1
|
Yang C, Song Y, Luo M, Wang Q, Zhang Y, Cen J, Du G, Shi J. Exosomes-encapsulated biomimetic polydopamine carbon dots with dual-targeting effect alleviate motor and non-motor symptoms of Parkinson's disease via anti-neuroinflammation. Int J Biol Macromol 2025; 296:139724. [PMID: 39809402 DOI: 10.1016/j.ijbiomac.2025.139724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/22/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
Currently, the clinical drugs for Parkinson's disease (PD) only focus on motor symptoms, while non-motor symptoms like depression are usually neglected. Even though, the efficacy of existing neurotherapeutic drugs is extremely poor which is due to the blood brain barrier (BBB). Therefore, a biomimetic polydopamine carbon dots (PDA C-dots) at 2-4 nm was synthesized, while exosomes from macrophages were applied to encapsulate PDA C-dots for improving their BBB-crossing ability and inflammation-targeting effect. Importantly, the prepared PDA C-dots@Exosomes (PEs) significantly alleviated both motor and non-motor symptoms of PD mice. Further mechanism research revealed that PEs eliminated oxidant stress and alleviated neuroinflammation to restore the injured neurons. The content of α-syn was markedly reduced, and the neural viability was dramatically improved on the areas of substantia nigra, striata, and prefrontal cortex. In summary, this work reported a mild synthetic approach to produce a kind of PDA C-dots, which had a fantastic neuroprotective effect. After being encapsulated with exosomes of macrophages, the obtained PEs could be utilized as a neuroprotective drug with great penetration ability of BBB and targeting ability into inflammatory zone. The great therapeutic effect on both motor and non-motor symptoms of PD indicates that PEs could become a promising drug for PD treatment.
Collapse
Affiliation(s)
- Chen Yang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan 475004, China; State Key Laboratory of Antiviral Drugs Henan University, Henan University, Kaifeng, Henan 475004, China
| | - Yanhao Song
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan 475004, China; State Key Laboratory of Antiviral Drugs Henan University, Henan University, Kaifeng, Henan 475004, China
| | - Mingkai Luo
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan 475004, China; State Key Laboratory of Antiviral Drugs Henan University, Henan University, Kaifeng, Henan 475004, China
| | - Qiuli Wang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan 475004, China; State Key Laboratory of Antiviral Drugs Henan University, Henan University, Kaifeng, Henan 475004, China
| | - Yumei Zhang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan 475004, China; State Key Laboratory of Antiviral Drugs Henan University, Henan University, Kaifeng, Henan 475004, China
| | - Juan Cen
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan 475004, China; State Key Laboratory of Antiviral Drugs Henan University, Henan University, Kaifeng, Henan 475004, China.
| | - Guanhua Du
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan 475004, China; State Key Laboratory of Antiviral Drugs Henan University, Henan University, Kaifeng, Henan 475004, China.
| | - Jiahua Shi
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan 475004, China; State Key Laboratory of Antiviral Drugs Henan University, Henan University, Kaifeng, Henan 475004, China.
| |
Collapse
|
2
|
Boziki M, Theotokis P, Kesidou E, Nella M, Bakirtzis C, Karafoulidou E, Tzitiridou-Chatzopoulou M, Doulberis M, Kazakos E, Deretzi G, Grigoriadis N, Kountouras J. Impact of Mast Cell Activation on Neurodegeneration: A Potential Role for Gut-Brain Axis and Helicobacter pylori Infection. Neurol Int 2024; 16:1750-1778. [PMID: 39728753 DOI: 10.3390/neurolint16060127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/08/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND The innate immune response aims to prevent pathogens from entering the organism and/or to facilitate pathogen clearance. Innate immune cells, such as macrophages, mast cells (MCs), natural killer cells and neutrophils, bear pattern recognition receptors and are thus able to recognize common molecular patterns, such as pathogen-associated molecular patterns (PAMPs), and damage-associated molecular patterns (DAMPs), the later occurring in the context of neuroinflammation. An inflammatory component in the pathology of otherwise "primary cerebrovascular and neurodegenerative" disease has recently been recognized and targeted as a means of therapeutic intervention. Activated MCs are multifunctional effector cells generated from hematopoietic stem cells that, together with dendritic cells, represent first-line immune defense mechanisms against pathogens and/or tissue destruction. METHODS This review aims to summarize evidence of MC implication in the pathogenesis of neurodegenerative diseases, namely, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, and multiple sclerosis. RESULTS In view of recent evidence that the gut-brain axis may be implicated in the pathogenesis of neurodegenerative diseases and the characterization of the neuroinflammatory component in the pathology of these diseases, this review also focuses on MCs as potential mediators in the gut-brain axis bi-directional communication and the possible role of Helicobacter pylori, a gastric pathogen known to alter the gut-brain axis homeostasis towards local and systemic pro-inflammatory responses. CONCLUSION As MCs and Helicobacter pylori infection may offer targets of intervention with potential therapeutic implications for neurodegenerative disease, more clinical and translational evidence is needed to elucidate this field.
Collapse
Affiliation(s)
- Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Maria Nella
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Christos Bakirtzis
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Eleni Karafoulidou
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Maria Tzitiridou-Chatzopoulou
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
- Midwifery Department, School of Healthcare Sciences, University of West Macedonia, Koila, 50100 Kozani, Greece
| | - Michael Doulberis
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
- Gastroklinik, Private Gastroenterological Practice, 8810 Horgen, Switzerland
- Division of Gastroenterology and Hepatology, Medical University Department, 5001 Aarau, Switzerland
| | - Evangelos Kazakos
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| | - Georgia Deretzi
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
- Department of Neurology, Papageorgiou General Hospital, 54629 Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Jannis Kountouras
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece
| |
Collapse
|
3
|
Liu Z, Xiang S, Chen B, Li J, Zhu D, Xu H, Hu S. Parkinson Disease -Targeted Nanocapsules for Synergistic Treatment: Combining Dopamine Replacement and Neuroinflammation Mitigation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404717. [PMID: 39431293 PMCID: PMC11633476 DOI: 10.1002/advs.202404717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/07/2024] [Indexed: 10/22/2024]
Abstract
Parkinson's disease (PD) is characterized by dopamine (DA) neuron loss and neuroinflammation. This study develops carrier-free nanocapsules (NCs) for targeted delivery of DA and catalase (CAT) to the PD brain, addressing both DA depletion and neuroinflammation simultaneously. The NCs are engineered by DA and 4-formylphenylboronic acid co-loading with cRGD-modified CAT (CAT-cRGD) and surface-modifying with Angiopep-2 (Ang). Ang targets the blood-brain barrier (BBB), enhancing brain delivery, while cRGD targets upregulated integrin receptors in the PD-affected BBB. The NCs showed a 1.4-fold increase in parkinsonian brain targeting efficiency compared to normal mice. In PD mice models, NCs demonstrated a stable increase in learning and memory, enhanced locomotor activity, and improved motor coordination. DA supplementation significantly enhanced dopaminergic signaling, increasing DA levels 1.8- and 3.5-fold in the striatum and substantia nigra, respectively. Additionally, delivered CAT effectively reduced neuroinflammation by mitigating endoplasmic reticulum stress, slowing disease progression, and protecting DA from oxidation. This innovative approach using PD-targeted NCs represents a synergistic strategy for PD treatment, combining symptomatic relief with disease progression intervention.
