1
|
Gu J, Cai X, Raza F, Zafar H, Chu B, Yuan H, Wang T, Wang J, Feng X. Preparation of a minocycline polymer micelle thermosensitive gel and its application in spinal cord injury. NANOSCALE ADVANCES 2024:d4na00625a. [PMID: 39355839 PMCID: PMC11440374 DOI: 10.1039/d4na00625a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/15/2024] [Indexed: 10/03/2024]
Abstract
Neuroprotection is an important approach for the treatment of spinal cord injury (SCI). Minocycline (MC), a known neuroprotective agent, has been utilized for SCI treatment, but its therapeutic effect is limited by instability and low bioavailability. Herein, we developed an innovative micellar thermosensitive hydrogel (MCPP-M-gel) that encapsulates MC in polyethylene glycol (PEG)-poly(lactide-co-glycolic acid) (PLGA) micelles to enhance its therapeutic efficacy in a rat model of SCI. The micelles were synthesized via the thin-film hydration method and characterized for encapsulation efficiency, particle size, zeta potential, and polydispersity index (PDI). MCPP-M-gel demonstrated favorable physico-mechanical properties and extended MC release over 72 hours in vitro without cytotoxic effects on neural crest-derived ectoderm mesenchymal stem cells (EMSCs). Thereafter, MC, MCPP-M, MCPP-M-gel and a blank micellar thermosensitive gel were injected into the injured site of SCI rats. Histopathological evaluation demonstrated that MCPP-M-gel could promote neuronal regeneration at the injured site of the SC after 28 days. Immunofluorescence techniques revealed that MCPP-M-gel increased the expression of neuronal class III β-tubulin (Tuj1), myelin basic protein (MBP), growth-associated protein 43 (GAP43), neurofilament protein-200 (NF-200) and nestin as well as reduced glial-fibrillary acidic protein (GFAP) expression in damaged areas of the SC. In conclusion, this study innovatively developed MCPP-M-gel based on a PEG-PLGA copolymer as a biomaterial, laying a solid foundation for further research and application of MCPP-M-gel in SCI models or other neurodegenerative diseases.
Collapse
Affiliation(s)
- Jun Gu
- School of Medicine, Yangzhou University Yangzhou Jiangsu 225009 China
- Department of Orthopedics, Xishan People's Hospital Wuxi Jiangsu 204105 China
| | - Xiaohu Cai
- School of Medicine, Yangzhou University Yangzhou Jiangsu 225009 China
- Department of Rehabilitation, Xishan People's Hospital Wuxi Jiangsu 204105 China
| | - Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University Shanghai 200240 China
| | - Hajra Zafar
- School of Pharmacy, Shanghai Jiao Tong University Shanghai 200240 China
| | - Bo Chu
- Department of Orthopedics, Xishan People's Hospital Wuxi Jiangsu 204105 China
| | - Haitao Yuan
- Department of Orthopedics, Xishan People's Hospital Wuxi Jiangsu 204105 China
| | - Tianqi Wang
- Department of Orthopedics, Xishan People's Hospital Wuxi Jiangsu 204105 China
| | - Jiapeng Wang
- School of Pharmacy, Jiangsu University Zhenjiang Jiangsu 212013 China
| | - Xiaojun Feng
- School of Medicine, Yangzhou University Yangzhou Jiangsu 225009 China
- Department of Orthopedics, Xishan People's Hospital Wuxi Jiangsu 204105 China
| |
Collapse
|
2
|
Wang D, Wang J, Yan D, Wang M, Yang L, Demin KA, de Abreu MS, Kalueff AV. Minocycline reduces neurobehavioral deficits evoked by chronic unpredictable stress in adult zebrafish. Brain Res 2024; 1845:149209. [PMID: 39233136 DOI: 10.1016/j.brainres.2024.149209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/11/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024]
Abstract
Chronic stress-related brain disorders are widespread and debilitating, and often cause lasting neurobehavioral deficits. Minocycline, a common antibiotic and an established inhibitor of microglia, emerges as potential treatment of these disorders. The zebrafish (Danio rerio) is an important emerging model organism in translational neuroscience and stress research. Here, we evaluated the potential of minocycline to correct microglia-mediated behavioral, genomic and neuroimmune responses induced by chronic unpredictable stress (CUS) in adult zebrafish. We demonstrated that CUS evoked overt behavioral deficits in the novel tank, light-dark box and shoaling tests, paralleled by elevated stress hormones (CRH, ACTH and cortisol), and upregulated brain expression of the 'neurotoxic M1' microglia-specific biomarker gene (MHC-2) and pro-inflammatory cytokine genes (IL-1β, IL-6 and IFN-γ). CUS also elevated peripheral (whole-body) pro-inflammatory (IL-1β, IFN-γ) and lowered anti-inflammatory cytokines (IL-4 and IL-10), as well as reduced whole-brain serotonin, dopamine and norepinephrine levels, and increased brain dopamine and serotonin turnover. In contrast, minocycline attenuated most of these effects, also reducing CUS-elevated peripheral levels of IL-6 and IFN-γ. Collectively, this implicates microglia in zebrafish responses to chronic stress, and suggests glial pathways as potential evolutionarily conserved drug targets for treating stress-evoked neuropathogenesis. Our findings also support the growing translational value of zebrafish models for understanding complex molecular mechanisms of brain pathogenesis and its therapy.
