1
|
Batallas D, Gallego JJ, Casanova-Ferrer F, López-Gramaje A, Rivas-Diaz P, Megías J, Escudero-García D, Durbán L, Benlloch S, Urios A, Hidalgo V, Salvador A, Montoliu C. Sex differences in the mediating role of brain-derived neurotrophic factor between inflammation and memory in cirrhotic patients with minimal hepatic encephalopathy. Brain Behav Immun Health 2025; 46:100998. [PMID: 40343108 PMCID: PMC12060516 DOI: 10.1016/j.bbih.2025.100998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/12/2025] [Accepted: 04/21/2025] [Indexed: 05/11/2025] Open
Abstract
Minimal hepatic encephalopathy (MHE) affects attention, visuo-motor coordination, and visual perception, with mixed evidence on its impact on memory. Brain-derived neurotrophic factor (BDNF) is associated with memory dysfunction, and plays a crucial role in modulating neuroplasticity. This study investigates the mediating role of BDNF in the relationship between pro-inflammatory cytokines (IL-6, IL-15, IL-18), and declarative memory performance, and the moderating effects of sex. Sixty-eight cirrhotic patients and 22 healthy volunteers performed the Psychometric Hepatic Encephalopathy Score for MHE diagnosis and logical memory subtest (Wechsler Memory Scale-III). Moderated mediation analysis using bias-corrected bootstrapping and multiple regression was performed. Results showed that increased levels of IL-18 and IL-15 were significantly associated with lower BDNF levels (p = 0.03 and p = 0.02 respectively). However, no direct effect was observed between IL-18 and IL-15 and memory. The conditional effects of BDNF on memory were significant only for women with and without MHE, and lower BDNF levels were associated with lower memory performance (without MHE: p = 0.002; MHE: p = 0.001). Moreover, BDNF mediated indirectly the relationship between pro-inflammatory cytokines and memory. IL-18 and IL-15 impacted memory through reduced BDNF levels only in women with and without MHE, whereas IL-6 showed no significant effect on BDNF or memory across groups. These findings underscore the important role of BDNF in memory in cirrhotic patients, especially women with MHE, by mediating the IL-18 and IL-15 effects. The study highlights the role of IL-18 and IL-15 cytokines in neuroplasticity-related memory decline, positioning BDNF as a key biomarker for inflammation-associated cognitive impairment in this population.
Collapse
Affiliation(s)
- Daniela Batallas
- Laboratory of Social Cognitive Neuroscience, Department of Psychobiology and IDOCAL, University of Valencia, 46010, Valencia, Spain
| | - Juan José Gallego
- Fundación Investigación Hospital Clínico Universitario de Valencia. INCLIVA, 46010, Valencia, Spain
- Department of Pathology. University of Valencia, 46010, Valencia, Spain
| | - Franc Casanova-Ferrer
- Fundación Investigación Hospital Clínico Universitario de Valencia. INCLIVA, 46010, Valencia, Spain
| | - Adriá López-Gramaje
- Fundación Investigación Hospital Clínico Universitario de Valencia. INCLIVA, 46010, Valencia, Spain
- Department of Pathology. University of Valencia, 46010, Valencia, Spain
| | - Pablo Rivas-Diaz
- Laboratory of Social Cognitive Neuroscience, Department of Psychobiology and IDOCAL, University of Valencia, 46010, Valencia, Spain
| | - Javier Megías
- Department of Pathology. University of Valencia, 46010, Valencia, Spain
| | - Desamparados Escudero-García
- Servicio de Medicina Digestiva, Hospital Clínico Universitario de Valencia, Spain
- Departamento de Medicina. University of Valencia, 46010 Valencia, Spain
| | - Lucía Durbán
- Servicio de Medicina Digestiva, Hospital Arnau de Vilanova, 46015, Valencia, Spain
| | - Salvador Benlloch
- Servicio de Medicina Digestiva, Hospital Arnau de Vilanova, 46015, Valencia, Spain
- CIBERehd, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Amparo Urios
- Fundación Investigación Hospital Clínico Universitario de Valencia. INCLIVA, 46010, Valencia, Spain
| | - Vanesa Hidalgo
- Laboratory of Social Cognitive Neuroscience, Department of Psychobiology and IDOCAL, University of Valencia, 46010, Valencia, Spain
- Department of Psychology and Sociology, Area of Psychobiology, University of Zaragoza, Teruel, Spain
| | - Alicia Salvador
- Laboratory of Social Cognitive Neuroscience, Department of Psychobiology and IDOCAL, University of Valencia, 46010, Valencia, Spain
- Spanish National Network for Research in Mental Health CIBERSAM, 28029, Madrid, Spain
| | - Carmina Montoliu
- Fundación Investigación Hospital Clínico Universitario de Valencia. INCLIVA, 46010, Valencia, Spain
- Department of Pathology. University of Valencia, 46010, Valencia, Spain
| |
Collapse
|
2
|
Tavakkoli M, Dadkhah M, Saadati H, Afshari S, Mostafalou S. Neurobehavioral toxicity of cypermethrin in association with oxidative, inflammatory and neurotrophic changes in the hippocampus of rats. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2025:1-12. [PMID: 40338169 DOI: 10.1080/09603123.2025.2503472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 05/02/2025] [Indexed: 05/09/2025]
Abstract
Pyrethroid pesticides can induce neurotoxicity in target and non-target organisms. We examined the effects of sub-chronic exposure to cypermethrin on anxiety-like behaviors, memory function and molecular changes in the hippocampus of rats. Twenty-four male Wistar rats (weighing 200-250 g) were randomly divided into three groups (n = 8) and received orally corn oil, cypermethrin 2 and cypermethrin 4 mg/kg/day, for 30 days. The elevated plus maze (EPM), open field maze (OFM) and novel object recognition memory (NORM) tests were conducted. At the end, brain derived neurotrophic factor (BDNF), glycogen synthase kinase-3 β (GSK-3β), cholinesterase activity, oxidative stress markers and inflammatory mediators were evaluated in the hippocampus. The results of OFM and EPM indicated higher level of anxiety and motor activity alterations in the cypermethrin treated rats. NORM test revealed impairment of cognitive memory and decreased discrimination ratio in cypermethrin treated rats. Cypermethrin decreased BDNF and increased GSK-3β, oxidative stress and mRNA expression of IL-1β and TNF-α and NF-κB in the hippocampus. The activity of cholinesterase enzyme was not changed. Cypermethrin can impair learning and memory along with provoking oxidative stress and inflammation, and dysregulation of BDNF and GSK-3β pathway may be involved in neurocognitive and neurobehavioral disorders due to long-time exposures.
Collapse
Affiliation(s)
- Marjaneh Tavakkoli
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Masoomeh Dadkhah
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hakimeh Saadati
- Department of Physiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Salva Afshari
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Sara Mostafalou
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
3
|
Yulug B, Kilic E, Oğuz T, Orhan C, Er B, Tuzcu M, Ozercan IH, Sahin N, Canpolat S, Komorowski J, Ojalvo SP, Sylla S, Cankaya S, Sahin K. Dose-Dependent Effect of a New Biotin Compound in Hippocampal Remyelination in Rats. Mol Neurobiol 2025; 62:6503-6520. [PMID: 39821844 PMCID: PMC11953097 DOI: 10.1007/s12035-025-04686-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 01/01/2025] [Indexed: 01/19/2025]
Abstract
Demyelination is commonly observed in neurodegenerative disorders, including multiple sclerosis (MS). Biotin supplementation is known to stabilize MS progression. To reduce the effective dose of biotin, we synthesized a new and superior form of biotin, a complex of magnesium ionically bound to biotin (MgB) and compared its dose-dependent effect with biotin alone after inducing demyelination using lysolecithin (LPC) in rats. Myelination was assessed using luxol fast blue staining and immunostaining against MBP protein, revealing that the most significant remyelination occurred in the MgB groups. Additionally, both biotin and MgB-treated animals showed dose-dependent improvements in spatial memory. Moreover, we detected a decrease in inflammatory proteins in both treatment groups, which was more prominent in high-dose MgB-treated animals and correlated with decreased expression of NF-κB p65, OP, and MMP-9 proteins. Further analysis of biotin-related proteins demonstrated that both biotin and, notably, MgB reversed the demyelination-dependent reduction of these proteins. Furthermore, biotin, particularly MgB, improved neuronal transmission proteins, Synapsin-1, PSD-93, and PSD-95. Additionally, both treatment groups exhibited increased BDNF, GAP43, and ICAM levels, with significant increments observed in high-dose MgB-treated animals. Increased GFAP, indicative of reactive gliosis, was observed in LPC-treated animals, and this effect was notably reversed by high-dose MgB treatment. The current data emphasize the dose-dependent beneficial effect on the remyelination process. Furthermore, the combination of biotin with Mg resulted in a more potent effect compared to biotin by itself. The strong influence of MgB encourages proof-of-concept studies using MgB in patients with MS.
Collapse
Affiliation(s)
- Burak Yulug
- Department of Neurology, School of Medicine, Alaaddin Keykubat University, Alanya, Turkey
| | - Ertugrul Kilic
- Department of Physiology, Istanbul Medipol University, Istanbul, Turkey
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Tuba Oğuz
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
- Department of Neuroscience, Graduate School of Health Sciences, Istanbul Medipol University, Istanbul, Turkey
| | - Cemal Orhan
- Department of Nutrition, Faculty of Veterinary Medicine, Firat University, Elazig, Turkey
| | - Besir Er
- Department of Biology, Faculty of Science, Firat University, Elazig, Turkey
| | - Mehmet Tuzcu
- Department of Biology, Faculty of Science, Firat University, Elazig, Turkey
| | | | - Nurhan Sahin
- Department of Nutrition, Faculty of Veterinary Medicine, Firat University, Elazig, Turkey
| | - Sinan Canpolat
- Department of Physiology, Faculty of Medicine, Firat University, Elazig, Turkey
| | - James Komorowski
- Research and Development, JDS Therapeutics, LLC, Purchase, NY, 10577, USA
| | - Sara Perez Ojalvo
- Research and Development, JDS Therapeutics, LLC, Purchase, NY, 10577, USA
| | - Sarah Sylla
- Research and Development, JDS Therapeutics, LLC, Purchase, NY, 10577, USA
| | - Seyda Cankaya
- Department of Neurology, School of Medicine, Alaaddin Keykubat University, Alanya, Turkey
| | - Kazim Sahin
- Department of Nutrition, Faculty of Veterinary Medicine, Firat University, Elazig, Turkey.
| |
Collapse
|
4
|
Shi L, Wang M, Yu R, An Y, Wang X, Zhang Y, Shi Y, Han C, Liu J. Sigma-1 receptor agonist PRE-084 increases BDNF by activating the ERK/CREB pathway to rescue learning and memory impairment caused by type II diabetes. Behav Brain Res 2025; 484:115493. [PMID: 39986614 DOI: 10.1016/j.bbr.2025.115493] [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: 10/08/2024] [Revised: 02/15/2025] [Accepted: 02/19/2025] [Indexed: 02/24/2025]
Abstract
Sigma-1 receptor (Sig-1R) agonists has therapeutic effects in neurological disorders and possesses properties that can reverse cognitive dysfunction. This study investigated the therapeutic efficacy of Sig-1R activation on cognitive dysfunction in streptozotocin (STZ) combined with high fat and high sugar diet (HFD)-induced type 2 diabetic rats. By employing morris water maze (MWM) testing and computed tomography (CT) imaging, we observed that activation of Sig-1R effectively mitigated brain atrophy and cognitive impairment in diabetes-induced cognitive impairment (DCI) rats. Given the fundamental role of intact hippocampal synaptic plasticity in maintaining cognitive function, we investigated the correlation between Sig-1R and Brain-Derived Neurotrophic Factor (BDNF), a well-established neurotrophic factor. And we also analyzed the expression of Postsynaptic density protein-95 (PSD95) protein. Golgi staining, Haematoxylin-eosin (HE) staining, Nissl staining, and immunofluorescence results show that activating Sig-1R can upregulate BDNF expression and reducing synaptic damage in hippocampal neurons. To elucidate the mechanism by which Sig-1R activation leads to increased BDNF levels, we investigated the Extracellular Signal-Regulated Kinase/Cyclic AMP Response Element-Binding Protein(ERK/CREB) protein pathway. In vitro and in vivo, we observed that Sig-1R activates the ERK/CREB signaling pathway, thereby stimulating BDNF release and increased PSD95 expression. Further intervention with BD1047 antagonist and Tropomyosin-Related Kinase B (TrkB) antagonist ANA-12 confirmed our conclusion that Sig-1R activation upregulated p-ERK and p-CREB protein expression, promoted BDNF transcription, the expression of PSD95 protein was up-regulated, reduces synaptic damage in damaged hippocampal neurons, and rescued cognitive impairment in DCI rats.
Collapse
Affiliation(s)
- Leilei Shi
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China.
| | - Mingmei Wang
- College of Biology & Food sciences, Changshu Institute of Technology, Suzhou 215123, PR China.
| | - Ruixuan Yu
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China.
| | - Yuyu An
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China.
| | - Xin Wang
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China.
| | - Yuhan Zhang
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China.
| | - Yongheng Shi
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; Key Laboratory of Pharmacodynamic Mechanism and Material Basis of Traditional Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xianyang 712046, PR China.
| | - Chaojun Han
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; Key Laboratory of Pharmacodynamic Mechanism and Material Basis of Traditional Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xianyang 712046, PR China.
| | - Jiping Liu
- Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; Key Laboratory of Pharmacodynamic Mechanism and Material Basis of Traditional Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xianyang 712046, PR China.
| |
Collapse
|
5
|
Jhan KY, Chen KY, Chang PK, Chiu CH, Chou CJ, Wang LC. 7,8-Dihydroxyflavone provides neuroprotection and rescues behavioral deficits in Angiostrongylus cantonensis-infected mice by ameliorating synaptic loss. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2025:S1684-1182(25)00081-7. [PMID: 40287337 DOI: 10.1016/j.jmii.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 02/26/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Angiostrongylus cantonensis triggers eosinophilic meningitis and/or eosinophilic meningoencephalitis in humans, potentially causing permanent central nervous system damage. While corticosteroids may ease infection-induced headaches, they prove ineffective against A. cantonensis larvae or neuronal injuries. Conversely, anthelmintics and neuroprotective agents like 7,8-Dihydroxyflavone (7,8-DHF), activating BDNF/TrkB signaling, offer promise. METHODS This study aimed to assess neural structure, brain function impairment and recovery, and pathological symptoms and cell death in A. cantonensis-infected C57BL/6 mice before and after treatment with 7,8-DHF or Albendazole alone, and in combination with 7,8-DHF/Albendazole. RESULTS All treatment groups effectively mitigated infection-induced weight loss in mice. Those receiving 7,8-DHF or combined treatment displayed significant improvements in motor and balance abilities during the rotarod test. Post-treatment, infected mice showed enhanced spatial learning and memory in the Morris water maze test. Golgi staining revealed that strategies involving 7,8-DHF reduced infection-induced synaptic loss. A. cantonensis infection induced severe inflammatory responses and brain damage in mice. Albendazole alone and the combined 7,8-DHF/Albendazole treatment effectively alleviated inflammation-related pathological phenomena. Analysis of neuronal functional protein expression across different brain regions showed increased presynaptic proteins following 7,8-DHF treatment. Moreover, 7,8-DHF treatment alone or combined with Albendazole prevented brain injury without inducing cell death in infected mice. CONCLUSION These findings suggested that 7,8-DHF could confer neuroprotective effects, particularly in mitigating synaptic loss in the presynaptic region, and alleviating behavioral deficits from infection without inducing cell death. We anticipate these results will aid in the treatment of angiostrongyliasis-induced neurological damage.