Collapse
Affiliation(s)
- Ziyao Liu
- Department of Nuclear MedicineXiangya HospitalCentral South UniversityChangsha410008China
- Key Laboratory of Biological Nanotechnology of National Health CommissionXiangya Hospital, Central South UniversityChangsha410008China
| | - Shijun Xiang
- Department of Nuclear MedicineXiangya HospitalCentral South UniversityChangsha410008China
| | - Bei Chen
- Department of Nuclear MedicineXiangya HospitalCentral South UniversityChangsha410008China
| | - Jian Li
- Department of Nuclear MedicineXiangya HospitalCentral South UniversityChangsha410008China
| | - Dingcheng Zhu
- College of MaterialChemistry and Chemical EngineeringKey Laboratory of Organosilicon Chemistry and Material TechnologyMinistry of EducationHangzhou Normal UniversityHangzhou311121China
| | - Hongjuan Xu
- Department of Nuclear MedicineXiangya HospitalCentral South UniversityChangsha410008China
- Key Laboratory of Biological Nanotechnology of National Health CommissionXiangya Hospital, Central South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DiseasesXiangya HospitalCentral South UniversityChangsha410008China
| | - Shuo Hu
- Department of Nuclear MedicineXiangya HospitalCentral South UniversityChangsha410008China
- Key Laboratory of Biological Nanotechnology of National Health CommissionXiangya Hospital, Central South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DiseasesXiangya HospitalCentral South UniversityChangsha410008China
| |
Collapse
|
4
|
Zeng Z, Jin W, Huang K, Xiong L, Luo Y, Li G, Zhang W, Hong G, Mao F, Xiong K, Luo X. Examining the relationship between CDAI and frailty and its manifestation in Parkinson's disease: a cross-sectional study. Front Nutr 2024; 11:1502748. [PMID: 39668905 PMCID: PMC11635959 DOI: 10.3389/fnut.2024.1502748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 11/15/2024] [Indexed: 12/14/2024] Open
Abstract
Background Higher intake of antioxidants is associated with reduced risk of various chronic diseases. However, the relationship between composite dietary antioxidants and frailty has not been characterized, especially in neurodegenerative conditions like Parkinson's disease (PD) where frailty is highly prevalent. This study aimed to investigate the association between composite dietary antioxidant index (CDAI), a composite score reflecting antioxidant vitamin and mineral intakes, and frailty risk in the general United States (US) population and PD patients. Methods Data from 21,354 participants ≥40 years in the National Health and Nutrition Examination Survey (NHANES) 2003-2018 represented the general population sample, while 268 PD patients were analyzed separately. Frailty was defined using a validated index. Weighted logistic regression and restricted cubic splines (RCS) examined overall and nonlinear CDAI-frailty associations, adjusting for sociodemographics, lifestyle factors, and comorbidities. Results In the general population, each unit increase in CDAI was associated with a 3.7% lower likelihood of frailty after full adjustments. Vitamin A, C, E, selenium and carotenoids exhibited J-shaped relationships where frailty risk decreased below intake thresholds of 1093.04 μg, 161.53 mg, 13.66 mg, 109.99 μg, and 5057.50 μg, respectively. In contrast, the CDAI- frailty inverse association was weaker among PD patients and only vitamin C (threshold 52.45 mg) and zinc (9.35 mg) showed nonlinear links. Conclusion Higher dietary antioxidant intake was associated with lower frailty prevalence in the general US population, with vitamins A, C, E, selenium, and carotenoids exhibiting nonlinear J-shaped relationships. In contrast, these associations were weaker and less consistent among PD patients, with only vitamins C and zinc showing nonlinear correlations. These findings highlight population-specific differences in the role of dietary antioxidants in frailty and suggest the need for personalized nutritional strategies in PD frailty management.
Collapse
Affiliation(s)
- Zhaohao Zeng
- Department of Neurology, Shenzhen People’s Hospital, The First Affiliated Hospital, Southern University of Science and Technology, The Second Clinical Medical College, Jinan University, Shenzhen, China
- The Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Wen Jin
- Department of Neurology, Shenzhen People’s Hospital, The First Affiliated Hospital, Southern University of Science and Technology, The Second Clinical Medical College, Jinan University, Shenzhen, China
- The Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, China
- Post-doctoral Scientific Research Station of Basic Medicine, Jinan University, Guangzhou, China
| | - Kunyu Huang
- Department of Pharmacy, Shenshan Medical Center, Memorial Hospital of SUN YAT-SEN University, Shanwei, China
| | - Lijiao Xiong
- The Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, China
- Department of Geriatrics, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Yu Luo
- Department of Neurology, Shenzhen People’s Hospital, The First Affiliated Hospital, Southern University of Science and Technology, The Second Clinical Medical College, Jinan University, Shenzhen, China
- The Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Guoyang Li
- Department of Neurology, Shenzhen People’s Hospital, The First Affiliated Hospital, Southern University of Science and Technology, The Second Clinical Medical College, Jinan University, Shenzhen, China
- The Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Wenli Zhang
- Department of Neurology, Shenzhen People’s Hospital, The First Affiliated Hospital, Southern University of Science and Technology, The Second Clinical Medical College, Jinan University, Shenzhen, China
- The Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Guo Hong
- Department of Neurology, Shenzhen People’s Hospital, The First Affiliated Hospital, Southern University of Science and Technology, The Second Clinical Medical College, Jinan University, Shenzhen, China
- The Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Fengju Mao
- Department of Neurology, Shenzhen People’s Hospital, The First Affiliated Hospital, Southern University of Science and Technology, The Second Clinical Medical College, Jinan University, Shenzhen, China
- The Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Kaifen Xiong
- Department of Neurology, Shenzhen People’s Hospital, The First Affiliated Hospital, Southern University of Science and Technology, The Second Clinical Medical College, Jinan University, Shenzhen, China
- The Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Xiaoguang Luo
- Department of Neurology, Shenzhen People’s Hospital, The First Affiliated Hospital, Southern University of Science and Technology, The Second Clinical Medical College, Jinan University, Shenzhen, China
- The Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, China
| |
Collapse
|
5
|
Kempuraj D, Dourvetakis KD, Cohen J, Valladares DS, Joshi RS, Kothuru SP, Anderson T, Chinnappan B, Cheema AK, Klimas NG, Theoharides TC. Neurovascular unit, neuroinflammation and neurodegeneration markers in brain disorders. Front Cell Neurosci 2024; 18:1491952. [PMID: 39526043 PMCID: PMC11544127 DOI: 10.3389/fncel.2024.1491952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Neurovascular unit (NVU) inflammation via activation of glial cells and neuronal damage plays a critical role in neurodegenerative diseases. Though the exact mechanism of disease pathogenesis is not understood, certain biomarkers provide valuable insight into the disease pathogenesis, severity, progression and therapeutic efficacy. These markers can be used to assess pathophysiological status of brain cells including neurons, astrocytes, microglia, oligodendrocytes, specialized microvascular endothelial cells, pericytes, NVU, and blood-brain barrier (BBB) disruption. Damage or derangements in tight junction (TJ), adherens junction (AdJ), and gap junction (GJ) components of the BBB lead to increased permeability and neuroinflammation in various brain disorders including neurodegenerative disorders. Thus, neuroinflammatory markers can be evaluated in blood, cerebrospinal fluid (CSF), or brain tissues to determine neurological disease severity, progression, and therapeutic responsiveness. Chronic inflammation is common in age-related neurodegenerative disorders including Alzheimer's disease (AD), Parkinson's disease (PD), and dementia. Neurotrauma/traumatic brain injury (TBI) also leads to acute and chronic neuroinflammatory responses. The expression of some markers may also be altered many years or even decades before the onset of neurodegenerative disorders. In this review, we discuss markers of neuroinflammation, and neurodegeneration associated with acute and chronic brain disorders, especially those associated with neurovascular pathologies. These biomarkers can be evaluated in CSF, or brain tissues. Neurofilament light (NfL), ubiquitin C-terminal hydrolase-L1 (UCHL1), glial fibrillary acidic protein (GFAP), Ionized calcium-binding adaptor molecule 1 (Iba-1), transmembrane protein 119 (TMEM119), aquaporin, endothelin-1, and platelet-derived growth factor receptor beta (PDGFRβ) are some important neuroinflammatory markers. Recent BBB-on-a-chip modeling offers promising potential for providing an in-depth understanding of brain disorders and neurotherapeutics. Integration of these markers in clinical practice could potentially enhance early diagnosis, monitor disease progression, and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- Dr. Kiran C. Patel College of Osteopathic Medicine, Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States