Collapse
Affiliation(s)
- Dongmei Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - Jingtao Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - Dongni Yan
- School of Pharmacy, Southwest University, Chongqing, China
| | - Mengyao Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - Longen Yang
- School of Pharmacy, Southwest University, Chongqing, China; Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, China; Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, China
| | - Konstantin A Demin
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Murilo S de Abreu
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil; Western Caspian University, Baku, Azerbaijan; Moscow Institute of Physics and Technology, Dolgoprudny, Russia.
| | - Allan V Kalueff
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Center, St. Petersburg, Russia; Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, China; Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou, China; Moscow Institute of Physics and Technology, Dolgoprudny, Russia.
| |
Collapse
|
3
|
Saha P, Saha R, Datta Chaudhuri R, Sarkar R, Sarkar M, Koley H, Chawla-Sarkar M. Unveiling the Antiviral Potential of Minocycline: Modulation of Nuclear Export of Viral Ribonuclear Proteins during Influenza Virus Infection. Viruses 2024; 16:1317. [PMID: 39205291 PMCID: PMC11359333 DOI: 10.3390/v16081317] [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: 07/11/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
Influenza A virus (IAV) poses a global threat worldwide causing pandemics, epidemics, and seasonal outbreaks. Annual modification of vaccines is costly due to continual shifts in circulating genotypes, leading to inadequate coverage in low- and middle-income countries like India. Additionally, IAVs are evolving resistance to approved antivirals, necessitating a search for alternative treatments. In this study, the antiviral role of the FDA-approved antibiotic minocycline against IAV strains was evaluated in vitro and in vivo by quantifying viral gene expression by qRT-PCR, viral protein levels by Western blotting, and viral titers. Our findings demonstrate that minocycline at a non-toxic dose effectively inhibits IAV replication, regardless of viral strain or cell line. Its antiviral mechanism operates independently of interferon signaling by targeting the MEK/ERK signaling pathway, which is crucial for the export of viral ribonucleoproteins (vRNPs). Minocycline prevents the assembly and release of infectious viral particles by causing the accumulation of vRNPs within the nucleus. Moreover, minocycline also inhibits IAV-induced late-stage apoptosis, further suppressing viral propagation. The antiviral activity of minocycline against IAVs could offer a promising solution amidst the challenges posed by influenza and the limitations of current treatments.
