1
|
Kaur R, Kumar S, Singh L. A comprehensive review: neuroinflammation and immune communication between the central nervous system and the periphery. Cytokine 2025; 192:156974. [PMID: 40449035 DOI: 10.1016/j.cyto.2025.156974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/30/2025] [Accepted: 05/27/2025] [Indexed: 06/02/2025]
Abstract
Immunity in the central nervous system (CNS) is generally attributed to neuron-associated microglia in the parenchyma. Microglial cells are specialized macrophages that interact closely with neurons to monitor them for signs of infection or injury. In addition to microglia, several other specialized macrophage populations are located at the borders of the CNS, including dural, leptomeningeal, perivascular, and choroid plexus macrophages. Collectively, these are CNS-associated macrophages (CAMs), but how these cells maintain the balance between the segregation of the CNS and the information transfer between the CNS parenchyma and the peripheral system is not well understood. The interaction between the immune system and the CNS is a newly emerging field of study that focuses on the functions of resident microglia and specialized macrophages, including leptomeningeal, choroid plexus, and perivascular macrophages. This review will help to improve understanding of the regulatory mechanisms of microglia and specialized macrophages and their involvement in the communication with the peripheral immune system. It could also advance neurological disease therapies that selectively target specific immune function parameters more effectively for managing neurodegenerative diseases.
Collapse
Affiliation(s)
- Ramandeep Kaur
- Animal Biotechnology, ICAR-National Dairy Research Institute, Karnal, India.
| | - Satish Kumar
- Animal Biochemistry, ICAR-National Dairy Research Institute, Karnal, India
| | - Lakhwinder Singh
- Centre for Drug Discovery, Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|
2
|
Gómez-Guerrero E, Debray-García Y, Aztatzi-Aguilar OG, Colchero-Amateco FA, Amador-Muñoz O, Poblano-Bata J, Poblete-Naredo I, Albores A. A549 cells exposed to a marijuana smoke extract affect mono-layer integrity related to oxidative stress state. Toxicol In Vitro 2025; 107:106072. [PMID: 40250736 DOI: 10.1016/j.tiv.2025.106072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 03/14/2025] [Accepted: 04/09/2025] [Indexed: 04/20/2025]
Abstract
Marijuana smoke contains several toxic compounds that may induce dysregulation of oxidative mechanisms and barrier system in airway alveolar cells. This study aimed to assess whether marijuana smoke extract (MSE) modifies mono-layer integrity and antioxidant effects on the alveolar epithelial cells. A549 cells were exposed to MSE (0.1 to 5 μg/mL) or cannabinoid (+)-WIN 55,212-2 (WIN; 0.01 to 1 μM) for 24 h. Epithelial integrity and protein expression of claudin (Cl)-2, Cl-5, and occludin (Ocl) were evaluated by transepithelial electrical resistance (TEER), permeability assay, Western blot, immunofluorescence, and qPCR. TEER decreased after MSE or WIN exposure, whereas the monolayer permeability increased. Protein expression and localization of Cl-2 and Ocl were reduced after MSE treatment. However, WIN increased Cl-2 protein and decreased Cl-5 and Ocl. Differential gene expressions were observed between treatments. Malondialdehyde (MDA) production and advanced oxidation protein products (AOPP) determination showed that MSE increased AOPP, whereas WIN augmented the MDA production and decreased AOPP levels. The activity of antioxidant enzymes shows an increase in catalase, glutathione-S-transferase, γ-glutamyl transferase and arginase after MSE treatment and a decrease with WIN. Data indicates that MSE exposure alters epithelial integrity and the alveolar cells antioxidant response that, finally, may lead to lung damage.
Collapse
Affiliation(s)
- Elvira Gómez-Guerrero
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (Cinvestav), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360 Ciudad de México, Mexico
| | - Yazmín Debray-García
- Departamento de Investigación de Toxicología y Medicina Ambiental, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, Mexico
| | - Octavio Gamaliel Aztatzi-Aguilar
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (Cinvestav), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360 Ciudad de México, Mexico
| | - Fanny Azuzena Colchero-Amateco
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (Cinvestav), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360 Ciudad de México, Mexico
| | - Omar Amador-Muñoz
- Instituto de Ciencias de la Atmósfera y Cambio Climático, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, Mexico
| | - Josefina Poblano-Bata
- Instituto de Ciencias de la Atmósfera y Cambio Climático, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México, Mexico
| | - Irais Poblete-Naredo
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (Cinvestav), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360 Ciudad de México, Mexico
| | - Arnulfo Albores
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN (Cinvestav), Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360 Ciudad de México, Mexico.
| |
Collapse
|
3
|
Dudek KA, Paton SEJ, Binder LB, Collignon A, Dion-Albert L, Cadoret A, Lebel M, Lavoie O, Bouchard J, Kaufmann FN, Clavet-Fournier V, Manca C, Guzmán M, Campbell M, Turecki G, Mechawar N, Flamand N, Lavoie-Cardinal F, Silvestri C, Di Marzo V, Menard C. Astrocytic cannabinoid receptor 1 promotes resilience by dampening stress-induced blood-brain barrier alterations. Nat Neurosci 2025; 28:766-782. [PMID: 40016352 PMCID: PMC11976283 DOI: 10.1038/s41593-025-01891-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 01/08/2025] [Indexed: 03/01/2025]
Abstract
Blood-brain barrier (BBB) alterations contribute to stress vulnerability and the development of depressive behaviors. In contrast, neurovascular adaptations underlying stress resilience remain unclear. Here we report that high expression of astrocytic cannabinoid receptor 1 (CB1) in the nucleus accumbens (NAc) shell, particularly in the end-feet ensheathing blood vessels, is associated with resilience during chronic social stress in adult male mice. Viral-mediated overexpression of Cnr1 in astrocytes of the NAc shell results in baseline anxiolytic effects and dampens stress-induced anxiety- and depression-like behaviors in male mice. It promotes the expression of vascular-related genes and reduces astrocyte inflammatory response and morphological changes following an immune challenge with the cytokine interleukin-6, linked to stress susceptibility and mood disorders. Physical exercise and antidepressant treatment increase the expression of astrocytic Cnr1 in the perivascular region in male mice. In human tissue from male donors with major depressive disorder, we observe loss of CNR1 in the NAc astrocytes. Our findings suggest a role for the astrocytic endocannabinoid system in stress responses via modulation of the BBB.
Collapse
Affiliation(s)
- Katarzyna A Dudek
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Sam E J Paton
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Luisa Bandeira Binder
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Adeline Collignon
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Laurence Dion-Albert
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Alice Cadoret
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Manon Lebel
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Olivier Lavoie
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Jonathan Bouchard
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Fernanda Neutzling Kaufmann
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Valerie Clavet-Fournier
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Claudia Manca
- Faculty of Medicine and Quebec Heart and Lung Institute, Université Laval, Quebec City, Quebec, Canada
| | - Manuel Guzmán
- Department of Biochemistry and Molecular Biology, Complutense University of Madrid, CIBERNED and IRYCIS, Madrid, Spain
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - Gustavo Turecki
- Department of Psychiatry, McGill University and Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Naguib Mechawar
- Department of Psychiatry, McGill University and Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Nicolas Flamand
- Faculty of Medicine and Quebec Heart and Lung Institute, Université Laval, Quebec City, Quebec, Canada
| | - Flavie Lavoie-Cardinal
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Cristoforo Silvestri
- Faculty of Medicine and Quebec Heart and Lung Institute, Université Laval, Quebec City, Quebec, Canada
| | - Vincenzo Di Marzo
- Faculty of Medicine and Quebec Heart and Lung Institute, Université Laval, Quebec City, Quebec, Canada
- Faculty of Agricultural and Food Sciences, INAF and NUTRISS Center, Quebec City, Quebec, Canada
- Joint International Research Unit on Chemical and Biomolecular Research on the Microbiome and its Impact on Metabolic Health and Nutrition between Université Laval, Quebec City, Quebec, Canada
- Consiglio Nazionale Delle Ricerche, Institute of Biomolecular Chemistry, Pozzuoli, Italy
| | - Caroline Menard
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Quebec, Canada.
| |
Collapse
|
4
|
Mac CH, Nguyen GLT, Nguyen DTM, Huang SM, Peng HH, Chang Y, Lo SK, Chiang HHK, Yang YZ, Song HL, Chia WT, Lin YJ, Sung HW. Noninvasive Vagus Nerve Electrical Stimulation for Immune Modulation in Sepsis Therapy. J Am Chem Soc 2025; 147:8406-8421. [PMID: 40033812 PMCID: PMC11912339 DOI: 10.1021/jacs.4c16367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/05/2025]
Abstract
Sepsis presents a significant medical challenge due to its intense inflammatory response to infection, often resulting in high mortality rates. A promising therapeutic strategy targets the cholinergic anti-inflammatory pathway (CAIP), which regulates immune responses. This study investigates the ingestion of piezoelectric particles that adhere to the stomach lining, specifically targeting TRPV1 receptors. In a mouse model of sepsis, these particles, when activated by low-intensity pulsed ultrasound, generate mild electrical pulses. These pulses stimulate vagal afferent fibers, transmitting signals to the brain and modulating the neural-immune network via the CAIP. Consequently, this leads to a reduction in systemic inflammation, mitigating weight loss, alleviating multiple tissue injuries, and preventing death by modulating immune cells in the spleen. This approach addresses the critical need for noninvasive sepsis therapies, potentially improving patient outcomes. Utilizing portable ultrasound equipment with minimal thermal effects, this technique offers a safe and convenient treatment option, even for home use.
Collapse
Affiliation(s)
- Cam-Hoa Mac
- Department
of Chemical Engineering, National Tsing
Hua University, Hsinchu 300044, Taiwan
| | - Giang Le Thi Nguyen
- Department
of Chemical Engineering, National Tsing
Hua University, Hsinchu 300044, Taiwan
| | - Dien Thi My Nguyen
- Department
of Chemical Engineering, National Tsing
Hua University, Hsinchu 300044, Taiwan
| | - Sheng-Min Huang
- Department
of Pharmacology, College of Medicine, National
Cheng Kung University, Tainan 701401, Taiwan
| | - Hsu-Hsia Peng
- Department
of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Yen Chang
- Taipei
Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation and School of
Medicine, Tzu Chi University, Hualien 970473, Taiwan
| | - Shih-Kai Lo
- Department
of Chemical Engineering, National Tsing
Hua University, Hsinchu 300044, Taiwan
| | - Hui-Hua Kenny Chiang
- Institute
of Biomedical Engineering, National Yang-Ming
Chiao Tung University, Taipei 112304, Taiwan
| | - Yuan-Zhen Yang
- Institute
of Biomedical Engineering, National Yang-Ming
Chiao Tung University, Taipei 112304, Taiwan
| | - Hsiang-Lin Song
- Department
of Pathology, National Taiwan University
Hospital, Hsinchu Branch, Hsinchu 302058, Taiwan
| | - Wei-Tso Chia
- Department
of Orthopedics, National Taiwan University
Hospital, Hsinchu Branch, Hsinchu 302058, Taiwan
| | - Yu-Jung Lin
- Research
Center for Applied Sciences, Academia Sinica, Taipei 115201, Taiwan
| | - Hsing-Wen Sung
- Department
of Chemical Engineering, National Tsing
Hua University, Hsinchu 300044, Taiwan
| |
Collapse
|
5
|
Ferrara AL, Palestra F, Piscitelli F, Petraroli A, Suffritti C, Firinu D, López-Lera A, Caballero T, Bork K, Spadaro G, Marone G, Di Marzo V, Bova M, Loffredo S. Altered levels of phospholipases C, diacylglycerols, endocannabinoids, and N-acylethanolamines in patients with hereditary angioedema due to FXII mutation. Allergy 2025; 80:287-296. [PMID: 38935036 DOI: 10.1111/all.16197] [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: 01/30/2024] [Revised: 05/31/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Hereditary angioedema (HAE) is a rare genetic disorder characterized by local, self-limiting edema due to temporary increase in vascular permeability. HAE with normal C1 esterase inhibitor (C1INH) activity includes the form with mutations in the F12 gene encoding for coagulation factor XII (FXII-HAE) causing an overproduction of bradykinin (BK) leading to angioedema attack. BK binding to B2 receptors (BK2R) leads to an activation of phospholipase C (PLC) and subsequent generation of second messengers: diacylglycerols (DAGs) and possibly the endocannabinoids (eCBs), 2-arachidonoylglycerol (2-AG) and anandamide (AEA), and eCB-related N-acylethanolamines [palmitoylethanolamide (PEA) and oleoylethanolamide (OEA)]. To date, there are no data on the role of these lipid mediators in FXII-HAE. METHODS Here, we analyzed plasma levels of PLC, DAGs, and eCBs in 40 patients with FXII-HAE and 40 sex- and age-matched healthy individuals. RESULTS Plasma PLC activity was increased in FXII-HAE patients compared to controls. Concentrations of DAG 18:1-20:4, a lipid second messenger produced by PLC, were higher in FXII-HAE compared to controls, and positively correlated with PLC activity and cleaved high molecular kininogen (cHK). Also the concentrations of the DAG metabolite, 2-AG were altered in FXII-HAE. AEA and OEA were decreased in FXII-HAE patients compared to controls; by contrast, PEA, was increased. The levels of all tested mediators did not differ between symptomatic and asymptomatic patients. Moreover, C1INH-HAE patients had elevated plasma levels of PLC, which correlated with cHK, but the levels of DAGs and eCBs were the same as controls. CONCLUSIONS BK overproduction and BKR2 activation are linked to alteration of PLCs and their metabolites in patients with FXII-HAE. Our results may pave way to investigations on the functions of these mediators in the pathophysiology of FXII-HAE, and provide new potential biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Anne Lise Ferrara
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research, University of Naples Federico II, WAO Center of Excellence, Naples, Italy
| | - Francesco Palestra
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research, University of Naples Federico II, WAO Center of Excellence, Naples, Italy
| | - Fabiana Piscitelli
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare-Consiglio Nazionale delle Ricerche, Pozzuoli, Italy
| | - Angelica Petraroli
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research, University of Naples Federico II, WAO Center of Excellence, Naples, Italy
- Italian Network for Hereditary and Acquired Angioedema, Napoli, Italy
| | - Chiara Suffritti
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy
| | - Davide Firinu
- Italian Network for Hereditary and Acquired Angioedema, Napoli, Italy
- Internal Medicine, Allergy and Clinical Immunology, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Alberto López-Lera
- Hospital La Paz Institute for Health Research (IdiPAZ), CIBERER (U754), Madrid, Spain
| | - Teresa Caballero
- Allergy Department, Hospital Universitario La Paz, Hospital La Paz Institute for Health Research (IdiPAZ), CIBERER (U754), Madrid, Spain
| | - Konrad Bork
- Department of Dermatology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Giuseppe Spadaro
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research, University of Naples Federico II, WAO Center of Excellence, Naples, Italy
- Italian Network for Hereditary and Acquired Angioedema, Napoli, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research, University of Naples Federico II, WAO Center of Excellence, Naples, Italy
- Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Research Council, Naples, Italy
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare-Consiglio Nazionale delle Ricerche, Pozzuoli, Italy
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Quèbec, and Centre NUTRISS, Institut sur la Nutrition et les Aliments Fonctionnels, Université Laval, Québec City, Canada
| | - Maria Bova
- Department of Internal Medicine, A.O.R.N. Antonio Cardarelli, Naples, Italy
| | - Stefania Loffredo
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research, University of Naples Federico II, WAO Center of Excellence, Naples, Italy
- Italian Network for Hereditary and Acquired Angioedema, Napoli, Italy
- Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Research Council, Naples, Italy
| |
Collapse
|
6
|
Kanjanasirirat P, Saengsawang W, Ketsawatsomkron P, Asavapanumas N, Borwornpinyo S, Soodvilai S, Hongeng S, Charoensutthivarakul S. GDNF and cAMP significantly enhance in vitro blood-brain barrier integrity in a humanized tricellular transwell model. Heliyon 2024; 10:e39343. [PMID: 39492921 PMCID: PMC11530796 DOI: 10.1016/j.heliyon.2024.e39343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 10/11/2024] [Accepted: 10/11/2024] [Indexed: 11/05/2024] Open
Abstract
Blood-brain barrier (BBB) is a crucial membrane safeguarding neural tissue by controlling the molecular exchange between blood and the brain. However, assessing BBB permeability presents challenges for central nervous system (CNS) drug development. In vitro studies of BBB-permeable agents before animal testing are essential to mitigate failures. Improved in vitro models are needed to mimic physiologically relevant BBB integrity. Here, we established an in vitro human-derived triculture BBB model, coculturing hCMEC/D3 with primary astrocytes and pericytes in a transwell format. This study found that the triculture BBB model exhibited significantly higher paracellular tightness (TEER 147.6 ± 6.5 Ω × cm2) than its monoculture counterpart (106.3 ± 1.0 Ω × cm2). Additionally, BBB permeability in the triculture model was significantly lower. While GDNF and cAMP have been shown to promote BBB integrity in monoculture models, their effect in our model was previously unreported. Our study demonstrates that both GDNF and cAMP increased TEER values (around 200 Ω × cm2 for each; 237.6 ± 17.7 Ω × cm2 for co-treatment) compared to untreated control, and decreased BBB permeability, mediated by increased claudin-5 expression. In summary, this humanized triculture BBB model, enhanced by GDNF and cAMP, offers an alternative for exploring in vitro drug penetration into the human brain.