| | - Kirk D. Dourvetakis
- Dr. Kiran C. Patel College of Osteopathic Medicine, Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States
| | - Jessica Cohen
- Dr. Kiran C. Patel College of Osteopathic Medicine, Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States
| | - Daniel Seth Valladares
- Dr. Kiran C. Patel College of Osteopathic Medicine, Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States
| | - Rhitik Samir Joshi
- Dr. Kiran C. Patel College of Osteopathic Medicine, Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States
| | - Sai Puneeth Kothuru
- Dr. Kiran C. Patel College of Osteopathic Medicine, Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States
- College of Psychology, Nova Southeastern University, Ft. Lauderdale, FL, United States
| | - Tristin Anderson
- Dr. Kiran C. Patel College of Osteopathic Medicine, Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States
| | - Baskaran Chinnappan
- Dr. Kiran C. Patel College of Osteopathic Medicine, Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States
| | - Amanpreet K. Cheema
- Dr. Kiran C. Patel College of Osteopathic Medicine, Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States
| | - Nancy G. Klimas
- Dr. Kiran C. Patel College of Osteopathic Medicine, Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States
- Miami VA Geriatric Research Education and Clinical Center (GRECC), Miami Veterans Affairs Healthcare System, Miami, FL, United States
| | - Theoharis C. Theoharides
- Dr. Kiran C. Patel College of Osteopathic Medicine, Institute for Neuro-Immune Medicine, Nova Southeastern University, Ft. Lauderdale, FL, United States
- Department of Immunology, Tufts, University School of Medicine, Boston, MA, United States
| |
Collapse
|
6
|
Nelke A, García-López S, Caso JR, Pereira MP. The therapeutic use of clonal neural stem cells in experimental Parkinson´s disease. Stem Cell Res Ther 2024; 15:356. [PMID: 39385216 PMCID: PMC11465761 DOI: 10.1186/s13287-024-03965-0] [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: 02/23/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Parkinson´s disease (PD), the second most common neurodegenerative disease in the world, is characterized by the death or impairment of dopaminergic neurons (DAn) in the substantia nigra pars compacta and dopamine depletion in the striatum. Currently, there is no cure for PD, and treatments only help to reduce the symptoms of the disease, and do not repair or replace the DAn damaged or lost in PD. Cell replacement therapy (CRT) seeks to relieve both pathological and symptomatic PD manifestations and has been shown to have beneficial effects in experimental PD models as well as in PD patients, but an apt cell line to be used in the treatment of PD has yet to be established. The purpose of this study was to examine the effects of the transplantation of hVM1 clone 32 cells, a bankable line of human neural stem cells (hNSCs), in a PD mouse model at four months post-transplant. METHODS Adult (five month-old) C57BL/6JRccHsd male mice were injected with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and subsequently transplanted with hVM1 clone 32 cells, or buffer, in the left striatum. Four months post-transplant, behavioral effects were explored using the open field and paw print tests, and histological analyses were performed. RESULTS Transplantation of hVM1 clone 32 cells rescued dopaminergic nigrostriatal populations in adult Parkinsonian mice. Motor and neurological deterioration were observed in buffer-treated mice, the latter of which had a tendency to improve in hNSC-transplanted mice. Detection of mast cell migration to the superficial cervical lymph nodes in cell-transplanted mice denoted a peripheral effect. Transplantation of hNSCs also rescued neuroblast neurogenesis in the subgranular zone, which was correlated with dopaminergic recovery and is indicative of local recovery mechanisms. CONCLUSIONS In this proof-of-concept study, the transplantation of hVM1 clone 32 cells provided neuroprotection in adult Parkinsonian mice by restoring the dopaminergic nigrostriatal pathway and hippocampal neurogenesis, demonstrating the efficacy of cell replacement therapy as a treatment for PD.
Collapse
Affiliation(s)
- Anna Nelke
- Unit of Molecular Neuropathology, Physiological and pathological processes Program, Centro de Biología Molecular Severo Ochoa UAM-CSIC, Calle Nicolás Cabrera, 1, Madrid, 28049, Spain.
- Department of Molecular Biology, Faculty of Science, Universidad Autónoma de Madrid, Ciudad Universitaria de Cantoblanco, Madrid, 28049, Spain.
- Institute for Molecular Biology - IUBM (Universidad Autónoma de Madrid), Madrid, Spain.
| | - Silvia García-López
- Unit of Molecular Neuropathology, Physiological and pathological processes Program, Centro de Biología Molecular Severo Ochoa UAM-CSIC, Calle Nicolás Cabrera, 1, Madrid, 28049, Spain
- Department of Molecular Biology, Faculty of Science, Universidad Autónoma de Madrid, Ciudad Universitaria de Cantoblanco, Madrid, 28049, Spain
- Institute for Molecular Biology - IUBM (Universidad Autónoma de Madrid), Madrid, Spain
| | - Javier R Caso
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto Universitario de Investigación Neuroquímica (IUIN-UCM), Avda. Complutense s/n, Madrid, 28040, Spain
| | - Marta P Pereira
- Unit of Molecular Neuropathology, Physiological and pathological processes Program, Centro de Biología Molecular Severo Ochoa UAM-CSIC, Calle Nicolás Cabrera, 1, Madrid, 28049, Spain.
- Department of Molecular Biology, Faculty of Science, Universidad Autónoma de Madrid, Ciudad Universitaria de Cantoblanco, Madrid, 28049, Spain.
- Institute for Molecular Biology - IUBM (Universidad Autónoma de Madrid), Madrid, Spain.
| |
Collapse
|
7
|
Stevenson M, Hebron ML, Liu X, Balaraman K, Wolf C, Moussa C. c-KIT inhibitors reduce pathology and improve behavior in the Tg(SwDI) model of Alzheimer's disease. Life Sci Alliance 2024; 7:e202402625. [PMID: 39009412 PMCID: PMC11249953 DOI: 10.26508/lsa.202402625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024] Open
Abstract
Treatments for Alzheimer's disease have primarily focused on removing brain amyloid plaques to improve cognitive outcomes in patients. We developed small compounds, known as BK40143 and BK40197, and we hypothesize that these drugs alleviate microglial-mediated neuroinflammation and induce autophagic clearance of neurotoxic proteins to improve behavior in models of neurodegeneration. Specificity binding assays of BK40143 and BK40197 showed primary binding to c-KIT/Platelet Derived Growth Factor Receptors (PDGFR)α/β, whereas BK40197 also differentially binds to FYVE finger-containing phosphoinositide kinase (PIKFYVE). Both compounds penetrate the CNS, and treatment with these drugs inhibited the maturation of peripheral mast cells in transgenic mice, correlating with cognitive improvements on measures of memory and anxiety. In the brain, microglial activation was profoundly attenuated and amyloid-beta and tau were reduced via autophagy. Multi-kinase inhibition, including c-KIT, exerts multifunctional effects to reduce neurodegenerative pathology via autophagy and microglial activity and may represent a potential therapeutic option for neurodegeneration.
Collapse
Affiliation(s)
- Max Stevenson
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington DC, USA
| | - Michaeline L Hebron
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington DC, USA
| | - Xiaoguang Liu
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington DC, USA
| | - Kaluvu Balaraman
- Medicinal Chemistry Shared Resource, Department of Chemistry, Georgetown University Medical Center, Washington DC, USA
| | - Christian Wolf
- Medicinal Chemistry Shared Resource, Department of Chemistry, Georgetown University Medical Center, Washington DC, USA
| | - Charbel Moussa
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington DC, USA
| |
Collapse
|
8
|
Stevenson M, Algarzae NK, Moussa C. Tyrosine kinases: multifaceted receptors at the intersection of several neurodegenerative disease-associated processes. FRONTIERS IN DEMENTIA 2024; 3:1458038. [PMID: 39221072 PMCID: PMC11361951 DOI: 10.3389/frdem.2024.1458038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024]
Abstract
Tyrosine kinases (TKs) are catalytic enzymes activated by auto-phosphorylation that function by phosphorylating tyrosine residues on downstream substrates. Tyrosine kinase inhibitors (TKIs) have been heavily exploited as cancer therapeutics, primarily due to their role in autophagy, blood vessel remodeling and inflammation. This suggests tyrosine kinase inhibition as an appealing therapeutic target for exploiting convergent mechanisms across several neurodegenerative disease (NDD) pathologies. The overlapping mechanisms of action between neurodegeneration and cancer suggest that TKIs may play a pivotal role in attenuating neurodegenerative processes, including degradation of misfolded or toxic proteins, reduction of inflammation and prevention of fibrotic events of blood vessels in the brain. In this review, we will discuss the distinct roles that select TKs have been shown to play in various disease-associated processes, as well as identify TKs that have been explored as targets for therapeutic intervention and associated pharmacological agents being investigated as treatments for NDDs.