Collapse
Affiliation(s)
- Priyanka Saha
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata 700010, India
| | - Ritubrita Saha
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata 700010, India
| | - Ratul Datta Chaudhuri
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata 700010, India
| | - Rakesh Sarkar
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata 700010, India
| | - Mehuli Sarkar
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata 700010, India
| | - Hemanta Koley
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata 700010, India
| | - Mamta Chawla-Sarkar
- Division of Virology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata 700010, India
| |
Collapse
|
4
|
Hosseini M, Bardaghi Z, Askarpour H, Jafari MM, Golkar A, Shirzad S, Rajabian A, Salmani H. Minocycline mitigated enduring neurological consequences in the mice model of sepsis. Behav Brain Res 2024; 461:114856. [PMID: 38199318 DOI: 10.1016/j.bbr.2024.114856] [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: 12/14/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 01/12/2024]
Abstract
AIM Sepsis-associated encephalopathy is a frequently observed consequence of sepsis, often resulting in chronic brain inflammation and injury, ultimately leading to a range of behavioral abnormalities. This study explores the potential preventive effects of minocycline on the long-lasting outcome of sepsis in a mice model of sepsis. METHODS Adult male C57 mice were subjected to experimental sepsis through a single intraperitoneal injection of 5 mg/kg lipopolysaccharide (LPS). Minocycline administration via oral gavage (12.5, 25, and 50 mg/kg) commenced three days before sepsis induction and continued on the day of induction. Mice underwent behavioral assessments one month post-sepsis, with subsequent brain tissue analysis to investigate oxidative stress markers and cholinergic function. KEY FINDINGS One month following sepsis induction, mice exhibited significant anxiety- and depressive-like behaviors as determined by assessments in the elevated plus maze (EPM), open field, and tail suspension test (TST). Additionally, they displayed impaired recognition memory in the novel object recognition (NOR) test. Brain tissue analysis revealed a notable increase in oxidative stress markers and acetylcholinesterase (AChE) activity in septic mice. Notably, minocycline treatment effectively mitigated the long-term behavioral abnormalities resulting from sepsis, attenuated oxidative stress markers, and reduced AChE activity. SIGNIFICANCE These findings underscore the potential of minocycline as a therapeutic intervention during sepsis induction to prevent the enduring behavioral and neurological consequences of experimental sepsis.
Collapse
Affiliation(s)
- Mahmoud Hosseini
- Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Bardaghi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hedyeh Askarpour
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran
| | | | - Ahmad Golkar
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Shima Shirzad
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arezoo Rajabian
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Salmani
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran; Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran; Department of Physiology and Pharmacology, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran.
| |
Collapse
|
5
|
Bergold PJ, Furhang R, Lawless S. Treating Traumatic Brain Injury with Minocycline. Neurotherapeutics 2023; 20:1546-1564. [PMID: 37721647 PMCID: PMC10684850 DOI: 10.1007/s13311-023-01426-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2023] [Indexed: 09/19/2023] Open
Abstract
Traumatic brain injury (TBI) results in both rapid and delayed brain damage. The speed, complexity, and persistence of TBI present large obstacles to drug development. Preclinical studies from multiple laboratories have tested the FDA-approved anti-microbial drug minocycline (MINO) to treat traumatic brain injury. At concentrations greater than needed for anti-microbial action, MINO readily inhibits microglial activation. MINO has additional pleotropic effects including anti-inflammatory, anti-oxidant, and anti-apoptotic activities. MINO inhibits multiple proteins that promote brain injury including metalloproteases, caspases, calpain, and polyADP-ribose-polymerase-1. At these elevated doses, MINO is well tolerated and enters the brain even when the blood-brain barrier is intact. Most preclinical studies with a first dose of MINO at less than 1 h after injury have shown improved multiple outcomes after TBI. Fewer studies with more delayed dosing have yielded similar results. A small number of clinical trials for TBI have established the safety of MINO and suggested some drug efficacy. Studies are also ongoing that either improve MINO pharmacology or combine MINO with other drugs to increase its therapeutic efficacy against TBI. This review builds upon a previous, recent review by some of the authors (Lawless and Bergold, Neural Regen Res 17:2589-92, 2022). The present review includes the additional preclinical studies examining the efficacy of minocycline in preclinical TBI models. This review also includes recommendations for a clinical trial to test MINO to treat TBI.
Collapse
Affiliation(s)
- Peter J Bergold
- Graduate Programs in Neural and Behavioral Sciences, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, New York, NY, 11203, USA.
