1
|
Guo L, Fan W, Li D, Hao Z, Liu P, Liu C, Cui K, Zhang W, Liu X, Zhang Q, Mao J, Xie J. Metabolic pathways, pharmacokinetic, and brain neurochemicals effects of capsaicin: Comprehensively insights from in vivo studies. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156212. [PMID: 39522253 DOI: 10.1016/j.phymed.2024.156212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/13/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Capsaicin (CAP), a prominent component of chili pepper known for its potent agonistic effects on TRPV1, has attracted significant attention for its diverse physiological effects. Nevertheless, there remains a paucity of data concerning its in vivo distribution, metabolism, pharmacodynamic properties, and influence on the metabolic profile of the brain. METHODS Stable isotope tracing, in vitro enzyme incubation, microdialysis coupled with UHPLC-MS/MS techniques were employed to investigate the in vivo metabolic pathways, distribution, and pharmacokinetic properties of CAP, and the potential biases in metabolic pathways was elucidate through molecular docking. Furthermore, the effect of CAP on brain metabolic profiles was assessed using untargeted metabolomics, and spatial visualization analysis was conducted through mass spectrometry imaging. RESULTS CAP was distributed predominantly in the kidneys, with lower content in the liver, heart, lungs, brain, and spleen following peripheral administration, and the absorption half-life in the body was about 20 min. CAP primarily underwent alkyl terminal dehydrogenation, hydroxylation, and macrocyclization metabolic pathways under the action of CYP2C9, CYP2C19 and CYP2D6, resulting in at least four metabolites. Among them, the hydroxylation products were main metabolites and the dehydrogenation product 16,17-dihydrocapsaicin could interact with the key binding sites Leu515 and Thr550 of TRPV1 like CAP. CAP quickly diffused to various brain regions and the metabolic characteristics in the striatum were relatively different from that in the blood. The distribution of CAP in the brain primarily triggered the release of neurotransmitters in areas associated with reward, cognition, and memory. Both acute and chronic exposure to CAP elevated amino acid levels in cortical regions, while producing contrasting effects on nucleotide metabolites. CONCLUSION This study offers an initial in-depth analysis of the distribution patterns, metabolic pathways and pharmacodynamic properties of CAP in the body and brain. These findings established a basis for further studies on CAP's pharmacology properties and its influence on the central nervous system.
Collapse
Affiliation(s)
- Lulu Guo
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100080, PR China; Beijing Life Science Academy, Beijing 102209, PR China
| | - Wu Fan
- Beijing Life Science Academy, Beijing 102209, PR China; Flavour Science Research Center, Food Laboratory of Zhongyuan, Zhengzhou University, Zhengzhou 450001, PR China
| | - Die Li
- Flavour Science Research Center, Food Laboratory of Zhongyuan, Zhengzhou University, Zhengzhou 450001, PR China
| | - Zhilin Hao
- Beijing Life Science Academy, Beijing 102209, PR China
| | - Pingping Liu
- Flavour Science Research Center, Food Laboratory of Zhongyuan, Zhengzhou University, Zhengzhou 450001, PR China
| | - Chang Liu
- Beijing Life Science Academy, Beijing 102209, PR China
| | - Kun Cui
- Beijing Life Science Academy, Beijing 102209, PR China
| | - Wenjuan Zhang
- Beijing Life Science Academy, Beijing 102209, PR China
| | - Xingyu Liu
- Beijing Life Science Academy, Beijing 102209, PR China
| | - Qidong Zhang
- Beijing Life Science Academy, Beijing 102209, PR China; Flavour Science Research Center, Food Laboratory of Zhongyuan, Zhengzhou University, Zhengzhou 450001, PR China
| | - Jian Mao
- Beijing Life Science Academy, Beijing 102209, PR China; Flavour Science Research Center, Food Laboratory of Zhongyuan, Zhengzhou University, Zhengzhou 450001, PR China.
| | - Jianping Xie
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100080, PR China; Beijing Life Science Academy, Beijing 102209, PR China; Flavour Science Research Center, Food Laboratory of Zhongyuan, Zhengzhou University, Zhengzhou 450001, PR China.