Collapse
Affiliation(s)
- Phongthon Kanjanasirirat
- School of Bioinnovation and Bio-Based Product Intelligence, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Excellent Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Witchuda Saengsawang
- Department of Basic Biomedical Sciences, Dr. William M. Scholl College of Podiatric Medicine, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
| | - Pimonrat Ketsawatsomkron
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakarn, 10540, Thailand
| | - Nithi Asavapanumas
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakarn, 10540, Thailand
| | - Suparerk Borwornpinyo
- Excellent Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Department of Biotechnology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Sunhapas Soodvilai
- Excellent Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Suradej Hongeng
- Excellent Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | - Sitthivut Charoensutthivarakul
- School of Bioinnovation and Bio-Based Product Intelligence, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Excellent Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
- Center for Neuroscience, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| |
Collapse
|
7
|
Socała K, Jakubiec M, Abram M, Mlost J, Starowicz K, Kamiński RM, Ciepiela K, Andres-Mach M, Zagaja M, Metcalf CS, Zawadzki P, Wlaź P, Kamiński K. TRPV1 channel in the pathophysiology of epilepsy and its potential as a molecular target for the development of new antiseizure drug candidates. Prog Neurobiol 2024; 240:102634. [PMID: 38834133 DOI: 10.1016/j.pneurobio.2024.102634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 04/26/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024]
Abstract
Identification of transient receptor potential cation channel, subfamily V member 1 (TRPV1), also known as capsaicin receptor, in 1997 was a milestone achievement in the research on temperature sensation and pain signalling. Very soon after it became evident that TRPV1 is implicated in a wide array of physiological processes in different peripheral tissues, as well as in the central nervous system, and thereby could be involved in the pathophysiology of numerous diseases. Increasing evidence suggests that modulation of TRPV1 may also affect seizure susceptibility and epilepsy. This channel is localized in brain regions associated with seizures and epilepsy, and its overexpression was found both in animal models of seizures and in brain samples from epileptic patients. Moreover, modulation of TRPV1 on non-neuronal cells (microglia, astrocytes, and/or peripheral immune cells) may have an impact on the neuroinflammatory processes that play a role in epilepsy and epileptogenesis. In this paper, we provide a comprehensive and critical overview of currently available data on TRPV1 as a possible molecular target for epilepsy management, trying to identify research gaps and future directions. Overall, several converging lines of evidence implicate TRPV1 channel as a potentially attractive target in epilepsy research but more studies are needed to exploit the possible role of TRPV1 in seizures/epilepsy and to evaluate the value of TRPV1 ligands as candidates for new antiseizure drugs.
Collapse
Affiliation(s)
- Katarzyna Socała
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, Lublin PL 20-033, Poland.
| | - Marcin Jakubiec
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland
| | - Michał Abram
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland
| | - Jakub Mlost
- Department of Neurochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, Cracow PL 31-343, Poland
| | - Katarzyna Starowicz
- Department of Neurochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, Cracow PL 31-343, Poland
| | - Rafał M Kamiński
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland
| | - Katarzyna Ciepiela
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland; Selvita S.A., Bobrzyńskiego 14, Cracow PL 30-348, Poland
| | - Marta Andres-Mach
- Department of Experimental Pharmacology, Institute of Rural Health, Jaczewskiego 2, Lublin PL 20-090, Poland
| | - Mirosław Zagaja
- Department of Experimental Pharmacology, Institute of Rural Health, Jaczewskiego 2, Lublin PL 20-090, Poland
| | - Cameron S Metcalf
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA
| | - Przemysław Zawadzki
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland
| | - Piotr Wlaź
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, Lublin PL 20-033, Poland
| | - Krzysztof Kamiński
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland
| |
Collapse
|
8
|
Malik JR, Modebelu UO, Fletcher CV, Podany AT, Scarsi KK, Byrareddy SN, Anand RK, Buch S, Sil S, Le J, Bradley JS, Brown AN, Sutar D, Avedissian SN. Establishment of a Four-Cell In Vitro Blood-Brain Barrier Model With Human Primary Brain Cells. Curr Protoc 2024; 4:e1067. [PMID: 38857108 PMCID: PMC11783448 DOI: 10.1002/cpz1.1067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
The blood-brain barrier (BBB) constitutes a crucial protective anatomical layer with a microenvironment that tightly controls material transit. Constructing an in vitro BBB model to replicate in vivo features requires the sequential layering of constituent cell types. Maintaining heightened integrity in the observed tight junctions during both the establishment and post-experiment phases is crucial to the success of these models. We have developed an in vitro BBB model that replicates the cellular composition and spatial orientation of in vivo BBB observed in humans. The experiment includes comprehensive procedures and steps aimed at enhancing the integration of the four-cell model. Departing from conventional in vitro BBB models, our methodology eliminates the necessity for pre-coated plates to facilitate cell adhesion, thereby improving cell visualization throughout the procedure. An in-house coating strategy and a simple yet effective approach significantly reduce costs and provides superior imaging of cells and corresponding tight junction protein expression. Also, our BBB model includes all four primary cell types that are structural parts of the human BBB. With its innovative and user-friendly features, our in-house optimized in vitro four-cell-based BBB model showcases novel methodology and provides a promising experimental platform for drug screening processes. © 2024 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Coating and culture system Basic Protocol 2: Cell seeding and Transwell insert handling Basic Protocol 3: Assessment of model functionality.
Collapse
Affiliation(s)
- Johid R. Malik
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska
- These authors contributed equally to this work
| | - Ukamaka O. Modebelu
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska
- These authors contributed equally to this work
| | - Courtney V. Fletcher
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska
- Division of Infectious Diseases, Department of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Anthony T. Podany
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska
| | - Kimberly K. Scarsi
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska
- Division of Infectious Diseases, Department of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Siddappa N. Byrareddy
- Department of Pharmacology & Experimental Neurosciences, University of Nebraska Medical Center, Omaha, Nebraska
| | | | - Shilpa Buch
- Department of Pharmacology & Experimental Neurosciences, University of Nebraska Medical Center, Omaha, Nebraska
| | - Susmia Sil
- Department of Pharmacology & Experimental Neurosciences, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jennifer Le
- University of California San Diego, Skaggs School of Pharmacy and Pharmaceutical Science, La Jolla, California
| | - John S. Bradley
- University of California San Diego School of Medicine, Department of Pediatrics, San Diego, California
| | - Ashley N. Brown
- Institute for Therapeutic Innovation, College of Medicine, University of Florida, Orlando, Florida
| | - Debapriya Sutar
- Department of Pharmacology & Experimental Neurosciences, University of Nebraska Medical Center, Omaha, Nebraska
| | - Sean N. Avedissian
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
9
|
Zeng J, Lu Y, Chu H, Lu L, Chen Y, Ji K, Lin Y, Li J, Wang S. Research trends and frontier hotspots of TRPV1 based on bibliometric and visualization analyses. Heliyon 2024; 10:e24153. [PMID: 38293347 PMCID: PMC10827456 DOI: 10.1016/j.heliyon.2024.e24153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/30/2023] [Accepted: 01/04/2024] [Indexed: 02/01/2024] Open
Abstract
Background Transient receptor potential vanilloid type1 (TRPV1) is a non-selective cation channel with multiple activation mechanisms, which has received increasing attention since it was first cloned in 1997. Methods We used bibliometric and visualization analyses to evaluate the theme trends and knowledge structure of TRPV1 research-papers on TRPV1 from 2002 to 2022 obtained from the Web of Science Core Collection. VOSviewer and CiteSpace were used to analyze authors, institutions, countries, co-cited references, and keywords. Results A total of 7413 papers were included. The main research area of TRPV1 was neuroscience; the most published country was the United States, and the University of California, San Francisco, had the highest centrality. Two major collaborative sub-networks were formed between the authors. The distribution of keywords shows that TRPV1 was initially studied extensively, and the recent studies focused on TRPV1 structure and diseases. "Oxidative stress," "TRPV1 structure," "cancer," and "model" have been the research hotspots in recent years. Conclusions This research provides valuable information for the study of TRPV1. Disease research was focused on pain, cancer, and neurodegenerative diseases. Both agonists and antagonists of TRPV1 are gradually being used in clinical practice, and acupuncture was effective in treating TRPV1-mediated inflammatory pain. TRPV1 is involved in classical endogenous cannabis system signaling, and new signaling pathways continue to be revealed.
Collapse
Affiliation(s)
- Jingchun Zeng
- Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yiqian Lu
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Hui Chu
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Liming Lu
- Clinical Research and Data Center, South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yuexuan Chen
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Kaisong Ji
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yeze Lin
- Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jingjing Li
- Bao'an Traditional Chinese Medicine Hospital//Seventh Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, Shenzhen, Guangdong, China
| | - Shuxin Wang
- Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
10
|
Pérez-Martín E, Pérez-Revuelta L, Barahona-López C, Pérez-Boyero D, Alonso JR, Díaz D, Weruaga E. Oleoylethanolamide Treatment Modulates Both Neuroinflammation and Microgliosis, and Prevents Massive Leukocyte Infiltration to the Cerebellum in a Mouse Model of Neuronal Degeneration. Int J Mol Sci 2023; 24:ijms24119691. [PMID: 37298639 DOI: 10.3390/ijms24119691] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/29/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Neurodegenerative diseases involve an exacerbated neuroinflammatory response led by microglia that triggers cytokine storm and leukocyte infiltration into the brain. PPARα agonists partially dampen this neuroinflammation in some models of brain insult, but neuronal loss was not the triggering cause in any of them. This study examines the anti-inflammatory and immunomodulatory properties of the PPARα agonist oleoylethanolamide (OEA) in the Purkinje Cell Degeneration (PCD) mouse, which exhibits striking neuroinflammation caused by aggressive loss of cerebellar Purkinje neurons. Using real-time quantitative polymerase chain reaction and immunostaining, we quantified changes in pro- and anti-inflammatory markers, microglial density and marker-based phenotype, and overall leukocyte recruitment at different time points after OEA administration. OEA was found to modulate cerebellar neuroinflammation by increasing the gene expression of proinflammatory mediators at the onset of neurodegeneration and decreasing it over time. OEA also enhanced the expression of anti-inflammatory and neuroprotective factors and the Pparα gene. Regarding microgliosis, OEA reduced microglial density-especially in regions where it is preferentially located in PCD mice-and shifted the microglial phenotype towards an anti-inflammatory state. Finally, OEA prevented massive leukocyte infiltration into the cerebellum. Overall, our findings suggest that OEA may change the environment to protect neurons from degeneration caused by exacerbated inflammation.
Collapse
Affiliation(s)
- Ester Pérez-Martín
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Laura Pérez-Revuelta
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Cristina Barahona-López
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain
| | - David Pérez-Boyero
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - José R Alonso
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - David Díaz
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Eduardo Weruaga
- Laboratory of Neuronal Plasticity and Neurorepair, Institute of Neuroscience of Castile and Leon (INCyL), Universidad de Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
| |
Collapse
|
11
|
Gräfe EL, Reid HMO, Shkolnikov I, Conway K, Kit A, Acosta C, Christie BR. Women are Taking the Hit: Examining the Unique Consequences of Cannabis Use Across the Female Lifespan. Front Neuroendocrinol 2023; 70:101076. [PMID: 37217080 DOI: 10.1016/j.yfrne.2023.101076] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/28/2023] [Accepted: 05/15/2023] [Indexed: 05/24/2023]
Abstract
Cannabis use has risen dramatically in recent years due to global decriminalization and a resurgence in the interest of potential therapeutic benefits. While emerging research is shaping our understanding of the benefits and harms of cannabis, there remains a paucity of data specifically focused on how cannabis affects the female population. The female experience of cannabis use is unique, both in the societal context and because of the biological ramifications. This is increasingly important given the rise in cannabis potency, as well as the implications this has for the prevalence of Cannabis Use Disorder (CUD). Therefore, this scoping review aims to discuss the prevalence of cannabis use and CUD in women throughout their lifespan and provide a balanced prospective on the positive and negative consequences of cannabis use. In doing so, this review will highlight the necessity for continued research that goes beyond sex differences.
Collapse
Affiliation(s)
- E L Gräfe
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - H M O Reid
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - I Shkolnikov
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - K Conway
- Island Medical Program, University of British Columbia, Victoria, British Columbia, Canada
| | - A Kit
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - C Acosta
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - B R Christie
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada; Island Medical Program, University of British Columbia, Victoria, British Columbia, Canada.
| |
Collapse
|
12
|
Malik JR, Fletcher CV, Podany AT, Dyavar SR, Scarsi KK, Pais GM, Scheetz MH, Avedissian SN. A novel 4-cell in-vitro blood-brain barrier model and its characterization by confocal microscopy and TEER measurement. J Neurosci Methods 2023; 392:109867. [PMID: 37116621 PMCID: PMC10275325 DOI: 10.1016/j.jneumeth.2023.109867] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/18/2023] [Accepted: 04/25/2023] [Indexed: 04/30/2023]
Abstract
The blood-brain barrier (BBB) is a protective cellular anatomical layer with a dynamic micro-environment, tightly regulating the transport of materials across it. To achieve in-vivo characteristics, an in-vitro BBB model requires the constituent cell types to be layered in an appropriate order. A cost-effective in-vitro BBB model is desired to facilitate central nervous system (CNS) drug penetration studies. Enhanced integrity of tight junctions observed during the in-vitro BBB establishment and post-experiment is essential in these models. We successfully developed an in-vitro BBB model mimicking the in-vivo cell composition and a distinct order of seeding primary human brain cells. Unlike other in-vitro BBB models, our work avoids the need for pre-coated plates for cell adhesion and provides better cell visualization during the procedure. We found that using bovine collagen-I coating, followed by bovine fibronectin coating and poly-L-lysine coating, yields better adhesion and layering of cells on the transwell membrane compared to earlier reported use of collagen and poly-L-lysine only. Our results indicated better cell visibility and imaging with the polyester transwell membrane as well as point to a higher and more stable Trans Endothelial Electrical Resistance values in this plate. In addition, we found that the addition of zinc induced higher claudin 5 expressions in neuronal cells. Dolutegravir, a drug used in the treatment of HIV, is known to appear in moderate concentrations in the CNS. Thus, dolutegravir was used to assess the functionality of the final model and cells. Using primary cells and an in-house coating strategy substantially reduces costs and provides superior imaging of cells and their tight junction protein expression. Our 4-cell-based BBB model is a suitable experimental model for the drug screening process.