- Department of Physiology and Pharmacology, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, New York, NY, 11203, USA.
| | - Rachel Furhang
- Graduate Programs in Neural and Behavioral Sciences, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, New York, NY, 11203, USA
| | - Siobhán Lawless
- Graduate Programs in Neural and Behavioral Sciences, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, New York, NY, 11203, USA
| |
Collapse
|
6
|
Wang Y, Yang X, Cao Y, Li X, Xu R, Yan J, Guo Z, Sun S, Sun X, Wu Y. Electroacupuncture alleviates early brain injury via modulating microglia polarization and suppressing neuroinflammation in a rat model of subarachnoid hemorrhage. Heliyon 2023; 9:e14475. [PMID: 36967957 PMCID: PMC10036651 DOI: 10.1016/j.heliyon.2023.e14475] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 02/02/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Subarachnoid hemorrhage refers to an uncommon but severe subtype of stroke leading to high mortality and disability rates. Electroacupuncture, a traditional Chinese medical therapy combined with modern technology, shows evident curative effects on cerebral vascular diseases. This study attempts to investigate the possible treatment effects and mechanisms of EA on early brain injury after SAH. Data were gathered among sham group, SAH-induced group, and EA-treated group of male SD rats, concerning mortality rates, weight loss, rotarod latencies, cerebral blood flow, cell apoptosis, pro-inflammatory cytokines releasing, apoptotic protein level, microglia activation and related signal pathway. All results were collected 24-72 h after SAH induction. EA treatment demonstrated significant improvement on motor function 24 h after SAH without significant changes in mortality rate, weight loss, and cerebral blood flow. Another important finding was that EA regulated Bax and Bcl-2 imbalance and reduced cleaved casepase-3 caused by SAH. Additionally, levels of TNF-α, IL-1β, IL-6 were suppressed. The neuron apoptosis was suppressed by EA. The M1 polarization of activated microglia decreased while M2 polarized phenotype increased after EA treatment. Furthermore, pSTAT3-NOX2 signal axis, the M1 phenotype related activation pathway, was depressed after EA treatment. These findings suggested that EA improved motor deficits and ameliorated early brain injury after SAH probably via decreasing neuron apoptosis and anti-inflammation, which may involve modulation of microglia polarization. Taken together, EA may be a potential therapy for SAH treatment.
Collapse
|
7
|
YTHDF1 Attenuates TBI-Induced Brain-Gut Axis Dysfunction in Mice. Int J Mol Sci 2023; 24:ijms24044240. [PMID: 36835655 PMCID: PMC9966860 DOI: 10.3390/ijms24044240] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/05/2023] [Accepted: 02/07/2023] [Indexed: 02/23/2023] Open
Abstract
The brain-gut axis (BGA) is a significant bidirectional communication pathway between the brain and gut. Traumatic brain injury (TBI) induced neurotoxicity and neuroinflammation can affect gut functions through BGA. N6-methyladenosine (m6A), as the most popular posttranscriptional modification of eukaryotic mRNA, has recently been identified as playing important roles in both the brain and gut. However, whether m6A RNA methylation modification is involved in TBI-induced BGA dysfunction is not clear. Here, we showed that YTHDF1 knockout reduced histopathological lesions and decreased the levels of apoptosis, inflammation, and oedema proteins in brain and gut tissues in mice after TBI. We also found that YTHDF1 knockout improved fungal mycobiome abundance and probiotic (particularly Akkermansia) colonization in mice at 3 days post-CCI. Then, we identified the differentially expressed genes (DEGs) in the cortex between YTHDF1-knockout and WT mice. These genes were primarily enriched in the regulation of neurotransmitter-related neuronal signalling pathways, inflammatory signalling pathways, and apoptotic signalling pathways. This study reveals that the ITGA6-mediated cell adhesion molecule signalling pathway may be the key feature of m6A regulation in TBI-induced BGA dysfunction. Our results suggest that YTHDF1 knockout could attenuate TBI-induced BGA dysfunction.
Collapse
|
8
|
Understanding CNS Effects of Antimicrobial Drugs Using Zebrafish Models. Vet Sci 2023; 10:vetsci10020096. [PMID: 36851400 PMCID: PMC9964482 DOI: 10.3390/vetsci10020096] [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: 12/13/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/31/2023] Open
Abstract
Antimicrobial drugs represent a diverse group of widely utilized antibiotic, antifungal, antiparasitic and antiviral agents. Their growing use and clinical importance necessitate our improved understanding of physiological effects of antimicrobial drugs, including their potential effects on the central nervous system (CNS), at molecular, cellular, and behavioral levels. In addition, antimicrobial drugs can alter the composition of gut microbiota, and hence affect the gut-microbiota-brain axis, further modulating brain and behavioral processes. Complementing rodent studies, the zebrafish (Danio rerio) emerges as a powerful model system for screening various antimicrobial drugs, including probing their putative CNS effects. Here, we critically discuss recent evidence on the effects of antimicrobial drugs on brain and behavior in zebrafish, and outline future related lines of research using this aquatic model organism.