Collapse
Affiliation(s)
- Kai-Yuan Jhan
- Department of Parasitology, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Kuang-Yao Chen
- Department of Parasitology, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan; Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan
| | - Pi-Kai Chang
- Department of Parasitology, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Cheng-Hsun Chiu
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan
| | - Chih-Jen Chou
- Department of Parasitology, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan.
| | - Lian-Chen Wang
- Department of Parasitology, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan; Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan.
| |
Collapse
|
6
|
Wu CYC, Zhang Y, Howard P, Huang F, Lee RHC. ACSL3 is a promising therapeutic target for alleviating anxiety and depression in Alzheimer's disease. GeroScience 2025; 47:2383-2397. [PMID: 39532829 PMCID: PMC11978576 DOI: 10.1007/s11357-024-01424-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD), the leading cause of dementia, affects over 55 million people worldwide and is often accompanied by depression and anxiety. Both significantly impact patients' quality of life and impose substantial societal and economic burdens on healthcare systems. Identifying the complex regulatory mechanisms that contribute to the psychological and emotional deficits in AD will provide promising therapeutic targets. Biosynthesis of omega-3 (ω3) and omega-6 fatty acids (ω6-FA) through long-chain acyl-CoA synthetases (ACSL) is crucial for cell function and survival. This is due to ω3/6-FA's imperative role in modulating the plasma membrane, energy production, and inflammation. While ACSL dysfunction is known to cause heart, liver, and kidney diseases, the role of ACSL in pathological conditions in the central nervous system (e.g., depression and anxiety) remains largely unexplored. The impact of ACSLs on AD-related depression and anxiety was investigated in a mouse model of Alzheimer's disease (3xTg-AD). ACSL3 levels were significantly reduced in the hippocampus of aged 3xTg-AD mice (via capillary-based immunoassay). This reduction in ACAL3 was closely associated with increased depression and anxiety-like behavior (via forced swim, tail suspension, elevated plus maze, and sucrose preference test). Upregulation of ACSL3 via adenovirus in aged 3xTg-AD mice led to increased protein levels of brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor C (VEGF-C) (via brain histology, capillary-based immunoassay), resulting in alleviation of depression and anxiety symptoms. The present study highlights a novel neuroprotective role of ACSL3 in the brain. Targeting ACSL3 will offer an innovative approach for treating AD-related depression and anxiety.
Collapse
Affiliation(s)
- Celeste Yin-Chieh Wu
- Department of Neurology, Louisiana State University Health, LSU Health Sciences Center Shreveport, 1501 Kings Hwy, Shreveport, LA, 71103-3932, USA.
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA.
| | - Yulan Zhang
- Department of Neurology, Louisiana State University Health, LSU Health Sciences Center Shreveport, 1501 Kings Hwy, Shreveport, LA, 71103-3932, USA
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA
| | - Peyton Howard
- Department of Neurology, Louisiana State University Health, LSU Health Sciences Center Shreveport, 1501 Kings Hwy, Shreveport, LA, 71103-3932, USA
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA
| | - Fang Huang
- Department of Neurology, Louisiana State University Health, LSU Health Sciences Center Shreveport, 1501 Kings Hwy, Shreveport, LA, 71103-3932, USA
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA
| | - Reggie Hui-Chao Lee
- Department of Neurology, Louisiana State University Health, LSU Health Sciences Center Shreveport, 1501 Kings Hwy, Shreveport, LA, 71103-3932, USA
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA
- Department of Cellular Biology and Anatomy, Louisiana State University Health, Shreveport, LA, USA
| |
Collapse
|
7
|
Zhu S, Yu Q, Xue Y, Li J, Huang Y, Liu W, Wang G, Wang L, Zhai Q, Zhao J, Zhang H, Chen W. Bifidobacterium bifidum CCFM1163 alleviates cathartic colon by activating the BDNF-TrkB-PLC/IP 3 pathway to reconstruct the intestinal nerve and barrier. Food Funct 2025; 16:2057-2072. [PMID: 39963068 DOI: 10.1039/d4fo05835f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
Introduction: Cathartic colon (CC) is a type of slow-transit constipation caused by a patient's long-term use of irritating laxatives. Probiotics play a crucial role in managing constipation. Objectives: This study aims to identify probiotics that can alleviate CC and explore their specific mechanisms of action. Methods: The CC-model was constructed using senna leaf extract. Bifidobacterium bifidum was applied to the mice for intervention. Relevant marker changes were then examined using ELISA and RT-qPCR. Furthermore, 16S rDNA sequencing was utilized for functional prediction of intestinal microorganisms, while GC-MS analysis was performed to determine the content of short-chain fatty acids (SCFAs) in feces. Results: Senna damages the intestinal nerve and the intestinal barrier while inducing CC. In contrast, Bifidobacterium bifidum CCFM1163 may enhance the brain-derived neurotrophic factor (BDNF) expression in the colon by altering the intestinal microbiota composition (e.g., increasing Lactobacillus and Bacteroides, and decreasing Faecalibaculum) and by elevating SCFA levels (e.g., acetic and isobutyric acid). Subsequently, elevated BDNF expression activates the BDNF-tyrosine kinase receptor B-phospholipase C/inositol trisphosphate (BDNF-TrkB-PLC/IP3) pathway, which upregulates the gene expression of Uchl1, S100β, and Acta2; repairs the enteric nervous system-interstitial cells of Cajal-smooth muscle cells (ENS-ICC-SMC) network; upregulates the gene expression of Ocln and Tjp1; improves intestinal permeability in CC mice; and modulates the immune response by upregulating Tlr4, downregulating Il1b, and upregulating Il10, ultimately alleviating CC. Conclusion: Bifidobacterium bifidum CCFM1163 was identified as a probiotic that can promote BDNF expression in the colon, activate the BDNF-TrkB-PLC/IP3 signaling pathway, and effectively alleviate CC.
Collapse
Affiliation(s)
- Shengnan Zhu
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qiangqing Yu
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yifan Xue
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jiazhen Li
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yin Huang
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wenxu Liu
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Gang Wang
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Linlin Wang
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Wei Chen
- State Key Laboratory of Food Science and resources, Jiangnan University, Wuxi, Jiangsu 214122, P. R China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
8
|
Wu X, Hao J, Jiang K, Wu M, Zhao X, Zhang X. Neuroinflammation and pathways that contribute to tourette syndrome. Ital J Pediatr 2025; 51:63. [PMID: 40022157 PMCID: PMC11871796 DOI: 10.1186/s13052-025-01874-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 01/26/2025] [Indexed: 03/03/2025] Open
Abstract
Tourette syndrome (TS), a neurological and psychological disease, typically exhibit motor and phonic tics. The pathophysiology of TS remains controversial. Currently, the recognized pathogenesis of TS is the imbalance of neurotransmitters, involving abnormality of the cortex-striatum-thalamus-cortex circuit. Recently, clinical researches demonstrate that triggers such as infection and allergic reaction could lead to the onset or exacerbation of tic symptoms. Current studies have also suggested that neural-immune crosstalk caused by inflammation is also associated with TS, potentially leading to the occurrence of tics by inducing neurotransmitter abnormalities. Herein, we review inflammation-related factors contributing to the occurrence of TS as well as the mechanisms by which immune-inflammatory pathways mediate the onset of TS. This aims to clarify the pathogenesis of TS and provide a theoretical basis for the treatment of TS.
Collapse
Affiliation(s)
- Xinnan Wu
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juanjuan Hao
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Keyu Jiang
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Wu
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Zhao
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Zhang
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
9
|
Chaki T, Horiguchi Y, Tachibana S, Sato S, Hirahata T, Nishihara N, Kii N, Yoshikawa Y, Hayamizu K, Yamakage M. Gut Microbiota Influences Developmental Anesthetic Neurotoxicity in Neonatal Rats. Anesth Analg 2025:00000539-990000000-01140. [PMID: 39899452 DOI: 10.1213/ane.0000000000007410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2025]
Abstract
BACKGROUND Anesthetic exposure during childhood is significantly associated with impairment of neurodevelopmental outcomes; however, the causal relationship and detailed mechanism of developmental anesthetic neurotoxicity remain unclear. Gut microbiota produces various metabolites and influences the brain function and development of the host. This relationship is referred to as the gut-brain axis. Gut microbiota may influence developmental anesthetic neurotoxicity caused by sevoflurane exposure. This study investigated the effect of changes in the composition of gut microbiota after fecal microbiota transplantation on spatial learning disability caused by developmental anesthetic neurotoxicity in neonatal rats. METHODS Neonatal rats were allocated into the Control (n = 10) and Sevo (n = 10) groups in Experiment 1 and the Sevo (n = 20) and Sevo+FMT (n = 20) groups in Experiment 2, according to the randomly allocated mothers' group. The rats in Sevo and Sevo+FMT groups were exposed to 2.1% sevoflurane for 2 hours on postnatal days 7 to 13. Neonatal rats in the Sevo+FMT group received fecal microbiota transplantation immediately after sevoflurane exposure on postnatal days 7 to 13. The samples for fecal microbiota transplantation were obtained from nonanesthetized healthy adult rats. Behavioral tests, including Open field, Y-maze, Morris water maze, and reversal Morris water maze tests, were performed to evaluate spatial learning ability on postnatal days 26 to 39. RESULTS Experiment 1 revealed that sevoflurane exposure significantly altered the gut microbiota composition. The relative abundance of Roseburia (effect value: 1.01) and Bacteroides genus (effect value: 1.03) increased significantly after sevoflurane exposure, whereas that of Lactobacillus (effect value: -1.20) decreased significantly. Experiment 2 revealed that fecal microbiota transplantation improved latency to target (mean ± SEM; Sevo group: 9.7 ± 8.2 seconds vs, Sevo+FMT group: 2.7 ± 2.4 seconds, d=1.16, 95% confidence interval: -12.7 to -1.3 seconds, P = .019) and target zone crossing times (Sevo group: 2.4 ± 1.6 vs, Sevo+FMT group: 5.4 ± 1.4, d=1.99, 95% confidence interval: 2.0-5.0, P < .001) in the reversal Morris water maze test. Microbiota analysis revealed that the α-diversity of gut microbiota increased after fecal microbiota transplantation. Similarly, the relative abundance of the Firmicutes phylum (effect value: 1.44), Ruminococcus genus (effect value: 1.69), and butyrate-producing bacteria increased after fecal microbiota transplantation. Furthermore, fecal microbiota transplantation increased the fecal concentration of butyrate and induced histone acetylation and the mRNA expression of brain-derived neurotrophic factor in the hippocampus, thereby suppressing neuroinflammation and neuronal apoptosis. CONCLUSIONS The alternation of gut microbiota after fecal microbiota transplantation influenced spatial learning ability in neonatal rats with developmental anesthetic neurotoxicity. Modulation of the gut microbiota may be an effective prophylaxis for developmental anesthetic neurotoxicity in children.
Collapse
Affiliation(s)
- Tomohiro Chaki
- From the Department of Anesthesiology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Lee JH, Lee KH, Ryu JH, Kim MJ, Kim E, Lee SY, Han SC, Choi BT, Shin YI, Shin HK. LM22A-4-loaded smart mesoporous balls enhance neuroprotection and functional recovery after ischemic stroke. Biomed Pharmacother 2025; 183:117863. [PMID: 39842267 DOI: 10.1016/j.biopha.2025.117863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/23/2024] [Accepted: 01/18/2025] [Indexed: 01/24/2025] Open
Abstract
Stroke is globally recognized as the second leading cause of death, significantly impairing both motor and cognitive functions. Enhancing regeneration after stroke is crucial for restoring these functions and necessitates strategies to promote neuroregeneration to achieve better post-stroke outcomes. Brain-derived neurotrophic factor (BDNF) plays a key role in neuroregeneration by influencing motor ability, learning, memory, and rehabilitation after stroke. However, challenges such as the substantial protein size, short half-life of BDNF, and blood-brain barrier hinder its efficient delivery to the brain. In this study, LM22A-4, a BDNF mimetic, was utilized and delivered through a Smart Mesoporous Ball (SMB-3) system to target the ischemic injured brain and explore its potential therapeutic effects in a mouse ischemic stroke model. Treatment with LM22A-4-loaded SMB-3 (LM22A-4-SMB-3) markedly restored neurological, motor, and cognitive deficits following ischemic stroke compared to LM22A-4 alone. Additionally, administration of LM22A-4-SMB-3 reduced apoptotic cell death and glial activation, as evidenced by the TUNEL assay results, and decreased GFAP and Iba-1 expression levels. Furthermore, the phosphorylation of TrkB and Akt, but not that of Erk, was considerably increased in the LM22A-4-SMB-3-treated group. Treatment also enhanced the number of BrdU+/NeuN+ cells, with a marked reduction in post-stroke brain atrophy. These findings suggest that LM22A-4-SMB-3 can attenuate ischemic damage and recover neurological, motor, and cognitive functions by increasing p-TrkB and p-Akt levels and promoting neurogenesis. Therefore, SMB-3-mediated delivery of LM22A-4 presents a potentially applicable delivery system, and LM22A-4-SMB-3 use could be considered a novel therapeutic strategy to improve post-stroke outcomes.
Collapse
Affiliation(s)
- Jae Ho Lee
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Graduate Training Program of Korean Medical Therapeutics for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea
| | - Kyeong Hyeon Lee
- Department of Rehabilitation Medicine, School of Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea
| | - Ji Hyeon Ryu
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Gyeongnam 50612, Republic of Korea
| | - Min Jae Kim
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Graduate Training Program of Korean Medical Therapeutics for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea
| | - Eunji Kim
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Graduate Training Program of Korean Medical Therapeutics for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea
| | - Seo-Yeon Lee
- Department of Pharmacology, Wonkwang University School of Medicine, Iksan, Jeonbuk 54538, Republic of Korea
| | - Sang-Cheol Han
- CEN Co., Ltd. Nano-Convergence Center, 761 Muan-ro, Miryang 50404, Republic of Korea
| | - Byung Tae Choi
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Graduate Training Program of Korean Medical Therapeutics for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea
| | - Yong-Il Shin
- Department of Rehabilitation Medicine, School of Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea.
| | - Hwa Kyoung Shin
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea; Graduate Training Program of Korean Medical Therapeutics for Healthy-Aging, Pusan National University, Yangsan, Gyeongnam 50612, Republic of Korea.
| |
Collapse
|
11
|
Malange KF, de Souza DM, Lemes JBP, Fagundes CC, Oliveira ALL, Pagliusi MO, Carvalho NS, Nishijima CM, da Silva CRR, Consonni SR, Sartori CR, Tambeli CH, Parada CA. The Implications of Brain-Derived Neurotrophic Factor in the Biological Activities of Platelet-Rich Plasma. Inflammation 2025; 48:426-446. [PMID: 38904872 DOI: 10.1007/s10753-024-02072-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 05/16/2024] [Accepted: 05/31/2024] [Indexed: 06/22/2024]
Abstract
Platelet-rich plasma (PRP) is a biological blood-derived therapeutic obtained from whole blood that contains higher levels of platelets. PRP has been primarily used to mitigate joint degeneration and chronic pain in osteoarthritis (OA). This clinical applicability is based mechanistically on the release of several proteins by platelets that can restore joint homeostasis. Platelets are the primary source of brain-derived neurotrophic factor (BDNF) outside the central nervous system. Interestingly, BDNF and PRP share key biological activities with clinical applicability for OA management, such as anti-inflammatory, anti-apoptotic, and antioxidant. However, the role of BDNF in PRP therapeutic activities is still unknown. Thus, this work aimed to investigate the implications of BDNF in therapeutic outcomes provided by PRP therapy in vitro and in-vivo, using the MIA-OA animal model in male Wistar rats. Initially, the PRP was characterized, obtaining a leukocyte-poor-platelet-rich plasma (LP-PRP). Our assays indicated that platelets activated by Calcium release BDNF, and suppression of M1 macrophage polarization induced by LP-PRP depends on BDNF full-length receptor, Tropomyosin Kinase-B (TrkB). OA animals were given LP-PRP intra-articular and showed functional recovery in gait, joint pain, inflammation, and tissue damage caused by MIA. Immunohistochemistry for activating transcriptional factor-3 (ATF-3) on L4/L5 dorsal root ganglia showed the LP-PRP decreased the nerve injury induced by MIA. All these LP-PRP therapeutic activities were reversed in the presence of TrkB receptor antagonist. Our results suggest that the therapeutic effects of LP-PRP in alleviating OA symptoms in rats depend on BDNF/TrkB activity.