Collapse
Affiliation(s)
- Max Stevenson
- The Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, Department of Neurology, Georgetown University Medical Center, Washington, DC, United States
| | - Norah K. Algarzae
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Charbel Moussa
- The Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, Department of Neurology, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|
9
|
Theoharides TC, Twahir A, Kempuraj D. Mast cells in the autonomic nervous system and potential role in disorders with dysautonomia and neuroinflammation. Ann Allergy Asthma Immunol 2024; 132:440-454. [PMID: 37951572 DOI: 10.1016/j.anai.2023.10.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/16/2023] [Accepted: 10/06/2023] [Indexed: 11/14/2023]
Abstract
Mast cells (MC) are ubiquitous in the body, and they are critical for not only in allergic diseases but also in immunity and inflammation, including having potential involvement in the pathophysiology of dysautonomias and neuroinflammatory disorders. MC are located perivascularly close to nerve endings and sites such as the carotid bodies, heart, hypothalamus, the pineal gland, and the adrenal gland that would allow them not only to regulate but also to be affected by the autonomic nervous system (ANS). MC are stimulated not only by allergens but also many other triggers including some from the ANS that can affect MC release of neurosensitizing, proinflammatory, and vasoactive mediators. Hence, MC may be able to regulate homeostatic functions that seem to be dysfunctional in many conditions, such as postural orthostatic tachycardia syndrome, autism spectrum disorder, myalgic encephalomyelitis/chronic fatigue syndrome, and Long-COVID syndrome. The evidence indicates that there is a possible association between these conditions and diseases associated with MC activation. There is no effective treatment for any form of these conditions other than minimizing symptoms. Given the many ways MC could be activated and the numerous mediators released, it would be important to develop ways to inhibit stimulation of MC and the release of ANS-relevant mediators.
Collapse
Affiliation(s)
- Theoharis C Theoharides
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, Florida; Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts.
| | - Assma Twahir
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, Florida
| | - Duraisamy Kempuraj
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, Florida
| |
Collapse
|
10
|
Pang P, Zhang S, Fan X, Zhang S. Knockdown of fat mass and obesity alleviates the ferroptosis in Parkinson's disease through m6A-NRF2-dependent manner. Cell Biol Int 2024; 48:431-439. [PMID: 38180302 DOI: 10.1002/cbin.12118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 11/21/2023] [Accepted: 12/02/2023] [Indexed: 01/06/2024]
Abstract
Emerging evidence has suggested that N6 -methyladenosine (m6 A) regulates the pathology of Parkinson's disease (PD). Nevertheless, the function of demethylase fat mass and obesity (FTO) associated pathogenesis is still not fully elucidated. Here, this research findings revealed that m6 A-modification was decreased in PD models, meanwhile, the FTO level upregulated in the PD models. Functionally, in N-methyl-4-phenylpyridinium (MPP+) treated SH-SY5Y cells, the ferroptosis significantly upregulated and FTO silencing mitigated the ferroptosis phenotype. Moreover, in silico assays indicated that nuclear factor erythroid 2-related factor-2 (NRF2) acted as the target of FTO, and FTO demethylated the m6 A modification from NRF2 mRNA. Furthermore, FTO impaired the NRF2 mRNA stability via m6 A-dependent pathway. Thus, our findings illustrated an important role of FTO on PD through m6 A-NRF2-ferroptosis manner. Taken together, the study revealed the potential function of FTO on PD nervous system diseases.
Collapse
Affiliation(s)
- Pengfei Pang
- Department of Neurosurgery, The Affiliated Hospital of Northwest University, Xi'an, China
| | - Shirong Zhang
- Department of Neurosurgery, The Affiliated Hospital of Northwest University, Xi'an, China
| | - Xinxin Fan
- Department of Neurosurgery, The Affiliated Hospital of Northwest University, Xi'an, China
| | - Shitao Zhang
- Department of Neurosurgery, The Affiliated Hospital of Northwest University, Xi'an, China
| |
Collapse
|
11
|
Cohen J, Mathew A, Dourvetakis KD, Sanchez-Guerrero E, Pangeni RP, Gurusamy N, Aenlle KK, Ravindran G, Twahir A, Isler D, Sosa-Garcia SR, Llizo A, Bested AC, Theoharides TC, Klimas NG, Kempuraj D. Recent Research Trends in Neuroinflammatory and Neurodegenerative Disorders. Cells 2024; 13:511. [PMID: 38534355 PMCID: PMC10969521 DOI: 10.3390/cells13060511] [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: 12/25/2023] [Revised: 03/03/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Neuroinflammatory and neurodegenerative disorders including Alzheimer's disease (AD), Parkinson's disease (PD), traumatic brain injury (TBI) and Amyotrophic lateral sclerosis (ALS) are chronic major health disorders. The exact mechanism of the neuroimmune dysfunctions of these disease pathogeneses is currently not clearly understood. These disorders show dysregulated neuroimmune and inflammatory responses, including activation of neurons, glial cells, and neurovascular unit damage associated with excessive release of proinflammatory cytokines, chemokines, neurotoxic mediators, and infiltration of peripheral immune cells into the brain, as well as entry of inflammatory mediators through damaged neurovascular endothelial cells, blood-brain barrier and tight junction proteins. Activation of glial cells and immune cells leads to the release of many inflammatory and neurotoxic molecules that cause neuroinflammation and neurodegeneration. Gulf War Illness (GWI) and myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) are chronic disorders that are also associated with neuroimmune dysfunctions. Currently, there are no effective disease-modifying therapeutic options available for these diseases. Human induced pluripotent stem cell (iPSC)-derived neurons, astrocytes, microglia, endothelial cells and pericytes are currently used for many disease models for drug discovery. This review highlights certain recent trends in neuroinflammatory responses and iPSC-derived brain cell applications in neuroinflammatory disorders.
Collapse
Affiliation(s)
- Jessica Cohen
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Annette Mathew
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Kirk D Dourvetakis
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Estella Sanchez-Guerrero
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Rajendra P Pangeni
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Narasimman Gurusamy
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Kristina K Aenlle
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
- Miami VA Geriatric Research Education and Clinical Center (GRECC), Miami Veterans Affairs Healthcare System, Miami, FL 33125, USA
| | - Geeta Ravindran
- Cell Therapy Institute, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Assma Twahir
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Dylan Isler
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Sara Rukmini Sosa-Garcia
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Axel Llizo
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Alison C Bested
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Theoharis C Theoharides
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Nancy G Klimas
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
- Miami VA Geriatric Research Education and Clinical Center (GRECC), Miami Veterans Affairs Healthcare System, Miami, FL 33125, USA
| | - Duraisamy Kempuraj
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| |
Collapse
|
12
|
Wu Y, Meng X, Cheng WY, Yan Z, Li K, Wang J, Jiang T, Zhou F, Wong KH, Zhong C, Dong Y, Gao S. Can pluripotent/multipotent stem cells reverse Parkinson's disease progression? Front Neurosci 2024; 18:1210447. [PMID: 38356648 PMCID: PMC10864507 DOI: 10.3389/fnins.2024.1210447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 01/02/2024] [Indexed: 02/16/2024] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by continuous and selective degeneration or death of dopamine neurons in the midbrain, leading to dysfunction of the nigrostriatal neural circuits. Current clinical treatments for PD include drug treatment and surgery, which provide short-term relief of symptoms but are associated with many side effects and cannot reverse the progression of PD. Pluripotent/multipotent stem cells possess a self-renewal capacity and the potential to differentiate into dopaminergic neurons. Transplantation of pluripotent/multipotent stem cells or dopaminergic neurons derived from these cells is a promising strategy for the complete repair of damaged neural circuits in PD. This article reviews and summarizes the current preclinical/clinical treatments for PD, their efficacies, and the advantages/disadvantages of various stem cells, including pluripotent and multipotent stem cells, to provide a detailed overview of how these cells can be applied in the treatment of PD, as well as the challenges and bottlenecks that need to be overcome in future translational studies.