| |
Collapse
|
2
|
Fan Y, Wang P, Jiang C, Chen J, Zhao M, Liu J. Tet1-mediated activation of the Ampk signaling by Trpv1 DNA hydroxymethylation exerts neuroprotective effects in a rat model of Parkinson's disease. Funct Integr Genomics 2024; 24:161. [PMID: 39285026 DOI: 10.1007/s10142-024-01446-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/23/2024] [Accepted: 09/05/2024] [Indexed: 09/26/2024]
Abstract
Epigenetic regulation plays a role in Parkinson's disease (PD), and ten-eleven translocation methylcytosine dioxygenase 1 (TET1) catalyzes the first step in DNA demethylation by converting 5-methylcytosine to 5-hydroxymethylcytosine. We investigated whether TET1 binds to the promoter of the transient receptor potential cation channel subfamily V member 1 (TRPV1) and regulates its expression, thereby controlling oxidative stress in PD. TRPV1 was identified as an oxidative stress-associated gene in the GSE20186 dataset including substantia nigra from 14 patients with PD and 14 healthy controls and the Genecards database. Lentiviral vectors were used to manipulate Trpv1 expression in rats, followed by 6-hydroxydopamine hydrochloride (6-OHDA) injection for modeling. Behavioral tests, immunofluorescence, Nissl staining, western blot assays, DHE fluorescent probe, biochemical analysis, and ELISA were conducted to assess oxidative stress and neurotoxicity. Trpv1 expression was significantly reduced in the brain tissues of 6-OHDA-treated Parkinsonian rats. Trpv1 alleviated behavioral dysfunction, oxidative stress, and dopamine neuron loss in rats. TET1 mediated TRPV1 hydroxymethylation to promote its expression, and Trpv1 inhibition reversed the mitigating effect of Tet1 on oxidative stress and behavioral dysfunction in PD. TRPV1 activated the AMPK signaling by promoting AMPK phosphorylation to alleviate neurotoxicity and oxidative stress in SH-SY5Y cells. Tet1-mediated Trpv1 hydroxymethylation modification promotes the Ampk signaling activation, thereby eliciting neuroprotection in 6-OHDA-treated Parkinsonian rats. These findings provide experimental evidence that targeting the TET1/TRPV1 axis may be neuroprotective for PD by acting on the AMPK signaling.
Collapse
Affiliation(s)
- Yu Fan
- Department of Neurology, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, People's Republic of China
| | - Po Wang
- Department of Neurology, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou Workers Hospital, Liuzhou, 545005, Guangxi, People's Republic of China
| | - Changchun Jiang
- Department of Neurology, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, People's Republic of China
| | - Jinyu Chen
- Department of Neurology, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, People's Republic of China
| | - Meili Zhao
- Department of Neurology, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, People's Republic of China
| | - Jiahui Liu
- Department of Neurology, Baotou Central Hospital, Baotou, 014040, Inner Mongolia, People's Republic of China.
| |
Collapse
|
3
|
Chen CC, Ke CH, Wu CH, Lee HF, Chao Y, Tsai MC, Shyue SK, Chen SF. Transient receptor potential vanilloid 1 inhibition reduces brain damage by suppressing neuronal apoptosis after intracerebral hemorrhage. Brain Pathol 2024; 34:e13244. [PMID: 38308041 PMCID: PMC11328348 DOI: 10.1111/bpa.13244] [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: 11/17/2023] [Accepted: 01/19/2024] [Indexed: 02/04/2024] Open
Abstract
Intracerebral hemorrhage (ICH) induces a complex sequence of apoptotic cascades and inflammatory responses, leading to neurological impairment. Transient receptor potential vanilloid 1 (TRPV1), a nonselective cation channel with high calcium permeability, has been implicated in neuronal apoptosis and inflammatory responses. This study used a mouse ICH model and neuronal cultures to examine whether TRPV1 activation exacerbates brain damage and neurological deficits by promoting neuronal apoptosis and neuroinflammation. ICH was induced by injecting collagenase in both wild-type (WT) C57BL/6 mice and TRPV1-/- mice. Capsaicin (CAP; a TRPV1 agonist) or capsazepine (a TRPV1 antagonist) was administered by intracerebroventricular injection 30 min before ICH induction in WT mice. The effects of genetic deletion or pharmacological inhibition of TRPV1 using CAP or capsazepine on motor deficits, histological damage, apoptotic responses, blood-brain barrier (BBB) permeability, and neuroinflammatory reactions were explored. The antiapoptotic mechanisms and calcium influx induced by TRPV1 inactivation were investigated in cultured hemin-stimulated neurons. TRPV1 expression was upregulated in the hemorrhagic brain, and TRPV1 was expressed in neurons, microglia, and astrocytes after ICH. Genetic deletion of TRPV1 significantly attenuated motor deficits and brain atrophy for up to 28 days. Deletion of TRPV1 also reduced brain damage, neurodegeneration, microglial activation, cytokine expression, and cell apoptosis at 1 day post-ICH. Similarly, the administration of CAP ameliorated brain damage, neurodegeneration, brain edema, BBB permeability, and cytokine expression at 1 day post-ICH. In primary neuronal cultures, pharmacological inactivation of TRPV1 by CAP attenuated neuronal vulnerability to hemin-induced injury, suppressed apoptosis, and preserved mitochondrial integrity in vitro. Mechanistically, CAP reduced hemin-stimulated calcium influx and prevented the phosphorylation of CaMKII in cultured neurons, which was associated with reduced activation of P38 and c-Jun NH2-terminal kinase mitogen-activated protein kinase signaling. Our results suggest that TRPV1 inhibition may be a potential therapy for ICH by suppressing mitochondria-related neuronal apoptosis.