Collapse
Affiliation(s)
- Kaue Franco Malange
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Carl Von Linnaeus, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, 13083-864, Brazil
| | - Douglas Menezes de Souza
- Department of Pharmacology, School of Medical Sciences, University of Campinas (UNICAMP), Rua Tessália Vieira de Camargo, 126, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, 13083-887, Brazil
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, 255, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, CEP 13083-862, Brazil
| | - Julia Borges Paes Lemes
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Carl Von Linnaeus, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, 13083-864, Brazil
| | - Cecilia Costa Fagundes
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Carl Von Linnaeus, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, 13083-864, Brazil
| | - Anna Lethicia Lima Oliveira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Carl Von Linnaeus, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, 13083-864, Brazil
| | - Marco Oreste Pagliusi
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Carl Von Linnaeus, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, 13083-864, Brazil
| | - Nathalia Santos Carvalho
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Carl Von Linnaeus, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, 13083-864, Brazil
| | - Catarine Massucato Nishijima
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Carl Von Linnaeus, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, 13083-864, Brazil
| | - Cintia Rizoli Ruiz da Silva
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, 255, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, CEP 13083-862, Brazil
| | - Silvio Roberto Consonni
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, 255, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, CEP 13083-862, Brazil
| | - Cesar Renato Sartori
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Carl Von Linnaeus, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, 13083-864, Brazil
| | - Claudia Herrera Tambeli
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Carl Von Linnaeus, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, 13083-864, Brazil
| | - Carlos Amilcar Parada
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Carl Von Linnaeus, Cidade Universitária Zeferino Vaz, Campinas, São Paulo, 13083-864, Brazil.
| |
Collapse
|
12
|
Van den Bos J, Janssens I, Vermeulen M, Dams A, De Reu H, Peeters S, Faghel C, Ouaamari YE, Wens I, Cools N. The Efficiency of Brain-Derived Neurotrophic Factor Secretion by mRNA-Electroporated Regulatory T Cells Is Highly Impacted by Their Activation Status. Eur J Immunol 2025; 55:e202451005. [PMID: 39703060 PMCID: PMC11830389 DOI: 10.1002/eji.202451005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
Genetic engineering of regulatory T cells (Tregs) presents a promising avenue for advancing immunotherapeutic strategies, particularly in autoimmune diseases and transplantation. This study explores the modification of Tregs via mRNA electroporation, investigating the influence of T-cell activation status on transfection efficiency, phenotype, and functionality. For this CD45RA+ Tregs were isolated, expanded, and modified to overexpress brain-derived neurotrophic factor (BDNF). Kinetics of BDNF expression and secretion were explored. Treg activation state was assessed by checking the expression of activation markers CD69, CD71, and CD137. Our findings show that only activated Tregs secrete BDNF post-genetic engineering, even though both activated and resting Tregs express BDNF intracellularly. Notably, the mTOR pathway and CD137 are implicated in the regulation of protein secretion in activated Tregs, indicating a complex interplay of signalling pathways. This study contributes to understanding the mechanisms governing protein expression and secretion in engineered Tregs, offering insights for optimizing cell-based therapies and advancing immune regulation strategies.
Collapse
Affiliation(s)
- Jasper Van den Bos
- Laboratory of Experimental HematologyVaccine and Infections Disease Institute (VAXINFECTIO)Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
| | - Ibo Janssens
- Laboratory of Experimental HematologyVaccine and Infections Disease Institute (VAXINFECTIO)Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
| | - Morgane Vermeulen
- Laboratory of Experimental HematologyVaccine and Infections Disease Institute (VAXINFECTIO)Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
| | - Amber Dams
- Laboratory of Experimental HematologyVaccine and Infections Disease Institute (VAXINFECTIO)Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
| | - Hans De Reu
- Laboratory of Experimental HematologyVaccine and Infections Disease Institute (VAXINFECTIO)Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
- Flow Cytometry and Sorting Core Facility (FACSUA)University of AntwerpAntwerpBelgium
| | - Stefanie Peeters
- Laboratory of Experimental HematologyVaccine and Infections Disease Institute (VAXINFECTIO)Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
| | - Carole Faghel
- Laboratory of Experimental HematologyVaccine and Infections Disease Institute (VAXINFECTIO)Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
| | - Yousra El Ouaamari
- Laboratory of Experimental HematologyVaccine and Infections Disease Institute (VAXINFECTIO)Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
| | - Inez Wens
- Laboratory of Experimental HematologyVaccine and Infections Disease Institute (VAXINFECTIO)Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
| | - Nathalie Cools
- Laboratory of Experimental HematologyVaccine and Infections Disease Institute (VAXINFECTIO)Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
- Flow Cytometry and Sorting Core Facility (FACSUA)University of AntwerpAntwerpBelgium
| |
Collapse
|
13
|
Liampas I, Tsirelis D, Dastamani M, Pariou SI, Papasavva M, Katsarou MS, Tsolakou A, Tsatsakis A, Bogdanos DP, Drakoulis N, Dardiotis E, Siokas V. Lack of Association between BDNF rs6265 and Multiple Sclerosis: A Case-Control Study. J Mol Neurosci 2024; 75:1. [PMID: 39690366 DOI: 10.1007/s12031-024-02301-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/08/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND AND OBJECTIVES Data on the association between BDNF rs6265 and multiple sclerosis (MS) are scarce and heterogeneous. MATERIALS AND METHODS We undertook a case-control study design. Newly diagnosed individuals with MS based on the 2017 revision of the McDonald criteria were recruited from the Neurology Department of the General University Hospital of Larissa. Healthy controls with a free medical and family history were also recruited. The relationship between BDNF rs6265 and MS was defined as the primary outcome. The association between rs6265 and age of MS onset, spinal lesions, and clinical manifestations at the time of MS onset were defined as the secondary outcomes. RESULTS We genotyped a total of 200 patients with MS and 205 healthy controls, yielding a sample power of approximately 80%. BDNF rs6265 was in Hardy-Weinberg Equilibrium among healthy participants (p = 0.64). No significant relationship was revealed between rs6265 and MS [log-additive OR = 0.83 (0.57,1.21), over-dominant OR = 0.73 (0.48,1.14), recessive OR = 1.24 (0.37,4.12), dominant OR = 0.77 (0.50,1.17), co-dominant OR1 = 0.74 (0.48,1.14) and co-dominant OR2 = 1.13 (0.34,3.80)]. Additionally, rs6265 was unrelated to the age of MS onset according to both unadjusted and sex-adjusted cox-proportional models. Finally, rs6265 was not associated with the presence of spinal lesions (cervical or thoracic) at MS onset, according to both unadjusted and age and sex-adjusted logistic regression models. CONCLUSIONS We failed to establish an association between BDNF rs6265 and the risk of MS, the age of onset, the presence of spinal lesions, and the clinical manifestations at the onset.
Collapse
Affiliation(s)
- Ioannis Liampas
- Department of Neurology, School of Medicine, University Hospital of Larissa, University of Thessaly, 41110, Larissa, Greece
- Laboratory of Neurogenetics, School of Medicine, University of Thessaly, 41110, Larissa, Greece
| | - Daniil Tsirelis
- Department of Neurology, School of Medicine, University Hospital of Larissa, University of Thessaly, 41110, Larissa, Greece
- Laboratory of Neurogenetics, School of Medicine, University of Thessaly, 41110, Larissa, Greece
| | - Metaxia Dastamani
- Department of Neurology, School of Medicine, University Hospital of Larissa, University of Thessaly, 41110, Larissa, Greece
- Laboratory of Neurogenetics, School of Medicine, University of Thessaly, 41110, Larissa, Greece
| | - Stavroula-Ioanna Pariou
- Research Group of Clinical Pharmacology and Pharmacogenomics, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771, Athens, Greece
| | - Maria Papasavva
- Research Group of Clinical Pharmacology and Pharmacogenomics, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771, Athens, Greece
- Department of Pharmacy, School of Health Sciences, Frederick University, Nicosia, Cyprus
| | - Martha-Spyridoula Katsarou
- Research Group of Clinical Pharmacology and Pharmacogenomics, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771, Athens, Greece
| | - Annia Tsolakou
- Research Group of Clinical Pharmacology and Pharmacogenomics, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771, Athens, Greece
| | - Aristidis Tsatsakis
- Laboratory of Toxicology, School of Medicine, University of Crete, 71003, Heraklion, Greece
| | - Dimitrios P Bogdanos
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University General Hospital of Larissa, University of Thessaly, Viopolis 40500, Larissa, Greece
| | - Nikolaos Drakoulis
- Research Group of Clinical Pharmacology and Pharmacogenomics, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771, Athens, Greece
| | - Efthimios Dardiotis
- Department of Neurology, School of Medicine, University Hospital of Larissa, University of Thessaly, 41110, Larissa, Greece
- Laboratory of Neurogenetics, School of Medicine, University of Thessaly, 41110, Larissa, Greece
| | - Vasileios Siokas
- Department of Neurology, School of Medicine, University Hospital of Larissa, University of Thessaly, 41110, Larissa, Greece.
- Laboratory of Neurogenetics, School of Medicine, University of Thessaly, 41110, Larissa, Greece.
- Department of Neurology, Laboratory of Neurogenetics, Faculty of Medicine, University Hospital of Larissa, University of Thessaly, Mezourlo Hill, 41100, Biopolis, Greece.
| |
Collapse
|
14
|
Jakkamsetti MS, Kolusu AS, Rongala S, Arakareddy BP, Nori LP, Samudrala PK. Saroglitazar, a PPAR α/γ agonist alleviates 3-Nitropropionic acid induced neurotoxicity in rats: Unveiling the underlying mechanisms. Neurotoxicology 2024; 105:131-146. [PMID: 39326639 DOI: 10.1016/j.neuro.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
Saroglitazar (SGZ), a peroxisomal proliferated activated receptor α/γ agonist showed neuroprotective effects in various neurodegenerative disorders like Alzheimer's and Parkinson's. However, no studies were performed on Huntington's, so the goal of the current study is to examine the effect of SGZ on Huntington's disease like symptoms induced by 3-Nitropropionic acid. In this protocol, twenty-four rats were divided into four groups, each group consisting of 6 animals. Group 1: The control group received 1 % CMC 10 mg/kg, p.o. for 14 days. Groups 2, 3, and 4 received 3-NP 15 mg/kg, i.p. from Day 1 to Day 7. Groups 3 and 4 received SGZ 5 mg/kg, p.o. and 10 mg/kg, p.o. respectively once daily from day 1 to day 14. Various behavioral tests like OFT, rotarod, hanging wire, narrow beam walk, MWM, and Y-maze were performed. On day-15, the animals were euthanised by cervical dislocation and brain sample were isolated for biochemical and histopathological analysis. Administration of 3-NP showed a significant decrease in motor coordination and cognitive function. Furthermore, 3-NP altered the activity of acetylcholinesterase, anti-oxidant enzymes, Nrf-2, NF-κB, BDNF, CREB levels, and histological features. However, treatment with SGZ showed ameliorative effects in the 3-NP induced neurotoxicity via PPAR α/γ pathway by reducing motor dysfunction, memory impairment, cholinesterase levels, oxidative stress, neuroinflammation. It also enhanced the levels of Nrf-2, BDNF, and CREB expression and improved histological features. In conclusion, treatment with Saroglitazar attenuated Huntington's disease-like symptoms in rats which are induced by 3-NP via activation of PPAR α/γ pathway.
Collapse
Affiliation(s)
- Madhuri Suma Jakkamsetti
- Department of Pharmacology, Shri Vishnu College of Pharmacy (SVCP), Vishnupur, Bhimavaram, West Godavari, Andhra Pradesh 534202, India
| | - Aravinda Sai Kolusu
- Department of Pharmacology, Shri Vishnu College of Pharmacy (SVCP), Vishnupur, Bhimavaram, West Godavari, Andhra Pradesh 534202, India
| | - Suma Rongala
- Department of Pharmacology, Shri Vishnu College of Pharmacy (SVCP), Vishnupur, Bhimavaram, West Godavari, Andhra Pradesh 534202, India
| | - Bhanu Prakash Arakareddy
- Department of Pharmacology, Shri Vishnu College of Pharmacy (SVCP), Vishnupur, Bhimavaram, West Godavari, Andhra Pradesh 534202, India
| | - Lakshmi Prashanthi Nori
- Department of Pharmaceutics, Shri Vishnu College of Pharmacy (SVCP), Vishnupur, Bhimavaram, West Godavari, Andhra Pradesh 534202, India
| | - Pavan Kumar Samudrala
- Department of Pharmacology, Shri Vishnu College of Pharmacy (SVCP), Vishnupur, Bhimavaram, West Godavari, Andhra Pradesh 534202, India.
| |
Collapse
|
15
|
Vastrad B, Vastrad C. Screening and identification of key biomarkers associated with endometriosis using bioinformatics and next-generation sequencing data analysis. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2024; 25:116. [DOI: 10.1186/s43042-024-00572-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/23/2024] [Indexed: 01/04/2025] Open
Abstract
Abstract
Background
Endometriosis is a common cause of endometrial-type mucosa outside the uterine cavity with symptoms such as painful periods, chronic pelvic pain, pain with intercourse and infertility. However, the early diagnosis of endometriosis is still restricted. The purpose of this investigation is to identify and validate the key biomarkers of endometriosis.
Methods
Next-generation sequencing dataset GSE243039 was obtained from the Gene Expression Omnibus database, and differentially expressed genes (DEGs) between endometriosis and normal control samples were identified. After screening of DEGs, gene ontology (GO) and REACTOME pathway enrichment analyses were performed. Furthermore, a protein–protein interaction (PPI) network was constructed and modules were analyzed using the Human Integrated Protein–Protein Interaction rEference database and Cytoscape software, and hub genes were identified. Subsequently, a network between miRNAs and hub genes, and network between TFs and hub genes were constructed using the miRNet and NetworkAnalyst tool, and possible key miRNAs and TFs were predicted. Finally, receiver operating characteristic curve analysis was used to validate the hub genes.
Results
A total of 958 DEGs, including 479 upregulated genes and 479 downregulated genes, were screened between endometriosis and normal control samples. GO and REACTOME pathway enrichment analyses of the 958 DEGs showed that they were mainly involved in multicellular organismal process, developmental process, signaling by GPCR and muscle contraction. Further analysis of the PPI network and modules identified 10 hub genes, including vcam1, snca, prkcb, adrb2, foxq1, mdfi, actbl2, prkd1, dapk1 and actc1. Possible target miRNAs, including hsa-mir-3143 and hsa-mir-2110, and target TFs, including tcf3 (transcription factor 3) and clock (clock circadian regulator), were predicted by constructing a miRNA-hub gene regulatory network and TF-hub gene regulatory network.
Conclusions
This investigation used bioinformatics techniques to explore the potential and novel biomarkers. These biomarkers might provide new ideas and methods for the early diagnosis, treatment and monitoring of endometriosis.
Collapse
|
16
|
Mousavi-Nasab K, Amani M, Mostafalou S. The Effect of Trientine on AlCl3-Induced Cognitive Dysfunction and Biochemical Changes in the Hippocampus of Rats. Drug Res (Stuttg) 2024; 74:405-414. [PMID: 39173674 DOI: 10.1055/a-2381-6882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Cognitive impairments affect millions of people worldwide with an increasing prevalence. Research on their etiology and treatment is developing, nevertheless significant gaps remain. Trientine (TETA), as a copper chelator, has been shown to have beneficial effects in different human chronic diseases such as diabetic cardiomyopathy and neuropathy. Here, we examined the impact of TETA on AlCl3-induced neurocognitive dysfunctions and molecular changes in the hippocampus of rats.Thirty-six male Wistar rats (weighing 200-250 g) were randomly divided into four groups including control, TETA (100 mg/kg/day), AlCl3 (100 mg/kg/day), and AlCl3 (100 mg/kg/day)+TETA (100 mg/kg/day), and received chemicals by gavage for 30 days. At the end of the treatment, the open field maze, elevated plus maze, novel object recognition memory test, and shuttle box test were done. Then after, brain-derived neurotrophic factor (BDNF), glycogen synthase kinase-3 β (GSK-3β), acetylcholinesterase activity, oxidative stress markers, and inflammatory mediators were measured in the hippocampus.AlCl3 increased anxiety-like behaviors and impaired recognition and short-term memory. TETA was able to improve AlCl3-induced anxiety-like behaviors and short-term memory dysfunction. In the AlCl3-treated group, there was a significant increase in GSK-3β, oxidative stress, pro-inflammatory and pro-apoptotic markers, and decreased BDNF in the hippocampus. Co-administration of TETA was able to decrease lipid peroxidation, inflammation, GSK-3β, and acetylcholinesterase activity, and increase BDNF in the hippocampus compared with AlCl3-treated rats.It can be concluded that TETA was able to improve neurobehavioral and neurocognitive functions by alleviating oxidative stress, inflammation, and pro-apoptotic pathways leading to the normalization of BDNF and GSK-3β.
Collapse
Affiliation(s)
- Kian Mousavi-Nasab
- Department of Pharmacology & Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Amani
- Department of Physiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Sara Mostafalou
- Department of Pharmacology & Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
17
|
Lai G, Malavolta M, Marcozzi S, Bigossi G, Giuliani ME, Casoli T, Balietti M. Late-onset major depressive disorder: exploring the therapeutic potential of enhancing cerebral brain-derived neurotrophic factor expression through targeted microRNA delivery. Transl Psychiatry 2024; 14:352. [PMID: 39227372 PMCID: PMC11371930 DOI: 10.1038/s41398-024-02935-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 09/05/2024] Open
Abstract
Major depressive disorder (MDD) is a severe psychiatric condition that significantly impacts the overall quality of life. Although MDD can occur across all age groups, it is notably prevalent among older individuals, with the aggravating circumstance that the clinical condition is frequently overlooked and undertreated. Furthermore, older adults often encounter resistance to standard treatments, experience adverse events, and face challenges associated with polypharmacy. Given that late-life MDD is associated with heightened rates of disability and mortality, as well as imposing a significant economic and logistical burden on healthcare systems, it becomes imperative to explore novel therapeutic approaches. These could serve as either supplements to standard guidelines or alternatives for non-responsive patients, potentially enhancing the management of geriatric MDD patients. This review aims to delve into the potential of microRNAs targeting Brain-Derived Neurotrophic Factor (BDNF). In MDD, a significant decrease in both central and peripheral BDNF has been well-documented, raising implications for therapy response. Notably, BDNF appears to be a key player in the intricate interplay between microRNA-induced neuroplasticity deficits and neuroinflammation, both processes deeply implicated in the onset and progression of the disease. Special emphasis is placed on delivery methods, with a comprehensive comparison of the strengths and weaknesses of each proposed approach. Our hypothesis proposes that employing multiple microRNAs concurrently, with the ability to directly influence BDNF and activate closely associated pathways, may represent the most promising strategy. Regarding vehicles, although the perfect nanoparticle remains elusive, considering the trade-offs, liposomes emerge as the most suitable option.
