroimmune Pharmacol 2018; 14:278-294. [DOI: 10.1007/s11481-018-9824-3] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/15/2018] [Indexed: 01/02/2023]
|
39
|
Anthocyanins Improve Hippocampus-Dependent Memory Function and Prevent Neurodegeneration via JNK/Akt/GSK3β Signaling in LPS-Treated Adult Mice. Mol Neurobiol 2018; 56:671-687. [PMID: 29779175 DOI: 10.1007/s12035-018-1101-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/29/2018] [Indexed: 10/16/2022]
Abstract
Microglia plays a critical role in the brain and protects neuronal cells from toxins. However, over-activation of microglia leads to deleterious effects. Lipopolysaccharide (LPS) has been reported to affect neuronal cells via activation of microglia as well as directly to initiate neuroinflammation. In the present study, we evaluated the anti-inflammatory and anti-oxidative effect of anthocyanins against LPS-induced neurotoxicity in an animal model and in cell cultures. Intraperitoneal injections of LPS (250 μg/kg/day for 1 week) induce ROS production and promote neuroinflammation and neurodegeneration which ultimately leads to memory impairment. However, anthocyanins treatment at a dose of 24 mg/kg/day for 2 weeks (1 week before and 1 week co-treated with LPS) prevented ROS production, inhibited neuroinflammation and neurodegeneration, and improved memory functions in LPS-treated mice. Both histological and immunoblot analysis indicated that anthocyanins reversed the activation of JNK, prevented neuroinflammation by lowering the levels of inflammatory markers (p-NF-kB, TNF-α, and IL-1β), and reduced neuronal apoptosis by reducing the expression of Bax, cytochrome c, cleaved caspase-3, and cleaved PARP-1, while increasing the level of survival proteins p-Akt, p-GSK3β, and anti-apoptotic Bcl-2 protein. Anthocyanins treatment increased the levels of memory-related pre- and post-synaptic proteins and improved the hippocampus-dependent memory in the LPS-treated mice. Overall, this data suggested that consumption of naturally derived anti-oxidant agent such as anthocyanins ameliorated several pathological events in the LPS-treated animal model and we believe that anthocyanins would be a safe therapeutic agent for slowing the inflammation-induced neurodegeneration in the brain against several diseases such as Alzheimer's disease and Parkinson's disease.
Collapse
|
40
|
Mohammad Jafari R, Ghahremani MH, Rahimi N, Shadboorestan A, Rashidian A, Esmaeili J, Ejtemaei Mehr S, Dehpour AR. The anticonvulsant activity and cerebral protection of chronic lithium chloride via NMDA receptor/nitric oxide and phospho-ERK. Brain Res Bull 2018; 137:1-9. [DOI: 10.1016/j.brainresbull.2017.10.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/30/2017] [Accepted: 10/31/2017] [Indexed: 02/08/2023]
|
41
|
Fang Y, Kang Y, Zou H, Cheng X, Xie T, Shi L, Zhang H. β-elemene attenuates macrophage activation and proinflammatory factor production via crosstalk with Wnt/β-catenin signaling pathway. Fitoterapia 2018; 124:92-102. [PMID: 29066299 DOI: 10.1016/j.fitote.2017.10.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 10/10/2017] [Accepted: 10/20/2017] [Indexed: 11/27/2022]
Abstract
β-elemene, extracted from Rhizoma zedoariae, has been widely used as a traditional medicine for its antitumor activity against a broad range of cancers. However, the effect of β-elemene in inflammation disorders has yet to be determined. The present study was designed to investigate the anti-inflammatory effects and potential molecular mechanisms of β-elemene in lipopolysaccharide (LPS)-induced murine macrophage cells RAW264.7. We found that the production of pro-inflammatory mediators, including interleukin-6(IL-6), tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β), induced by LPS was significantly suppressed by β-elemene in a dose-dependent manner in RAW264.7 macrophage cell line. Also, β-elemene inhibited LPS-induced nitric oxide synthase (iNOS) and interleukin-10 (IL-10) expression by RAW264.7, which was related to the down-regulation of Wnt/β-catenin signaling pathway. Importantly, this study demonstrates that β-catenin was significantly inhibited by β-elemene, which appeared to be largely responsible for the down-regulation of Wnt/β-catenin signaling pathway. Accordingly, the deletion of β-catenin in primary macrophages reversed β-catenin-elicited inhibition of immune response. Furthermore, β-catenin expression and Wnt/β-catenin signaling pathway induced by LPS in RAW264.7 was also significantly inhibited by α-humulene, one isomeric sesquiterpene of β-elemene. α-humulene was also found to significantly inhibit LPS-induced production of proinflammatory cytokines. However, α-humulene showed more cytotoxic ability than β-elemene. Collectively, our data illustrated that β-elemene exerted a potent inhibitory effect on pro-inflammatory meditator and cytokines production via the inactivation of β-catenin, and also demonstrated the protective functions of β-elemene in endotoxin-induced inflammation. β-elemene may serve as potential nontoxic modulatory agents for the prevention and treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Yangyi Fang
- Key Laboratory of Immunology and Molecular Medicine, Division of Basical Medicine, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yanhua Kang
- Key Laboratory of Immunology and Molecular Medicine, Division of Basical Medicine, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China; Department of Immunology, School of Basic Medical Science, Nanjing University of Chinese Medicine, Nanjing, China
| | - Han Zou
- Key Laboratory of Immunology and Molecular Medicine, Division of Basical Medicine, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xiaxuan Cheng
- Key Laboratory of Immunology and Molecular Medicine, Division of Basical Medicine, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China; Department of Biochemistry and Molecular Biology, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Tian Xie
- Institute of Holistic Integrative Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China; Institute of Holistic Integrative Oncology, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Liyun Shi
- Key Laboratory of Immunology and Molecular Medicine, Division of Basical Medicine, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China; Department of Immunology, School of Basic Medical Science, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Hang Zhang
- Key Laboratory of Immunology and Molecular Medicine, Division of Basical Medicine, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang, China; Institute of Holistic Integrative Oncology, Hangzhou Normal University, Hangzhou, Zhejiang, China.
| |
Collapse
|