Collapse
Affiliation(s)
- Yongkang Wu
- Key Laboratory of Adolescent Health Evaluation and Sports Intervention, Ministry of Education, East China Normal University, Shanghai, China
| | - Xiangtian Meng
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wai-Yin Cheng
- Research Institute for Future Food, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Zhichao Yan
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Keqin Li
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jian Wang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tianfang Jiang
- Department of Neurology, Shanghai Eighth People’s Hospital Affiliated to Jiangsu University, Shanghai, China
| | - Fei Zhou
- Department of Neurology, Third Affiliated Hospital of Navy Military Medical University, Shanghai, China
| | - Ka-Hing Wong
- Research Institute for Future Food, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Chunlong Zhong
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yi Dong
- Key Laboratory of Adolescent Health Evaluation and Sports Intervention, Ministry of Education, East China Normal University, Shanghai, China
| | - Shane Gao
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
13
|
Ali T, Klein AN, McDonald K, Johansson L, Mukherjee PG, Hallbeck M, Doh-Ura K, Schatzl HM, Gilch S. Cellulose ether treatment inhibits amyloid beta aggregation, neuroinflammation and cognitive deficits in transgenic mouse model of Alzheimer's disease. J Neuroinflammation 2023; 20:177. [PMID: 37507761 PMCID: PMC10375631 DOI: 10.1186/s12974-023-02858-y] [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: 03/08/2023] [Accepted: 07/23/2023] [Indexed: 07/30/2023] Open
Abstract
Alzheimer's disease (AD) is an incurable, progressive and devastating neurodegenerative disease. Pathogenesis of AD is associated with the aggregation and accumulation of amyloid beta (Aβ), a major neurotoxic mediator that triggers neuroinflammation and memory impairment. Recently, we found that cellulose ether compounds (CEs) have beneficial effects against prion diseases by inhibiting protein misfolding and replication of prions, which share their replication mechanism with Aβ. CEs are FDA-approved safe additives in foods and pharmaceuticals. Herein, for the first time we determined the therapeutic effects of the representative CE (TC-5RW) in AD using in vitro and in vivo models. Our in vitro studies showed that TC-5RW inhibits Aβ aggregation, as well as neurotoxicity and immunoreactivity in Aβ-exposed human and murine neuroblastoma cells. In in vivo studies, for the first time we observed that single and weekly TC-5RW administration, respectively, improved memory functions of transgenic 5XFAD mouse model of AD. We further demonstrate that TC-5RW treatment of 5XFAD mice significantly inhibited Aβ oligomer and plaque burden and its associated neuroinflammation via regulating astrogliosis, microgliosis and proinflammatory mediator glial maturation factor beta (GMFβ). Additionally, we determined that TC-5RW reduced lipopolysaccharide-induced activated gliosis and GMFβ in vitro. In conclusion, our results demonstrate that CEs have therapeutic effects against Aβ pathologies and cognitive impairments, and direct, potent anti-inflammatory activity to rescue neuroinflammation. Therefore, these FDA-approved compounds are effective candidates for developing therapeutics for AD and related neurodegenerative diseases associated with protein misfolding.
Collapse
Affiliation(s)
- Tahir Ali
- Calgary Prion Research Unit, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4Z6, Canada.
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.
| | - Antonia N Klein
- Calgary Prion Research Unit, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4Z6, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Keegan McDonald
- Calgary Prion Research Unit, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4Z6, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Lovisa Johansson
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, 58185, Linköping, Sweden
| | | | - Martin Hallbeck
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, 58185, Linköping, Sweden
| | - Katsumi Doh-Ura
- Department of Neurochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hermann M Schatzl
- Calgary Prion Research Unit, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4Z6, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Sabine Gilch
- Calgary Prion Research Unit, Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4Z6, Canada.
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
14
|
Theoharides TC, Kempuraj D. Role of SARS-CoV-2 Spike-Protein-Induced Activation of Microglia and Mast Cells in the Pathogenesis of Neuro-COVID. Cells 2023; 12:688. [PMID: 36899824 PMCID: PMC10001285 DOI: 10.3390/cells12050688] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/07/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19). About 45% of COVID-19 patients experience several symptoms a few months after the initial infection and develop post-acute sequelae of SARS-CoV-2 (PASC), referred to as "Long-COVID," characterized by persistent physical and mental fatigue. However, the exact pathogenetic mechanisms affecting the brain are still not well-understood. There is increasing evidence of neurovascular inflammation in the brain. However, the precise role of the neuroinflammatory response that contributes to the disease severity of COVID-19 and long COVID pathogenesis is not clearly understood. Here, we review the reports that the SARS-CoV-2 spike protein can cause blood-brain barrier (BBB) dysfunction and damage neurons either directly, or via activation of brain mast cells and microglia and the release of various neuroinflammatory molecules. Moreover, we provide recent evidence that the novel flavanol eriodictyol is particularly suited for development as an effective treatment alone or together with oleuropein and sulforaphane (ViralProtek®), all of which have potent anti-viral and anti-inflammatory actions.
Collapse
Affiliation(s)
- Theoharis C. Theoharides
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Duraisamy Kempuraj
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| |
Collapse
|
15
|
Wang Q, Zheng J, Pettersson S, Reynolds R, Tan EK. The link between neuroinflammation and the neurovascular unit in synucleinopathies. SCIENCE ADVANCES 2023; 9:eabq1141. [PMID: 36791205 PMCID: PMC9931221 DOI: 10.1126/sciadv.abq1141] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 01/19/2023] [Indexed: 05/28/2023]
Abstract
The neurovascular unit (NVU) is composed of vascular cells, glial cells, and neurons. As a fundamental functional module in the central nervous system, the NVU maintains homeostasis in the microenvironment and the integrity of the blood-brain barrier. Disruption of the NVU and interactions among its components are involved in the pathophysiology of synucleinopathies, which are characterized by the pathological accumulation of α-synuclein. Neuroinflammation contributes to the pathophysiology of synucleinopathies, including Parkinson's disease, multiple system atrophy, and dementia with Lewy bodies. This review aims to summarize the neuroinflammatory response of glial cells and vascular cells in the NVU. We also review neuroinflammation in the context of the cross-talk between glial cells and vascular cells, between glial cells and pericytes, and between microglia and astroglia. Last, we discuss how α-synuclein affects neuroinflammation and how neuroinflammation influences the aggregation and spread of α-synuclein and analyze different properties of α-synuclein in synucleinopathies.
Collapse
Affiliation(s)
- Qing Wang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Jialing Zheng
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Sven Pettersson
- ASEAN Microbiome Nutrition Centre, National Neuroscience Institute, Singapore 308433, Singapore
- Karolinska Institutet, Department of Odontology, 171 77 Solna, Sweden
- Faculty of Medical Sciences, Sunway University, Subang Jaya, 47500 Selangor, Malaysia
- Department of Microbiology and Immunology, National University Singapore, Singapore 117545, Singapore
| | - Richard Reynolds
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, Du Cane Road, London W12 0NN, UK
- Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Eng-King Tan
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
16
|
Balasubramanian N, James TD, Selvakumar GP, Reinhardt J, Marcinkiewcz CA. Repeated ethanol exposure and withdrawal alters angiotensin-converting enzyme 2 expression in discrete brain regions: Implications for SARS-CoV-2 neuroinvasion. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:219-239. [PMID: 36529893 PMCID: PMC9878009 DOI: 10.1111/acer.15000] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 11/18/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND People with alcohol use disorder (AUD) may be at higher risk for COVID-19. Angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2) are required for cellular entry by SARS-CoV-2, but information on their expression in specific brain regions after alcohol exposure is limited. We sought to clarify how chronic alcohol exposure affects ACE2 expression in monoaminergic brainstem circuits and other putative SARS-CoV-2 entry points. METHODS Brains were examined for ACE2 using immunofluorescence after 4 weeks of chronic intermittent ethanol (CIE) vapor inhalation. We also examined TMPRSS2, Cathepsin L, and ADAM17 by Western blot and RAS pathway mediators and pro-inflammatory markers via RT-qPCR. RESULTS ACE2 was increased in most brain regions following CIE including the olfactory bulb (OB), hypothalamus (HT), raphe magnus (RMG), raphe obscurus (ROB), locus coeruleus (LC), and periaqueductal gray (PAG). We also observed increased colocalization of ACE2 with monoaminergic neurons in brainstem nuclei. Moreover, soluble ACE2 (sACE2) was elevated in OB, HT, and LC. The increase in sACE2 in OB and HT was accompanied by upregulation of ADAM17, an ACE2 sheddase, while TMPRSS2 increased in HT and LC. Cathepsin L, an endosomal receptor involved in viral entry, was also increased in OB. Alcohol can increase Angiotensin II, which triggers a pro-inflammatory response that may upregulate ACE2 via activation of RAS pathway receptors AT1R/AT2R. ACE2 then metabolizes Angiotensin II to Angiotensin (1-7) and provokes an anti-inflammatory response via MAS1. Accordingly, we report that AT1R/AT2R mRNA decreased in OB and increased in the LC, while MAS1 mRNA increased in both OB and LC. Other mRNAs for pro-inflammatory markers were also dysregulated in OB, HT, raphe, and LC. CONCLUSIONS Our results suggest that alcohol triggers a compensatory upregulation of ACE2 in the brain due to disturbed RAS and may increase the risk or severity of SARS-CoV-2 infection.