Collapse
Affiliation(s)
- Johid R Malik
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Courtney V Fletcher
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA; Division of Infectious Diseases, Department of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Anthony T Podany
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Kimberly K Scarsi
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA; Division of Infectious Diseases, Department of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Gwendolyn M Pais
- Department of Pharmacy Practice, Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, USA; Midwestern University, College of Pharmacy Center of Pharmacometric Excellence, Downers Grove, IL, USA
| | - Marc H Scheetz
- Department of Pharmacy Practice, Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, USA; Midwestern University, College of Pharmacy Center of Pharmacometric Excellence, Downers Grove, IL, USA
| | - Sean N Avedissian
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
13
|
Rodgers TM, Muzzio N, Valero A, Ahmad I, Lüdtke TU, Moya SE, Romero G. Poly (β-amino Ester) Nanoparticles Modified with a Rabies Virus-derived peptide for the Delivery of ASCL1 Across a 3D In Vitro Model of the Blood Brain Barrier. ACS APPLIED NANO MATERIALS 2023; 6:6299-6311. [PMID: 37274933 PMCID: PMC10234607 DOI: 10.1021/acsanm.3c00651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Gene editing has emerged as a therapeutic approach to manipulate the genome for killing cancer cells, protecting healthy tissues, and improving immune response to a tumor. The gene editing tool achaete-scute family bHLH transcription factor 1 CRISPR guide RNA (ASCL1-gRNA) is known to restore neuronal lineage potential, promote terminal differentiation, and attenuate tumorigenicity in glioblastoma tumors. Here, we fabricated a polymeric nonviral carrier to encapsulate ASCL1-gRNA by electrostatic interactions and deliver it into glioblastoma cells across a 3D in vitro model of the blood-brain barrier (BBB). To mimic rabies virus (RV) neurotropism, gene-loaded poly (β-amino ester) nanoparticles are surface functionalized with a peptide derivative of rabies virus glycoprotein (RVG29). The capability of the obtained NPs, hereinafter referred to as RV-like NPs, to travel across the BBB, internalize into glioblastoma cells and deliver ASCL1-gRNA are investigated in a 3D BBB in vitro model through flow cytometry and CLSM microscopy. The formation of nicotinic acetylcholine receptors in the 3D BBB in vitro model is confirmed by immunochemistry. These receptors are known to bind to RVG29. Unlike Lipofectamine that primarily internalizes and transfects endothelial cells, RV-like NPs are capable to travel across the BBB, preferentially internalize glioblastoma cells and deliver ASCL1-gRNA at an efficiency of 10 % causing non-cytotoxic effects.
Collapse
Affiliation(s)
- Tina M Rodgers
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, Texas, 78249, USA
| | - Nicolas Muzzio
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, Texas, 78249, USA
| | - Andrea Valero
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, Texas, 78249, USA
| | - Ikram Ahmad
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, Texas, 78249, USA
| | - Tanja Ursula Lüdtke
- Soft Matter Nanotechnology, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramon 182, Donostia/San Sebastian, Gipuzkoa, 20014 Spain
| | - Sergio E Moya
- Soft Matter Nanotechnology, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramon 182, Donostia/San Sebastian, Gipuzkoa, 20014 Spain
| | - Gabriela Romero
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, Texas, 78249, USA
| |
Collapse
|
14
|
Gonzales-Aloy E, Ahmed-Cox A, Tsoli M, Ziegler DS, Kavallaris M. From cells to organoids: The evolution of blood-brain barrier technology for modelling drug delivery in brain cancer. Adv Drug Deliv Rev 2023; 196:114777. [PMID: 36931346 DOI: 10.1016/j.addr.2023.114777] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/13/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023]
Abstract
Brain cancer remains the deadliest cancer. The blood-brain barrier (BBB) is impenetrable to most drugs and is a complex 3D network of multiple cell types including endothelial cells, astrocytes, and pericytes. In brain cancers, the BBB becomes disrupted during tumor progression and forms the blood-brain tumor barrier (BBTB). To advance therapeutic development, there is a critical need for physiologically relevant BBB in vitro models. 3D cell systems are emerging as valuable preclinical models to accelerate discoveries for diseases. Given the versatility and capability of 3D cell models, their potential for modelling the BBB and BBTB is reviewed. Technological advances of BBB models and challenges of in vitro modelling the BBTB, and application of these models as tools for assessing therapeutics and nano drug delivery, are discussed. Quantitative, in vitro BBB models that are predictive of effective brain cancer therapies will be invaluable for accelerating advancing new treatments to the clinic.
Collapse
Affiliation(s)
- Estrella Gonzales-Aloy
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, NSW, Australia; Australian Center for NanoMedicine, UNSW Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, NSW, Australia
| | - Aria Ahmed-Cox
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, NSW, Australia; Australian Center for NanoMedicine, UNSW Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, NSW, Australia; Katharina Gaus Light Microscopy Facility, Mark Wainright Analytical Center, UNSW Sydney, NSW, Australia
| | - Maria Tsoli
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, NSW, Australia
| | - David S Ziegler
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, NSW, Australia; Kids Cancer Center, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Maria Kavallaris
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, NSW, Australia; Australian Center for NanoMedicine, UNSW Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, NSW, Australia; UNSW RNA Institute, UNSW Sydney, NSW, Australia.
| |
Collapse
|
15
|
Rani V, Prabhu A. In vitro blood brain barrier models: Molecular aspects and therapeutic strategies in glioma management. Curr Res Transl Med 2023; 71:103376. [PMID: 36580825 DOI: 10.1016/j.retram.2022.103376] [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: 07/22/2022] [Revised: 10/19/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Glioma management is the most challenging task in clinical oncology due to numerous reasons. One of the major hurdles in glioma therapy is the presence of blood brain barrier which resists the entry of most of the drugs into the brain. However, in case of tumors, blood brain barrier integrity is compromised, which in turn can be advantageous in delivering the drugs, if the therapeutic module is strategically modified. For such improvised therapeutic strategy, it is necessary to understand the molecular composition and profiling of blood brain barrier and blood brain tumor barrier. This review mainly focuses on the composition, markers expressed on the blood brain barrier which will help the readers to understand its basic environment. It also gives a detailed account of the various in vitro models that are used to study the nature of the blood brain barrier and describes various strategies in improvising the drug delivery in glioma management.
Collapse
Affiliation(s)
- Vinitha Rani
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, 575018 Karnataka, India
| | - Ashwini Prabhu
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Deralakatte, Mangalore, 575018 Karnataka, India.
| |
Collapse
|
16
|
Yao E, Luo L, Lin C, Wen J, Li Y, Ren T, Chen Y, Huang J, Jin X. OEA alleviates apoptosis in diabetic rats with myocardial ischemia/reperfusion injury by regulating the PI3K/Akt signaling pathway through activation of TRPV1. Front Pharmacol 2022; 13:964475. [PMID: 36452230 PMCID: PMC9701823 DOI: 10.3389/fphar.2022.964475] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 10/27/2022] [Indexed: 07/22/2023] Open
Abstract
Reperfusion therapy after myocardial infarction may lead to myocardial injury, which can be complicated and exacerbated by diabetes. The existing therapeutic methods for myocardial ischemia-reperfusion injury (MIRI) in diabetic patients are not ideal. Oleoylethanolamide (OEA) has been found to have protective effects on diabetes and acute cerebral ischemia. This study aimed to determine whether OEA can alleviate MIRI in diabetic rats, and to explore the underlying mechanism. The model of diabetic rats with MIRI was established by blocking the left coronary artery for 30 min, followed by restoring blood flow stability for 120 min. The myocardial enzyme spectrum, area of MIRI, and expression levels of apoptosis-related proteins were detected. The results showed that OEA pretreatment could reduce myocardial infarction area, protect myocardial tissue structure, and reduce myocardial cell apoptosis in diabetic rats with MIRI. Meanwhile, the levels of creatine kinase (CK)-MB (CK-MB), lactate dehydrogenase (LDH), and malondialdehyde (MDA) were reduced, while superoxide dismutase (SOD) level was elevated. H9C2 cells were treated with high glucose and oxygen-glucose deprivation/reperfusion (OGD/R) to establish an in vitro model. Capsazepine (CPZ), an antagonist of transient receptor potential vanilloid subtype 1 (TRPV1), and LY294002, an inhibitor of PI3K, were used to treat H9C2 cells in vitro. Apoptosis level and the expression levels of apoptosis-related proteins were measured. It was found that OEA activated TRPV1 and the PI3K/Akt signaling pathway, downregulated the expression levels of apoptosis-related proteins (Bcl-2 and cleaved caspase-3), and ameliorated the apoptosis of H9C2 cells treated with high glucose and OGD/R. This study clarified that OEA, as a TRPV1 agonist, could reduce myocardial cell apoptosis by activating the PI3K/Akt signaling pathway in diabetic rats with MIRI. The findings may provide a theoretical basis for administration of OEA as a potential therapeutic agent into diabetic patients with MIRI.
Collapse
Affiliation(s)
- Enhui Yao
- Department of Cardiology, Fujian Medical University Union Hospital, Fujian Institute of Coronary Artery Disease, Fujian Heart Medical Center, Fuzhou, China
| | - Lili Luo
- Xiamen Key Laboratory of Chiral Drugs, School of Medicine, Xiamen University, Xiamen, China
| | - Chenxi Lin
- Department of Pediatrics, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jing Wen
- Xiamen Key Laboratory of Chiral Drugs, School of Medicine, Xiamen University, Xiamen, China
| | - Yanglongfei Li
- Department of Cardiology, Fujian Medical University Union Hospital, Fujian Institute of Coronary Artery Disease, Fujian Heart Medical Center, Fuzhou, China
| | - Tong Ren
- Xiamen Key Laboratory of Chiral Drugs, School of Medicine, Xiamen University, Xiamen, China
| | - Yujie Chen
- Department of Cardiology, Fujian Medical University Union Hospital, Fujian Institute of Coronary Artery Disease, Fujian Heart Medical Center, Fuzhou, China
| | - Jinhua Huang
- Department of Cardiology, Fujian Medical University Union Hospital, Fujian Institute of Coronary Artery Disease, Fujian Heart Medical Center, Fuzhou, China
| | - Xin Jin
- Xiamen Key Laboratory of Chiral Drugs, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
17
|
Nicolicht-Amorim P, Delgado-Garcia LM, Nakamura TKE, Courbassier NR, Mosini AC, Porcionatto MA. Simple and efficient protocol to isolate and culture brain microvascular endothelial cells from newborn mice. Front Cell Neurosci 2022; 16:949412. [PMID: 36313615 PMCID: PMC9606660 DOI: 10.3389/fncel.2022.949412] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/06/2022] [Indexed: 10/09/2023] Open
Abstract
The neurovascular unit (NVU) is a multicellular structure comprising of neurons, glial cells, and non-neural cells, and it is supported by a specialized extracellular matrix, the basal lamina. Astrocytes, brain microvascular endothelial cells (BMECs), pericytes, and smooth muscle cells constitute the blood-brain barrier (BBB). BMECs have a mesodermal origin and invade the nervous system early in neural tube development, forming the BBB anatomical core. BMECs are connected by adherent junction complexes composed of integral membrane and cytoplasmic proteins. In vivo and in vitro studies have shown that, given the proximity and relationship with neural cells, BMECs acquire a unique gene expression profile, proteome, and specific mechanical and physical properties compared to endothelial cells from the general vasculature. BMECs are fundamental in maintaining brain homeostasis by regulating transcellular and paracellular transport of fluids, molecules, and cells. Therefore, it is essential to gain in-depth knowledge of the dynamic cellular structure of the cells in the NVU and their interactions with health and disease. Here we describe a significantly improved and simplified protocol using C57BL/6 newborn mice at postnatal day 1 (PND1) to isolate, purify, and culture BMECs monolayers in two different substrates (glass coverslips and transwell culture inserts). In vitro characterization and validation of the BMEC primary culture monolayers seeded on glass or insert included light microscopy, immunolabeling, and gene expression profile. Transendothelial electrical resistance (TEER) measurement and diffusion test were used as functional assays for adherent junction complexes and integrity and permeability of BMECs monolayers. The protocol presented here for the isolation and culture of BMECs is more straightforward than previously published protocols and yields a high number of purified cells. Finally, we tested BMECs function using the oxygen-glucose deprivation (OGD) model of hypoxia. This protocol may be suitable as a bioscaffold for secondary cell seeding allowing the study and better understanding of the NVU.
Collapse
Affiliation(s)
- Priscila Nicolicht-Amorim
- Laboratory of Molecular Neurobiology, Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Lina M. Delgado-Garcia
- Laboratory of Molecular Neurobiology, Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Natália Rodrigues Courbassier
- Laboratory of Molecular Neurobiology, Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Amanda Cristina Mosini
- Laboratory of Neurobiology, Department of Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Marimelia A. Porcionatto
- Laboratory of Molecular Neurobiology, Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
18
|
Hagan K, Varelas P, Zheng H. Endocannabinoid System of the Blood-Brain Barrier: Current Understandings and Therapeutic Potentials. Cannabis Cannabinoid Res 2022; 7:561-568. [PMID: 34918950 PMCID: PMC9587775 DOI: 10.1089/can.2021.0101] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The endocannabinoid system (ECS) has been found at the blood-brain barrier (BBB), as Cannabinoid receptors were characterized in human brain microvascular endothelial cells and astrocytes. In several in vitro and in vivo studies, cannabinoids decreased BBB permeability and enhanced membrane integrity, which may be achieved through endothelial tight junctions and other mechanisms. These permeability regulation effects of cannabinoids suggested that the ECS may protect the brain by enhancing barrier integrity. Related questions about cannabinoid-drug interaction and drug distribution across the BBB are also raised. Specifically, can cannabinoids significantly reduce drug bioavailability to the brain? More in-depth and systematic investigations are needed to characterize and quantify these effects of cannabinoids on brain microvasculature physiopathology. Therefore, this review summarizes literatures from different disciplines to promote more research on assessing the therapeutic benefits and risks of using cannabinoids to protect BBB from dysfunctions or breakdown, and to avoid consequent brain damages due to inflammation, neurodegenerations, hemorrhage, ischemia, or other causes.