Collapse
|
9
|
Zheng Y, Fan L, Xia S, Yang Q, Zhang Z, Chen H, Zeng H, Fu X, Peng Y, Xu C, Yu K, Liu F, Cao S. Role of complement C1q/C3-CR3 signaling in brain injury after experimental intracerebral hemorrhage and the effect of minocycline treatment. Front Immunol 2022; 13:919444. [PMID: 36189326 PMCID: PMC9520460 DOI: 10.3389/fimmu.2022.919444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
AimThe complement cascade is activated and may play an important pathophysiologic role in brain injury after experimental intracerebral hemorrhage (ICH). However, the exact mechanism of specific complement components has not been well studied. This study determined the role of complement C1q/C3-CR3 signaling in brain injury after ICH in mice. The effect of minocycline on C1q/C3-CR3 signaling-induced brain damage was also examined.MethodsThere were three parts to the study. First, the natural time course of C1q and CR3 expression was determined within 7 days after ICH. Second, mice had an ICH with CR3 agonists, LA-1 or vehicle. Behavioral score, neuronal cell death, hematoma volume, and oxidative stress response were assessed at 7 days after ICH. Third, the effect of minocycline on C1q/C3-CR3 signaling and brain damage was examined.ResultsThere were increased numbers of C1q-positive and CR3-positive cells after ICH. Almost all perihematomal C1q-positive and CR3-positive cells were microglia/macrophages. CR3 agonist LA-1 aggravated neurological dysfunction, neuronal cell death, and oxidative stress response on day 7 after ICH, as well as enhancing the expression of the CD163/HO-1 pathway and accelerating hematoma resolution. Minocycline treatment exerted neuroprotective effects on brain injury following ICH, partly due to the inhibition of C1q/C3-CR3 signaling, and that could be reversed by LA-1.ConclusionsThe complement C1q/C3-CR3 signaling is upregulated after ICH. The activation of C1q/C3-CR3 signaling by LA-1 aggravates brain injury following ICH. The neuroprotection of minocycline, at least partly, is involved with the repression of the C1q/C3-CR3 signaling pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Fuyi Liu
- *Correspondence: Fuyi Liu, ; Shenglong Cao,
| | | |
Collapse
|
10
|
2, 3, 5, 4'-tetrahydroxystilbene-2-O-beta-D-glucoside protects against neuronal cell death and traumatic brain injury-induced pathophysiology. Aging (Albany NY) 2022; 14:2607-2627. [PMID: 35314517 PMCID: PMC9004580 DOI: 10.18632/aging.203958] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 03/01/2022] [Indexed: 11/25/2022]
Abstract
Traumatic brain injury (TBI) is a global health issue that affects at least 10 million people per year. Neuronal cell death and brain injury after TBI, including apoptosis, inflammation, and excitotoxicity, have led to detrimental effects in TBI. 2, 3, 5, 4’-tetrahydroxystilbene-2-O-beta-D-glucoside (THSG), a water-soluble compound extracted from the Chinese herb Polygonum multiflorum, has been shown to exert various biological functions. However, the effects of THSG on TBI is still poorly understood. THSG reduced L-glutamate-induced DNA fragmentation and protected glial and neuronal cell death after L-glutamate stimulation. Our results also showed that TBI caused significant behavioral deficits in the performance of beam walking, mNSS, and Morris water maze tasks in a mouse model. Importantly, daily administration of THSG (60 mg/kg/day) after TBI for 21 days attenuated the injury severity score, promoted motor coordination, and improved cognitive performance post-TBI. Moreover, administration of THSG also dramatically decreased the brain lesion volume. THSG reduced TBI-induced neuronal apoptosis in the brain cortex 24 h after TBI. Furthermore, THSG increased the number of immature neurons in the subgranular zone (SGZ) of the dentate gyrus (DG) of the hippocampus. Our results demonstrate that THSG exerts neuroprotective effects on glutamate-induced excitotoxicity and glial and neuronal cell death. The present study also demonstrated that THSG effectively protects against TBI-associated motor and cognitive impairment, at least in part, by inhibiting TBI-induced apoptosis and promoting neurogenesis.
Collapse
|