Collapse
Affiliation(s)
| | - Thomas D James
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa, USA
| | | | - Jessica Reinhardt
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa, USA
| | | |
Collapse
|
17
|
Yu Z, Huang L, Xia Y, Cheng S, Yang C, Chen C, Zou Z, Wang X, Tian X, Jiang X, Zhou L. Analysis of m6A modification regulators in the substantia nigra and striatum of MPTP-induced Parkinson’s disease mice. Neurosci Lett 2022; 791:136907. [DOI: 10.1016/j.neulet.2022.136907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/14/2022] [Accepted: 10/02/2022] [Indexed: 10/31/2022]
|
18
|
Wang B, Wang L, Qu Y, Lu J, Xia W. Chitosan oligosaccharides exert neuroprotective effects via modulating the PI3K/Akt/Bcl-2 pathway in a Parkinsonian model. Food Funct 2022; 13:5838-5853. [PMID: 35545086 DOI: 10.1039/d1fo04374a] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD), the second most common neurodegenerative disease, is a threat to patients due to the inability to prevent or decelerate disease progression. Currently, most clinical drugs for the treatment of PD are synthetic drugs that always present undesirable adverse or toxic effects. Chitosan oligosaccharide (COS) is a natural oligosaccharide that has been considered relatively safe and studied in the therapeutic effects on different types of neuronal disorders. In this study, we separated four COS monomers (COSs) including chitobiose (COS2), chitotriose (COS3), chitotetraose (COS4) and chitopentaose (COS5) to explore their structure-activity relationship in PD mice induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Techniques including TLC, HPLC, MS, and NMR were applied to investigate the purity and structure of the COSs. After the oral administration of COSs, behavior indexes, pathological indexes, cytokines, and expression of proteins in the nigrostriatal pathway of the mice were analyzed. The results showed that the four COSs were fully deacetylated and the purity was >90%. Additionally, the neurobehavioral deficits of the PD mice were improved by treatment with COSs. The results further proved that COSs could protect the TH-labelled dopaminergic neurons via reducing the overexpression of α-synuclein, alleviating neuroinflammation, and activating the PI3K/Akt/Bcl-2 pathway to reduce apoptosis. COS3 exhibited a better effect on protecting dopaminergic neurons; however, COS2 provided a better effect on reducing the overexpression of α-synuclein. To conclude, the neuroprotective activity makes COSs a viable candidate as an ingredient for healthcare products.
Collapse
Affiliation(s)
- Bin Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China. .,Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Ling Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China. .,Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Yufei Qu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China.
| | - Jingyu Lu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China.
| | - Wenshui Xia
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China. .,Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
19
|
Zhang X, Deng R, Zhang S, Deng J, Jia JJ, Sun B, Zhou X, Bai J. Thioredoxin-1 regulates calcium homeostasis in MPP + /MPTP-induced Parkinson's disease models. Eur J Neurosci 2021; 54:4827-4837. [PMID: 34132424 DOI: 10.1111/ejn.15355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 06/04/2021] [Accepted: 06/12/2021] [Indexed: 11/30/2022]
Abstract
Disturbance in calcium (Ca2+ ) homeostasis has been involved in a variety of neuropathological conditions including Parkinson's disease (PD). The Ca2+ channel, transient receptor potential channel 1 (TRPC1), plays a protective role in regulating entry of Ca2+ activated by store depletion of Ca2+ in endoplasmic reticulum (ER). We have showed that thioredoxin-1 (Trx-1) plays a role in suppressing ER stress in PD. However, whether Trx-1 regulates TRPC1 expression in PD is still unknown. In the present study, we demonstrated that treatment of 1-methyl-4-phenylpyridinum ion (MPP+ ) significantly reduced the expression of TRPC1 in PC12 cells, which was restored by Trx-1 overexpression, and further decreased significantly by Trx-1 siRNA. Moreover, we found that Ca2+ entered into the cells was decreased by MPP+ in PC 12 cells, which was restored by Trx-1 overexpression, and further decreased by Trx-1 siRNA. MPP+ significantly increased calcium-dependent cysteine protease calpain1 expression in PC12 cells, which was suppressed by Trx-1 overexpression. Calpain1 expression was increased by Trx-1 siRNA or SKF96365, an inhibitor of TRPC1. Moreover, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) decreased TRPC1 expression in the substantia nigra pars compacta region (SNpc), which was restored in mice overexpressing Trx-1, and further decreased in mice of knockdown Trx-1. Inversely, the expression of calpain1 was increased by MPTP, which was suppressed in mice overexpressing Trx-1, and further increased in mice of knockdown Trx-1. In conclusion, Trx-1 regulates the Ca2+ entry through regulating TRPC1 expression after treatment of MPP+ /MPTP.
Collapse
Affiliation(s)
- Xianwen Zhang
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Ruhua Deng
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Se Zhang
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Juan Deng
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Jing Jing Jia
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Bo Sun
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Xiaoshuang Zhou
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| | - Jie Bai
- Laboratory of molecular neurobiology, Medical Faculty, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
20
|
Liu X, Liu W, Wang C, Chen Y, Liu P, Hayashi T, Mizuno K, Hattori S, Fujisaki H, Ikejima T. Silibinin attenuates motor dysfunction in a mouse model of Parkinson's disease by suppression of oxidative stress and neuroinflammation along with promotion of mitophagy. Physiol Behav 2021; 239:113510. [PMID: 34181930 DOI: 10.1016/j.physbeh.2021.113510] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/14/2021] [Accepted: 06/23/2021] [Indexed: 12/14/2022]
Abstract
Silybum marianum (L.) Gaertn has been widely used to obtain a drug for the treatment of hepatic diseases. Silibinin (silybin), a flavonoid extracted and isolated from the fruit of S. marianumis investigated in our study to explore its motor protective potential on Parkinson's disease (PD) model mice induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). PD is a neurodegenerative disease that causes a debilitating movement disorder, characterized by a progressive loss of nigrostriatal (substantia nigra and striatum) dopaminergic neurons. Several studies have proven that neurodegeneration is aggravated by neuroinflammation, oxidative stress and/or the presence of α-synuclein (α-syn) aggregation. Essentially no causal therapy for PD exists at present. Our results demonstrate that silibinin significantly attenuates MPTP-induced movement disorder in behavioral tests. Immunohistochemical analysis shows that MPTP injection results in the loss of dopaminergic neurons in the substantia nigra, and the decrease of the striatal tyrosine hydroxylase. However, MPTP-injected mice were protected against dopaminergic neuronal loss by oral administration of silibinin (280 mg/kg) that increased expressions of PTEN-induced putative kinase 1 (PINK1) and Parkin, suggesting mitophagy activation. The neuroprotective mechanism of silibinin involves not only reduction of mitochondrial damage by repressing proinflammatory response and α-syn aggregation, but also enhancement of oxidative defense system. Namely, protection of dopaminergic nerves is due to promotion of mitophagy, leading to clearance of the toxic effects of damaged mitochondria. These findings suggest that silibinin has a potential to be further developed as a therapeutic candidate for PD.
Collapse
Affiliation(s)
- Xiumin Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China
| | - Weiwei Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China
| | - Chenkang Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China
| | - Yinzhe Chen
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China
| | - Panwen Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China
| | - Toshihiko Hayashi
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China; Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, 2665-1, Nakanomachi, Hachioji, Tokyo, 192-0015, Japan; Nippi Research Institute of Biomatrix, Toride, Ibaraki 302-0017, Japan
| | - Kazunori Mizuno
- Nippi Research Institute of Biomatrix, Toride, Ibaraki 302-0017, Japan
| | - Shunji Hattori
- Nippi Research Institute of Biomatrix, Toride, Ibaraki 302-0017, Japan
| | - Hitomi Fujisaki
- Nippi Research Institute of Biomatrix, Toride, Ibaraki 302-0017, Japan
| | - Takashi Ikejima
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China; Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China.