Collapse
Affiliation(s)
- Kofi Hagan
- Department of Pharmaceutical Sciences, Albany College of Pharmacy and Health Sciences, Albany, New York, USA
| | | | - HaiAn Zheng
- Department of Pharmaceutical Sciences, Albany College of Pharmacy and Health Sciences, Albany, New York, USA
| |
Collapse
|
19
|
Walker CCF, Sordillo LM, Contreras GA. Anandamide Alters Barrier Integrity of Bovine Vascular Endothelial Cells during Endotoxin Challenge. Antioxidants (Basel) 2022; 11:antiox11081461. [PMID: 36009180 PMCID: PMC9405077 DOI: 10.3390/antiox11081461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 01/27/2023] Open
Abstract
Vascular endothelial cells are crucial mediators of inflammation during infectious diseases, due to their ability to produce lipid-based inflammatory mediators and facilitate leukocyte migration and translocation to infected tissues. Mastitis is the costliest infectious disease in North America, with over two billion dollars in annual costs due to loss of milk production, medical treatment, and potential loss of the animal. Infections caused by coliform bacteria are particularly deleterious, causing a negative impact on cow well-being and a high mortality rate. Dysfunction and breakdown of the endothelial barrier is a key part of the pathology of coliform mastitis. The endocannabinoid system (ECS), shown to modulate inflammatory responses of vascular endothelial cells in humans and rodents, may be a novel target for inflammatory modulation in dairy cows. The endocannabinoid (EC) arachidonoylethanolamide (AEA) is a potent anti- or pro-inflammatory mediator in endothelial cells, depending on location, timing, and concentration. We hypothesized that elevated AEA during LPS challenge will impair endothelial barrier integrity via increased production of reactive oxygen species (ROS) and activation of apoptotic pathways. Challenge of bovine aortic endothelial cells (BAEC) with 25 ng/mL lipopolysaccharide (LPS) for 8 h induced AEA synthesis, increased expression of cannabinoid receptor 1 and 2 (CB1/2) and the AEA synthesizing enzyme N-acyl phosphatidylethanolamine phospholipase D (NAPE-PLD), while decreasing gene expression of the AEA degradation enzyme fatty acid amide hydrolase (FAAH). Trans endothelial resistance (TER), measured through electrical resistance across the monolayer, increased 2 h after 0.5 µM AEA treatment and decreased with 5 µM AEA, compared to LPS alone. Addition of AEA to BAEC challenged with LPS induced mitochondrial dysfunction via increased ROS production, cytochrome-C release, and activation of caspase 3/7. Antagonism of CB1 by 1 µM AM251 ameliorated AEA induced ROS production and cytochrome-C release. Addition of AM251 also eliminated 2 h TER increase and improved TER following 5 µM AEA. Doses of 0.5, 1, and 5 µM AEA delayed endothelial barrier recovery, which was eliminated by the addition of AM251. Mitochondrial dysfunction and activation of apoptotic pathways in response to AEA treatment during LPS challenge of BAEC may act to delay inflammatory resolution and contribute to endothelial dysfunction.
Collapse
|
20
|
Vicente-Acosta A, Ceprian M, Sobrino P, Pazos MR, Loría F. Cannabinoids as Glial Cell Modulators in Ischemic Stroke: Implications for Neuroprotection. Front Pharmacol 2022; 13:888222. [PMID: 35721207 PMCID: PMC9199389 DOI: 10.3389/fphar.2022.888222] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
Stroke is the second leading cause of death worldwide following coronary heart disease. Despite significant efforts to find effective treatments to reduce neurological damage, many patients suffer from sequelae that impair their quality of life. For this reason, the search for new therapeutic options for the treatment of these patients is a priority. Glial cells, including microglia, astrocytes and oligodendrocytes, participate in crucial processes that allow the correct functioning of the neural tissue, being actively involved in the pathophysiological mechanisms of ischemic stroke. Although the exact mechanisms by which glial cells contribute in the pathophysiological context of stroke are not yet completely understood, they have emerged as potentially therapeutic targets to improve brain recovery. The endocannabinoid system has interesting immunomodulatory and protective effects in glial cells, and the pharmacological modulation of this signaling pathway has revealed potential neuroprotective effects in different neurological diseases. Therefore, here we recapitulate current findings on the potential promising contribution of the endocannabinoid system pharmacological manipulation in glial cells for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Andrés Vicente-Acosta
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.,Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| | - Maria Ceprian
- ERC Team, PGNM, INSERM U1315, CNRS UMR5261, University of Lyon 1, University of Lyon, Lyon, France
| | - Pilar Sobrino
- Departamento de Neurología, Hospital Universitario Fundación Alcorcón, Alcorcón, Spain
| | - Maria Ruth Pazos
- Laboratorio de Apoyo a la Investigación, Hospital Universitario Fundación Alcorcón, Alcorcón, Spain
| | - Frida Loría
- Laboratorio de Apoyo a la Investigación, Hospital Universitario Fundación Alcorcón, Alcorcón, Spain
| |
Collapse
|
21
|
Zubrzycki M, Zubrzycka M, Wysiadecki G, Szemraj J, Jerczynska H, Stasiolek M. Release of Endocannabinoids into the Cerebrospinal Fluid during the Induction of the Trigemino-Hypoglossal Reflex in Rats. Curr Issues Mol Biol 2022; 44:2401-2416. [PMID: 35678693 PMCID: PMC9164053 DOI: 10.3390/cimb44050164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 11/16/2022] Open
Abstract
The endocannabinoid system (ECS) plays an important role in pain processing and modulation. Since the specific effects of endocannabinoids within the orofacial area are largely unknown, we aimed to determine whether an increase in the endocannabinoid concentration in the cerebrospinal fluid (CSF) caused by the peripheral administration of the FAAH inhibitor URB597 and tooth pulp stimulation would affect the transmission of impulses between the sensory and motor centers localized in the vicinity of the third and fourth cerebral ventricles. The study objectives were evaluated on rats using a method that allowed the recording of the amplitude of evoked tongue jerks (ETJ) in response to noxious tooth pulp stimulation and URB597 treatment. The amplitude of ETJ was a measure of the effect of endocannabinoids on the neural structures. The concentrations of the endocannabinoids tested (AEA and 2-AG) were determined in the CSF, along with the expression of the cannabinoid receptors (CB1 and CB2) in the tissues of the mesencephalon, thalamus, and hypothalamus. We demonstrated that anandamide (AEA), but not 2-arachidonoylglycerol (2-AG), was significantly increased in the CSF after treatment with a FAAH inhibitor, while tooth pulp stimulation had no effect on the AEA and 2-AG concentrations in the CSF. We also found positive correlations between the CSF AEA concentration and cannabinoid receptor type 1 (CB1R) expression in the brain, and between 2-AG and cannabinoid receptor type 2 (CB2R), and negative correlations between the CSF concentration of AEA and brain CB2R expression, and between 2-AG and CB1R. Our study shows that endogenous AEA, which diffuses through the cerebroventricular ependyma into CSF and exerts a modulatory effect mediated by CB1Rs, alters the properties of neurons in the trigeminal sensory nuclei, interneurons, and motoneurons of the hypoglossal nerve. In addition, our findings may be consistent with the emerging concept that AEA and 2-AG have different regulatory mechanisms because they are involved differently in orofacial pain. We also suggest that FAAH inhibition may offer a therapeutic approach to the treatment of orofacial pain.
Collapse
Affiliation(s)
- Marek Zubrzycki
- Department of Cardiac Surgery and Transplantology, The Cardinal Stefan Wyszynski Institute of Cardiology, Alpejska 42, 04-628 Warsaw, Poland
| | - Maria Zubrzycka
- Department of Clinical Physiology, Faculty of Medicine, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland;
| | - Grzegorz Wysiadecki
- Department of Normal and Clinical Anatomy, Chair of Anatomy and Histology, Medical University of Lodz, Żeligowskiego 7/9, 90-752 Lodz, Poland;
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland;
| | - Hanna Jerczynska
- Central Scientific Laboratory (CoreLab), Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland;
| | - Mariusz Stasiolek
- Department of Neurology, Medical University of Lodz, Kopcinskiego 22, 90-153 Lodz, Poland;
| |
Collapse
|
22
|
Vasincu A, Rusu RN, Ababei DC, Larion M, Bild W, Stanciu GD, Solcan C, Bild V. Endocannabinoid Modulation in Neurodegenerative Diseases: In Pursuit of Certainty. BIOLOGY 2022; 11:biology11030440. [PMID: 35336814 PMCID: PMC8945712 DOI: 10.3390/biology11030440] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/04/2022] [Accepted: 03/10/2022] [Indexed: 01/13/2023]
Abstract
Simple Summary Neurodegenerative diseases represent an important cause of morbidity and mortality worldwide. Existing therapeutic options are limited and focus mostly on improving symptoms and reducing exacerbations. The endocannabinoid system is involved in the pathophysiology of such disorders, an idea which has been highlighted by recent scientific work. The current work focusses its attention on the importance and implications of this system and its synthetic and natural ligands in disorders such as Alzheimer’s, Parkinson’s, Huntington’s and multiple sclerosis. Abstract Neurodegenerative diseases are an increasing cause of global morbidity and mortality. They occur in the central nervous system (CNS) and lead to functional and mental impairment due to loss of neurons. Recent evidence highlights the link between neurodegenerative and inflammatory diseases of the CNS. These are typically associated with several neurological disorders. These diseases have fundamental differences regarding their underlying physiology and clinical manifestations, although there are aspects that overlap. The endocannabinoid system (ECS) is comprised of receptors (type-1 (CB1R) and type-2 (CB2R) cannabinoid-receptors, as well as transient receptor potential vanilloid 1 (TRPV1)), endogenous ligands and enzymes that synthesize and degrade endocannabinoids (ECBs). Recent studies revealed the involvement of the ECS in different pathological aspects of these neurodegenerative disorders. The present review will explore the roles of cannabinoid receptors (CBRs) and pharmacological agents that modulate CBRs or ECS activity with reference to Alzheimer’s Disease (AD), Parkinson’s Disease (PD), Huntington’s Disease (HD) and multiple sclerosis (MS).
Collapse
Affiliation(s)
- Alexandru Vasincu
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (A.V.); (D.-C.A.); (V.B.)
| | - Răzvan-Nicolae Rusu
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (A.V.); (D.-C.A.); (V.B.)
- Correspondence:
| | - Daniela-Carmen Ababei
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (A.V.); (D.-C.A.); (V.B.)
| | - Mădălina Larion
- Department of Anaesthesiology Intensive Therapy, Regional Institute of Gastroenterology and Hepatology “Prof. Dr. Octavian Fodor”, 19 Croitorilor Street, 400162 Cluj-Napoca, Romania;
- Department of Anaesthetics, Midland Regional Hospital, Longford Road, Mullingar, N91 NA43 Co. Westmeath, Ireland
| | - Walther Bild
- Department of Physiology, “Grigore T Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania;
- Center of Biomedical Research of the Romanian Academy, 700506 Iasi, Romania
| | - Gabriela Dumitrița Stanciu
- Center for Advanced Research and Development in Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania;
| | - Carmen Solcan
- Preclinics Department, “Ion Ionescu de la Brad” University of Life Sciences, 8 M. Sadoveanu Alley, 700489 Iasi, Romania;
| | - Veronica Bild
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (A.V.); (D.-C.A.); (V.B.)
- Center of Biomedical Research of the Romanian Academy, 700506 Iasi, Romania
- Center for Advanced Research and Development in Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania;
| |
Collapse
|
23
|
Li Y, Zhou P, Hu T, Ren J, Xu Y, Qiu Y, Lu C, Li Y. NAAA inhibitor F96 attenuates BBB disruption and secondary injury after traumatic brain injury (TBI). Eur J Pharmacol 2021; 912:174561. [PMID: 34655598 DOI: 10.1016/j.ejphar.2021.174561] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/29/2021] [Accepted: 10/11/2021] [Indexed: 11/19/2022]
Abstract
Traumatic brain injury (TBI) is a leading cause of death worldwide, for which there is currently no comprehensive treatment available. Preventing blood-brain barrier (BBB) disruption is crucial for TBI treatment. N-acylethanolamine acid amidase (NAAA)-regulated palmitoylethanolamide (PEA) signaling play an important role in the control of inflammation. However, the role of NAAA in BBB dysfunction following TBI remains unclear. In the present study, we found that TBI induces the increase of PEA levels in the injured cortex, which prevent the disruption of BBB after TBI. TBI also induces the infiltration of NAAA-contained neutrophils, increasing the contribution of NAAA to the PEA degradation. Neutrophil-derived NAAA weakens PEA/PPARα-mediated BBB protective effects after TBI, facilitates the accumulation of immune cells, leading to secondary expansion of tissue injury. Inactivation of NAAA increased PEA levels in injured site, prevents early BBB damage and improves secondary injury, thereby eliciting long-term functional improvements after TBI. This study identified a new role of NAAA in TBI, suggesting that NAAA is a new important target for BBB dysfunction related CNS diseases.
Collapse
Affiliation(s)
- Yitian Li
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, And Fujian Provincial Key Laboratory of Nanomaterials, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, China; Medical College, Xiamen University, Xiamen, Fujian, 361102, China; Department of Clinical Pharmacy, The Third Hospital of Mianyang/Sichuan Mental Health Center, Mianyang, 621000, Sichuan, China
| | - Pan Zhou
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, And Fujian Provincial Key Laboratory of Nanomaterials, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, China; Medical College, Xiamen University, Xiamen, Fujian, 361102, China; Department of Pathology, Qilu Hospital, Shandong University, Jinan, 250012 China
| | - Ting Hu
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, And Fujian Provincial Key Laboratory of Nanomaterials, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, China; Xiamen Institute of Rare-earth Materials, Haixi Institutes, Chinese Academy of Sciences, Fujian, 361005, China
| | - Jie Ren
- Medical College, Xiamen University, Xiamen, Fujian, 361102, China; Eye Institute of Xiamen University, Xiamen, Fujian, 361102, China
| | - Yaping Xu
- Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen Medical College, China
| | - Yan Qiu
- Medical College, Xiamen University, Xiamen, Fujian, 361102, China; Eye Institute of Xiamen University, Xiamen, Fujian, 361102, China
| | - Canzhong Lu
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, And Fujian Provincial Key Laboratory of Nanomaterials, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, China; Xiamen Institute of Rare-earth Materials, Haixi Institutes, Chinese Academy of Sciences, Fujian, 361005, China
| | - Yuhang Li
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, And Fujian Provincial Key Laboratory of Nanomaterials, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, China; Xiamen Institute of Rare-earth Materials, Haixi Institutes, Chinese Academy of Sciences, Fujian, 361005, China; Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen Medical College, China.
| |
Collapse
|
24
|
Lowe H, Toyang N, Steele B, Bryant J, Ngwa W. The Endocannabinoid System: A Potential Target for the Treatment of Various Diseases. Int J Mol Sci 2021; 22:9472. [PMID: 34502379 PMCID: PMC8430969 DOI: 10.3390/ijms22179472] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/23/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023] Open
Abstract
The Endocannabinoid System (ECS) is primarily responsible for maintaining homeostasis, a balance in internal environment (temperature, mood, and immune system) and energy input and output in living, biological systems. In addition to regulating physiological processes, the ECS directly influences anxiety, feeding behaviour/appetite, emotional behaviour, depression, nervous functions, neurogenesis, neuroprotection, reward, cognition, learning, memory, pain sensation, fertility, pregnancy, and pre-and post-natal development. The ECS is also involved in several pathophysiological diseases such as cancer, cardiovascular diseases, and neurodegenerative diseases. In recent years, genetic and pharmacological manipulation of the ECS has gained significant interest in medicine, research, and drug discovery and development. The distribution of the components of the ECS system throughout the body, and the physiological/pathophysiological role of the ECS-signalling pathways in many diseases, all offer promising opportunities for the development of novel cannabinergic, cannabimimetic, and cannabinoid-based therapeutic drugs that genetically or pharmacologically modulate the ECS via inhibition of metabolic pathways and/or agonism or antagonism of the receptors of the ECS. This modulation results in the differential expression/activity of the components of the ECS that may be beneficial in the treatment of a number of diseases. This manuscript in-depth review will investigate the potential of the ECS in the treatment of various diseases, and to put forth the suggestion that many of these secondary metabolites of Cannabis sativa L. (hereafter referred to as "C. sativa L." or "medical cannabis"), may also have potential as lead compounds in the development of cannabinoid-based pharmaceuticals for a variety of diseases.
Collapse
Affiliation(s)
- Henry Lowe
- Biotech R & D Institute, University of the West Indies, Mona 99999, Jamaica; (H.L.); (J.B.)