Collapse
Affiliation(s)
- Chien-Cheng Chen
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
- Graduate Institute of Gerontology and Health Care Management, Chang Gung University of Science and Technology, Taoyuan, Taiwan, Republic of China
| | - Chia-Hua Ke
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
| | - Chun-Hu Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Hung-Fu Lee
- Department of Neurosurgery, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
- National Taipei University of Nursing and Health Sciences, Taipei, Taiwan, Republic of China
| | - Yuan Chao
- Department of Medical Education, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan, Republic of China
| | - Min-Chien Tsai
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Song-Kun Shyue
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, Republic of China
| | - Szu-Fu Chen
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, Republic of China
| |
Collapse
|
4
|
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
|
5
|
Lian B, Zhang J, Yin X, Wang J, Li L, Ju Q, Wang Y, Jiang Y, Liu X, Chen Y, Tang X, Sun C. SIRT1 improves lactate homeostasis in the brain to alleviate parkinsonism via deacetylation and inhibition of PKM2. Cell Rep Med 2024; 5:101684. [PMID: 39128469 PMCID: PMC11384727 DOI: 10.1016/j.xcrm.2024.101684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 04/15/2024] [Accepted: 07/22/2024] [Indexed: 08/13/2024]
Abstract
Sirtuin 1 (SIRT1) is a histone deacetylase and plays diverse functions in various physiological events, from development to lifespan regulation. Here, in Parkinson's disease (PD) model mice, we demonstrated that SIRT1 ameliorates parkinsonism, while SIRT1 knockdown further aggravates PD phenotypes. Mechanistically, SIRT1 interacts with and deacetylates pyruvate kinase M2 (PKM2) at K135 and K206, thus leading to reduced PKM2 enzyme activity and lactate production, which eventually results in decreased glial activation in the brain. Administration of lactate in the brain recapitulates PD-like phenotypes. Furthermore, increased expression of PKM2 worsens PD symptoms, and, on the contrary, inhibition of PKM2 by shikonin or PKM2-IN-1 alleviates parkinsonism in mice. Collectively, our data indicate that excessive lactate in the brain might be involved in the progression of PD. By improving lactate homeostasis, SIRT1, together with PKM2, are likely drug targets for developing agents for the treatment of neurodegeneration in PD.
Collapse
Affiliation(s)
- Bolin Lian
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China; School of Life Sciences, Nantong University, 9 Seyuan Road, Nantong, Jiangsu 226019, China
| | - Jing Zhang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China
| | - Xiang Yin
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China
| | - Jiayan Wang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China
| | - Li Li
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China
| | - Qianqian Ju
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China
| | - Yuejun Wang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China
| | - Yuhui Jiang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China
| | - Xiaoyu Liu
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China
| | - Yu Chen
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong 226001, China.
| | - Xin Tang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China.
| | - Cheng Sun
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China.
| |
Collapse
|
6
|
Issa S, Fayoud H, Shaimardanova A, Sufianov A, Sufianova G, Solovyeva V, Rizvanov A. Growth Factors and Their Application in the Therapy of Hereditary Neurodegenerative Diseases. Biomedicines 2024; 12:1906. [PMID: 39200370 PMCID: PMC11351319 DOI: 10.3390/biomedicines12081906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/11/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Hereditary neurodegenerative diseases (hNDDs) such as Alzheimer's, Parkinson's, Huntington's disease, and others are primarily characterized by their progressive nature, severely compromising both the cognitive and motor abilities of patients. The underlying genetic component in hNDDs contributes to disease risk, creating a complex genetic landscape. Considering the fact that growth factors play crucial roles in regulating cellular processes, such as proliferation, differentiation, and survival, they could have therapeutic potential for hNDDs, provided appropriate dosing and safe delivery approaches are ensured. This article presents a detailed overview of growth factors, and explores their therapeutic potential in treating hNDDs, emphasizing their roles in neuronal survival, growth, and synaptic plasticity. However, challenges such as proper dosing, delivery methods, and patient variability can hinder their clinical application.
Collapse
Affiliation(s)
- Shaza Issa
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia; (S.I.); (H.F.)
| | - Haidar Fayoud
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia; (S.I.); (H.F.)
| | - Alisa Shaimardanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.S.); (V.S.)
| | - Albert Sufianov
- Department of Neurosurgery, Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia;
- The Research and Educational Institute of Neurosurgery, Peoples’ Friendship University of Russia (RUDN), 117198 Moscow, Russia
| | - Galina Sufianova
- Department of Pharmacology, Tyumen State Medical University, 625023 Tyumen, Russia;
| | - Valeriya Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.S.); (V.S.)
| | - Albert Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (A.S.); (V.S.)
- Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, 420111 Kazan, Russia
| |
Collapse
|
7
|
Urmeneta-Ortíz MF, Tejeda-Martínez AR, González-Reynoso O, Flores-Soto ME. Potential Neuroprotective Effect of the Endocannabinoid System on Parkinson's Disease. PARKINSON'S DISEASE 2024; 2024:5519396. [PMID: 39104613 PMCID: PMC11300097 DOI: 10.1155/2024/5519396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/21/2024] [Accepted: 07/01/2024] [Indexed: 08/07/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by alterations in motor capacity resulting from a decrease in the neurotransmitter dopamine due to the selective death of dopaminergic neurons of the nigrostriatal pathway. Unfortunately, conventional pharmacological treatments fail to halt disease progression; therefore, new therapeutic strategies are needed, and currently, some are being investigated. The endocannabinoid system (ECS), highly expressed in the basal ganglia (BG) circuit, undergoes alterations in response to dopaminergic depletion, potentially contributing to motor symptoms and the etiopathogenesis of PD. Substantial evidence supports the neuroprotective role of the ECS through various mechanisms, including anti-inflammatory, antioxidative, and antiapoptotic effects. Therefore, the ECS emerges as a promising target for PD treatment. This review provides a comprehensive summary of current clinical and preclinical evidence concerning ECS alterations in PD, along with potential pharmacological targets that may exert the protection of dopaminergic neurons.