| |
Collapse
|
21
|
Hu L, Si L, Dai X, Dong H, Ma Z, Sun Z, Li N, Sha H, Chen Y, Qian Y, Zhang Z. Exosomal miR-409-3p secreted from activated mast cells promotes microglial migration, activation and neuroinflammation by targeting Nr4a2 to activate the NF-κB pathway. J Neuroinflammation 2021; 18:68. [PMID: 33750404 PMCID: PMC7945321 DOI: 10.1186/s12974-021-02110-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 02/16/2021] [Indexed: 02/07/2023] Open
Abstract
Objective Neuroinflammation plays a critical role in central nervous system diseases. Exosomal miRNAs released from various cells are implicated in cell-to-cell communication. Prior studies have placed substantial emphasis on the role of cytokines in mast cell-microglia interactions during neuroinflammation. However, it has never been clearly determined whether exosomal miRNAs participate in the interaction between mast cells and microglia and thus mediate neuroinflammation. Methods The characteristics of exosomes isolated from cell culture supernatants were confirmed by transmission electron microscopy (TEM), nanoparticle-tracking analysis (NTA) and Western blot. The transfer of PKH67-labelled exosomes and Cy3-labelled miR-409-3p was observed by fluorescence microscopy. Migration and activation of murine BV-2 microglial cells were evaluated through Transwell assays and immunofluorescence staining for Iba1 and CD68. CD86, IL-1β, IL-6 and TNF-α were assessed via qRT-PCR and ELISA. MiR-409-3p was detected by qRT-PCR. Nr4a2 and NF-κB levels were measured by western blot. Regulatory effects were identified by luciferase reporter assays. Results Lipopolysaccharide (LPS)-stimulated murine P815 mast cells secreted exosomes that were efficiently taken up by murine BV-2 cells, which promoted murine BV-2 cell migration and activation. LPS-P815 exosomes increased the CD86, IL-1β, IL-6 and TNF-α levels in murine BV-2 microglia. Furthermore, activated mast cells delivered exosomal miR-409-3p to murine BV-2 microglia. Upregulated miR-409-3p promoted murine BV-2 microglial migration, activation and neuroinflammation by targeting Nr4a2 to activate the NF-κB pathway. Conclusion Exosomal miR-409-3p secreted from activated mast cells promotes microglial migration, activation and neuroinflammation by targeting Nr4a2 to activate the NF-κB pathway, which provides evidence that not only cytokines but also exosomal miRNAs participate in neuroinflammation. In the future, targeting exosomal miRNAs may provide new insights into neuroinflammation.
Collapse
Affiliation(s)
- Liuqing Hu
- Department of Anesthesiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Linjie Si
- Department of Cardiovascular Surgery, the First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Xiaonan Dai
- Department of Obstetrics, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, 210004, China
| | - Hongquan Dong
- Department of Anesthesiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Zijian Ma
- Department of Thoracic Surgery, the First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Zhaochu Sun
- Department of Anesthesiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Nana Li
- Department of Anesthesiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Huanhuan Sha
- Department of Anesthesiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Yinan Chen
- Department of Anesthesiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Yanning Qian
- Department of Anesthesiology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China.
| | - Zhiyuan Zhang
- Department of Pathology, Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
22
|
Kempuraj D, Thangavel R, Kempuraj DD, Ahmed ME, Selvakumar GP, Raikwar SP, Zaheer SA, Iyer SS, Govindarajan R, Chandrasekaran PN, Zaheer A. Neuroprotective effects of flavone luteolin in neuroinflammation and neurotrauma. Biofactors 2021; 47:190-197. [PMID: 33098588 DOI: 10.1002/biof.1687] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/27/2020] [Accepted: 09/29/2020] [Indexed: 12/13/2022]
Abstract
Neuroinflammation leads to neurodegeneration, cognitive defects, and neurodegenerative disorders. Neurotrauma/traumatic brain injury (TBI) can cause activation of glial cells, neurons, and neuroimmune cells in the brain to release neuroinflammatory mediators. Neurotrauma leads to immediate primary brain damage (direct damage), neuroinflammatory responses, neuroinflammation, and late secondary brain damage (indirect) through neuroinflammatory mechanism. Secondary brain damage leads to chronic inflammation and the onset and progression of neurodegenerative diseases. Currently, there are no effective and specific therapeutic options to treat these brain damages or neurodegenerative diseases. Flavone luteolin is an important natural polyphenol present in several plants that show anti-inflammatory, antioxidant, anticancer, cytoprotective, and macrophage polarization effects. In this short review article, we have reviewed the neuroprotective effects of luteolin in neurotrauma and neurodegenerative disorders and pathways involved in this mechanism. We have collected data for this study from publications in the PubMed using the keywords luteolin and mast cells, neuroinflammation, neurodegenerative diseases, and TBI. Recent reports suggest that luteolin suppresses systemic and neuroinflammatory responses in Coronavirus disease 2019 (COVID-19). Studies have shown that luteolin exhibits neuroprotective effects through various mechanisms, including suppressing immune cell activation, such as mast cells, and inflammatory mediators released from these cells. In addition, luteolin can suppress neuroinflammatory response, activation of microglia and astrocytes, oxidative stress, neuroinflammation, and the severity of neuroinflammatory diseases such as Alzheimer's disease, Parkinson's disease, multiple sclerosis, and TBI pathogenesis. In conclusion, luteolin can improve cognitive decline and enhance neuroprotection in neurodegenerative diseases, TBI, and stroke.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- The Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, Missouri, USA
| | - Ramasamy Thangavel
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- The Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, Missouri, USA
| | - Deepak D Kempuraj
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- David H. Hickman High School, Columbia Public Schools, Columbia, Missouri, USA
| | - Mohammad Ejaz Ahmed
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- The Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, Missouri, USA
| | - Govindhasamy Pushpavathi Selvakumar
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- The Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, Missouri, USA
| | - Sudhanshu P Raikwar
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- The Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, Missouri, USA
| | - Smita A Zaheer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- The Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Shankar S Iyer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- The Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, Missouri, USA
| | - Raghav Govindarajan
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | | | - Asgar Zaheer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- The Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, Missouri, USA
| |
Collapse
|
23
|
Chen Y, Su Z, Liu F. Effects of functionally diverse calpain system on immune cells. Immunol Res 2021; 69:8-17. [PMID: 33483937 DOI: 10.1007/s12026-021-09177-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/18/2021] [Indexed: 12/12/2022]
Abstract
Calpains are a family of nonlysosomal cysteine proteases, which play important roles in numerous physiological and pathological processes. Locations of them dictates the functions so that they are classified as ubiquitously expressed calpains and tissue-specific calpains. Recent studies are mainly focused on conventional calpains (calpain-1,2) in development and diseases, and increasing people pay attention to other subtypes of calpains but may not been summarized appropriately. Growing evidence suggests that calpains are also involved in immune regulation. However, seldom articles review the regulation of calpains on immune cells. The aim of this article is to review the research progress of each calpain isozyme and the effect of calpains on immune cells, especially the promotion effect of calpains on the immune response of macrophage, neutrophils, dendritic cells, mast cells, natural killed cells, and lymphocytes. These effects would hold great promise for the clinical application of calpains as a practicable therapeutic option in the treatment of immune related diseases.
Collapse
Affiliation(s)
- Yueqi Chen
- International Genome Center, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.,Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | - Zhaoliang Su
- International Genome Center, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.,Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | - Fang Liu
- International Genome Center, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
24
|
Ahmed S, Kwatra M, Ranjan Panda S, Murty USN, Naidu VGM. Andrographolide suppresses NLRP3 inflammasome activation in microglia through induction of parkin-mediated mitophagy in in-vitro and in-vivo models of Parkinson disease. Brain Behav Immun 2021; 91:142-158. [PMID: 32971182 DOI: 10.1016/j.bbi.2020.09.017] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 01/08/2023] Open
Abstract
Cellular communication linking microglia activation and dopaminergic neuronal loss play an imperative role in the progression of Parkinson's disease (PD); however, underlying molecular mechanisms are not precise and require further elucidation. NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome activation is extensively studied in context to microglial activation and progressive dopaminergic neuronal loss in PD. Several pathophysiological factors such as oxidative stress, mitochondrial dysfunction impaired mitophagy plays a crucial role in activating NLRP3 inflammasome complex. Hence, regulation of microglial activation through mitophagy could be a valuable strategy in controlling microglia mediated neurodegeneration. In this study we have developed a model of inflammasome activation by combining LPS with a mitochondrial complex-I inhibitor MPP+. The idea of using MPP+ after priming mouse microglia with LPS was to disrupt mitochondria and release reactive oxygen species, which act as Signal 2 in augmenting NLRP3 assembly, thereby releasing potent inflammatory mediators such as active interleukin-1 beta (IL-1β) and IL-18. LPS-MPP+ combination was seen to impaired the mitophagy by inhibiting the initial step of autophagosome formation as evidenced by protein expression and confocal imaging data. Treatment with Andrographolide promoted the parkin-dependent autophagic flux formation in microglia; resulting in the removal of defective mitochondria which in turn inhibit NLRP3 inflammasome activation. Additionally, the neuroprotective role of Andrographolide in inhibiting NLRP3 activation together with salvage ATP level via promoting parkin-dependent mitophagy was seen in the substantial nigra par compacta (SNpc) region of mice brain. Furthermore, Andrographolide rescued the dopaminergic neuron loss and improved the behavioural parameters in animal model. Collectively, our results reveal the role of mitophagy in the regulation of NLRP3 inflammasome by removing defective mitochondria. In addition, andrographolide was seen to abate NLRP3 inflammasome activation in microglia and rescue dopaminergic neuron loss.