- Vilotos Pharmaceuticals Inc., Baltimore, MD 21202, USA;
- Flavocure Biotech Inc., Baltimore, MD 21202, USA
- Department of Medicine, University of Maryland Medical School, Baltimore, MD 21202, USA
| | - Ngeh Toyang
- Vilotos Pharmaceuticals Inc., Baltimore, MD 21202, USA;
- Flavocure Biotech Inc., Baltimore, MD 21202, USA
| | - Blair Steele
- Biotech R & D Institute, University of the West Indies, Mona 99999, Jamaica; (H.L.); (J.B.)
| | - Joseph Bryant
- Biotech R & D Institute, University of the West Indies, Mona 99999, Jamaica; (H.L.); (J.B.)
| | - Wilfred Ngwa
- Brigham and Women’s Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA;
- Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| |
Collapse
|
25
|
Coronado-Álvarez A, Romero-Cordero K, Macías-Triana L, Tatum-Kuri A, Vera-Barrón A, Budde H, Machado S, Yamamoto T, Imperatori C, Murillo-Rodríguez E. The synthetic CB 1 cannabinoid receptor selective agonists: Putative medical uses and their legalization. Prog Neuropsychopharmacol Biol Psychiatry 2021; 110:110301. [PMID: 33741446 DOI: 10.1016/j.pnpbp.2021.110301] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/17/2021] [Accepted: 03/08/2021] [Indexed: 02/07/2023]
Abstract
More than 500 molecules have been identified as components of Cannabis sativa (C. sativa), of which the most studied is Δ9-tetrahydrocannabinol (Δ9-THC). Several studies have suggested that Δ9-THC exerts diverse biological effects, ranging from fragmentation of DNA to behavioral disruptions. Currently, it is accepted that most of the pharmacological properties of Δ9-THC engage the activation of the cannabinoid receptors, named CB1 and CB2. Interestingly, multiple pieces of evidence have suggested that the cannabinoid receptors play an active role in the modulation of several diseases leading to the design of synthetic cannabinoid-like compounds. Advances in the development of synthetic CB1 cannabinoid receptor selective agonists as therapeutical approaches are, however, limited. This review focuses on available evidence searched in PubMed regarding the synthetic CB1 cannabinoid receptor selective agonists such as AM-1235, arachidonyl-2' chloroethylamide (ACEA), CP 50,556-1 (Levonantradol), CP-55,940, HU-210, JWH-007, JWH-018, JWH-200 (WIN 55,225), methanandamide, nabilone, O-1812, UR-144, WIN 55,212-2, nabiximols, and dronabinol. Indeed, it would be ambitious to describe all available evidence related to the synthetic CB1 cannabinoid receptor selective agonists. However, and despite the positive evidence on the positive results of using these compounds in experimental models of health disturbances and preclinical trials, we discuss evidence in regards some concerns due to side effects.
Collapse
Affiliation(s)
- Astrid Coronado-Álvarez
- Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud, Universidad Anáhuac Mayab, Mérida, Yucatán, Mexico; Intercontinental Neuroscience Research Group, Mexico
| | - Karen Romero-Cordero
- Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud, Universidad Anáhuac Mayab, Mérida, Yucatán, Mexico; Intercontinental Neuroscience Research Group, Mexico
| | - Lorena Macías-Triana
- Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud, Universidad Anáhuac Mayab, Mérida, Yucatán, Mexico; Intercontinental Neuroscience Research Group, Mexico
| | - Agnes Tatum-Kuri
- Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud, Universidad Anáhuac Mayab, Mérida, Yucatán, Mexico; Intercontinental Neuroscience Research Group, Mexico
| | - Alba Vera-Barrón
- Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud, Universidad Anáhuac Mayab, Mérida, Yucatán, Mexico; Intercontinental Neuroscience Research Group, Mexico
| | - Henning Budde
- Intercontinental Neuroscience Research Group, Mexico; Medical School Hamburg, Hamburg, Germany
| | - Sérgio Machado
- Intercontinental Neuroscience Research Group, Mexico; Laboratory of Physical Activity Neuroscience, Physical Activity Sciences Postgraduate Program, Salgado de Oliveira University, Niterói, Brazil
| | - Tetsuya Yamamoto
- Intercontinental Neuroscience Research Group, Mexico; Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima, Japan
| | - Claudio Imperatori
- Intercontinental Neuroscience Research Group, Mexico; Cognitive and Clinical Psychology Laboratory, Department of Human Sciences, European University of Rome, Rome, Italy
| | - Eric Murillo-Rodríguez
- Laboratorio de Neurociencias Moleculares e Integrativas, Escuela de Medicina, División Ciencias de la Salud, Universidad Anáhuac Mayab, Mérida, Yucatán, Mexico; Intercontinental Neuroscience Research Group, Mexico.
| |
Collapse
|
26
|
Shen G, Hu S, Zhao Z, Zhang L, Ma Q. C-Type Natriuretic Peptide Ameliorates Vascular Injury and Improves Neurological Outcomes in Neonatal Hypoxic-Ischemic Brain Injury in Mice. Int J Mol Sci 2021; 22:ijms22168966. [PMID: 34445671 PMCID: PMC8396645 DOI: 10.3390/ijms22168966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/10/2021] [Accepted: 08/17/2021] [Indexed: 01/01/2023] Open
Abstract
C-type natriuretic peptide (CNP) is an important vascular regulator that is present in the brain. Our previous study demonstrated the innate neuroprotectant role of CNP in the neonatal brain after hypoxic-ischemic (HI) insults. In this study, we further explored the role of CNP in cerebrovascular pathology using both in vivo and in vitro models. In a neonatal mouse HI brain injury model, we found that intracerebroventricular administration of recombinant CNP dose-dependently reduces brain infarct size. CNP significantly decreases brain edema and immunoglobulin G (IgG) extravasation into the brain tissue, suggesting a vasculoprotective effect of CNP. Moreover, in primary brain microvascular endothelial cells (BMECs), CNP dose-dependently protects BMEC survival and monolayer integrity against oxygen-glucose deprivation (OGD). The vasculoprotective effect of CNP is mediated by its innate receptors NPR2 and NPR3, in that inhibition of either NPR2 or NPR3 counteracts the protective effect of CNP on IgG leakage after HI insult and BMEC survival under OGD. Of importance, CNP significantly ameliorates brain atrophy and improves neurological deficits after HI insults. Altogether, the present study indicates that recombinant CNP exerts vascular protection in neonatal HI brain injury via its innate receptors, suggesting a potential therapeutic target for the treatment of neonatal HI brain injury.
Collapse
Affiliation(s)
- Guofang Shen
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (G.S.); (S.H.); (L.Z.)
| | - Shirley Hu
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (G.S.); (S.H.); (L.Z.)
| | - Zhen Zhao
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
| | - Lubo Zhang
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (G.S.); (S.H.); (L.Z.)
| | - Qingyi Ma
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (G.S.); (S.H.); (L.Z.)
- Correspondence: ; Tel.: +1-909-558-4325; Fax: +1-909-558-4029
| |
Collapse
|
27
|
Inampudi C, Ciccotosto GD, Cappai R, Crack PJ. Genetic Modulators of Traumatic Brain Injury in Animal Models and the Impact of Sex-Dependent Effects. J Neurotrauma 2021; 37:706-723. [PMID: 32027210 DOI: 10.1089/neu.2019.6955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Traumatic brain injury (TBI) is a major health problem causing disability and death worldwide. There is no effective treatment, due in part to the complexity of the injury pathology and factors affecting its outcome. The extent of brain injury depends on the type of insult, age, sex, lifestyle, genetic risk factors, socioeconomic status, other co-injuries, and underlying health problems. This review discusses the genes that have been directly tested in TBI models, and whether their effects are known to be sex-dependent. Sex differences can affect the incidence, symptom onset, pathology, and clinical outcomes following injury. Adult males are more susceptible at the acute phase and females show greater injury in the chronic phase. TBI is not restricted to a single sex; despite variations in the degree of symptom onset and severity, it is important to consider both female and male animals in TBI pre-clinical research studies.
Collapse
Affiliation(s)
- Chaitanya Inampudi
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| | - Giuseppe D Ciccotosto
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| | - Roberto Cappai
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| | - Peter J Crack
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
28
|
Estrada JA, Contreras I. Endocannabinoid Receptors in the CNS: Potential Drug Targets for the Prevention and Treatment of Neurologic and Psychiatric Disorders. Curr Neuropharmacol 2021; 18:769-787. [PMID: 32065105 PMCID: PMC7536826 DOI: 10.2174/1570159x18666200217140255] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/14/2019] [Accepted: 02/11/2020] [Indexed: 12/15/2022] Open
Abstract
The endocannabinoid system participates in the regulation of CNS homeostasis and functions, including neurotransmission, cell signaling, inflammation and oxidative stress, as well as neuronal and glial cell proliferation, differentiation, migration and survival. Endocannabinoids are produced by multiple cell types within the CNS and their main receptors, CB1 and CB2, are expressed in both neurons and glia. Signaling through these receptors is implicated in the modulation of neuronal and glial alterations in neuroinflammatory, neurodegenerative and psychiatric conditions, including Alzheimer’s, Parkinson’s and Huntington’s disease, multiple sclerosis, amyotrophic lateral sclerosis, stroke, epilepsy, anxiety and depression. The therapeutic potential of endocannabinoid receptors in neurological disease has been hindered by unwelcome side effects of current drugs used to target them; however, due to their extensive expression within the CNS and their involvement in physiological and pathological process in nervous tissue, they are attractive targets for drug development. The present review highlights the potential applications of the endocannabinoid system for the prevention and treatment of neurologic and psychiatric disorders.
Collapse
Affiliation(s)
- José Antonio Estrada
- Neurochemistry Laboratory, Faculty of Medicine, Universidad Autónoma del Estado de México, Toluca, Mexico
| | - Irazú Contreras
- Neurochemistry Laboratory, Faculty of Medicine, Universidad Autónoma del Estado de México, Toluca, Mexico
| |
Collapse
|
29
|
Morris G, Walder K, Kloiber S, Amminger P, Berk M, Bortolasci CC, Maes M, Puri BK, Carvalho AF. The endocannabinoidome in neuropsychiatry: Opportunities and potential risks. Pharmacol Res 2021; 170:105729. [PMID: 34119623 DOI: 10.1016/j.phrs.2021.105729] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/31/2021] [Accepted: 06/09/2021] [Indexed: 02/08/2023]
Abstract
The endocannabinoid system (ECS) comprises two cognate endocannabinoid receptors referred to as CB1R and CB2R. ECS dysregulation is apparent in neurodegenerative/neuro-psychiatric disorders including but not limited to schizophrenia, major depressive disorder and potentially bipolar disorder. The aim of this paper is to review mechanisms whereby both receptors may interact with neuro-immune and neuro-oxidative pathways, which play a pathophysiological role in these disorders. CB1R is located in the presynaptic terminals of GABAergic, glutamatergic, cholinergic, noradrenergic and serotonergic neurons where it regulates the retrograde suppression of neurotransmission. CB1R plays a key role in long-term depression, and, to a lesser extent, long-term potentiation, thereby modulating synaptic transmission and mediating learning and memory. Optimal CB1R activity plays an essential neuroprotective role by providing a defense against the development of glutamate-mediated excitotoxicity, which is achieved, at least in part, by impeding AMPA-mediated increase in intracellular calcium overload and oxidative stress. Moreover, CB1R activity enables optimal neuron-glial communication and the function of the neurovascular unit. CB2R receptors are detected in peripheral immune cells and also in central nervous system regions including the striatum, basal ganglia, frontal cortex, hippocampus, amygdala as well as the ventral tegmental area. CB2R upregulation inhibits the presynaptic release of glutamate in several brain regions. CB2R activation also decreases neuroinflammation partly by mediating the transition from a predominantly neurotoxic "M1" microglial phenotype to a more neuroprotective "M2" phenotype. CB1R and CB2R are thus novel drug targets for the treatment of neuro-immune and neuro-oxidative disorders including schizophrenia and affective disorders.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Ken Walder
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, Centre for Molecular and Medical Research, School of Medicine, Geelong, Australia
| | - Stefan Kloiber
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 33 Ursula Franklin Street, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Paul Amminger
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Michael Berk
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, Centre for Youth Mental Health, Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, The University of Melbourne, Melbourne, Australia
| | - Chiara C Bortolasci
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Michael Maes
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
| | | | - Andre F Carvalho
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia.
| |
Collapse
|
30
|
Kong D, Xie B, Li Y, Xu Y. PEA prevented early BBB disruption after cerebral ischaemic/reperfusion (I/R) injury through regulation of ROCK/MLC signaling. Biochem Biophys Res Commun 2021; 566:164-169. [PMID: 34126347 DOI: 10.1016/j.bbrc.2021.06.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 06/03/2021] [Indexed: 12/29/2022]
Abstract
Palmitoylethanolamide (PEA) offers a strong protection against BBB disruption and neurological deficits after cerebral ischaemic/reperfusion (I/R) injury. To date, these BBB protective effects of PEA are mainly attributed to PPARα-mediated actions. However, whether PEA protects against BBB disruption through direct regulation of cytoskeletal microfilaments remains unknown. Here, we identified PEA as a Rho-associated protein kinase (ROCK2) inhibitor (IC50 = 38.4 ± 4.8 μM). In vitro data suggested that PEA reduced the activation of ROCK/MLC signaling and stress fiber formation within microvascular endothelial cells (ECs) after oxygen-glucose deprivation (OGD), and consequently attenuated early (0-4 h) EC barrier disruption. These actions of PEA could not be blocked by the PPARα antagonist GW6471. In summary, the present study described a previously unexplored role of PEA as a ROCK2 inhibitor, and propose a PPARα-independent mechanism for pharmacological effects of PEA.
Collapse
Affiliation(s)
- Dequan Kong
- Emergency Medicine Department, Xiang'an Hospital of Xiamen University, China
| | - Baoying Xie
- School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yuhang Li
- Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen Medical College, Xiamen, Fujian, 361002, China; CAS Key Laboratory of Design and Assembly of Functional Nanostructures, And Fujian Provincial Key Laboratory of Nanomaterials, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, China; Xiamen Institute of Rare-earth Materials, Haixi Institutes, Chinese Academy of Sciences, Fujian, 361005, China
| | - Yaping Xu
- Institute of Respiratory Diseases Xiamen Medical College, Xiamen, Fujian, 361002, China; Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen Medical College, Xiamen, Fujian, 361002, China.
| |
Collapse
|
31
|
Kasatkina LA, Rittchen S, Sturm EM. Neuroprotective and Immunomodulatory Action of the Endocannabinoid System under Neuroinflammation. Int J Mol Sci 2021; 22:ijms22115431. [PMID: 34063947 PMCID: PMC8196612 DOI: 10.3390/ijms22115431] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 12/17/2022] Open
Abstract
Endocannabinoids (eCBs) are lipid-based retrograde messengers with a relatively short half-life that are produced endogenously and, upon binding to the primary cannabinoid receptors CB1/2, mediate multiple mechanisms of intercellular communication within the body. Endocannabinoid signaling is implicated in brain development, memory formation, learning, mood, anxiety, depression, feeding behavior, analgesia, and drug addiction. It is now recognized that the endocannabinoid system mediates not only neuronal communications but also governs the crosstalk between neurons, glia, and immune cells, and thus represents an important player within the neuroimmune interface. Generation of primary endocannabinoids is accompanied by the production of their congeners, the N-acylethanolamines (NAEs), which together with N-acylneurotransmitters, lipoamino acids and primary fatty acid amides comprise expanded endocannabinoid/endovanilloid signaling systems. Most of these compounds do not bind CB1/2, but signal via several other pathways involving the transient receptor potential cation channel subfamily V member 1 (TRPV1), peroxisome proliferator-activated receptor (PPAR)-α and non-cannabinoid G-protein coupled receptors (GPRs) to mediate anti-inflammatory, immunomodulatory and neuroprotective activities. In vivo generation of the cannabinoid compounds is triggered by physiological and pathological stimuli and, specifically in the brain, mediates fine regulation of synaptic strength, neuroprotection, and resolution of neuroinflammation. Here, we review the role of the endocannabinoid system in intrinsic neuroprotective mechanisms and its therapeutic potential for the treatment of neuroinflammation and associated synaptopathy.