Collapse
Affiliation(s)
- María Fernanda Urmeneta-Ortíz
- Chemical Engineering Department, University Center for Exact and Engineering SciencesUniversity of Guadalajara, Blvd. M. García Barragán # 1451, Guadalajara C.P. 44430, Jalisco, Mexico
- Cellular and Molecular Neurobiology LaboratoryNeurosciences DivisionWestern Biomedical Research Center (CIBO)Mexican Social Security Institute, Sierra Mojada #800, Independencia Oriente, Guadalajara 44340, Jalisco, Mexico
| | - Aldo Rafael Tejeda-Martínez
- Cellular and Molecular Neurobiology LaboratoryNeurosciences DivisionWestern Biomedical Research Center (CIBO)Mexican Social Security Institute, Sierra Mojada #800, Independencia Oriente, Guadalajara 44340, Jalisco, Mexico
| | - Orfil González-Reynoso
- Chemical Engineering Department, University Center for Exact and Engineering SciencesUniversity of Guadalajara, Blvd. M. García Barragán # 1451, Guadalajara C.P. 44430, Jalisco, Mexico
| | - Mario Eduardo Flores-Soto
- Cellular and Molecular Neurobiology LaboratoryNeurosciences DivisionWestern Biomedical Research Center (CIBO)Mexican Social Security Institute, Sierra Mojada #800, Independencia Oriente, Guadalajara 44340, Jalisco, Mexico
| |
Collapse
|
8
|
Hu B, Wang H, Liang H, Ma N, Wu D, Zhao R, Lv H, Xiao Z. Multiple effects of spicy flavors on neurological diseases through the intervention of TRPV1: a critical review. Crit Rev Food Sci Nutr 2024:1-20. [PMID: 39041177 DOI: 10.1080/10408398.2024.2381689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
The spicy properties of foods are contributed by various spicy flavor substances (SFs) such as capsaicin, piperine, and allicin. Beyond their distinctive sensory characteristics, SFs also influence health conditions and numerous studies have associated spicy flavors with disease treatment. In this review, we enumerate different types of SFs and describe their role in food processing, with a specific emphasis on critically examining their influence on human wellness. Particularly, detailed insights into the mechanisms through which SFs enhance physiological balance and alleviate neurological diseases are provided, and a systematic analysis of the significance of transient receptor potential vanilloid type-1 (TRPV1) in regulating metabolism and nervous system homeostasis is presented. Moreover, enhancing the accessibility and utilization of SFs can potentially amplify the physiological effects. This review aims to provide compelling evidence for the integration of food flavor and human health.
Collapse
Affiliation(s)
- Boyong Hu
- Department of Food Science & Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Heng Wang
- Department of Food Science & Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Hong Liang
- Department of Food Science & Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Ning Ma
- Department of Food Science & Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Diyi Wu
- Department of Food Science & Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Ruotong Zhao
- Department of Food Science & Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Haoming Lv
- Department of Food Science & Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Zuobing Xiao
- Department of Food Science & Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
9
|
Yu S, Chen X, Yang T, Cheng J, Liu E, Jiang L, Song M, Shu H, Ma Y. Revealing the mechanisms of blood-brain barrier in chronic neurodegenerative disease: an opportunity for therapeutic intervention. Rev Neurosci 2024; 0:revneuro-2024-0040. [PMID: 38967133 DOI: 10.1515/revneuro-2024-0040] [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: 03/16/2024] [Accepted: 05/30/2024] [Indexed: 07/06/2024]
Abstract
The brain microenvironment is tightly regulated, and the blood-brain barrier (BBB) plays a pivotal role in maintaining the homeostasis of the central nervous system. It effectively safeguards brain tissue from harmful substances in peripheral blood. However, both acute pathological factors and age-related biodegradation have the potential to compromise the integrity of the BBB and are associated with chronic neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD), as well as Epilepsy (EP). This association arises due to infiltration of peripheral foreign bodies including microorganisms, immune-inflammatory mediators, and plasma proteins into the central nervous system when the BBB is compromised. Nevertheless, these partial and generalized understandings do not prompt a shift from passive to active treatment approaches. Therefore, it is imperative to acquire a comprehensive and in-depth understanding of the intricate molecular mechanisms underlying vascular disease alterations associated with the onset and progression of chronic neurodegenerative disorders, as well as the subsequent homeostatic changes triggered by BBB impairment. The present article aims to systematically summarize and review recent scientific work with a specific focus on elucidating the fundamental mechanisms underlying BBB damage in AD, PD, and EP as well as their consequential impact on disease progression. These findings not only offer guidance for optimizing the physiological function of the BBB, but also provide valuable insights for developing intervention strategies aimed at early restoration of BBB structural integrity, thereby laying a solid foundation for designing drug delivery strategies centered around the BBB.