Collapse
Affiliation(s)
- Giovanni Lai
- Advanced Technology Center for Aging Research and Geriatric Mouse Clinic, IRCCS INRCA, Ancona, Italy
| | - Marco Malavolta
- Advanced Technology Center for Aging Research and Geriatric Mouse Clinic, IRCCS INRCA, Ancona, Italy.
| | - Serena Marcozzi
- Advanced Technology Center for Aging Research and Geriatric Mouse Clinic, IRCCS INRCA, Ancona, Italy
| | - Giorgia Bigossi
- Advanced Technology Center for Aging Research and Geriatric Mouse Clinic, IRCCS INRCA, Ancona, Italy
| | - Maria Elisa Giuliani
- Advanced Technology Center for Aging Research and Geriatric Mouse Clinic, IRCCS INRCA, Ancona, Italy
| | - Tiziana Casoli
- Center of Neurobiology of Aging, IRCCS INRCA, Ancona, Italy
| | - Marta Balietti
- Center of Neurobiology of Aging, IRCCS INRCA, Ancona, Italy
| |
Collapse
|
18
|
Hedley KE, Gomez HM, Kecelioglu E, Carroll OR, Jobling P, Horvat JC, Tadros MA. Neonatal Chlamydia muridarum respiratory infection causes neuroinflammation within the brainstem during the early postnatal period. J Neuroinflammation 2024; 21:158. [PMID: 38879567 PMCID: PMC11179230 DOI: 10.1186/s12974-024-03150-3] [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: 01/29/2024] [Accepted: 06/10/2024] [Indexed: 06/19/2024] Open
Abstract
Respiratory infections are one of the most common causes of illness and morbidity in neonates worldwide. In the acute phase infections are known to cause wide-spread peripheral inflammation. However, the inflammatory consequences to the critical neural control centres for respiration have not been explored. Utilising a well characterised model of neonatal respiratory infection, we investigated acute responses within the medulla oblongata which contains key respiratory regions. Neonatal mice were intranasally inoculated within 24 h of birth, with either Chlamydia muridarum or sham-infected, and tissue collected on postnatal day 15, the peak of peripheral inflammation. A key finding of this study is that, while the periphery appeared to show no sex-specific effects of a neonatal respiratory infection, sex had a significant impact on the inflammatory response of the medulla oblongata. There was a distinct sex-specific response in the medulla coincident with peak of peripheral inflammation, with females demonstrating an upregulation of anti-inflammatory cytokines and males showing very few changes. Microglia also demonstrated sex-specificity with the morphology of females and males differing based upon the nuclei. Astrocytes showed limited changes during the acute response to neonatal infection. These data highlight the strong sex-specific impact of a respiratory infection can have on the medulla in the acute inflammatory phase.
Collapse
Affiliation(s)
- Kateleen E Hedley
- School of Biomedical Sciences & Pharmacy, The University of Newcastle Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Henry M Gomez
- School of Biomedical Sciences & Pharmacy, The University of Newcastle Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Eda Kecelioglu
- School of Biomedical Sciences & Pharmacy, The University of Newcastle Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Olivia R Carroll
- School of Biomedical Sciences & Pharmacy, The University of Newcastle Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Phillip Jobling
- School of Biomedical Sciences & Pharmacy, The University of Newcastle Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Jay C Horvat
- School of Biomedical Sciences & Pharmacy, The University of Newcastle Callaghan, NSW, 2308, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Melissa A Tadros
- School of Biomedical Sciences & Pharmacy, The University of Newcastle Callaghan, NSW, 2308, Australia.
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.
| |
Collapse
|
19
|
Balde A, Ramya CS, Nazeer RA. A review on current advancement in zebrafish models to study chronic inflammatory diseases and their therapeutic targets. Heliyon 2024; 10:e31862. [PMID: 38867970 PMCID: PMC11167310 DOI: 10.1016/j.heliyon.2024.e31862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 04/02/2024] [Accepted: 05/22/2024] [Indexed: 06/14/2024] Open
Abstract
Chronic inflammatory diseases are caused due to prolonged inflammation at a specific site of the body. Among other inflammatory diseases, bacterial meningitis, chronic obstructive pulmonary disease (COPD), atherosclerosis and inflammatory bowel diseases (IBD) are primarily focused on because of their adverse effects and fatality rates around the globe in recent times. In order to come up with novel strategies to eradicate these diseases, a clear understanding of the mechanisms of the diseases is needed. Similarly, detailed insight into the mechanisms of commercially available drugs and potent lead compounds from natural sources are also important to establish efficient therapeutic effects. Zebrafish is widely accepted as a model to study drug toxicity and the pharmacokinetic effects of the drug. Moreover, researchers use various inducers to trigger inflammatory cascades and stimulate physiological changes in zebrafish. The effect of these inducers contrasts with the type of zebrafish used in the investigation. Hence, a thorough analysis is required to study the current advancements in the zebrafish model for chronic inflammatory disease suppression. This review presents the most common inflammatory diseases, commercially available drugs, novel therapeutics, and their mechanisms of action for disease suppression. The review also provides a detailed description of various zebrafish models for these diseases. Finally, the future prospects and challenges for the same are described, which can help the researchers understand the potency of the zebrafish model and its further exploration for disease attenuation.
Collapse
Affiliation(s)
- Akshad Balde
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - Cunnathur Saravanan Ramya
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - Rasool Abdul Nazeer
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| |
Collapse
|
20
|
Ma LY, Jia B, Geng H, Liang J, Huo L. Poly(rC)-binding protein 1 alleviates neurotoxicity in 6-OHDA-induced SH-SY5Y cells and modulates glial cells in neuroinflammation. Brain Res 2024; 1832:148863. [PMID: 38492841 DOI: 10.1016/j.brainres.2024.148863] [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: 12/13/2023] [Revised: 03/03/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND Parkinson's disease (PD) is a debilitating neurodegenerative condition characterized by the loss of dopaminergic neurons and neuroinflammation. Previous research has identified the involvement of Poly (rC)-binding protein 1 (PCBP1) in certain degenerative diseases; however, its specific mechanisms in PD remain incompletely understood. METHODS In this study, 6-OHDA-induced neurotoxicity in the cell lines SH-SY5Y, BV-2 and HA, was used to evaluate the protective effects of PCBP1. We assessed alterations in BDNF levels in SY5Y cells, changes in GDNF expression in glial cells, as well as variations in HSP70 and NF-κB activation. Additionally, glial cells were used as the in vitro model for neuroinflammation mechanisms. RESULTS The results indicate that the overexpression of PCBP1 significantly enhances cell growth compared to the control plasmid pEGFP/N1 group. Overexpression of PCBP1 leads to a substantial reduction in early apoptosis rates in SH-SY5Y, HA, and BV-2 cells, with statistically significant differences (p < 0.05). Furthermore, the overexpression of PCBP1 in cells results in a marked increase in the expression of HSP70, GDNF, and BDNF, while reducing NF-κB expression. Additionally, in SH-SY5Y, HA, and BV-2 cells overexpressing PCBP1, there is a decrease in the inflammatory factor IL-6 compared to the control plasmid pEGFP/N1 group, while BV-2 cells exhibit a significant increase in the anti-inflammatory factor IL-10. CONCLUSION Our findings suggest that PCBP1 plays a substantial role in promoting cell growth and modulating the balance of neuroprotective and inflammatory factors. These results offer valuable insights into the potential therapeutic utility of PCBP1 in mitigating neuroinflammation and enhancing neuronal survival in PD.
Collapse
Affiliation(s)
- Ling-Yun Ma
- Central Laboratory, Department of Neurology, Fuxing Hospital, Capital Medical University, Beijing 100038, China
| | - Bingbing Jia
- Central Laboratory, Department of Neurology, Fuxing Hospital, Capital Medical University, Beijing 100038, China; Department of Neurology, Beijing Children's Hospital, Capital Medical University, Beijing 100045, China
| | - Haoming Geng
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Jiantao Liang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Lirong Huo
- Central Laboratory, Department of Neurology, Fuxing Hospital, Capital Medical University, Beijing 100038, China.
| |
Collapse
|
21
|
Ma J, Xie H, Yuan C, Shen J, Chen J, Chen Q, Liu J, Tong Q, Sun J. The gut microbial signatures of patients with lacunar cerebral infarction. Nutr Neurosci 2024; 27:620-636. [PMID: 37538045 DOI: 10.1080/1028415x.2023.2242121] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
BACKGROUND Emerging evidence revealed that gut microbial dysbiosis is involved in the pathogenesis of multiple neurological diseases, but there is little available data on the relationship between gut microbiota and lacunar cerebral infarction (LCI). METHODS Fecal samples from acute LCI patients (n = 65) and matched healthy controls (n = 65) were collected. The compositions and potential functions of the gut microbiota were estimated. RESULTS The results showed that there were significant gut microbial differences between LCI and control groups. Patients with LCI had higher abundances of genus Lactobacillus, Streptococcus, Veillonella, Acidaminococcus, Bacillus, Peptoclostridium, Intestinibacter, Alloscardovia and Cloacibacillus but lower proportions of genus Agathobacter and Lachnospiraceae_UCG-004. Investigating further these microbes such as Lactobacillus and Veillonella were correlated with clinical signs. Moreover, we found that 9 gene functions of gut microbiota were different between LCI patients and controls, which were associated with amino acid metabolism and inflammatory signal transduction. Notably, four optimal microbial markers were determined, and the combination of Streptococcus, Lactobacillus, Agathobacter, Lachnospiraceae_UCG-004 and the three risk factors achieved an area under the curve (AUC) value of 0.854 to distinguish LCI from controls. CONCLUSION These findings revealed the characterizing of gut microbiota in LCI patients and provided potential microbial biomarkers for clinical diagnosis of LCI.
Collapse
Affiliation(s)
- Jiaying Ma
- Department of Geriatrics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Huijia Xie
- Department of Geriatrics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Chengxiang Yuan
- Department of Neurology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Jie Shen
- Department of Neurology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Jiaxin Chen
- Department of Geriatrics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Qionglei Chen
- Department of Geriatrics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Jiaming Liu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Qiuling Tong
- Department of Neurology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Jing Sun
- Department of Geriatrics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, People's Republic of China
| |
Collapse
|
22
|
Seasons GM, Pellow C, Kuipers HF, Pike GB. Ultrasound and neuroinflammation: immune modulation via the heat shock response. Theranostics 2024; 14:3150-3177. [PMID: 38855178 PMCID: PMC11155413 DOI: 10.7150/thno.96270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/09/2024] [Indexed: 06/11/2024] Open
Abstract
Current pharmacological therapeutic approaches targeting chronic inflammation exhibit transient efficacy, often with adverse effects, limiting their widespread use - especially in the context of neuroinflammation. Effective interventions require the consideration of homeostatic function, pathway dysregulation, and pleiotropic effects when evaluating therapeutic targets. Signalling molecules have multiple functions dependent on the immune context, and this complexity results in therapeutics targeting a single signalling molecule often failing in clinical translation. Additionally, the administration of non-physiologic levels of neurotrophic or anti-inflammatory factors can alter endogenous signalling, resulting in unanticipated effects. Exacerbating these challenges, the central nervous system (CNS) is isolated by the blood brain barrier (BBB), restricting the infiltration of many pharmaceutical compounds into the brain tissue. Consequently, there has been marked interest in therapeutic techniques capable of modulating the immune response in a pleiotropic manner; ultrasound remains on this frontier. While ultrasound has been used therapeutically in peripheral tissues - accelerating healing in wounds, bone fractures, and reducing inflammation - it is only recently that it has been applied to the CNS. The transcranial application of low intensity pulsed ultrasound (LIPUS) has successfully mitigated neuroinflammation in vivo, in models of neurodegenerative disease across a broad spectrum of ultrasound parameters. To date, the underlying biological effects and signalling pathways modulated by ultrasound are poorly understood, with a diverse array of reported molecules implicated. The distributed nature of the beneficial response to LIPUS implies the involvement of an, as yet, undetermined upstream signalling pathway, homologous to the protective effect of febrile range hyperthermia in chronic inflammation. As such, we review the heat shock response (HSR), a protective signalling pathway activated by thermal and mechanical stress, as the possible upstream regulator of the anti-inflammatory effects of ultrasound.
Collapse
Affiliation(s)
- Graham M. Seasons
- Hotchkiss Brain Institute, University of Calgary, Alberta, T2N 4N1, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada
| | - Carly Pellow
- Hotchkiss Brain Institute, University of Calgary, Alberta, T2N 4N1, Canada
- Department of Radiology, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada
| | - Hedwich F. Kuipers
- Hotchkiss Brain Institute, University of Calgary, Alberta, T2N 4N1, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada
- Department of Cell Biology & Anatomy, Hotchkiss Brain Institute and Snyder Institute for Chronic Diseases, University of Calgary, Alberta, T2N 1N4, Canada
| | - G. Bruce Pike
- Hotchkiss Brain Institute, University of Calgary, Alberta, T2N 4N1, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada
- Department of Radiology, Cumming School of Medicine, University of Calgary, Alberta, T2N 1N4, Canada
| |
Collapse
|
23
|
Mohammadinia F, Esmaeili‐Mahani S, Abbasnejad M, Dogani M, Poorrahimi AM. Methyl jasmonate ameliorates pain-induced learning and memory impairments through regulating the expression of genes involved in neuroinflammation. Brain Behav 2024; 14:e3502. [PMID: 38680072 PMCID: PMC11056706 DOI: 10.1002/brb3.3502] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 03/27/2024] [Accepted: 04/06/2024] [Indexed: 05/01/2024] Open
Abstract
OBJECTIVE Orofacial pain with high prevalence is one of the substantial human health issues. The importance of this matter became more apparent when it was revealed that orofacial pain, directly and indirectly, affects cognition performances. Currently, researchers have focused on investigating pharmaceutics to alleviate pain and ameliorate its subsequent cognitive impairments. DESIGN In this study, the rats were first treated with the central administration of methyl jasmonate (MeJA), which is an antioxidant and anti-inflammatory bio-compound. After 20 min, orofacial pain was induced in the rats by the injection of capsaicin in their dental pulp. Subsequently, the animals' pain behaviors were analyzed, and the effects of pain and MeJA treatments on rats learning and memory were evaluated/compared using the Morris water maze (MWM) test. In addition, the expression of tumor necrosis factor-α (TNF-α), IL-1β, BDNF, and COX-2 genes in the rats' hippocampus was evaluated using real-time polymerase chain reaction. RESULTS Experiencing orofacial pain resulted in a significant decline in the rats learning and memory. However, the central administration of 20 μg/rat of MeJA effectively mitigated these impairments. In the MWM, the performance of the MeJA-treated rats showed a two- to threefold improvement compared to the nontreated ones. Moreover, in the hippocampus of pain-induced rats, the expression of pro-inflammatory factors TNF-α, IL-1β, and COX-2 significantly increased, whereas the BDNF expression decreased. In contrast, MeJA downregulated the pro-inflammatory factors and upregulated the BDNF by more than 50%. CONCLUSIONS These findings highlight the notable antinociceptive potential of MeJA and its ability to inhibit pain-induced learning and memory dysfunction through its anti-inflammatory effect.