Collapse
Affiliation(s)
- Ludmila A. Kasatkina
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria; (L.A.K.); (S.R.)
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Sonja Rittchen
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria; (L.A.K.); (S.R.)
| | - Eva M. Sturm
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria; (L.A.K.); (S.R.)
- Correspondence:
| |
Collapse
|
32
|
Transient Receptor Potential Vanilloid in the Brain Gliovascular Unit: Prospective Targets in Therapy. Pharmaceutics 2021; 13:pharmaceutics13030334. [PMID: 33806707 PMCID: PMC7999963 DOI: 10.3390/pharmaceutics13030334] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/20/2021] [Accepted: 02/22/2021] [Indexed: 12/25/2022] Open
Abstract
The gliovascular unit (GVU) is composed of the brain microvascular endothelial cells forming blood–brain barrier and the neighboring surrounding “mural” cells (e.g., pericytes) and astrocytes. Modulation of the GVU/BBB features could be observed in a variety of vascular, immunologic, neuro-psychiatric diseases, and cancers, which can disrupt the brain homeostasis. Ca2+ dynamics have been regarded as a major factor in determining BBB/GVU properties, and previous studies have demonstrated the role of transient receptor potential vanilloid (TRPV) channels in modulating Ca2+ and BBB/GVU properties. The physiological role of thermosensitive TRPV channels in the BBB/GVU, as well as their possible therapeutic potential as targets in treating brain diseases via preserving the BBB are reviewed. TRPV2 and TRPV4 are the most abundant isoforms in the human BBB, and TRPV2 was evidenced to play a main role in regulating human BBB integrity. Interspecies differences in TRPV2 and TRPV4 BBB expression complicate further preclinical validation. More studies are still needed to better establish the physiopathological TRPV roles such as in astrocytes, vascular smooth muscle cells, and pericytes. The effect of the chronic TRPV modulation should also deserve further studies to evaluate their benefit and innocuity in vivo.
Collapse
|
33
|
Van Hove L, Kim KR, Arrick DM, Mayhan WG. A cannabinoid type 2 (CB2) receptor agonist augments NOS-dependent responses of cerebral arterioles during type 1 diabetes. Microvasc Res 2021; 133:104077. [PMID: 32979391 PMCID: PMC7704564 DOI: 10.1016/j.mvr.2020.104077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/14/2020] [Accepted: 09/19/2020] [Indexed: 11/19/2022]
Abstract
While activation of cannabinoid (CB2) receptors has been shown to be neuroprotective, no studies have examined whether this neuroprotection is directed at cerebral arterioles and no studies have examined whether activation of CB2 receptors can rescue cerebrovascular dysfunction during a chronic disease state such as type 1 diabetes (T1D). Our goal was to test the hypothesis that administration of a CB2 agonist (JWH-133) would improve impaired endothelial (eNOS)- and neuronal (nNOS)-dependent dilation of cerebral arterioles during T1D. In vivo diameter of cerebral arterioles in nondiabetic and T1D rats was measured in response to an eNOS-dependent agonist (adenosine 5'-diphosphate; ADP), an nNOS-dependent agonist (N-methyl-d-aspartate; NMDA), and an NOS-independent agonist (nitroglycerin) before and 1 h following JWH-133 (1 mg/kg IP). Dilation of cerebral arterioles to ADP and NMDA was greater in nondiabetic than in T1D rats. Treatment with JWH-133 increased responses of cerebral arterioles to ADP and NMDA in both nondiabetic and T1D rats. Responses of cerebral arterioles to nitroglycerin were similar between nondiabetic and T1D rats, and JWH-133 did not influence responses to nitroglycerin in either group. The restoration in responses to the agonists by JWH-133 could be inhibited by treatment with a specific inhibitor of CB2 receptors (AM-630; 3 mg/kg IP). Thus, activation of CB2 receptors can potentiate reactivity of cerebral arterioles during physiologic and pathophysiologic states. We speculate that treatment with CB2 receptor agonists may have potential therapeutic benefits for the treatment of cerebral vascular diseases via a mechanism that can increase cerebral blood flow.
Collapse
MESH Headings
- Animals
- Arterioles/drug effects
- Arterioles/enzymology
- Brain/blood supply
- Cannabinoid Receptor Agonists/pharmacology
- Cannabinoids/pharmacology
- Cerebrovascular Disorders/enzymology
- Cerebrovascular Disorders/physiopathology
- Cerebrovascular Disorders/prevention & control
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/enzymology
- Diabetes Mellitus, Type 1/physiopathology
- Male
- Nitric Oxide Synthase Type I/metabolism
- Nitric Oxide Synthase Type III/metabolism
- Rats, Sprague-Dawley
- Receptor, Cannabinoid, CB2/agonists
- Receptor, Cannabinoid, CB2/metabolism
- Signal Transduction
- Vasodilation/drug effects
- Vasodilator Agents/pharmacology
- Rats
Collapse
Affiliation(s)
- Lauren Van Hove
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, Vermillion, SD 57069, United States of America
| | - Kirsten R Kim
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, Vermillion, SD 57069, United States of America
| | - Denise M Arrick
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, Vermillion, SD 57069, United States of America
| | - William G Mayhan
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, Vermillion, SD 57069, United States of America.
| |
Collapse
|
34
|
Gao J, Chen N, Li N, Xu F, Wang W, Lei Y, Shi J, Gong Q. Neuroprotective Effects of Trilobatin, a Novel Naturally Occurring Sirt3 Agonist from Lithocarpus polystachyus Rehd., Mitigate Cerebral Ischemia/Reperfusion Injury: Involvement of TLR4/NF-κB and Nrf2/Keap-1 Signaling. Antioxid Redox Signal 2020; 33:117-143. [PMID: 32212827 DOI: 10.1089/ars.2019.7825] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Aims: Neuroinflammation and oxidative stress are deemed the prime causes of brain injury after cerebral ischemia/reperfusion (I/R). Since the silent mating-type information regulation 2 homologue 3 (Sirt3) pathway plays an imperative role in protecting against neuroinflammation and oxidative stress, it has been verified as a target to treat ischemia stroke. Therefore, we attempted to seek novel Sirt3 agonist and explore its underlying mechanism for stroke treatment both in vivo and in vitro. Results: Trilobatin (TLB) not only dramatically suppressed neuroinflammation and oxidative stress injury after middle cerebral artery occlusion in rats, but also effectively mitigated oxygen and glucose deprivation/reoxygenation injury in primary cultured astrocytes. These beneficial effects, along with the reduced proinflammatory cytokines via suppressing Toll-like receptor 4 (TLR4) signaling pathway, lessened oxidative injury via activating nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathways, in keeping with the findings in vivo. Intriguingly, the TLB-mediated neuroprotection on cerebral I/R injury was modulated by reciprocity between TLR4-mediated neuroinflammatory responses and Nrf2 antioxidant responses as evidenced by molecular docking and silencing TLR4 and Nrf2, respectively. Most importantly, TLB not only directly bonded to Sirt3 but also increased Sirt3 expression and activity, indicating that Sirt3 might be a promising therapeutic target of TLB. Innovation: TLB is a naturally occurring Sirt3 agonist with potent neuroprotective effects via regulation of TLR4/nuclear factor-kappa B and Nrf2/Kelch-like ECH-associated protein 1 (Keap-1) signaling pathways both in vivo and in vitro. Conclusion: Our findings indicate that TLB protects against cerebral I/R-induced neuroinflammation and oxidative injury through the regulation of neuroinflammatory and oxidative responses via TLR4, Nrf2, and Sirt3, suggesting that TLB might be a promising Sirt3 agonist against ischemic stroke.
Collapse
Affiliation(s)
- Jianmei Gao
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Nana Chen
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Na Li
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Fan Xu
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Wei Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Yaying Lei
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Jingshan Shi
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Qihai Gong
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
| |
Collapse
|
35
|
Reddy V, Grogan D, Ahluwalia M, Salles ÉL, Ahluwalia P, Khodadadi H, Alverson K, Nguyen A, Raju SP, Gaur P, Braun M, Vale FL, Costigliola V, Dhandapani K, Baban B, Vaibhav K. Targeting the endocannabinoid system: a predictive, preventive, and personalized medicine-directed approach to the management of brain pathologies. EPMA J 2020; 11:217-250. [PMID: 32549916 PMCID: PMC7272537 DOI: 10.1007/s13167-020-00203-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023]
Abstract
Cannabis-inspired medical products are garnering increasing attention from the scientific community, general public, and health policy makers. A plethora of scientific literature demonstrates intricate engagement of the endocannabinoid system with human immunology, psychology, developmental processes, neuronal plasticity, signal transduction, and metabolic regulation. Despite the therapeutic potential, the adverse psychoactive effects and historical stigma, cannabinoids have limited widespread clinical application. Therefore, it is plausible to weigh carefully the beneficial effects of cannabinoids against the potential adverse impacts for every individual. This is where the concept of "personalized medicine" as a promising approach for disease prediction and prevention may take into the account. The goal of this review is to provide an outline of the endocannabinoid system, including endocannabinoid metabolizing pathways, and will progress to a more in-depth discussion of the therapeutic interventions by endocannabinoids in various neurological disorders.
Collapse
Affiliation(s)
- Vamsi Reddy
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA USA
| | - Dayton Grogan
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA USA
| | - Meenakshi Ahluwalia
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA USA
| | - Évila Lopes Salles
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA USA
| | - Pankaj Ahluwalia
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA USA
| | - Hesam Khodadadi
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA USA
| | - Katelyn Alverson
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA USA
| | - Andy Nguyen
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA USA
| | - Srikrishnan P. Raju
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA USA
- Brown University, Providence, RI USA
| | - Pankaj Gaur
- Georgia Cancer Center, Augusta University, Augusta, GA USA
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, USA
| | - Molly Braun
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, USA
- VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, USA
| | - Fernando L. Vale
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA USA
| | | | - Krishnan Dhandapani
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA USA
| | - Babak Baban
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA USA
| | - Kumar Vaibhav
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA USA
| |
Collapse
|
36
|
Negri S, Faris P, Rosti V, Antognazza MR, Lodola F, Moccia F. Endothelial TRPV1 as an Emerging Molecular Target to Promote Therapeutic Angiogenesis. Cells 2020; 9:cells9061341. [PMID: 32471282 PMCID: PMC7349285 DOI: 10.3390/cells9061341] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/26/2020] [Accepted: 05/26/2020] [Indexed: 02/06/2023] Open
Abstract
Therapeutic angiogenesis represents an emerging strategy to treat ischemic diseases by stimulating blood vessel growth to rescue local blood perfusion. Therefore, injured microvasculature may be repaired by stimulating resident endothelial cells or circulating endothelial colony forming cells (ECFCs) or by autologous cell-based therapy. Endothelial Ca2+ signals represent a crucial player in angiogenesis and vasculogenesis; indeed, several angiogenic stimuli induce neovessel formation through an increase in intracellular Ca2+ concentration. Several members of the Transient Receptor Potential (TRP) channel superfamily are expressed and mediate Ca2+-dependent functions in vascular endothelial cells and in ECFCs, the only known truly endothelial precursor. TRP Vanilloid 1 (TRPV1), a polymodal cation channel, is emerging as an important player in endothelial cell migration, proliferation, and tubulogenesis, through the integration of several chemical stimuli. Herein, we first summarize TRPV1 structure and gating mechanisms. Next, we illustrate the physiological roles of TRPV1 in vascular endothelium, focusing our attention on how endothelial TRPV1 promotes angiogenesis. In particular, we describe a recent strategy to stimulate TRPV1-mediated pro-angiogenic activity in ECFCs, in the presence of a photosensitive conjugated polymer. Taken together, these observations suggest that TRPV1 represents a useful target in the treatment of ischemic diseases.
Collapse
Affiliation(s)
- Sharon Negri
- Laboratory of General Physiology, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (S.N.); (P.F.)
| | - Pawan Faris
- Laboratory of General Physiology, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (S.N.); (P.F.)
| | - Vittorio Rosti
- Center for the Study of Myelofibrosis, Laboratory of Biochemistry, Biotechnology and Advanced Diagnosis, IRCCS Policlinico San Matteo Foundation, 27100 Pavia, Italy;
| | - Maria Rosa Antognazza
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia, via Pascoli 70/3, 20133 Milano, Italy; (M.R.A.); (F.L.)
| | - Francesco Lodola
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia, via Pascoli 70/3, 20133 Milano, Italy; (M.R.A.); (F.L.)
| | - Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (S.N.); (P.F.)
- Correspondence:
| |
Collapse
|
37
|
The Association Between Vascular Inflammation and Depressive Disorder. Causality, Biomarkers and Targeted Treatment. Pharmaceuticals (Basel) 2020; 13:ph13050092. [PMID: 32408603 PMCID: PMC7281196 DOI: 10.3390/ph13050092] [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: 04/22/2020] [Revised: 05/09/2020] [Accepted: 05/10/2020] [Indexed: 12/18/2022] Open
Abstract
Diabetes, obesity, atherosclerosis, and myocardial infarction are frequently co-morbid with major depressive disorder. In the current review, it is argued that vascular inflammation is a factor that is common to all disorders and that an endothelial dysfunction of the blood-brain barrier could be involved in the induction of depression symptoms. Biomarkers for vascular inflammation include a high plasma level of C-reactive protein, soluble cell-adhesion molecules, von Willebrand factor, aldosterone, and proinflammatory cytokines like interleukin-6 or tumor necrosis factor α. A further possible biomarker is flow-mediated dilation of the brachial artery. Treatment of vascular inflammation is expected to prevent or to reduce symptoms of depression. Several tentative treatments for this form of depression can be envisioned: eicosapentaenoic acid (EPA), valproate, Vagus-nerve stimulation, nicotinic α7 agonists, and agonists of the cannabinoid CB2-receptor.
Collapse
|
38
|
Mohammad Y, Fallah AB, Reynolds JNJ, Boyd BJ, Rizwan SB. Steric stabilisers govern the colloidal and chemical stability but not in vitro cellular toxicity of linoleoylethanolamide cubosomes. Colloids Surf B Biointerfaces 2020; 192:111063. [PMID: 32353710 DOI: 10.1016/j.colsurfb.2020.111063] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/08/2020] [Accepted: 04/15/2020] [Indexed: 11/21/2022]
Abstract
Linoleoylethanolamide (LEA) is an endogenous lipid with remarkable neuromodulatory properties. However, its therapeutic potential is limited by rapid clearance in vivo, targetability and solubility. This study aimed to formulate LEA into liquid crystalline nanoparticles (cubosomes) as a strategy to address the aforementioned challenges. The influence of three different steric stabilisers: Tween 80 and Pluronic F68, both of which have the potential to interact with receptors expressed at the blood-brain barrier and Pluronic F127 as a control, on colloidal stability, internal structure, chemical stability and cytotoxicity of the dispersions were investigated. We found that for effective stabilization of LEA dispersions, a higher concentration of Tween 80 was required compared to Pluronics. Freshly prepared dispersions showed mean particle size of <250 nm and low PDIs (<0.2), with an Im3m type cubic structure but with different lattice parameters. Upon storage at ambient temperature for a week, increased mean particle size and PDI, with a significant reduction in the concentration of LEA was observed in Tween 80-stabilised dispersions. Greater than 80% cell viability was observed at concentrations of up to 20 μg/mL LEA in the presence of all three stabilisers. Collectively, our results suggest that the stabiliser type influences colloidal and chemical stability but not cytotoxicity of LEA cubosomes. This study highlights the potential of endogenous bioactive lipids to be utilized as core cubosome forming lipids with the view to improving their solubility, rapid clearance and targetability to enable delivery of these bioactive molecules to the brain.