Collapse
Affiliation(s)
- Sixun Yu
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan Province, China
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan Province, China
| | - Xin Chen
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan Province, China
| | - Tao Yang
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan Province, China
| | - Jingmin Cheng
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan Province, China
| | - Enyu Liu
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan Province, China
| | - Lingli Jiang
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan Province, China
| | - Min Song
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan Province, China
| | - Haifeng Shu
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan Province, China
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan Province, China
| | - Yuan Ma
- Department of Neurosurgery, Western Theater General Hospital, Chengdu, Sichuan Province, China
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| |
Collapse
|
10
|
Li J, Ng KW, Sung CC, Chung KKK. The role of age-associated alpha-synuclein aggregation in a conditional transgenic mouse model of Parkinson's disease: Implications for Lewy body formation. J Neurochem 2024; 168:1215-1236. [PMID: 38693066 DOI: 10.1111/jnc.16122] [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: 11/08/2023] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 05/03/2024]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder that is affecting an increasing number of older adults. Although PD is mostly sporadic, genetic mutations have been found in cohorts of families with a history of familial PD (FPD). The first such mutation linked to FPD causes a point mutation (A53T) in α-synuclein (α-syn), a major component of Lewy bodies, which are a classical pathological hallmark of PD. These findings suggest that α-syn is an important contributor to the development of PD. In our previous study, we developed an adenoviral mouse model of PD and showed that the expression of wild-type (WT) α-syn or a mutant form with an increased propensity to aggregate, designated as WT-CL1 α-syn, could be used to study how α-syn aggregation contributes to PD. In this study, we established a transgenic mouse model that conditionally expresses WT or WT-CL1 α-syn in dopaminergic (DA) neurons and found that the expression of either WT or WT-CL1 α-syn was associated with an age-dependent degeneration of DA neurons and movement dysfunction. Using this model, we were able to monitor the process of α-syn aggregate formation and found a correlation between age and the number and sizes of α-syn aggregates formed. These results provide a potential mechanism by which age-dependent α-syn aggregation may lead to the formation of Lewy bodies in PD pathogenesis.
Collapse
Affiliation(s)
- Jiahua Li
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Ka Wai Ng
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Chun Chau Sung
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Kenny K K Chung
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| |
Collapse
|
11
|
Hanifa M, Suri M, Singh H, Gagnani R, Jaggi AS, Bali A. Dual Role of TRPV1 Channels in Cerebral Stroke: An Exploration from a Mechanistic and Therapeutic Perspective. Mol Neurobiol 2024:10.1007/s12035-024-04221-5. [PMID: 38760620 DOI: 10.1007/s12035-024-04221-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 05/05/2024] [Indexed: 05/19/2024]
Abstract
Transient receptor potential vanilloid subfamily member 1 (TRPV1) has been strongly implicated in the pathophysiology of cerebral stroke. However, the exact role and mechanism remain elusive. TPRV1 channels are exclusively present in the neurovascular system and involve many neuronal processes. Numerous experimental investigations have demonstrated that TRPV1 channel blockers or the lack of TRPV1 channels may prevent harmful inflammatory responses during ischemia-reperfusion injury, hence conferring neuroprotection. However, TRPV1 agonists such as capsaicin and some other non-specific TRPV1 activators may induce transient/slight degree of TRPV1 channel activation to confer neuroprotection through a variety of mechanisms, including hypothermia induction, improving vascular functions, inducing autophagy, preventing neuronal death, improving memory deficits, and inhibiting inflammation. Another factor in capsaicin-mediated neuroprotection could be the desensitization of TRPV1 channels. Based on the summarized evidence, it may be plausible to suggest that TPRV1 channels have a dual role in ischemia-reperfusion-induced cerebral injury, and thus, both agonists and antagonists may produce neuroprotection depending upon the dose and duration. The current review summarizes the dual function of TRPV1 in ischemia-reperfusion-induced cerebral injury models, explains its mechanism, and predicts the future.
Collapse
Affiliation(s)
- Mohd Hanifa
- Department of Pharmacology, Central University of Punjab, Bathinda, 151401, India
| | - Manisha Suri
- Department of Pharmacology, Central University of Punjab, Bathinda, 151401, India
| | - Harshita Singh
- Department of Pharmacology, Central University of Punjab, Bathinda, 151401, India
| | - Riya Gagnani
- Department of Pharmacology, Central University of Punjab, Bathinda, 151401, India
| | | | - Anjana Bali
- Department of Pharmacology, Central University of Punjab, Bathinda, 151401, India.