Collapse
Affiliation(s)
- Fatemeh Mohammadinia
- Kerman Neuroscience Research Center, Institute of NeuropharmacologyKerman University of Medical SciencesKermanIran
- Department of Biology, Faculty of SciencesShahid Bahonar University of KermanKermanIran
| | - Saeed Esmaeili‐Mahani
- Kerman Neuroscience Research Center, Institute of NeuropharmacologyKerman University of Medical SciencesKermanIran
- Department of Biology, Faculty of SciencesShahid Bahonar University of KermanKermanIran
| | - Mehdi Abbasnejad
- Department of Biology, Faculty of SciencesShahid Bahonar University of KermanKermanIran
| | - Manijeh Dogani
- Department of Biology, Faculty of SciencesShahid Bahonar University of KermanKermanIran
| | - Ali Mohammad Poorrahimi
- Kerman Neuroscience Research Center, Institute of NeuropharmacologyKerman University of Medical SciencesKermanIran
| |
Collapse
|
24
|
Balan I, Boero G, Chéry SL, McFarland MH, Lopez AG, Morrow AL. Neuroactive Steroids, Toll-like Receptors, and Neuroimmune Regulation: Insights into Their Impact on Neuropsychiatric Disorders. Life (Basel) 2024; 14:582. [PMID: 38792602 PMCID: PMC11122352 DOI: 10.3390/life14050582] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/18/2024] [Accepted: 04/28/2024] [Indexed: 05/26/2024] Open
Abstract
Pregnane neuroactive steroids, notably allopregnanolone and pregnenolone, exhibit efficacy in mitigating inflammatory signals triggered by toll-like receptor (TLR) activation, thus attenuating the production of inflammatory factors. Clinical studies highlight their therapeutic potential, particularly in conditions like postpartum depression (PPD), where the FDA-approved compound brexanolone, an intravenous formulation of allopregnanolone, effectively suppresses TLR-mediated inflammatory pathways, predicting symptom improvement. Additionally, pregnane neurosteroids exhibit trophic and anti-inflammatory properties, stimulating the production of vital trophic proteins and anti-inflammatory factors. Androstane neuroactive steroids, including estrogens and androgens, along with dehydroepiandrosterone (DHEA), display diverse effects on TLR expression and activation. Notably, androstenediol (ADIOL), an androstane neurosteroid, emerges as a potent anti-inflammatory agent, promising for therapeutic interventions. The dysregulation of immune responses via TLR signaling alongside reduced levels of endogenous neurosteroids significantly contributes to symptom severity across various neuropsychiatric disorders. Neuroactive steroids, such as allopregnanolone, demonstrate efficacy in alleviating symptoms of various neuropsychiatric disorders and modulating neuroimmune responses, offering potential intervention avenues. This review emphasizes the significant therapeutic potential of neuroactive steroids in modulating TLR signaling pathways, particularly in addressing inflammatory processes associated with neuropsychiatric disorders. It advances our understanding of the complex interplay between neuroactive steroids and immune responses, paving the way for personalized treatment strategies tailored to individual needs and providing insights for future research aimed at unraveling the intricacies of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Irina Balan
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Department of Psychiatry, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Giorgia Boero
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA;
| | - Samantha Lucenell Chéry
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Neuroscience Curriculum, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Minna H. McFarland
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Neuroscience Curriculum, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alejandro G. Lopez
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - A. Leslie Morrow
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Department of Psychiatry, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
25
|
Jiang D, Liu H, Li T, Zhao S, Yang K, Yao F, Zhou B, Feng H, Wang S, Shen J, Tang J, Zhang YX, Wang Y, Guo C, Tang TS. Agomirs upregulating carboxypeptidase E expression rescue hippocampal neurogenesis and memory deficits in Alzheimer's disease. Transl Neurodegener 2024; 13:24. [PMID: 38671492 PMCID: PMC11046780 DOI: 10.1186/s40035-024-00414-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Adult neurogenesis occurs in the subventricular zone (SVZ) and the subgranular zone of the dentate gyrus in the hippocampus. The neuronal stem cells in these two neurogenic niches respond differently to various physiological and pathological stimuli. Recently, we have found that the decrement of carboxypeptidase E (CPE) with aging impairs the maturation of brain-derived neurotrophic factor (BDNF) and neurogenesis in the SVZ. However, it remains unknown whether these events occur in the hippocampus, and what the role of CPE is in the adult hippocampal neurogenesis in the context of Alzheimer's disease (AD). METHODS In vivo screening was performed to search for miRNA mimics capable of upregulating CPE expression and promoting neurogenesis in both neurogenic niches. Among these, two agomirs were further assessed for their effects on hippocampal neurogenesis in the context of AD. We also explored whether these two agomirs could ameliorate behavioral symptoms and AD pathology in mice, using direct intracerebroventricular injection or by non-invasive intranasal instillation. RESULTS Restoration of CPE expression in the hippocampus improved BDNF maturation and boosted adult hippocampal neurogenesis. By screening the miRNA mimics targeting the 5'UTR region of Cpe gene, we developed two agomirs that were capable of upregulating CPE expression. The two agomirs significantly rescued adult neurogenesis and cognition, showing multiple beneficial effects against the AD-associated pathologies in APP/PS1 mice. Of note, noninvasive approach via intranasal delivery of these agomirs improved the behavioral and neurocognitive functions of APP/PS1 mice. CONCLUSIONS CPE may regulate adult hippocampal neurogenesis via the CPE-BDNF-TrkB signaling pathway. This study supports the prospect of developing miRNA agomirs targeting CPE as biopharmaceuticals to counteract aging- and disease-related neurological decline in human brains.
Collapse
Affiliation(s)
- Dongfang Jiang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hongmei Liu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Tingting Li
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Song Zhao
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Keyan Yang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Fuwen Yao
- Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Bo Zhou
- Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, 100101, China
| | - Haiping Feng
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Sijia Wang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jiaqi Shen
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jinglan Tang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Present Address: Department of Psychology, UC San Diego, La Jolla, CA, 92093, USA
| | - Yu-Xin Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yun Wang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Caixia Guo
- Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Tie-Shan Tang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
26
|
Feng X, Ma X, Li J, Zhou Q, Liu Y, Song J, Liu J, Situ Q, Wang L, Zhang J, Lin F. Inflammatory Pathogenesis of Post-stroke Depression. Aging Dis 2024; 16:AD.2024.0203. [PMID: 38377025 PMCID: PMC11745428 DOI: 10.14336/ad.2024.0203] [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: 12/12/2023] [Accepted: 02/03/2024] [Indexed: 02/22/2024] Open
Abstract
Post-stroke depression (PSD) is a complex mood disorder that emerges in individuals following a stroke, characterized by the development of depressive symptoms. The pathogensis of PSD is diverse, with inflammation playing a vital role in its onset and progression. Emerging evidence suggests that microglial activation, astrocyte responses, nuclear factor κB(NF-κB) signaling, dysregulation of the hypothalamic pituitary adrenal (HPA) axis, alterations in brain-derived neurotrophic factor (BDNF) expression, neurotransmitter imbalances, adenosine triphosphate (ATP) and its receptors and oxidative stress are intricately linked to the pathogenesis of PSD. The involvement of inflammatory cytokines in these processes highlights the significance of the inflammatory pathway. Integrating these hypotheses, the inflammatory mechanism offers a novel perspective to expand therapeutic strategies for PSD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Jingzhi Zhang
- School of Acupuncture and Tuina, School of Health and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Facai Lin
- School of Acupuncture and Tuina, School of Health and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| |
Collapse
|
27
|
Wang Y, Liang J, Xu B, Yang J, Wu Z, Cheng L. TrkB/BDNF signaling pathway and its small molecular agonists in CNS injury. Life Sci 2024; 336:122282. [PMID: 38008209 DOI: 10.1016/j.lfs.2023.122282] [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: 02/18/2023] [Revised: 10/19/2023] [Accepted: 11/18/2023] [Indexed: 11/28/2023]
Abstract
As one of the most prevalent neurotrophic factors in the central nervous system (CNS), brain-derived neurotrophic factor (BDNF) plays a significant role in CNS injury by binding to its specific receptor Tropomyosin-related kinase receptor B (TrkB). The BDNF/TrkB signaling pathway is crucial for neuronal survival, structural changes, and plasticity. BDNF acts as an axonal growth and extension factor, a pro-survival factor, and a synaptic modulator in the CNS. BDNF also plays an important role in the maintenance and plasticity of neuronal circuits. Several studies have demonstrated the importance of BDNF in the treatment and recovery of neurodegenerative and neurotraumatic disorders. By undertaking in-depth study on the mechanism of BDNF/TrkB function, important novel therapeutic strategies for treating neuropsychiatric disorders have been discovered. In this review, we discuss the expression patterns and mechanisms of the TrkB/BDNF signaling pathway in CNS damage and introduce several intriguing small molecule TrkB receptor agonists produced over the previous several decades.
Collapse
Affiliation(s)
- Yujin Wang
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; Medical School, Tongji University, Shanghai 200433, China
| | - Jing Liang
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; School of Stomatology, Tongji University, Shanghai 200072, China
| | - Boyu Xu
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; Medical School, Tongji University, Shanghai 200433, China
| | - Jin Yang
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China; Medical School, Tongji University, Shanghai 200433, China
| | - Zhourui Wu
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China.
| | - Liming Cheng
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China; Key Laboratory of Spine and Spinal cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai 200072, China.
| |
Collapse
|
28
|
Sun MK, Alkon DL. Treating Alzheimer's Disease: Focusing on Neurodegenerative Consequences. J Alzheimers Dis 2024; 101:S263-S274. [PMID: 39422958 DOI: 10.3233/jad-240479] [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] [Indexed: 10/19/2024]
Abstract
Neurodegenerative disorders involve progressive dysfunction and loss of synapses and neurons and brain atrophy, slowly declining memories and cognitive skills, throughout a long process. Alzheimer's disease (AD), the leading neurodegenerative disorder, suffers from a lack of effective therapeutic drugs. Decades of efforts targeting its pathologic hallmarks, amyloid plaques and neurofibrillary tangles, in clinical trials have produced therapeutics with marginal benefits that lack meaningful clinical improvements in cognition. Delivering meaningful clinical therapeutics to treat or prevent neurodegenerative disorders thus remains a great challenge to scientists and clinicians. Emerging evidence, however, suggests that dysfunction of various synaptogenic signaling pathways participates in the neurodegenerative progression, resulting in deterioration of operation/structure of the synaptic networks involved in cognition. These derailed endogenous signaling pathways and disease processes are potential pharmacological targets for the therapies. Therapeutics with meaningful clinical benefit in cognition may depend on the effectiveness of arresting and reversing the neurodegenerative process through these targets. In essence, promoting neuro-regeneration may represent the only option to recover degenerated synapses and neurons. These potential directions in clinical trials for AD therapeutics with meaningful clinical benefit in cognitive function are summarized and discussed.
Collapse
|
29
|
Balan I, Grusca A, O’Buckley TK, Morrow AL. Neurosteroid [3α,5α]-3-hydroxy-pregnan-20-one enhances IL-10 production via endosomal TRIF-dependent TLR4 signaling pathway. Front Endocrinol (Lausanne) 2023; 14:1299420. [PMID: 38179300 PMCID: PMC10765172 DOI: 10.3389/fendo.2023.1299420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/24/2023] [Indexed: 01/06/2024] Open
Abstract
Background Previous studies demonstrated the inhibitory effect of allopregnanolone (3α,5α-THP) on the activation of inflammatory toll-like receptor 4 (TLR4) signals in RAW264.7 macrophages and the brains of selectively bred alcohol-preferring (P) rats. In the current study, we investigated the impact of 3α,5α-THP on the levels of IL-10 and activation of the TRIF-dependent endosomal TLR4 pathway. Methods The amygdala and nucleus accumbens (NAc) of P rats, which exhibit innately activated TLR4 pathways as well as RAW264.7 cells, were used. Enzyme-linked immunosorbent assays (ELISA) and immunoblotting assays were used to ascertain the effects of 3α,5α-THP on the TRIF-dependent endosomal TLR4 pathway and endosomes were isolated to examine translocation of TLR4 and TRIF. Additionally, we investigated the effects of 3α,5α-THP and 3α,5α-THDOC (0.1, 0.3, and 1.0 µM) on the levels of IL-10 in RAW264.7 macrophages. Finally, we examined whether inhibiting TRIF (using TRIF siRNA) in RAW264.7 cells altered the levels of IL-10. Results 3α,5α-THP administration facilitated activation of the endosomal TRIF-dependent TLR4 pathway in males, but not female P rats. 3α,5α-THP increased IL-10 levels (+13.2 ± 6.5%) and BDNF levels (+21.1 ± 11.5%) in the male amygdala. These effects were associated with increases in pTRAM (+86.4 ± 28.4%), SP1 (+122.2 ± 74.9%), and PI(3)K-p110δ (+61.6 ± 21.6%), and a reduction of TIRAP (-13.7 ± 6.0%), indicating the activation of the endosomal TRIF-dependent TLR4 signaling pathway. Comparable effects were observed in NAc of these animals. Furthermore, 3α,5α-THP enhanced the accumulation of TLR4 (+43.9 ± 11.3%) and TRIF (+64.8 ± 32.8%) in endosomes, with no significant effect on TLR3 accumulation. Additionally, 3α,5α-THP facilitated the transition from early endosomes to late endosomes (increasing Rab7 levels: +35.8 ± 18.4%). In RAW264.7 cells, imiquimod (30 µg/mL) reduced IL-10 while 3α,5α-THP and 3α,5α-THDOC (0.1, 0.3, and 1.0 µM) restored IL-10 levels. To determine the role of the TRIF-dependent TLR4 signaling pathway in IL-10 production, the downregulation of TRIF (-62.9 ± 28.2%) in RAW264.7 cells led to a reduction in IL-10 levels (-42.3 ± 8.4%). TRIF (-62.9 ± 28.2%) in RAW264.7 cells led to a reduction in IL-10 levels (-42.3 ± 8.4%) and 3α,5α-THP (1.0 µM) no longer restored the reduced IL-10 levels. Conclusion The results demonstrate 3α,5α-THP enhancement of the endosomal TLR4-TRIF anti-inflammatory signals and elevations of IL-10 in male P rat brain that were not detected in female P rat brain. These effects hold significant implications for controlling inflammatory responses in both the brain and peripheral immune cells.
Collapse
Affiliation(s)
- Irina Balan
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Adelina Grusca
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Todd K. O’Buckley
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - A. Leslie Morrow
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
30
|
Petruso F, Giff A, Milano B, De Rossi M, Saccaro L. Inflammation and emotion regulation: a narrative review of evidence and mechanisms in emotion dysregulation disorders. Neuronal Signal 2023; 7:NS20220077. [PMID: 38026703 PMCID: PMC10653990 DOI: 10.1042/ns20220077] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023] Open
Abstract
Emotion dysregulation (ED) describes a difficulty with the modulation of which emotions are felt, as well as when and how these emotions are experienced or expressed. It is a focal overarching symptom in many severe and prevalent neuropsychiatric diseases, including bipolar disorders (BD), attention deficit/hyperactivity disorder (ADHD), and borderline personality disorder (BPD). In all these disorders, ED can manifest through symptoms of depression, anxiety, or affective lability. Considering the many symptomatic similarities between BD, ADHD, and BPD, a transdiagnostic approach is a promising lens of investigation. Mounting evidence supports the role of peripheral inflammatory markers and stress in the multifactorial aetiology and physiopathology of BD, ADHD, and BPD. Of note, neural circuits that regulate emotions appear particularly vulnerable to inflammatory insults and peripheral inflammation, which can impact the neuroimmune milieu of the central nervous system. Thus far, few studies have examined the link between ED and inflammation in BD, ADHD, and BPD. To our knowledge, no specific work has provided a critical comparison of the results from these disorders. To fill this gap in the literature, we review the known associations and mechanisms linking ED and inflammation in general, and clinically, in BD, ADHD, and BD. Our narrative review begins with an examination of the routes linking ED and inflammation, followed by a discussion of disorder-specific results accounting for methodological limitations and relevant confounding factors. Finally, we critically discuss both correspondences and discrepancies in the results and comment on potential vulnerability markers and promising therapeutic interventions.
Collapse
Affiliation(s)
| | - Alexis E. Giff
- Department of Neuroscience, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Switzerland
| | - Beatrice A. Milano
- Sant’Anna School of Advanced Studies, Pisa, Italy
- University of Pisa, Pisa, Italy
| | | | - Luigi Francesco Saccaro
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Switzerland
- Department of Psychiatry, Geneva University Hospital, Switzerland
| |
Collapse
|
31
|
Boahen A, Hu D, Adams MJ, Nicholls PK, Greene WK, Ma B. Bidirectional crosstalk between the peripheral nervous system and lymphoid tissues/organs. Front Immunol 2023; 14:1254054. [PMID: 37767094 PMCID: PMC10520967 DOI: 10.3389/fimmu.2023.1254054] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
The central nervous system (CNS) influences the immune system generally by regulating the systemic concentration of humoral substances (e.g., cortisol and epinephrine), whereas the peripheral nervous system (PNS) communicates specifically with the immune system according to local interactions/connections. An imbalance between the components of the PNS might contribute to pathogenesis and the further development of certain diseases. In this review, we have explored the "thread" (hardwiring) of the connections between the immune system (e.g., primary/secondary/tertiary lymphoid tissues/organs) and PNS (e.g., sensory, sympathetic, parasympathetic, and enteric nervous systems (ENS)) in health and disease in vitro and in vivo. Neuroimmune cell units provide an anatomical and physiological basis for bidirectional crosstalk between the PNS and the immune system in peripheral tissues, including lymphoid tissues and organs. These neuroimmune interactions/modulation studies might greatly contribute to a better understanding of the mechanisms through which the PNS possibly affects cellular and humoral-mediated immune responses or vice versa in health and diseases. Physical, chemical, pharmacological, and other manipulations of these neuroimmune interactions should bring about the development of practical therapeutic applications for certain neurological, neuroimmunological, infectious, inflammatory, and immunological disorders/diseases.