Collapse
Affiliation(s)
- Younus Mohammad
- University of Otago, 18 Frederick Street, 9054, Dunedin, New Zealand
| | - Anita B Fallah
- University of Otago, 18 Frederick Street, 9054, Dunedin, New Zealand
| | - John N J Reynolds
- University of Otago, 18 Frederick Street, 9054, Dunedin, New Zealand
| | - Ben J Boyd
- University of Otago, 18 Frederick Street, 9054, Dunedin, New Zealand
| | - Shakila B Rizwan
- University of Otago, 18 Frederick Street, 9054, Dunedin, New Zealand.
| |
Collapse
|
39
|
Jing N, Fang B, Li Z, Tian A. Exogenous activation of cannabinoid-2 receptor modulates TLR4/MMP9 expression in a spinal cord ischemia reperfusion rat model. J Neuroinflammation 2020; 17:101. [PMID: 32248810 PMCID: PMC7132899 DOI: 10.1186/s12974-020-01784-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 03/24/2020] [Indexed: 11/14/2022] Open
Abstract
Background Cannabinoid-2 receptor (CB2R) plays an important role in the cascading inflammation following ischemic injury. The toll-like receptors 4 (TLR4)/matrix metalloproteinase 9 (MMP9) signal pathway is involved in blood-brain barrier dysfunction induced by ischemia stroke. The aim of this study is to investigate the roles of exogenous activation of CB2R on attenuating neurological deficit and blood-spinal cord barrier (BSCB) disruption during rat spinal cord ischemia reperfusion (I/R) injury, through modulation of the TLR4/MMP9 axis. Methods Animals were intraperitoneally pretreated with TLR4 inhibitor TAK-242, CB2R agonist JWH-133 with or without CB2R antagonist AM630, or equivalent volume of vehicle 1 h before undergoing 14-min occlusion of descending aorta or sham operation. One, two, three, and 7 days after reperfusion, hindlimb locomotor function was evaluated with Basso, Beattie, and Bresnahan (BBB) Locomotor Scale, BSCB integrity was detected by measurement of Evans blue (EB) extravasation and spinal cord edema. The protein expression levels of CB2R, tight junction protein Zonula occluden-1 (ZO-1), TLR4, MMP9, MyD88, NF-κB p65, and NF-κB p-p65 were determined by western blot. The MMP9 activity was analyzed by gelatin zymography. Double immunofluorescence staining was used to identify the perivascular localization of CB2R, TLR4, MMP9, and reactive astrocytes, as well as the colocalization of CB2R, TLR4, and MMP9 with reactive astrocytes. Results JWH-133 pretreatment attenuated hindlimb motor functional deficit and BSCB leakage, along with preventing downregulation of ZO-1 and upregulation of TLR4/MMP9, similar to the effects of TAK-242 preconditioning. JWH-133 or TAK-242 pretreatment reduced the perivascular expression of TLR4/MMP9 and reactive astrocytes following injury. JWH-133 pretreatment also downregulated MyD88/NF-κB level, MMP9 activity, and the astrocytic TLR4/MMP9 after I/R injury. Conclusions Exogenous activation of CB2R by JWH-133 attenuated neurological deficit and BSCB disruption after spinal cord I/R injury via inhibition of TLR4/MMP9 expression.
Collapse
Affiliation(s)
- Na Jing
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, 155 North Nanjing Street, Shenyang, 110001, Liaoning, People's Republic of China
| | - Bo Fang
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, 155 North Nanjing Street, Shenyang, 110001, Liaoning, People's Republic of China
| | - Zhe Li
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, 155 North Nanjing Street, Shenyang, 110001, Liaoning, People's Republic of China
| | - Ayong Tian
- Department of Anesthesiology, First Affiliated Hospital, China Medical University, 155 North Nanjing Street, Shenyang, 110001, Liaoning, People's Republic of China.
| |
Collapse
|
40
|
ZUBRZYCKI M, STASIOLEK M, ZUBRZYCKA M. Opioid and Endocannabinoid System in Orofacial Pain. Physiol Res 2019; 68:705-715. [DOI: 10.33549/physiolres.934159] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Orofacial pain disorders are frequent in the general population and their pharmacological treatment is difficult and controversial. Therefore, the search for novel, safe and efficient analgesics is an important but still elusive goal for contemporary medicine. In the recent years, the antinociceptive potential of endocannabinoids and opioids has been emphasized. However, concerns for the safety of their use limit their clinical applications. the possibility of modulating the activity of endocannabinoids by regulation of their synthesis and/or degradation offers an innovative approach to the treatment of pain. A rat model of trigeminal pain, utilizing tongue jerks evoked by electrical tooth pulp stimulation during perfusion of the cerebral ventricles with various neurotransmitter solutions can be used in the pharmacological studies of nociception in the orofacial area. The aim of this review is to present the effects of pharmacological activity of opioids and endocannabinoids affecting the transmission of the sensory information from the orofacial area on the example of trigemino-hypoglossal reflex in rats.
Collapse
Affiliation(s)
- M. ZUBRZYCKI
- Department of Cardiovascular and Thoracic Surgery, University of Ulm, Ulm, Germany,
| | - M. STASIOLEK
- Department of Neurology, Medical University of Lodz, Lodz, Poland
| | - M. ZUBRZYCKA
- Department of Cardiovascular Physiology, Interdepartmental Chair of Experimental and Clinical Physiology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
41
|
Karwad MA, Couch DG, Wright KL, Tufarelli C, Larvin M, Lund J, O'Sullivan SE. Endocannabinoids and endocannabinoid-like compounds modulate hypoxia-induced permeability in CaCo-2 cells via CB 1, TRPV1, and PPARα. Biochem Pharmacol 2019; 168:465-472. [PMID: 31325449 DOI: 10.1016/j.bcp.2019.07.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 07/15/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND AND PURPOSE We have previously reported that endocannabinoids modulate permeability in Caco-2 cells under inflammatory conditions and hypothesised in the present study that endocannabinoids could also modulate permeability in ischemia/reperfusion. EXPERIMENTAL APPROACH Caco-2 cells were grown on cell culture inserts to confluence. Trans-epithelial electrical resistance (TEER) was used to measure permeability. To generate hypoxia (0% O2), a GasPak™ EZ anaerobe pouch system was used. Endocannabinoids were applied to the apical or basolateral membrane in the presence or absence of receptor antagonists. KEY RESULTS Complete hypoxia decreased TEER (increased permeability) by ~35% after 4 h (recoverable) and ~50% after 6 h (non-recoverable). When applied either pre- or post-hypoxia, apical application of N-arachidonoyl-dopamine (NADA, via TRPV1), oleamide (OA, via TRPV1) and oleoylethanolamine (OEA, via TRPV1) inhibited the increase in permeability. Apical administration of anandamide (AEA) and 2-arachidonoylglycerol (2-AG) worsened the permeability effect of hypoxia (both via CB1). Basolateral application of NADA (via TRPV1), OA (via CB1 and TRPV1), noladin ether (NE, via PPARα), and palmitoylethanolamine (PEA, via PPARα) restored permeability after 4 h hypoxia, whereas OEA increased permeability (via PPARα). After 6 h hypoxia, where permeability does not recover, only basolateral application PEA sustainably decreased permeability, and NE decreased permeability. CONCLUSIONS AND IMPLICATIONS A variety of endocannabinoids and endocannabinoid-like compounds modulate Caco-2 permeability in hypoxia/reoxygenation, which involves multiple targets, depending on whether the compounds are applied to the basolateral or apical membrane. CB1 antagonism and TRPV1 or PPARα agonism may represent novel therapeutic targets against several intestinal disorders associated with increased permeability.
Collapse
Affiliation(s)
- M A Karwad
- Division of Medical Sciences & Graduate Entry Medicine, School of Medicine, Royal Derby Hospital, University of Nottingham, Nottingham, UK
| | - D G Couch
- Division of Medical Sciences & Graduate Entry Medicine, School of Medicine, Royal Derby Hospital, University of Nottingham, Nottingham, UK
| | - K L Wright
- Division of Biomedical & Life Sciences, Faculty of Health & Medicine, Lancaster University, Lancaster, UK
| | - C Tufarelli
- Leicester Cancer Research Centre, Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - M Larvin
- Graduate Entry Medical School and Health Research Institute, University of Limerick, Limerick, Ireland
| | - J Lund
- Division of Medical Sciences & Graduate Entry Medicine, School of Medicine, Royal Derby Hospital, University of Nottingham, Nottingham, UK
| | - S E O'Sullivan
- Division of Medical Sciences & Graduate Entry Medicine, School of Medicine, Royal Derby Hospital, University of Nottingham, Nottingham, UK.
| |
Collapse
|
42
|
Greer JB, Magnuson JT, Hester K, Giroux M, Pope C, Anderson T, Liu J, Dang V, Denslow ND, Schlenk D. Effects of Chlorpyrifos on Cholinesterase and Serine Lipase Activities and Lipid Metabolism in Brains of Rainbow Trout (Oncorhynchus mykiss). Toxicol Sci 2019; 172:146-154. [PMID: 31359069 PMCID: PMC6813751 DOI: 10.1093/toxsci/kfz167] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/08/2019] [Accepted: 07/09/2019] [Indexed: 12/20/2022] Open
Abstract
Chlorpyrifos is an organophosphorus insecticide that elicits acute toxicity through inhibition of acetylcholinesterase (AChE), leading to acetylcholine accumulation and prolonged stimulation of cholinergic receptors throughout the central and peripheral nervous systems. Previous studies have indicated that neurodevelopment may also be impaired through alternative pathways, including reduction of cAMP catalyzed downstream events. The upstream initiating events that underlie non-cholinergic neurological actions of chlorpyrifos and other organophosphorus compounds remain unclear. To investigate the potential role of disruption of fatty acid signaling as a mechanism of toxicity, lipid metabolism and fatty acid profiles were examined to identify alterations that may play a critical role in upstream signaling in the CNS. Juvenile rainbow trout were treated for 7 days with nominal chlorpyrifos concentrations previously reported to diminish olfactory responses (10, 20, and 40 μg/L). While lethality was noted higher doses, measured chlorpyrifos concentrations of 1.38 μg/L (nominal concentration 10 μg/L) significantly reduced the activity of AChE and two serine lipases, monoacylglycerol lipase and fatty acid amide hydrolase in the brain. Reductions in lysophosphatidylethanolamines (16:0; 18:0, 18:1, and 22:6) derived from the phosphatidylethanolamines and free fatty acids (Palmitic acid16:0; Linolenic acid18:3; Eicosadienoic acid 20:2; Arachidonic acid 20:4; and Docosahexaenoic acid 22:6) were also noted, suggesting that chlorpyrifos inhibited the metabolism of selected phospholipid signaling precursors at sublethal concentrations. These results indicate that in addition to AChE inhibition, environmentally relevant chlorpyrifos exposure alters serine lipase activity and lipid metabolites in the trout brain, which may compromise neuronal signaling and impact neurobehavioral responses in aquatic animals.
Collapse
Affiliation(s)
- J B Greer
- Department of Environmental Sciences, University of California Riverside, 2460A Geology, Riverside, CA, United States
| | - J T Magnuson
- Department of Environmental Sciences, University of California Riverside, 2460A Geology, Riverside, CA, United States
| | - K Hester
- Center for Veterinary Health Sciences and Interdisciplinary Toxicology Program, Oklahoma State University, 264 McElroy Hall, Stillwater, OK, United States
| | - M Giroux
- Department of Environmental Sciences, University of California Riverside, 2460A Geology, Riverside, CA, United States
| | - C Pope
- Center for Veterinary Health Sciences and Interdisciplinary Toxicology Program, Oklahoma State University, 264 McElroy Hall, Stillwater, OK, United States
| | - T Anderson
- Center for Veterinary Health Sciences and Interdisciplinary Toxicology Program, Oklahoma State University, 264 McElroy Hall, Stillwater, OK, United States
| | - J Liu
- Center for Veterinary Health Sciences and Interdisciplinary Toxicology Program, Oklahoma State University, 264 McElroy Hall, Stillwater, OK, United States
| | - V Dang
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida, PO Box 110885, 2187 Mowry Rd., Gainesville, FL, United States
| | - N D Denslow
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida, PO Box 110885, 2187 Mowry Rd., Gainesville, FL, United States
| | - D Schlenk
- Department of Environmental Sciences, University of California Riverside, 2460A Geology, Riverside, CA, United States.,Institute of Environmental Health, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
43
|
Stone NL, England TJ, O'Sullivan SE. A Novel Transwell Blood Brain Barrier Model Using Primary Human Cells. Front Cell Neurosci 2019; 13:230. [PMID: 31244605 PMCID: PMC6563620 DOI: 10.3389/fncel.2019.00230] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/08/2019] [Indexed: 01/28/2023] Open
Abstract
Structural alterations and breakdown of the blood brain barrier (BBB) is often a primary or secondary consequence of disease, resulting in brain oedema and the transport of unwanted substances into the brain. It is critical that effective in vitro models are developed to model the in vivo environment to aid in clinically relevant research, especially regarding drug screening and permeability studies. Our novel model uses only primary human cells and includes four of the key cells of the BBB: astrocytes, pericytes, brain microvascular endothelial cells (HBMEC) and neurons. We show that using a larger membrane pore size (3.0 μM) there is an improved connection between the endothelial cells, astrocytes and pericytes. Compared to a two and three cell model, we show that when neurons are added to HBMECs, astrocytes and pericytes, BBB integrity was more sensitive to oxygen-glucose deprivation evidenced by increased permeability and markers of cell damage. Our data also show that a four cell model responds faster to the barrier tightening effects of glucocorticoid dexamethasone, when compared to a two cell and three cell model. These data highlight the important role that neurons play in response to ischaemia, particularly how they contribute to BBB maintenance and breakdown. We consider that this model is more representative of the interactions at the neurovascular unit than other transwell models and is a useful method to study BBB physiology.
Collapse
Affiliation(s)
- Nicole L Stone
- Division of Medical Sciences and Graduate Entry Medicine, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Timothy J England
- Division of Medical Sciences and Graduate Entry Medicine, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Saoirse E O'Sullivan
- Division of Medical Sciences and Graduate Entry Medicine, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
44
|
Davis MP, Behm B, Mehta Z, Fernandez C. The Potential Benefits of Palmitoylethanolamide in Palliation: A Qualitative Systematic Review. Am J Hosp Palliat Care 2019; 36:1134-1154. [PMID: 31113223 DOI: 10.1177/1049909119850807] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Palmitoylethanolamide (PEA) is a nutraceutical endocannabinoid that was retrospectively discovered in egg yolks. Feeding poor children with known streptococcal infections prevented rheumatic fever. Subsequently, it was found to alter the course of influenza. Unfortunately, there is little known about its pharmacokinetics. Palmitoylethanolamide targets nonclassical cannabinoid receptors rather than CB1 and CB2 receptors. Palmitoylethanolamide will only indirectly activate classical cannabinoid receptors by an entourage effect. There are a significant number of prospective and randomized trials demonstrating the pain-relieving effects of PEA. There is lesser evidence of benefit in patients with nonpain symptoms related to depression, Parkinson disease, strokes, and autism. There are no reported drug-drug interactions and very few reported adverse effects from PEA. Further research is needed to define the palliative benefits to PEA.