| |
Collapse
|
12
|
Huang Q, Wang Y, Chen S, Liang F. Glycometabolic Reprogramming of Microglia in Neurodegenerative Diseases: Insights from Neuroinflammation. Aging Dis 2024; 15:1155-1175. [PMID: 37611905 PMCID: PMC11081147 DOI: 10.14336/ad.2023.0807] [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: 06/06/2023] [Accepted: 08/07/2023] [Indexed: 08/25/2023] Open
Abstract
Neurodegenerative diseases (ND) are conditions defined by progressive deterioration of the structure and function of the nervous system. Some major examples include Alzheimer's disease (AD), Parkinson's disease (PD), and Amyotrophic lateral sclerosis (ALS). These diseases lead to various dysfunctions, like impaired cognition, memory, and movement. Chronic neuroinflammation may underlie numerous neurodegenerative disorders. Microglia, an important immunocell in the brain, plays a vital role in defending against neuroinflammation. When exposed to different stimuli, microglia are activated and assume different phenotypes, participating in immune regulation of the nervous system and maintaining tissue homeostasis. The immunological activity of activated microglia is affected by glucose metabolic alterations. However, in the context of chronic neuroinflammation, specific alterations of microglial glucose metabolism and their mechanisms of action remain unclear. Thus, in this paper, we review the glycometabolic reprogramming of microglia in ND. The key molecular targets and main metabolic pathways are the focus of this research. Additionally, this study explores the mechanisms underlying microglial glucose metabolism reprogramming in ND and offers an analysis of the most recent therapeutic advancements. The ultimate aim is to provide insights into the development of potential treatments for ND.
Collapse
Affiliation(s)
- Qi Huang
- Department of Rehabilitation, The Central Hospital of Wuhan, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China.
| | - Yanfu Wang
- Department of Rehabilitation, The Central Hospital of Wuhan, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China.
| | - Shanshan Chen
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Fengxia Liang
- Department of Acupuncture and Moxibustion, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
13
|
Maximiano TKE, Carneiro JA, Fattori V, Verri WA. TRPV1: Receptor structure, activation, modulation and role in neuro-immune interactions and pain. Cell Calcium 2024; 119:102870. [PMID: 38531262 DOI: 10.1016/j.ceca.2024.102870] [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: 11/30/2023] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024]
Abstract
In the 1990s, the identification of a non-selective ion channel, especially responsive to capsaicin, revolutionized the studies of somatosensation and pain that were to follow. The TRPV1 channel is expressed mainly in neuronal cells, more specifically, in sensory neurons responsible for the perception of noxious stimuli. However, its presence has also been detected in other non-neuronal cells, such as immune cells, β- pancreatic cells, muscle cells and adipocytes. Activation of the channel occurs in response to a wide range of stimuli, such as noxious heat, low pH, gasses, toxins, endocannabinoids, lipid-derived endovanilloid, and chemical agents, such as capsaicin and resiniferatoxin. This activation results in an influx of cations through the channel pore, especially calcium. Intracellular calcium triggers different responses in sensory neurons. Dephosphorylation of the TRPV1 channel leads to its desensitization, which disrupts its function, while its phosphorylation increases the channel's sensitization and contributes to the channel's rehabilitation after desensitization. Kinases, phosphoinositides, and calmodulin are the main signaling pathways responsible for the channel's regulation. Thus, in this review we provide an overview of TRPV1 discovery, its tissue expression as well as on the mechanisms by which TRPV1 activation (directly or indirectly) induces pain in different disease models.
Collapse
Affiliation(s)
- Thaila Kawane Euflazio Maximiano
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Jessica Aparecida Carneiro
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Victor Fattori
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital-Harvard Medical School, Karp Research Building, 300 Longwood Ave, 02115, Boston, Massachusetts, United States.
| | - Waldiceu A Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil.
| |
Collapse
|
14
|
Sánchez JC, Alemán A, Henao JF, Olaya JC, Ehrlich BE. NCS-1 protein regulates TRPA1 channel through the PI3K pathway in breast cancer and neuronal cells. J Physiol Biochem 2024; 80:451-463. [PMID: 38564162 PMCID: PMC11074019 DOI: 10.1007/s13105-024-01016-z] [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: 11/29/2023] [Accepted: 03/05/2024] [Indexed: 04/04/2024]
Abstract
The physical and functional interaction between transient receptor potential channel ankyrin 1 (TRPA1) and neuronal calcium sensor 1 (NCS-1) was assessed. NCS-1 is a calcium (Ca2+) sensor found in many tissues, primarily neurons, and TRPA1 is a Ca2+ channel involved not only in thermal and pain sensation but also in conditions such as cancer and chemotherapy-induced peripheral neuropathy, in which NCS-1 is also a regulatory component.We explored the interactions between these two proteins by employing western blot, qRT-PCR, co-immunoprecipitation, Ca2+ transient monitoring with Fura-2 spectrophotometry, and electrophysiology assays in breast cancer cells (MDA-MB-231) with different levels of NCS-1 expression and neuroblastoma cells (SH-SY5Y).Our findings showed that the expression of TRPA1 was directly correlated with NCS-1 levels at both the protein and mRNA levels. Additionally, we found a physical and functional association between these two proteins. Physically, the NCS-1 and TRPA1 co-immunoprecipitate. Functionally, NCS-1 enhanced TRPA1-dependent Ca2+ influx, current density, open probability, and conductance, where the functional effects depended on PI3K. Conclusion: NCS-1 appears to act not only as a Ca2+ sensor but also modulates TRPA1 protein expression and channel function in a direct fashion through the PI3K pathway. These results contribute to understanding how Ca2+ homeostasis is regulated and provides a mechanism underlying conditions where Ca2+ dynamics are compromised, including breast cancer. With a cellular pathway identified, targeted treatments can be developed for breast cancer and neuropathy, among other related diseases.