Collapse
Affiliation(s)
- Angela Boahen
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Seri-Kembangan, Selangor, Malaysia
| | - Dailun Hu
- Department of Pathogenic Biology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Murray J. Adams
- School of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA, Australia
| | - Philip K. Nicholls
- School of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA, Australia
| | - Wayne K. Greene
- School of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA, Australia
| | - Bin Ma
- School of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA, Australia
| |
Collapse
|
32
|
Lee J, Thomas Broome S, Jansen MI, Mandwie M, Logan GJ, Marzagalli R, Musumeci G, Castorina A. Altered Hippocampal and Striatal Expression of Endothelial Markers and VIP/PACAP Neuropeptides in a Mouse Model of Systemic Lupus Erythematosus. Int J Mol Sci 2023; 24:11118. [PMID: 37446298 DOI: 10.3390/ijms241311118] [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: 05/22/2023] [Revised: 07/01/2023] [Accepted: 07/02/2023] [Indexed: 07/15/2023] Open
Abstract
Neuropsychiatric systemic lupus erythematosus (NPSLE) is one of the most common and severe manifestations of lupus; however, its pathogenesis is still poorly understood. While there is sparse evidence suggesting that the ongoing autoimmunity may trigger pathogenic changes to the central nervous system (CNS) microvasculature, culminating in inflammatory/ischemic damage, further evidence is still needed. In this study, we used the spontaneous mouse model of SLE (NZBWF1 mice) to investigate the expression of genes and proteins associated with endothelial (dys)function: tissue and urokinase plasminogen activators (tPA and uPA), intercellular and vascular adhesion molecules 1 (ICAM-1 and VCAM-1), brain derived neurotrophic factor (BDNF), endothelial nitric oxide synthase (eNOS) and Krüppel-like factor 4 (KLF4) and neuroprotection/immune modulation: pituitary adenylate cyclase-activating peptide (PACAP), vasoactive intestinal peptide (VIP), PACAP receptor (PAC1), VIP receptors 1 and 2 (VPAC1 and VPAC2). Analyses were carried out both in the hippocampus and striatum of SLE mice of two different age groups (2 and 7 months old), since age correlates with disease severity. In the hippocampus, we identified a gene/protein expression profile indicative of mild endothelial dysfunction, which increased in severity in aged SLE mice. These alterations were paralleled by moderate alterations in the expression of VIP, PACAP and related receptors. In contrast, we report a robust upregulation of endothelial activation markers in the striatum of both young and aged mice, concurrent with significant induction of the VIP/PACAP system. These data identify molecular signatures of endothelial alterations in the hippocampus and striatum of NZBWF1 mice, which are accompanied by a heightened expression of endogenous protective/immune-modulatory neuropeptides. Collectively, our results support the idea that NPSLE may cause alterations of the CNS micro-vascular compartment that cannot be effectively counteracted by the endogenous activity of the neuropeptides PACAP and VIP.
Collapse
Affiliation(s)
- Jayden Lee
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Science, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Sarah Thomas Broome
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Science, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Margo Iris Jansen
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Science, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Mawj Mandwie
- Gene Therapy Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney and Sydney Children's Hospitals Network, Westmead, NSW 2145, Australia
| | - Grant J Logan
- Gene Therapy Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney and Sydney Children's Hospitals Network, Westmead, NSW 2145, Australia
| | - Rubina Marzagalli
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Science, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, Anatomy, Histology and Movement Sciences Section, School of Medicine, University of Catania, 95124 Catania, Italy
| | - Alessandro Castorina
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Science, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| |
Collapse
|
33
|
Zhang L, Lu RR, Xu RH, Wang HH, Feng WS, Zheng XK. Naringenin and apigenin ameliorates corticosterone-induced depressive behaviors. Heliyon 2023; 9:e15618. [PMID: 37215924 PMCID: PMC10192682 DOI: 10.1016/j.heliyon.2023.e15618] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 05/24/2023] Open
Abstract
Background Depression is a common kind of mental illness, and it becomes the main health burden in the world. Purpose The aim of this study was to investigate the antidepressant effects of naringin and apigenin isolated from Chrysanthemum morifolium Ramatis. Methods Firstly, 20 mg/kg corticosterone (CORT) was injected into mice to establish an in vivo model of depression. After treated with different dosages of naringenin and apigenin for 3 weeks, the mice underwent a series of behavioral experiments. Following this, all mice were sacrificed and biochemical analyses were performed. Subsequently, CORT (500 μM) induced PC12 cells was used as an in vitro model of depression, and lipopolysaccharide (LPS) (1 μg ml-1) induced N9 microglia cells was used as an in vitro model of neuroinflammation in N9 microglia cells, to investigate the neuroprotective mechanisms of naringenin and apigenin. Results Results showed that the naringenin and apigenin treatment ameliorated CORT-induced sucrose preference decrease and immobility time increase, elevated the 5-hydroxytryptamine(5-HT), dopamine (DA) and norepinephrine (NE) levels, and enhanced the cAMP-response element binding protein (CREB) and brain derived neurotrophic factor (BDNF) protein expressions in the hippocampus. The results showed that the naringenin and apigenin treatment improved the PC-12 cell viability through reducing apoptosis rate induced by CORT. Furthermore, naringenin and apigenin were able to inhibit the activation of N9 cells after LPS induction, and shift microglia from proinflammatory M1 microglia toward anti-inflammatory M2 microglia, as evidenced by the decreased ratio of M1 type microglia marker CD86 and M2 type microglia marker CD86. Conclusion These results suggested that naringenin and apigenin may improve depressive behaviors through promoting BDNF and inhibiting neuroinflammation and neuronal apoptosis.
Collapse
Affiliation(s)
- Li Zhang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of PR China, Zhengzhou 450046, China
| | - Ren-Rui Lu
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Rui-Hao Xu
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Hui-Hui Wang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Wei-Sheng Feng
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of PR China, Zhengzhou 450046, China
| | - Xiao-Ke Zheng
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
- Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of PR China, Zhengzhou 450046, China
| |
Collapse
|
34
|
Singh D, Preetam Ambati A, Aich P. Sex and Time: Important Variables for Understanding the Impact of Constant Darkness on Behavior, Brain, and Physiology. Neuroscience 2023; 519:73-89. [PMID: 36966879 DOI: 10.1016/j.neuroscience.2023.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 03/02/2023] [Accepted: 03/13/2023] [Indexed: 04/03/2023]
Abstract
The circadian clock can coordinate, regulate and predict physiology and behavior in response to the standard light-dark (LD: 12 h light and 12 h dark) cycle. If we alter the LD cycle by exposing mice to constant darkness (DD: 00 h light and 24 h dark), it can perturb behavior, the brain, and associated physiological parameters. The length of DD exposure and the sex of experimental animals are crucial variables that could alter the impact of DD on the brain, behavior, and physiology, which have not yet been explored. We exposed mice to DD for three and five weeks and studied their impact on (1) behavior, (2) hormones, (3) the prefrontal cortex, and (4) metabolites in male and female mice. We also studied the effect of three weeks of standard light-dark cycle restoration after five weeks of DD on the parameters mentioned above. We found that DD exposure was associated with anxiety-like behavior, increased corticosterone and pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β), downregulated neurotrophins (BDNF and NGF), and altered metabolites profile in a duration of DD exposure and sex-dependent manner. Females showed a more robust adaptation than males under DD exposure. Three weeks of restoration was adequate to establish homeostasis in both sexes. To the best of our knowledge, this study is the first of its kind to look at how DD exposure impacts physiology and behavior as a function of sex- and time. These findings would have translational value and may help in establishing sex-specific interventions for addressing DD-related psychological issues.
Collapse
Affiliation(s)
- Dhyanendra Singh
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, PO - Bhimpur-Padanpur, Jatni - 752050, Dist. -Khurda, Odisha, India; Centre for Interdisciplinary Science (CIS), National Institute of Science Education and Research (NISER), HBNI, PO - Bhimpur-Padanpur, Jatni - 752050, Dist. -Khurda, Odisha, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India
| | - Abhilash Preetam Ambati
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, PO - Bhimpur-Padanpur, Jatni - 752050, Dist. -Khurda, Odisha, India; Centre for Interdisciplinary Science (CIS), National Institute of Science Education and Research (NISER), HBNI, PO - Bhimpur-Padanpur, Jatni - 752050, Dist. -Khurda, Odisha, India
| | - Palok Aich
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, PO - Bhimpur-Padanpur, Jatni - 752050, Dist. -Khurda, Odisha, India; Centre for Interdisciplinary Science (CIS), National Institute of Science Education and Research (NISER), HBNI, PO - Bhimpur-Padanpur, Jatni - 752050, Dist. -Khurda, Odisha, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India.
| |
Collapse
|
35
|
Zhang Y, Ma R, Deng Q, Wang W, Cao C, Yu C, Li S, Shi L, Tian J. S-adenosylmethionine improves cognitive impairment in D-galactose-induced brain aging by inhibiting oxidative stress and neuroinflammation. J Chem Neuroanat 2023; 128:102232. [PMID: 36632907 DOI: 10.1016/j.jchemneu.2023.102232] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 12/29/2022] [Accepted: 01/06/2023] [Indexed: 01/09/2023]
Abstract
Oxidative stress and neuroinflammation play crucial roles in aging. S-adenosylmethionine (SAM), a popular supplement, is a potential antioxidant and candidate therapy for depression. This study aimed to evaluate the neuroprotective effects of SAM on D-galactose-induced brain aging and explore its underlying mechanisms. Brain aging model was established with D-galactose (180 mg/kg/day) for 8 weeks. During the last 4 weeks, SAM (16 mg/kg) was co-administrated with D-galactose. Behavior tests were used to assess cognitive function and depression-like behaviors of rats. Results showed that cognitive impairment and depression-like behaviors were reversed by SAM. SAM reduced neuronal cell loss, increased brain-derived neurotrophic factor level in the hippocampus, inhibited amyloid-β level and microglia activation, as well as pro-inflammatory factors levels in the hippocampus and serum. Further, SAM enhanced antioxidant capacity and attenuated cholinergic damage by reducing malondialdehyde levels, increasing acetylcholine levels, expression levels of α7 nicotinic acetylcholine receptor (α7nAChR), nuclear factor erythrocyte 2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1) in the hippocampus. Above all, SAM has a potential neuroprotective effect on ameliorating cognitive impairment in brain aging, which is related to inhibition of oxidative stress and neuroinflammation, as well as α7nAChR signals. DATA AVAILABILITY: Data will be made available on request.
Collapse
Affiliation(s)
- Yawen Zhang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Rui Ma
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Qian Deng
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Wencheng Wang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Chi Cao
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Chunyang Yu
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Shulin Li
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Lei Shi
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Jianying Tian
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China.
| |
Collapse
|
36
|
Xu QQ, Su ZR, Yang W, Zhong M, Xian YF, Lin ZX. Patchouli alcohol attenuates the cognitive deficits in a transgenic mouse model of Alzheimer's disease via modulating neuropathology and gut microbiota through suppressing C/EBPβ/AEP pathway. J Neuroinflammation 2023; 20:19. [PMID: 36717922 PMCID: PMC9887791 DOI: 10.1186/s12974-023-02704-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 01/22/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a chronic neurodegenerative disease characterized by progressive cognitive dysfunctions and behavioral impairments. Patchouli alcohol (PA), isolated from Pogostemonis Herba, exhibits multiple pharmacological properties, including neuroprotective effects. This study aimed to investigate the therapeutic effects of PA against AD using the TgCRND8 transgenic AD mouse model, and to explore the underlying mechanisms targeting CCAAT/enhancer-binding protein β/asparagine endopeptidase (C/EBPβ/AEP) signaling pathway. METHODS After genotyping to confirm the transgenicity, drug treatments were administered intragastrically once daily to 3-month-old TgCRND8 mice for 4 consecutive months. Several behavioral tests were applied to assess different aspects of neurological functions. Then the brain and colon tissues were harvested for in-depth mechanistic studies. To further verify whether PA exerts anti-AD effects via modulating C/EBPβ/AEP signaling pathway in TgCRND8 mice, adeno-associated virus (AAV) vectors encoding CEBP/β were bilaterally injected into the hippocampal CA1 region in TgCRND8 mice to overexpress C/EBPβ. Additionally, the fecal microbiota transplantation (FMT) experiment was performed to verify the potential role of gut microbiota on the anti-AD effects of PA. RESULTS Our results showed that PA treatment significantly improved activities of daily living (ADL), ameliorated the anxiety-related behavioral deficits and cognitive impairments in TgCRND8 mice. PA modulated the amyloid precursor protein (APP) processing. PA also markedly reduced the levels of beta-amyloid (Aβ) 40 and Aβ42, suppressed Aβ plaque burdens, inhibited tau protein hyperphosphorylation at several sites and relieved neuroinflammation in the brains of TgCRND8 mice. Moreover, PA restored gut dysbiosis and inhibited the activation of the C/EBPβ/AEP signaling pathway in the brain and colon tissues of TgCRND8 mice. Interestingly, PA strikingly alleviated the AD-like pathologies induced by the overexpression of C/EBPβ in TgCRND8 mice. Additionally, the FMT of fecal microbiota from the PA-treated TgCRND8 mice significantly alleviated the cognitive impairments and AD-like pathologies in the germ-free TgCRND8 mice. CONCLUSION All these findings amply demonstrated that PA could ameliorate the cognitive deficits in TgCRND8 mice via suppressing Aβ plaques deposition, hyperphosphorylation of tau protein, neuroinflammation and gut dysbiosis through inhibiting the activation of C/EBPβ/AEP pathway, suggesting that PA is a promising naturally occurring chemical worthy of further development into the pharmaceutical treatment of AD.
Collapse
Affiliation(s)
- Qing-Qing Xu
- grid.10784.3a0000 0004 1937 0482School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
| | - Zi-Ren Su
- grid.411866.c0000 0000 8848 7685Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Wen Yang
- grid.10784.3a0000 0004 1937 0482School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
| | - Mei Zhong
- grid.10784.3a0000 0004 1937 0482School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
| | - Yan-Fang Xian
- grid.10784.3a0000 0004 1937 0482School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
| | - Zhi-Xiu Lin
- grid.10784.3a0000 0004 1937 0482School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China ,grid.10784.3a0000 0004 1937 0482Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China ,grid.10784.3a0000 0004 1937 0482Li Dak Sum Yip Yio Chin R&D Centre for Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
| |
Collapse
|
37
|
Antidepressive-Like Effect of Aegle marmelos Leaf Extract in Chronic Unpredictable Mild Stress-Induced Depression-Like Behaviour in Rats. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6479953. [PMID: 36593774 PMCID: PMC9805397 DOI: 10.1155/2022/6479953] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/30/2022] [Accepted: 11/17/2022] [Indexed: 12/25/2022]
Abstract
Background Depression is a psychiatric disorder leading to anhedonia and lack of interest and motivation. Depressive symptoms are triggered by stressful life events, and patients with major depression are at significantly increased risk of attempting suicide. The crucial concern in depression treatment with antidepressant medications is that few weeks are required to show the therapeutic effect along with moderate side effects. The use of herbal medications is a new strategy for the treatment of depression which is often based on medicinal plants.Aegle marmelos (L.) Corr. (family: Rutaceae) is reported to have several actions on the central nervous system producing beneficial effects in anxiety, Alzheimer's disease, Parkinson's disease, epilepsy, and convulsion. Thus, the current investigation designed to assess the antidepressant activity of the standardized hydroethanolic extract of Aegle marmelos (EAM) leaves in male rats exposed to the chronic unpredictable mild stress (CUMS) paradigm. Methods Rats were divided in 5 groups. The control group was not subjected to experimental CUMS paradigm, while 4 other groups were subjected to CUMS paradigm to induce depression-like behaviour from day 1 to day 28. Following the CUMS paradigm, 4 groups were divided as CUMS disease control, CUMS+EAM (150 mg/kg, p.o.), CUMS+EAM (300 mg/kg, p.o.), and CUMS+imipramine (15 mg/kg, p.o.), and treatment was given for seven consecutive days to the respective groups (day 29 to day 35). Behavioural parameters such as open field test, forced swim test, sucrose feeding test, and tail suspension test on day 1, day 28, and day 35 were measured, and biochemical parameters such as plasma corticosterone level, serotonergic system (5-HT, 5-HIAA, and 5-HT/5-HIAA), mitochondrial function, and proinflammatory mediators (TNF-α, IL-1β, and IL-6) were estimated in hippocampus (HIP) and prefrontal cortex (PFC) regions of the brain on day 35, after the behavioural observations. On the other hand, phytochemical profile of Aegle marmelos was done. Results On day 35, EAM (300 mg/kg) significantly reduced the immobility time during the tail suspension test from 208.66 ± 4.72 s to 108.83 ± 4.81 s and forced swim test from 200.16 ± 4.12 s to 148.5 ± 4.58 s. It also enhanced the behavioural parameters in the open field test such as ambulation from 26.5 ± 2.14 to 56.5 ± 1.80, rearing from 8.33 ± 0.71 to 19 ± 0.57, time spent in centre from 9.16 ± 0.9 to 17.16 ± 0.79 s, total distance travelled from 2.36 ± 0.12 to 4.68 ± 0.10 m, and anhedonia in the sucrose feeding test from 109.33 ± 1.08 to 135.83 ± 3.91 mL. The stimulation of the HPA axis resulting elevated corticosterone level caused by CUMS was reduced by EAM (300 mg/kg) from 80.12 ± 2.020 to 48.25 ± 2.407 μg/dL. Furthermore, EAM (300 mg/kg) increase CUMS-induced changes in serotonin (5-HT) level in HIP and PFC from 3.132 ± 0.09586 to 4.518 ± 0.1812 and 4.308 ± 0.1593 to 5.262 ± 0.1014 ng/mg protein, respectively. EAM (300 mg/kg) significantly attenuated the CUMS-induced changes in proinflammatory cytokine production and mitochondrial function in HIP and PFC. One group used to determine the acute toxicity as per OECD-23 standard protocol which resulted that 300 mg/kg EAM has no significant acute toxicity. Total phenolic content and total flavonoid content of standardized hydroalcoholic extract of AM was found 95.024 ± 2.431 and 36.820 ± 3.41, respectively, and additional identification tests showed the presence of alkaloids, tannins, saponins, cardiac glycosides, flavonoids, and terpenoids. Conclusion On the basis of findings, EAM can be inferred as a potential antidepressant-like effect of this plan in preclinical research.