Collapse
|
45
|
Yao E, Zhang G, Huang J, Yang X, Peng L, Huang X, Luo X, Ren J, Huang R, Yang L, Zhou Y, Zhuo R, Zhao Y, Jin X. Immunomodulatory effect of oleoylethanolamide in dendritic cells via TRPV1/AMPK activation. J Cell Physiol 2019; 234:18392-18407. [PMID: 30895621 DOI: 10.1002/jcp.28474] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 02/15/2019] [Accepted: 02/19/2019] [Indexed: 12/26/2022]
Abstract
Oleoylethanolamide (OEA) is an endogenous lipid mediator involved in the control of feeding, body weight, and energy metabolism. However, whether OEA modulates maturation of dendritic cells (DCs) has never been addressed. Hence, we evaluated the effect of OEA on DCs maturation in bone marrow-derived DCs (BMDCs) in four aspects: (a) Cell surface markers were determined using flow cytometric analysis; (b) cell mobile ability was testified with the transwell assay; (c) stimulation of T cells proliferation was performed in a coculture system; and (d) cytokine production was measured using polymerase chain reaction (PCR). The result showed that, in mature BMDCs induced by lipopolysaccharides (LPS), the OEA treatment decreased expressions of cell surface markers, reduced cell migration, diminished the proliferation of cocultured T cells, and regulated cytokine production of BMDCs, indicating the modulatory effect of OEA on DCs maturation. Furthermore, to explore the underlying mechanism of the immunomodulatory effect of OEA, we used antagonists of transient receptor potential vanilloid-1 (TRPV1) and AMP-activated protein kinase (AMPK), determined the protein expressions of TRPV1/AMPK and Toll-like receptor 4 (TLR4)/nuclear factor-kappa B (NF-κB) using western blot, and measured the intracellular calcium concentration using calcium imaging. The result illustrated that OEA downregulated TLR4/NF-κB, the classical pathway leading to DCs maturation induced by LPS, through the activation of TRPV1 and AMPK. Collectively, the present study suggests that OEA suppresses DCs maturation through the activation of TRPV1/AMPK. These findings increase our understanding of this endogenous lipid OEA.
Collapse
Affiliation(s)
- Enhui Yao
- Medical College, Xiamen University, Xiamen, China.,Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Guixiang Zhang
- Medical College, Xiamen University, Xiamen, China.,Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jinhua Huang
- Medical College, Xiamen University, Xiamen, China.,Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiazhen Yang
- Medical College, Xiamen University, Xiamen, China.,Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Lu Peng
- Medical College, Xiamen University, Xiamen, China
| | - Xuefeng Huang
- Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Xiaoxin Luo
- Medical College, Xiamen University, Xiamen, China
| | - Jie Ren
- Medical College, Xiamen University, Xiamen, China
| | - Rui Huang
- Medical College, Xiamen University, Xiamen, China
| | - Lichao Yang
- Medical College, Xiamen University, Xiamen, China
| | - Yu Zhou
- Medical College, Xiamen University, Xiamen, China
| | - Rengong Zhuo
- Medical College, Xiamen University, Xiamen, China
| | - Yun Zhao
- Medical College, Xiamen University, Xiamen, China
| | - Xin Jin
- Medical College, Xiamen University, Xiamen, China
| |
Collapse
|
46
|
Dual Roles of Astrocyte-Derived Factors in Regulation of Blood-Brain Barrier Function after Brain Damage. Int J Mol Sci 2019; 20:ijms20030571. [PMID: 30699952 PMCID: PMC6387062 DOI: 10.3390/ijms20030571] [Citation(s) in RCA: 181] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 01/23/2019] [Accepted: 01/27/2019] [Indexed: 12/13/2022] Open
Abstract
The blood-brain barrier (BBB) is a major functional barrier in the central nervous system (CNS), and inhibits the extravasation of intravascular contents and transports various essential nutrients between the blood and the brain. After brain damage by traumatic brain injury, cerebral ischemia and several other CNS disorders, the functions of the BBB are disrupted, resulting in severe secondary damage including brain edema and inflammatory injury. Therefore, BBB protection and recovery are considered novel therapeutic strategies for reducing brain damage. Emerging evidence suggests key roles of astrocyte-derived factors in BBB disruption and recovery after brain damage. The astrocyte-derived vascular permeability factors include vascular endothelial growth factors, matrix metalloproteinases, nitric oxide, glutamate and endothelin-1, which enhance BBB permeability leading to BBB disruption. By contrast, the astrocyte-derived protective factors include angiopoietin-1, sonic hedgehog, glial-derived neurotrophic factor, retinoic acid and insulin-like growth factor-1 and apolipoprotein E which attenuate BBB permeability resulting in recovery of BBB function. In this review, the roles of these astrocyte-derived factors in BBB function are summarized, and their significance as therapeutic targets for BBB protection and recovery after brain damage are discussed.
Collapse
|
47
|
Horid’ko TM, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kyiv, Kosiakova HV, Berdyshev AG, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kyiv;, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kyiv;. Preventive effect of N-stearoylethanolamine on memory disorders, blood and brain biochemical parameters in rats with experimental scopolamine-induced cognitive impairment. UKRAINIAN BIOCHEMICAL JOURNAL 2018. [DOI: 10.15407/ubj90.06.097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
48
|
Krishnatreyya H, Hazarika H, Saha A, Chattopadhyay P. Capsaicin, the primary constituent of pepper sprays and its pharmacological effects on mammalian ocular tissues. Eur J Pharmacol 2018; 819:114-121. [DOI: 10.1016/j.ejphar.2017.11.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 11/20/2017] [Accepted: 11/24/2017] [Indexed: 01/07/2023]
|
49
|
Leonard MZ, Alapafuja SO, Ji L, Shukla VG, Liu Y, Nikas SP, Makriyannis A, Bergman J, Kangas BD. Cannabinoid CB 1 Discrimination: Effects of Endocannabinoids and Catabolic Enzyme Inhibitors. J Pharmacol Exp Ther 2017; 363:314-323. [PMID: 28947487 PMCID: PMC5683067 DOI: 10.1124/jpet.117.244392] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 09/14/2017] [Indexed: 11/22/2022] Open
Abstract
An improved understanding of the endocannabinoid system has provided new avenues of drug discovery and development toward the management of pain and other behavioral maladies. Exogenous cannabinoid type 1 (CB1) receptor agonists such as Δ9-tetrahydrocannabinol are increasingly used for their medicinal actions; however, their utility is constrained by concern regarding abuse-related subjective effects. This has led to growing interest in the clinical benefit of indirectly enhancing the activity of the highly labile endocannabinoids N-arachidonoylethanolamine [AEA (or anandamide)] and/or 2-arachidonoylglycerol (2-AG) via catabolic enzyme inhibition. The present studies were conducted to determine whether such actions can lead to CB1 agonist-like subjective effects, as reflected in CB1-related discriminative stimulus effects in laboratory subjects. Squirrel monkeys (n = 8) that discriminated the CB1 full agonist AM4054 (0.01 mg/kg) from vehicle were used to study, first, the inhibitors of fatty acid amide hydrolase (FAAH) or monoacylglycerol lipase (MGL) alone or in combination [FAAH (URB597, AM4303); MGL (AM4301); FAAH/MGL (JZL195, AM4302)] and, second, the ability of the endocannabinoids AEA and 2-AG to produce CB1 agonist-like effects when administered alone or after enzyme inhibition. Results indicate that CB1-related discriminative stimulus effects were produced by combined, but not selective, inhibition of FAAH and MGL, and that these effects were nonsurmountably antagonized by low doses of rimonabant. Additionally, FAAH or MGL inhibition revealed CB1-like subjective effects produced by AEA but not by 2-AG. Taken together, the present data suggest that therapeutic effects of combined, but not selective, enhancement of AEA or 2-AG activity via enzyme inhibition may be accompanied by CB1 receptor-mediated subjective effects.
Collapse
MESH Headings
- Adamantane/administration & dosage
- Adamantane/adverse effects
- Adamantane/analogs & derivatives
- Adamantane/pharmacology
- Amidohydrolases/antagonists & inhibitors
- Amidohydrolases/metabolism
- Animals
- Arachidonic Acids/administration & dosage
- Arachidonic Acids/agonists
- Arachidonic Acids/antagonists & inhibitors
- Arachidonic Acids/pharmacology
- Behavior, Animal/drug effects
- Cannabinoid Receptor Antagonists/administration & dosage
- Cannabinoid Receptor Antagonists/adverse effects
- Cannabinoid Receptor Antagonists/pharmacology
- Cannabinol/administration & dosage
- Cannabinol/adverse effects
- Cannabinol/analogs & derivatives
- Cannabinol/pharmacology
- Discrimination Learning/drug effects
- Dose-Response Relationship, Drug
- Drug Agonism
- Drug Antagonism
- Drugs, Investigational/administration & dosage
- Drugs, Investigational/adverse effects
- Drugs, Investigational/pharmacology
- Endocannabinoids/administration & dosage
- Endocannabinoids/agonists
- Endocannabinoids/antagonists & inhibitors
- Endocannabinoids/pharmacology
- Enzyme Inhibitors/administration & dosage
- Enzyme Inhibitors/adverse effects
- Enzyme Inhibitors/pharmacology
- Glycerides/administration & dosage
- Glycerides/agonists
- Glycerides/antagonists & inhibitors
- Glycerides/pharmacology
- Injections, Intramuscular
- Injections, Intravenous
- Ligands
- Male
- Monoacylglycerol Lipases/antagonists & inhibitors
- Monoacylglycerol Lipases/metabolism
- Nerve Tissue Proteins/agonists
- Nerve Tissue Proteins/antagonists & inhibitors
- Nerve Tissue Proteins/metabolism
- Polyunsaturated Alkamides
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/metabolism
- Saimiri
Collapse
Affiliation(s)
- Michael Z Leonard
- Harvard Medical School, Department of Psychiatry, Boston, Massachusetts (J.B., B.D.K.); McLean Hospital, Preclinical Pharmacology Laboratory, Belmont, Massachusetts (M.Z.L., J.B., B.D.K.); MakScientific LLC, Burlington, Massachusetts (S.O.A.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., V.G.S., Y.L., S.P.N., A.M.)
| | - Shakiru O Alapafuja
- Harvard Medical School, Department of Psychiatry, Boston, Massachusetts (J.B., B.D.K.); McLean Hospital, Preclinical Pharmacology Laboratory, Belmont, Massachusetts (M.Z.L., J.B., B.D.K.); MakScientific LLC, Burlington, Massachusetts (S.O.A.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., V.G.S., Y.L., S.P.N., A.M.)
| | - Lipin Ji
- Harvard Medical School, Department of Psychiatry, Boston, Massachusetts (J.B., B.D.K.); McLean Hospital, Preclinical Pharmacology Laboratory, Belmont, Massachusetts (M.Z.L., J.B., B.D.K.); MakScientific LLC, Burlington, Massachusetts (S.O.A.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., V.G.S., Y.L., S.P.N., A.M.)
| | - Vidyanand G Shukla
- Harvard Medical School, Department of Psychiatry, Boston, Massachusetts (J.B., B.D.K.); McLean Hospital, Preclinical Pharmacology Laboratory, Belmont, Massachusetts (M.Z.L., J.B., B.D.K.); MakScientific LLC, Burlington, Massachusetts (S.O.A.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., V.G.S., Y.L., S.P.N., A.M.)
| | - Yingpeng Liu
- Harvard Medical School, Department of Psychiatry, Boston, Massachusetts (J.B., B.D.K.); McLean Hospital, Preclinical Pharmacology Laboratory, Belmont, Massachusetts (M.Z.L., J.B., B.D.K.); MakScientific LLC, Burlington, Massachusetts (S.O.A.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., V.G.S., Y.L., S.P.N., A.M.)
| | - Spyros P Nikas
- Harvard Medical School, Department of Psychiatry, Boston, Massachusetts (J.B., B.D.K.); McLean Hospital, Preclinical Pharmacology Laboratory, Belmont, Massachusetts (M.Z.L., J.B., B.D.K.); MakScientific LLC, Burlington, Massachusetts (S.O.A.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., V.G.S., Y.L., S.P.N., A.M.)
| | - Alexandros Makriyannis
- Harvard Medical School, Department of Psychiatry, Boston, Massachusetts (J.B., B.D.K.); McLean Hospital, Preclinical Pharmacology Laboratory, Belmont, Massachusetts (M.Z.L., J.B., B.D.K.); MakScientific LLC, Burlington, Massachusetts (S.O.A.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., V.G.S., Y.L., S.P.N., A.M.)
| | - Jack Bergman
- Harvard Medical School, Department of Psychiatry, Boston, Massachusetts (J.B., B.D.K.); McLean Hospital, Preclinical Pharmacology Laboratory, Belmont, Massachusetts (M.Z.L., J.B., B.D.K.); MakScientific LLC, Burlington, Massachusetts (S.O.A.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., V.G.S., Y.L., S.P.N., A.M.)
| | - Brian D Kangas
- Harvard Medical School, Department of Psychiatry, Boston, Massachusetts (J.B., B.D.K.); McLean Hospital, Preclinical Pharmacology Laboratory, Belmont, Massachusetts (M.Z.L., J.B., B.D.K.); MakScientific LLC, Burlington, Massachusetts (S.O.A.); and Center for Drug Discovery, Northeastern University, Boston, Massachusetts (L.J., V.G.S., Y.L., S.P.N., A.M.)
| |
Collapse
|
50
|
Meng G, Ma X, Li L, Tan Y, Liu X, Liu X, Zhao Y. Predictors of early-onset post-ischemic stroke depression: a cross-sectional study. BMC Neurol 2017; 17:199. [PMID: 29149884 PMCID: PMC5693521 DOI: 10.1186/s12883-017-0980-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 11/13/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Post-stroke depression (PSD) seriously affects the rehabilitation of nerve function and quality of life. However, the pathogenesis of PSD is still not clear. This study aimed to investigate the demographic, clinical, and biochemical factors in patients with PSD. METHODS Patients with an acute ischemic stroke, who met the inclusion criteria at Shanghai Tenth People's Hospital from April 2016 to September 2016, were recruited for this study. The stroke severity was assessed using the National Institutes of Health Stroke Scale (NIHSS), and the mental state was assessed using Mini-Mental State Examination (MMSE), Hamilton Depression Scale (HAMD), and Hamilton Anxiety Scale (HAMA) at 1 week of admission. The patients were divided into PSD and non-PSD groups. The demographic and clinical characteristics, as well as the biochemical factors, were compared between the two groups. A logistic regression analysis was performed to identify the risk factors for depression following stroke. RESULTS A total of 83 patients with acute ischemic stroke were recruited. Of these, 36 (43.4%) developed depression. The multivariate logistic regression analysis indicated that high NIHSS [odds ratio (OR): 1.84, 95% confidence interval (CI): 1.09-3.12, P = 0.023] and high HAMD scores (OR: 2.38, 95% CI: 1.61-3.50, P < 0.001) were independent risk predictors for PSD and so were lower dopamine level (OR: 0.64, 95% CI: 0.45-0.91, P = 0.014), lower 5-hydroxytryptamine level (OR: 0.99, 95% CI: 0.98-1.00, P = 0.046), higher tumor necrosis factor-α level (OR: 1.05, 95% CI: 1.00-1.09, P = 0.044), and lower nerve growth factor level (OR: 0.06, 95% CI: 0.01-0.67, P = 0.022). CONCLUSIONS The identification of higher NIHSS scores, higher HAMD scores, lower dopamine level, lower 5-hydroxytryptamine level, higher tumor necrosis factor-α level, and lower nerve growth factor level might be useful for clinicians in recognizing and treating depression in patients after a stroke.
Collapse
Affiliation(s)
- Guilin Meng
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Xiaoye Ma
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Lei Li
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yan Tan
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Xiaohui Liu
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Xueyuan Liu
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yanxin Zhao
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| |
Collapse
|