Collapse
Affiliation(s)
- Julio C Sánchez
- Department of Basic Sciences, Laboratory of Cell Physiology, Faculty of Health Sciences, Universidad Tecnológica de Pereira, AA 97, La Julita, 660003, Pereira, Risaralda, Colombia.
| | - Alexander Alemán
- Department of Basic Sciences, Laboratory of Cell Physiology, Faculty of Health Sciences, Universidad Tecnológica de Pereira, AA 97, La Julita, 660003, Pereira, Risaralda, Colombia
| | - Juan F Henao
- Department of Basic Sciences, Laboratory of Cell Physiology, Faculty of Health Sciences, Universidad Tecnológica de Pereira, AA 97, La Julita, 660003, Pereira, Risaralda, Colombia
| | - Juan C Olaya
- Department of Basic Sciences, Laboratory of Cell Physiology, Faculty of Health Sciences, Universidad Tecnológica de Pereira, AA 97, La Julita, 660003, Pereira, Risaralda, Colombia
| | - Barbara E Ehrlich
- Departments of Pharmacology and Cellular and Molecular Physiology, Yale University, New Haven, CT, 06520, USA
| |
Collapse
|
15
|
Rezzani R, Favero G, Gianò M, Pinto D, Labanca M, van Noorden CJ, Rinaldi F. Transient Receptor Potential Channels in the Healthy and Diseased Blood-Brain Barrier. J Histochem Cytochem 2024; 72:199-231. [PMID: 38590114 PMCID: PMC11020746 DOI: 10.1369/00221554241246032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 03/18/2024] [Indexed: 04/10/2024] Open
Abstract
The large family of transient receptor potential (TRP) channels are integral membrane proteins that function as environmental sensors and act as ion channels after activation by mechanical (touch), physical (heat, pain), and chemical stimuli (pungent compounds such as capsaicin). Most TRP channels are localized in the plasma membrane of cells but some of them are localized in membranes of organelles and function as intracellular Ca2+-ion channels. TRP channels are involved in neurological disorders but their precise role(s) and relevance in these disorders are not clear. Endothelial cells of the blood-brain barrier (BBB) express TRP channels such as TRP vanilloid 1-4 and are involved in thermal detection by regulating BBB permeability. In neurological disorders, TRP channels in the BBB are responsible for edema formation in the brain. Therefore, drug design to modulate locally activity of TRP channels in the BBB is a hot topic. Today, the application of TRP channel antagonists against neurological disorders is still limited.
Collapse
Affiliation(s)
- Rita Rezzani
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Interdipartimental University Center of Research Adaption and Regeneration of Tissues and Organs - ARTO, University of Brescia, Brescia, Italy
- Italian Society for the Study of Orofacial Pain (Società Italiana Studio Dolore Orofacciale - SISDO), Brescia, Italy
| | - Gaia Favero
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Interdipartimental University Center of Research Adaption and Regeneration of Tissues and Organs - ARTO, University of Brescia, Brescia, Italy
| | - Marzia Gianò
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Daniela Pinto
- Human Microbiome Advanced Project Institute, Milan, Italy
| | - Mauro Labanca
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Italian Society for the Study of Orofacial Pain (Società Italiana Studio Dolore Orofacciale - SISDO), Brescia, Italy
| | - Cornelis J.F. van Noorden
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Fabio Rinaldi
- Human Microbiome Advanced Project Institute, Milan, Italy
| |
Collapse
|
16
|
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
|
17
|
Zhang K, Qin Z, Chen J, Guo G, Jiang X, Wang F, Zhuang J, Zhang Z. TRPV1 modulated NLRP3 inflammasome activation via calcium in experimental subarachnoid hemorrhage. Aging (Albany NY) 2024; 16:1096-1110. [PMID: 38180747 PMCID: PMC10866436 DOI: 10.18632/aging.205379] [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: 07/25/2023] [Accepted: 11/15/2023] [Indexed: 01/06/2024]
Abstract
Neuroinflammation plays a key role in early brain injury (EBI) of subarachnoid hemorrhage (SAH), and NLRP3 inflammasome plays an important role in the development of neuroinflammation after SAH, but the mechanism of NLRP3 inflammasome activation after SAH is still unclear. TRPV1 is a non-selective calcium channel that is involved in the pathology of neuroinflammation, but its role in SAH has not been revealed. Our study showed that TRPV1 was significantly upregulated after SAH and was predominantly expressed in microglia/macrophages. Antagonism of TRPV1 was effective in ameliorating neurological impairment, brain edema, neuronal damage, and reducing the inflammatory response (evidenced by reducing the number of CD16/32 positive microglia/macrophages, inhibiting the expression of CD16, CD32, CD86, IL-1b, TNF-a and blocking NLRP3 inflammasome activation). However, this effect can be abolished by NLRP3 inflammasome antagonist MCC950. In vitro experiment confirmed that TRPV1 activated NLRP3 inflammasome by increasing intracellular calcium levels. In conclusion, TRPV1 mediates EBI after SAH via calcium/NLRP3, and TRPV1 is a potential therapeutic target after SAH.