Collapse
|
38
|
Yanai S, Tago T, Toyohara J, Arasaki T, Endo S. Reversal of spatial memory impairment by phosphodiesterase 3 inhibitor cilostazol is associated with reduced neuroinflammation and increased cerebral glucose uptake in aged male mice. Front Pharmacol 2022; 13:1031637. [PMID: 36618932 PMCID: PMC9810637 DOI: 10.3389/fphar.2022.1031637] [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: 08/30/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
The nucleotide second messenger 3', 5'-cyclic adenosine monophosphate (cAMP) and 3', 5'-cyclic guanosine monophosphate (cGMP) mediate fundamental functions of the brain, including learning and memory. Phosphodiesterase 3 (PDE3) can hydrolyze both cAMP and cGMP and appears to be involved in the regulation of their contents in cells. We previously demonstrated that long-term administration of cilostazol, a PDE3 inhibitor, maintained good memory performance in aging mice. Here, we report on studies aimed at determining whether cilostazol also reverses already-impaired memory in aged male mice. One month of oral 1.5% cilostazol administration in 22-month-old mice reversed age-related declines in hippocampus-dependent memory tasks, including the object recognition and the Morris water maze. Furthermore, cilostazol reduced neuroinflammation, as evidenced by immunohistochemical staining, and increased glucose uptake in the brain, as evidence by positron emission tomography (PET) with 2-deoxy-2-[18F]fluoro-d-glucose ([18F]FDG). These results suggest that already-expressed memory impairment in aged male mice that depend on cyclic nucleotide signaling can be reversed by inhibition of PDE3. The reversal of age-related memory impairments may occur in the central nervous system, either through cilostazol-enhanced recall or strengthening of weak memories that otherwise may be resistant to recall.
Collapse
Affiliation(s)
- Shuichi Yanai
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Tetsuro Tago
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Jun Toyohara
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Tomoko Arasaki
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Shogo Endo
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan,*Correspondence: Shogo Endo,
| |
Collapse
|
39
|
Jakaria M, Belaidi AA, Southon A, Dent KA, Lane DJR, Bush AI, Ayton S. Receptor-Independent Anti-Ferroptotic Activity of TrkB Modulators. Int J Mol Sci 2022; 23:ijms232416205. [PMID: 36555849 PMCID: PMC9784883 DOI: 10.3390/ijms232416205] [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: 10/12/2022] [Revised: 12/05/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Dysregulated brain-derived neurotrophic factor (BDNF)/tropomyosin receptor kinase B (TrkB) signalling is implicated in several neurodegenerative diseases, including Alzheimer's disease. A failure of neurotrophic support may participate in neurodegenerative mechanisms, such as ferroptosis, which has likewise been implicated in this disease class. The current study investigated whether modulators of TrkB signalling affect ferroptosis. Cell viability, C11 BODIPY, and cell-free oxidation assays were used to observe the impact of TrkB modulators, and an immunoblot assay was used to detect TrkB expression. TrkB modulators such as agonist BDNF, antagonist ANA-12, and inhibitor K252a did not affect RSL3-induced ferroptosis sensitivity in primary cortical neurons expressing detectable TrkB receptors. Several other modulators of the TrkB receptor, including agonist 7,8-DHF, activator phenelzine sulphate, and inhibitor GNF-5837, conferred protection against a range of ferroptosis inducers in several immortalised neuronal and non-neuronal cell lines, such as N27 and HT-1080 cells. We found these immortalised cell lines lack detectable TrkB receptor expression, so the anti-ferroptotic activity of these TrkB modulators was most likely due to their inherent radical-trapping antioxidant properties, which should be considered when interpreting their experimental findings. These modulators or their variants could be potential anti-ferroptotic therapeutics for various diseases.
Collapse
|
40
|
Krishnan R, Jang YS, Kim JO, Yoon SY, Rajendran R, Oh MJ. Temperature dependent cellular, and epigenetic regulatory mechanisms underlying the antiviral immunity in sevenband grouper to nervous necrosis virus infection. FISH & SHELLFISH IMMUNOLOGY 2022; 131:898-907. [PMID: 36334701 DOI: 10.1016/j.fsi.2022.10.068] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/25/2022] [Accepted: 10/31/2022] [Indexed: 06/16/2023]
Abstract
Changes in the thermal optima of fish impacts changes in the physiology and immune response associated with infections. The present study showed that at suboptimal temperatures (17 °C), the host tries to evade viral infection by downregulating the inflammatory response through enhanced neuronal protection. There was significantly less abundance of IgM + B cells in the 17 °C group compared to that in the 25 °C group. An increased macrophage population (Iba1+) during the survival phase in fish challenged at 25 °C demonstrated inflammation. Optimal temperature challenge activated virus-induced senescence in brain cells, demonstrated with a heterochromatin-associated H3K9me3 histone mark. There was an abundant expression of anti-inflammatory cytokines in the brain of fish at the suboptimal challenge. Besides the cytokines, the expression of BDNF was significantly higher in the suboptimally challenged group, suggesting that its neuronal protection activity following NNV infection is mediated through TGFβ. The suboptimal challenge resulted in H3k9ac displaying transcriptional competency, activation of trained immunity H3K4me3, and enrichment of H3 histone-lysine-4 monomethylation (H3K4me1), resulting in a robust re-stimulatory immune response. The observations from the H4 modifications showed that besides H4K12ac and H4K20m3, all the assayed modifications were significantly higher in suboptimal convalescent fishes. The suboptimally challenged fish acquired more methylation along cytosine residues than the optimally infected fish. Together, these observations suggest that optimal temperature results in an immune priming effect, whereas the protection enabled in suboptimal convalescent fishes is operated through epigenetically controlled trained immune functions.
Collapse
Affiliation(s)
- Rahul Krishnan
- Department of Aqualife Medicine, Chonnam National University, Yeosu, 59629, Republic of Korea.
| | - Yo-Seb Jang
- Department of Aqualife Medicine, Chonnam National University, Yeosu, 59629, Republic of Korea.
| | - Jong-Oh Kim
- Department of Microbiology, Pukyong National University, Busan, Republic of Korea.
| | - Su-Young Yoon
- Department of Aqualife Medicine, Chonnam National University, Yeosu, 59629, Republic of Korea.
| | - Rahul Rajendran
- Department of Aqualife Medicine, Chonnam National University, Yeosu, 59629, Republic of Korea.
| | - Myung-Joo Oh
- Department of Aqualife Medicine, Chonnam National University, Yeosu, 59629, Republic of Korea.
| |
Collapse
|
41
|
Ding J, Lian J, Wang J, Yang S, Li H, Shen H, Sun Q, Li X, Chen G. The role of Tenascin C in intracerebral hemorrhage-induced secondary brain injury in rats via induction of neuronal cell death and neuroinflammation. J Chem Neuroanat 2022; 125:102147. [PMID: 36028204 DOI: 10.1016/j.jchemneu.2022.102147] [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: 03/15/2022] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Spontaneous intracerebral hemorrhage (ICH) is a major cause of stroke that causes high rates of disability and mortality in adults. Tenascin C (TNC) protein, one of the matricellular proteins associated with platelet-derived growth factor receptor (PDGFR) activation, has been reported to induce neuronal apoptosis. However, the role and underlying mechanisms of TNC in ICH-induced secondary brain injury (SBI) have not yet been fully explained. The main purpose of this study was to explore the role of TNC and its potential mechanisms in ICH. METHODS An ICH model was established by injecting autologous blood into the right basal ganglia in male Sprague Dawley (SD) rats, and imatinib, an inhibitor of PDGFR, was used to inhibit the release of TNC. RESULTS We found that TNC protein was significantly increased in the brain tissues after ICH and expressed in both neurons and microglia. We also found that the TNC level was elevated in the cerebrospinal fluid (CSF) after ICH. Additionally, we observed that the infiltration of activated microglia and the release of TNFα and IL-1β induced by ICH were decreased after inhibition of the protein levels of TNC and cleaved-TNC by a chemical inhibitor (imatinib). Furthermore, imatinib improved neuronal cell death and neurobehavioral abnormalities induced by ICH. CONCLUSION In summary, our study revealed that TNC protein plays an important role in ICH-induced SBI, and inhibition of TNC could alleviate ICH-induced neuroinflammation, neuronal cell death, and neurobehaviour. Therefore, TNC may be a potential therapeutic target for ICH-induced SBI.
Collapse
Affiliation(s)
- Jiasheng Ding
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, China
| | - Jinrong Lian
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, China
| | - Jiahe Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, China
| | - Siyuan Yang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, China
| | - Qing Sun
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, China.
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Institute of Stroke Research, Soochow University, China
| |
Collapse
|
42
|
Wang J, Dai GD. Comparative Effects of Brominated Flame Retardants BDE-209, TBBPA, and HBCD on Neurotoxicity in Mice. Chem Res Toxicol 2022; 35:1512-1518. [PMID: 35950316 DOI: 10.1021/acs.chemrestox.2c00126] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Brominated flame retardants (BFRs) are ubiquitous industrial chemicals. In China, BFRs that are applied in large quantities include decabromodiphenyl ether (BDE-209), tetrabromobisphenol A (TBBPA), and hexabromocyclododecane (HBCD). Although findings are not always unequivocal, mounting evidence in vivo suggests that the BFRs have potential neurotoxicity. The present study aimed to assess and compare the neurotoxic effects of these three BFRs' exposure. Male mice were orally exposed to BDE-209, TBBPA, or HBCD at 50 and 100 mg/kg bw/day for 28 days. The cognitive behavior, oxidative stress (ROS, MDA, and GSH), apoptosis-related genes (caspase-3, bax, and bcl-2), memory-related proteins (BDNF and PSD-95), and neurotransmitters (AChE and ChAT) were detected comparatively. Results showed that high doses of BDE-209, TBBPA, and HBCD exposure impaired spatial memory of mice, elevated ROS and MDA and reduced GSH levels of hippocampus, upregulated caspase-3 and bax expressions, decreased BDNF and PSD-95 levels, and disordered AChE and ChAT levels. Notably, BDE-209 caused greater adverse effects > HBCD > TBBPA. This study confirms and extends that these three BFRs had similar neurotoxic effects at current concentrations, although they may be more or less toxic.
Collapse
Affiliation(s)
- Juan Wang
- Clinical Nursing Department, Nursing College, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Guo-Dong Dai
- Department of Neurosurgery, Xianning Central Hospital, Xianning 437100, PR China
| |
Collapse
|
43
|
Ren Q, Sun J, Xu D, Xie H, Ye M, Zhao Y. A Dietary Supplement Containing Micronutrients, Phosphatidylserine, and Docosahexaenoic Acid Counteracts Cognitive Impairment in D-Galactose-Induced Aged Rats. Front Nutr 2022; 9:931734. [PMID: 35866081 PMCID: PMC9294405 DOI: 10.3389/fnut.2022.931734] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/13/2022] [Indexed: 11/17/2022] Open
Abstract
At present, it is a trend to use dietary supplements to prevent age-related cognitive impairment. This study aimed to investigate the effects of a dietary supplement enriched with micronutrients, phosphatidylserine, and docosahexaenoic acid on cognitive performance using a D-galactose (D-gal) induced aging rat model. Seven-month-old male Sprague-Dawley rats were randomly divided into five groups, including the control group, D-gal model group, and low-dose (2 g/kg body weight), medium-dose (6 g/kg body weight), and high-dose (10 g/kg body weight) dietary supplement intervention groups, which were investigated for 13 weeks. The dietary supplement intervention was found to improve cognitive performance in Morris water maze test, increase superoxidase dismutase activity, reduce malondialdehyde activity, decrease tumor necrosis factor-α and interleukin-6 concentrations, inhibit the activation of astrocytes, and elevate brain-derived neurotrophic factor protein and mRNA expression in the brains of D-gal-induced aged rats. This dietary supplement customized for the aged can be applied to the restoration of cognitive performance by enhancing antioxidant and anti-neuroinflammatory abilities, up-regulating neurotrophic factors, and inhibiting the activation of astrocytes. These results will be useful for future studies focused on implementation in humans.
Collapse
Affiliation(s)
- Qian Ren
- Department of Clinical Nutrition, Huadong Hospital Affiliated to Fudan University, Shanghai, China
- Department of Clinical Nutrition, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Jianqin Sun
- Department of Clinical Nutrition, Huadong Hospital Affiliated to Fudan University, Shanghai, China
- *Correspondence: Jianqin Sun,
| | - Danfeng Xu
- Department of Clinical Nutrition, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Hua Xie
- Department of Clinical Nutrition, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Mengyao Ye
- Department of Clinical Nutrition, Huadong Hospital Affiliated to Fudan University, Shanghai, China
- Department of Endocrinology and Metabolism, Wenzhou Integrated Traditional Chinese and Western Medicine Hospital, Wenzhou, China
| | - Yanfang Zhao
- Department of Clinical Nutrition, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
44
|
Xia F, Zeng Q, Chen J. Circulating brain-derived neurotrophic factor dysregulation and its linkage with lipid level, stenosis degree, and inflammatory cytokines in coronary heart disease. J Clin Lab Anal 2022; 36:e24546. [PMID: 35666604 PMCID: PMC9279961 DOI: 10.1002/jcla.24546] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/17/2022] [Accepted: 05/24/2022] [Indexed: 11/07/2022] Open
Abstract
Background Brain‐derived neurotrophic factor (BDNF) regulates the lipid metabolism, atherosclerosis plaque formation, and inflammatory process, while the study about its clinical role in coronary heart disease (CHD) is few. The present study intended to explore the expression of BDNF and its relationship with stenosis, inflammation, and adhesion molecules in CHD patients. Methods After serum samples were obtained from 207 CHD patients, BDNF, tumor necrosis factor‐alpha (TNF‐α), interleukin (IL)‐1β, IL‐6, IL‐8, IL‐17A, vascular cell adhesion molecule‐1 (VCAM‐1), and intercellular adhesion molecule‐1 (ICAM‐1) levels were determined using ELISA. Then, the BDNF level was also examined in 40 disease controls (DCs) and 40 healthy controls (HCs), separately. Results BDNF was lower in CHD patients than in DCs and HCs (median (95% confidential interval) value: 5.6 (3.5–9.6) ng/mL vs. 10.7 (6.1–17.0) ng/mL and 12.6 (9.4–18.2) ng/mL, both p < 0.001). BDNF could well distinguish CHD patients from DCs (area under the curve [AUC]: 0.739) and HCs (AUC: 0.857). BDNF was negatively associated with triglyceride (p = 0.014), total cholesterol (p = 0.037), and low‐density lipoprotein cholesterol (p = 0.008). BDNF was negatively associated with CRP (p < 0.001), TNF‐α (p < 0.001), IL‐1β (p = 0.008), and IL‐8 (p < 0.001). BDNF was negatively related to VCAM‐1 (p < 0.001) and ICAM‐1 (p = 0.003). BDNF was negatively linked with the Gensini score (p < 0.001). Conclusion BDNF reflects the lipid dysregulation, inflammatory status, and stenosis degree in CHD patients.