Collapse
Affiliation(s)
- Sahabuddin Ahmed
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Assam 781101, India
| | - Mohit Kwatra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Assam 781101, India
| | - Samir Ranjan Panda
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Assam 781101, India
| | - U S N Murty
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Assam 781101, India
| | - V G M Naidu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Assam 781101, India.
| |
Collapse
|
25
|
Ahmed ME, Selvakumar GP, Kempuraj D, Raikwar SP, Thangavel R, Bazley K, Wu K, Khan O, Khan A, Zaheer S, Iyer S, Burton C, James D, Zaheer A. Glia Maturation Factor (GMF) Regulates Microglial Expression Phenotypes and the Associated Neurological Deficits in a Mouse Model of Traumatic Brain Injury. Mol Neurobiol 2020; 57:4438-4450. [PMID: 32737763 DOI: 10.1007/s12035-020-02040-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 07/24/2020] [Indexed: 12/20/2022]
Abstract
Traumatic brain injury (TBI) induces inflammatory responses through microglial activation and polarization towards a more inflammatory state that contributes to the deleterious secondary brain injury. Glia maturation factor (GMF) is a pro-inflammatory protein that is responsible for neuroinflammation following insult to the brain, such as in TBI. We hypothesized that the absence of GMF in GMF-knockout (GMF-KO) mice would regulate microglial activation state and the M1/M2 phenotypes following TBI. We used the weight drop model of TBI in C57BL/6 mice wild-type (WT) and GMF-KO mice. Immunofluorescence staining, Western blot, and ELISA assays were performed to confirm TBI-induced histopathological and neuroinflammatory changes. Behavioral analysis was done to check motor coordination ability and cognitive function. We demonstrated that the deletion of GMF in GMF-KO mice significantly limited lesion volume, attenuated neuronal loss, inhibited gliosis, and activated microglia adopted predominantly anti-inflammatory (M2) phenotypes. Using an ELISA method, we found a gradual decrease in pro-inflammatory cytokines (TNF-α and IL-6) and upregulation of anti-inflammatory cytokines (IL-4 and IL-10) in GMF-KO mice compared with WT mice, thus, promoting the transition of microglia towards a more predominantly anti-inflammatory (M2) phenotype. GMF-KO mice showed significant improvement in motor ability, memory, and cognition. Overall, our results demonstrate that GMF deficiency regulates microglial polarization, which ameliorates neuronal injury and behavioral impairments following TBI in mice and concludes that GMF is a regulator of neuroinflammation and an ideal therapeutic target for the treatment of TBI.
Collapse
Affiliation(s)
- Mohammad Ejaz Ahmed
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | - Govindhasamy Pushpavathi Selvakumar
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | - Duraisamy Kempuraj
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | - Sudhanshu P Raikwar
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | - Ramasamy Thangavel
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | - Kieran Bazley
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Kristopher Wu
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Osaid Khan
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Asher Khan
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Smita Zaheer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Shankar Iyer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA
| | | | | | - Asgar Zaheer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.
| |
Collapse
|
26
|
Selvakumar GP, Ahmed ME, Iyer SS, Thangavel R, Kempuraj D, Raikwar SP, Bazley K, Wu K, Khan A, Kukulka K, Bussinger B, Zaheer S, Burton C, James D, Zaheer A. Absence of Glia Maturation Factor Protects from Axonal Injury and Motor Behavioral Impairments after Traumatic Brain Injury. Exp Neurobiol 2020; 29:230-248. [PMID: 32565489 PMCID: PMC7344375 DOI: 10.5607/en20017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/09/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) causes disability and death, accelerating the progression towards Alzheimer's disease and Parkinson's disease (PD). TBI causes serious motor and cognitive impairments, as seen in PD that arise during the period of the initial insult. However, this has been understudied relative to TBI induced neuroinflammation, motor and cognitive decline that progress towards PD. Neuronal ubiquitin-C-terminal hydrolase- L1 (UCHL1) is a thiol protease that breaks down ubiquitinated proteins and its level represents the severity of TBI. Previously, we demonstrated the molecular action of glia maturation factor (GMF); a proinflammatory protein in mediating neuroinflammation and neuronal loss. Here, we show that the weight drop method induced TBI neuropathology using behavioral tests, western blotting, and immunofluorescence techniques on sections from wild type (WT) and GMF-deficient (GMF-KO) mice. Results reveal a significant improvement in substantia nigral tyrosine hydroxylase and dopamine transporter expression with motor behavioral performance in GMF-KO mice following TBI. In addition, a significant reduction in neuroinflammation was manifested, as shown by activation of nuclear factor-kB, reduced levels of inducible nitric oxide synthase, and cyclooxygenase- 2 expressions. Likewise, neurotrophins including brain-derived neurotrophic factor and glial-derived neurotrophic factor were significantly improved in GMF-KO mice than WT 72 h post-TBI. Consistently, we found that TBI enhances GFAP and UCHL-1 expression and reduces the number of dopaminergic TH-positive neurons in WT compared to GMF-KO mice 72 h post-TBI. Interestingly, we observed a reduction of THpositive tanycytes in the median eminence of WT than GMF-KO mice. Overall, we found that absence of GMF significantly reversed these neuropathological events and improved behavioral outcome. This study provides evidence that PD-associated pathology progression can be initiated upon induction of TBI.
Collapse
Affiliation(s)
- Govindhasamy Pushpavathi Selvakumar
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Mohammad Ejaz Ahmed
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Shankar S Iyer
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Ramasamy Thangavel
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Duraisamy Kempuraj
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Sudhanshu P Raikwar
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Kieran Bazley
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Kristopher Wu
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Asher Khan
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Klaudia Kukulka
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Bret Bussinger
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | - Smita Zaheer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| | | | | | - Asgar Zaheer
- Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri 65211, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA.,Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri 65211, USA
| |
Collapse
|
27
|
Mast Cell Activation, Neuroinflammation, and Tight Junction Protein Derangement in Acute Traumatic Brain Injury. Mediators Inflamm 2020; 2020:4243953. [PMID: 32684835 PMCID: PMC7333064 DOI: 10.1155/2020/4243953] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/28/2020] [Accepted: 06/02/2020] [Indexed: 02/06/2023] Open
Abstract
Traumatic brain injury (TBI) is one of the major health problems worldwide that causes death or permanent disability through primary and secondary damages in the brain. TBI causes primary brain damage and activates glial cells and immune and inflammatory cells, including mast cells in the brain associated with neuroinflammatory responses that cause secondary brain damage. Though the survival rate and the neurological deficiencies have shown significant improvement in many TBI patients with newer therapeutic options, the underlying pathophysiology of TBI-mediated neuroinflammation, neurodegeneration, and cognitive dysfunctions is understudied. In this study, we analyzed mast cells and neuroinflammation in weight drop-induced TBI. We analyzed mast cell activation by toluidine blue staining, serum chemokine C-C motif ligand 2 (CCL2) level by enzyme-linked immunosorbent assay (ELISA), and proteinase-activated receptor-2 (PAR-2), a mast cell and inflammation-associated protein, vascular endothelial growth factor receptor 2 (VEGFR2), and blood-brain barrier tight junction-associated claudin 5 and Zonula occludens-1 (ZO-1) protein expression in the brains of TBI mice. Mast cell activation and its numbers increased in the brains of 24 h and 72 h TBI when compared with sham control brains without TBI. Mouse brains after TBI show increased CCL2, PAR-2, and VEGFR2 expression and derangement of claudin 5 and ZO-1 expression as compared with sham control brains. TBI can cause mast cell activation, neuroinflammation, and derangement of tight junction proteins associated with increased BBB permeability. We suggest that inhibition of mast cell activation can suppress neuroimmune responses and glial cell activation-associated neuroinflammation and neurodegeneration in TBI.
Collapse
|