Collapse
Affiliation(s)
- Feng Xia
- Department of Cardiology, Wuhan Asia General Hospital, Wuhan, China
| | - Qingrong Zeng
- Department of Cardiology, Wuhan Asia General Hospital, Wuhan, China
| | - Jing Chen
- Department of Critical Care Medicine, Wuhan Asia General Hospital, Wuhan, China
| |
Collapse
|
45
|
Mehterov N, Minchev D, Gevezova M, Sarafian V, Maes M. Interactions Among Brain-Derived Neurotrophic Factor and Neuroimmune Pathways Are Key Components of the Major Psychiatric Disorders. Mol Neurobiol 2022; 59:4926-4952. [PMID: 35657457 DOI: 10.1007/s12035-022-02889-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/17/2022] [Indexed: 10/25/2022]
Abstract
The purpose of this review is to summarize the current knowledge regarding the reciprocal associations between brain-derived neurotrophic factor (BDNF) and immune-inflammatory pathways and how these links may explain the involvement of this neurotrophin in the immune pathophysiology of mood disorders and schizophrenia. Toward this end, we delineated the protein-protein interaction (PPI) network centered around BDNF and searched PubMed, Scopus, Google Scholar, and Science Direct for papers dealing with the involvement of BDNF in the major psychosis, neurodevelopment, neuronal functions, and immune-inflammatory and related pathways. The PPI network was built based on the significant interactions of BDNF with neurotrophic (NTRK2, NTF4, and NGFR), immune (cytokines, STAT3, TRAF6), and cell-cell junction (CTNNB, CDH1) DEPs (differentially expressed proteins). Enrichment analysis shows that the most significant terms associated with this PPI network are the tyrosine kinase receptor (TRKR) and Src homology region two domain-containing phosphatase-2 (SHP2) pathways, tyrosine kinase receptor signaling pathways, positive regulation of kinase and transferase activity, cytokine signaling, and negative regulation of the immune response. The participation of BDNF in the immune response and its interactions with neuroprotective and cell-cell adhesion DEPs is probably a conserved regulatory process which protects against the many detrimental effects of immune activation and hyperinflammation including neurotoxicity. Lowered BDNF levels in mood disorders and schizophrenia (a) are associated with disruptions in neurotrophic signaling and activated immune-inflammatory pathways leading to neurotoxicity and (b) may interact with the reduced expression of other DEPs (CTNNB1, CDH1, or DISC1) leading to multiple aberrations in synapse and axonal functions.
Collapse
Affiliation(s)
- Nikolay Mehterov
- Department of Medical Biology, Medical University of Plovdiv, Plovdiv, Bulgaria.,Research Institute at Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Danail Minchev
- Department of Medical Biology, Medical University of Plovdiv, Plovdiv, Bulgaria.,Research Institute at Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Maria Gevezova
- Department of Medical Biology, Medical University of Plovdiv, Plovdiv, Bulgaria.,Research Institute at Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Victoria Sarafian
- Department of Medical Biology, Medical University of Plovdiv, Plovdiv, Bulgaria.,Research Institute at Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Michael Maes
- Faculty of Medicine, Department of Psychiatry, Chulalongkorn University, Bangkok, 10330, Thailand. .,Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria. .,Department of Psychiatry, IMPACT Strategic Research Centre, Deakin University, Geelong, VIC, Australia.
| |
Collapse
|
46
|
Maroofi A, Bagheri Rouch A, Naderi N, Damirchi A. Effects of two different exercise paradigms on cardiac function, BDNF-TrkB expression, and myocardial protection in the presence and absence of Western diet. IJC HEART & VASCULATURE 2022; 40:101022. [PMID: 35399608 PMCID: PMC8991101 DOI: 10.1016/j.ijcha.2022.101022] [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: 11/23/2021] [Revised: 03/17/2022] [Accepted: 03/30/2022] [Indexed: 12/11/2022]
Abstract
Background Brain-derived neurotrophic factor (BDNF) -tropomyosin-related kinase receptor B (TrkB) signaling is a vital regulator of myocardial performance. Here, we tested the impact of high-intensity interval training (HIIT) and moderate-intensity continuous training (MICT) on heart function, metabolic parameters, and serum/cardiac BDNF (with its TrkB receptor) in animals fed a Western (WD) or regular diet (ND). Further, myocardial expression of pro-inflammatory cytokine interleukin-18 (IL-18) and cardioprotective molecule heme oxygens-1 (HO-1) were monitored. Methods Wistar rats were divided into HIIT, MICT, and sedentary (SED), all fed a WD or ND, for 12 weeks. Heart function, protein expression, and serum factors were assessed via echocardiography, western blotting, and ELISA, respectively. Results WD plus SED caused insulin resistance, dyslipidemia, visceral fat deposition, serum BDNF depletion as well as cardiac upregulation of IL-18 and downregulation of HO-1, without affecting, heart function and BDNF-TrkB expression. The cardiometabolic risk factors, serum BDNF losses, and IL-18 overexpression were similarly obviated by HIIT and MICT, although HO-1 expression was boosted by HIIT exclusively (even in ND). HIIT enhanced heart function, regardless of the diet. HIIT augmented cardiac BDNF expression, with a significant difference between ND and WD. Likewise, HIIT instigated TrkB expression only in ND. Conclusions HIIT and MICT can cope with myocardial inflammation and cardiometabolic risk factors in WD consumers and, exclusively, HIIT may grant further protection by increasing heart function, BDNF-TrkB expression, and HO-1 expression. Thus, the HIIT paradigm should be considered as a preference for subjects who require heart function to be preserved or enhanced.
Collapse
Affiliation(s)
- Abdulbaset Maroofi
- Department of Exercise Physiology, Faculty of Physical Education & Sport Sciences, University of Guilan, Rasht, Iran
| | - Ahmadreza Bagheri Rouch
- Department of Exercise Physiology, Faculty of Physical Education & Sport Sciences, University of Guilan, Rasht, Iran
| | - Nasim Naderi
- Rajaie Cardiovascular, Medical & Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Arsalan Damirchi
- Department of Exercise Physiology, Faculty of Physical Education & Sport Sciences, University of Guilan, Rasht, Iran
| |
Collapse
|
47
|
Jin T, Zhang Y, Botchway BOA, Zhang J, Fan R, Zhang Y, Liu X. Curcumin can improve Parkinson's disease via activating BDNF/PI3k/Akt signaling pathways. Food Chem Toxicol 2022; 164:113091. [PMID: 35526734 DOI: 10.1016/j.fct.2022.113091] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 02/07/2023]
Abstract
Parkinson's disease is a common progressive neurodegenerative disease, and presently has no curative agent. Curcumin, as one of the natural polyphenols, has great potential in neurodegenerative diseases and other different pathological settings. The brain-derived neurotrophic factor (BDNF) and phosphatidylinositol 3 kinase (PI3k)/protein kinase B (Akt) signaling pathways are significantly involved nerve regeneration and anti-apoptotic activities. Currently, relevant studies have confirmed that curcumin has an optimistic impact on neuroprotection via regulating BDNF and PI3k/Akt signaling pathways in neurodegenerative disease. Here, we summarized the relationship between BDNF and PI3k/Akt signaling pathway, the main biological functions and neuroprotective effects of curcumin via activating BDNF and PI3k/Akt signaling pathways in Parkinson's disease. This paper illustrates that curcumin, as a neuroprotective agent, can delay the progression of Parkinson's disease by protecting nerve cells.
Collapse
Affiliation(s)
- Tian Jin
- Department of Histology and Embryology, Medical College, Shaoxing University, Zhejiang, China
| | - Yong Zhang
- Department of Histology and Embryology, Medical College, Shaoxing University, Zhejiang, China
| | - Benson O A Botchway
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Zhang
- Department of Pharmacology, Medical College, Shaoxing University, Zhejiang, China
| | - Ruihua Fan
- School of Life Science, Shaoxing University, Zhejiang, China
| | - Yufeng Zhang
- Department of Histology and Embryology, Medical College, Shaoxing University, Zhejiang, China
| | - Xuehong Liu
- Department of Histology and Embryology, Medical College, Shaoxing University, Zhejiang, China.
| |
Collapse
|
48
|
Sims SK, Wilken-Resman B, Smith CJ, Mitchell A, McGonegal L, Sims-Robinson C. Brain-Derived Neurotrophic Factor and Nerve Growth Factor Therapeutics for Brain Injury: The Current Translational Challenges in Preclinical and Clinical Research. Neural Plast 2022; 2022:3889300. [PMID: 35283994 PMCID: PMC8906958 DOI: 10.1155/2022/3889300] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 02/04/2022] [Indexed: 01/11/2023] Open
Abstract
Ischemic stroke and traumatic brain injury (TBI) are among the leading causes of death and disability worldwide with impairments ranging from mild to severe. Many therapies are aimed at improving functional and cognitive recovery by targeting neural repair but have encountered issues involving efficacy and drug delivery. As a result, therapeutic options for patients are sparse. Neurotrophic factors are one of the key mediators of neural plasticity and functional recovery. Neurotrophic factors such as brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) serve as potential therapeutic options to increase neural repair and recovery as they promote neuroprotection and regeneration. BDNF and NGF have demonstrated the ability to improve functional recovery in preclinical and to a lesser extent clinical studies. Direct and indirect methods to increase levels of neurotrophic factors in animal models have been successful in improving postinjury outcome measures. However, the translation of these studies into clinical trials has been limited. Preclinical experiments have largely failed to result in significant impacts in clinical research. This review will focus on the administration of these neurotrophic factors in preclinical and clinical stroke and TBI and the challenges in translating these therapies from the bench to the clinic.
Collapse
Affiliation(s)
- Serena-Kaye Sims
- Medical University of South Carolina, 173 Ashley Ave, Charleston, SC 29424, USA
| | | | - Crystal J. Smith
- Medical University of South Carolina, 173 Ashley Ave, Charleston, SC 29424, USA
| | - Ashley Mitchell
- Medical University of South Carolina, 173 Ashley Ave, Charleston, SC 29424, USA
| | - Lilly McGonegal
- College of Charleston, 66 George Street, Charleston, SC 29424, USA
| | | |
Collapse
|
49
|
Xie J, Liu L, Guo H, Bao Q, Hu P, Li H, Che H, Xie W. Orally administered melanin from Sepiapharaonis ink ameliorates depression-anxiety-like behaviors in DSS-induced colitis by mediating inflammation pathway and regulating apoptosis. Int Immunopharmacol 2022; 106:108625. [PMID: 35180627 DOI: 10.1016/j.intimp.2022.108625] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/12/2022] [Accepted: 02/10/2022] [Indexed: 12/20/2022]
Abstract
The effects of intestinal inflammation on the brain and behavior have received a lot of attention. Melanin (MSI) from Sepiapharaonis ink as an emerging functional food, it exhibited a significant protective effect on dextran sulfate sodium (DSS) induced colitis in previous study. In present study, C57BL/6J mice were free to drink 2.5% DSS solution to establish the colitis model. During the DSS treatment, mice were orally administrated with MSI once per day (75, 150, and 300 mg/kg, respectively). The results showed that MSI treatment ameliorated the depression and anxiety symptoms of colitis mice. Further mechanism studies indicated that MSI alleviated inflammatory response by adjusting cytokines TNF-α, IL-1β, IFN-γ, and IL-10, and proteins NLRP3/ASC/caspase-1 inflammasome), inhibited the activation of microglia, restored brain synaptic density, reduced oxidative stress (SOD, MDA) and regulated apoptosis (tunel staining, caspase-3). MSI could modulate depression-anxiety states by targeting inflammation, nerve tissue, oxidative stress and apoptosis. MSI administration could serve as an emerging blue food and nutrition strategy for the prevention of digestive tract inflammation and behavioral disorders.
Collapse
Affiliation(s)
- Jingwen Xie
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Shandong, Qingdao 266042, China
| | - Lin Liu
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Shandong, Qingdao 266042, China
| | - Hao Guo
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Shandong, Qingdao 266042, China
| | - Qi Bao
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Shandong, Qingdao 266042, China
| | - Penglong Hu
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Shandong, Qingdao 266042, China
| | - Hongyan Li
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Shandong, Qingdao 266042, China; Shandong Provincial Key Laboratory of Biochemical Engineering, Shandong, Qingdao 266042, China; Shandong Provincial Key Laboratory of Biochemical Engineering, Shandong, Qingdao 266042, China
| | - Hongxia Che
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Shandong, Qingdao 266042, China; Shandong Provincial Key Laboratory of Biochemical Engineering, Shandong, Qingdao 266042, China; Shandong Provincial Key Laboratory of Biochemical Engineering, Shandong, Qingdao 266042, China
| | - Wancui Xie
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Shandong, Qingdao 266042, China; Shandong Provincial Key Laboratory of Biochemical Engineering, Shandong, Qingdao 266042, China; Shandong Provincial Key Laboratory of Biochemical Engineering, Shandong, Qingdao 266042, China.
| |
Collapse
|
50
|
Dolcetti E, Bruno A, Azzolini F, Gilio L, Moscatelli A, De Vito F, Pavone L, Iezzi E, Gambardella S, Giardina E, Ferese R, Buttari F, Rizzo FR, Furlan R, Finardi A, Musella A, Mandolesi G, Guadalupi L, Centonze D, Stampanoni Bassi M. The BDNF Val66Met Polymorphism (rs6265) Modulates Inflammation and Neurodegeneration in the Early Phases of Multiple Sclerosis. Genes (Basel) 2022; 13:genes13020332. [PMID: 35205376 PMCID: PMC8871843 DOI: 10.3390/genes13020332] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 11/23/2022] Open
Abstract
The clinical course of multiple sclerosis (MS) is critically influenced by the interplay between inflammatory and neurodegenerative processes. The brain-derived neurotrophic factor (BDNF) Val66Met polymorphism (rs6265), one of the most studied single-nucleotide polymorphisms (SNPs), influences brain functioning and neurodegenerative processes in healthy individuals and in several neuropsychiatric diseases. However, the role of this polymorphism in MS is still controversial. In 218 relapsing–remitting (RR)-MS patients, we explored, at the time of diagnosis, the associations between the Val66Met polymorphism, clinical characteristics, and the cerebrospinal fluid (CSF) levels of a large set of pro-inflammatory and anti-inflammatory molecules. In addition, associations between Val66Met and structural MRI measures were assessed. We identified an association between the presence of Met and a combination of cytokines, identified by principal component analysis (PCA), including the pro-inflammatory molecules MCP-1, IL-8, TNF, Eotaxin, and MIP-1b. No significant associations emerged with clinical characteristics. Analysis of MRI measures evidenced reduced cortical thickness at the time of diagnosis in patients with Val66Met. We report for the first time an association between the Val66Met polymorphism and central inflammation in MS patients at the time of diagnosis. The role of this polymorphism in both inflammatory and neurodegenerative processes may explain its complex influence on the MS course.
Collapse
Affiliation(s)
| | - Antonio Bruno
- Neurology Unit, IRCSS Neuromed, 86077 Pozzilli, Italy
| | | | - Luana Gilio
- Neurology Unit, IRCSS Neuromed, 86077 Pozzilli, Italy
| | - Alessandro Moscatelli
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy
- Laboratory of Neuromotor Physiology, IRCSS Fondazione Santa Lucia, 00179 Rome, Italy
| | | | - Luigi Pavone
- Neurology Unit, IRCSS Neuromed, 86077 Pozzilli, Italy
| | - Ennio Iezzi
- Neurology Unit, IRCSS Neuromed, 86077 Pozzilli, Italy
| | - Stefano Gambardella
- Neurology Unit, IRCSS Neuromed, 86077 Pozzilli, Italy
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", 61029 Urbino, Italy
| | - Emiliano Giardina
- Genomic Medicine Laboratory, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", 00133 Rome, Italy
| | | | - Fabio Buttari
- Neurology Unit, IRCSS Neuromed, 86077 Pozzilli, Italy
| | | | - Roberto Furlan
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Annamaria Finardi
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology (INSpe), Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Alessandra Musella
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, 00163 Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, 00163 Rome, Italy
| | - Georgia Mandolesi
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, 00163 Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, 00163 Rome, Italy
| | - Livia Guadalupi
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, 00163 Rome, Italy
| | - Diego Centonze
- Neurology Unit, IRCSS Neuromed, 86077 Pozzilli, Italy
- Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy
| | | |
Collapse
|