Collapse
Affiliation(s)
- Keke Zhang
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Zhen Qin
- Department of Clinical Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250021, China
| | - Jinyan Chen
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Gengyin Guo
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Xiaokun Jiang
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Feng Wang
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co., Ltd., Nanjing 210023, China
| | - Jianfeng Zhuang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong 250021, China
| | - Zhen Zhang
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| |
Collapse
|
18
|
Song R, Chen H, Zhan R, Han M, Zhao L, Shen X. Vitamin E protects dopaminergic neurons against manganese-induced neurotoxicity through stimulation of CHRM1 and KCNJ4. J Trace Elem Med Biol 2024; 81:127326. [PMID: 37939525 DOI: 10.1016/j.jtemb.2023.127326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND Manganese (Mn) overexposure can induce neurotoxicity and lead to manganism. Vitamin E (Vit E) has neuroprotective effects by acting as an ROS scavenger, preventing mitochondrial dysfunction and neuronal apoptosis. However, the effects of Vit E on Mn-induced nigrostriatal system lesions remains unknown. OBJECTIVES We aim to investigate whether Vit E has protective effects on Mn-induced nigrostriatal system lesions and mRNA expression profiles in the SN of mice. METHODS Sixty 8-week-old C57BL/6 male mice were randomly divided into the Control, MnCl2, MnCl2 +Vit E, and Vit E group. Twenty-four hours after the last injection, the behaviour test was performed. The numbers of dopaminergic neurons in Substantia nigra (SN), the contents of dopamine and its metabolite levels in striatium, and the morphology of mitochondria and nuclei in the dopaminergic neurons in SN were detected by immunofluorescence staining, high-performance liquid chromatography, and transmission electron microscopy. Transcriptome analysis was used to analyze the signaling pathways and RT-PCR was used to verify the mRNA levels. RESULTS Vit E ameliorates behavioral disorders and attenuates the loss of nigral dopaminergic neurons in the Mn-induced mouse model. In addition, Vit E antagonized Mn-induced toxicity by restoring mitochondrial function. The results of transcriptome sequencing and RTPCR show that the protective effect of Vit E was related to the upregulation of CHRM1 and KCNJ4 mRNA in the SN. CONCLUSIONS Vit E has neuroprotective effects on Mn-induced neurodegeneration in the nigrostriatal system. This effect may be related to the upregulation of CHRM1 and KCNJ4 mRNA stimulated by Vit E in the SN.
Collapse
Affiliation(s)
- Ruihan Song
- Department of Epidemiology and Health Statistics, Medical School of Qingdao University, Qingdao, Shandong, China
| | - Huanhuan Chen
- Qingdao Municipal Center for Disease Control and Prevention/Qingdao Institute of Preventive Medicine, Qingdao, Shandong, China
| | - Runqing Zhan
- Qingdao University Affiliated Hiser Hospital, Qingdao, Shandong, China
| | - Miaomiao Han
- Department of Epidemiology and Health Statistics, Medical School of Qingdao University, Qingdao, Shandong, China
| | - Longzhu Zhao
- Department of Epidemiology and Health Statistics, Medical School of Qingdao University, Qingdao, Shandong, China
| | - Xiaoli Shen
- Department of Epidemiology and Health Statistics, Medical School of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
19
|
Wang W, Sun T. Impact of TRPV1 on Pathogenesis and Therapy of Neurodegenerative Diseases. Molecules 2023; 29:181. [PMID: 38202764 PMCID: PMC10779880 DOI: 10.3390/molecules29010181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 01/12/2024] Open
Abstract
Transient receptor potential vanilloid 1 (TRPV1) is a transmembrane and non-selective cation channel protein, which can be activated by various physical and chemical stimuli. Recent studies have shown the strong pathogenetic associations of TRPV1 with neurodegenerative diseases (NDs), in particular Alzheimer's disease (AD), Parkinson's disease (PD) and multiple sclerosis (MS) via regulating neuroinflammation. Therapeutic effects of TRPV1 agonists and antagonists on the treatment of AD and PD in animal models also are emerging. We here summarize the current understanding of TRPV1's effects and its agonists and antagonists as a therapeutic means in neurodegenerative diseases, and highlight future treatment strategies using natural TRPV1 agonists. Developing new targets and applying natural products are becoming a promising direction in the treatment of chronic disorders, especially neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Tao Sun
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen 361021, China;
| |
Collapse
|
20
|
Kolesova YS, Stroylova YY, Maleeva EE, Moysenovich AM, Pozdyshev DV, Muronetz VI, Andreev YA. Modulation of TRPV1 and TRPA1 Channels Function by Sea Anemones' Peptides Enhances the Viability of SH-SY5Y Cell Model of Parkinson's Disease. Int J Mol Sci 2023; 25:368. [PMID: 38203538 PMCID: PMC10779363 DOI: 10.3390/ijms25010368] [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: 11/17/2023] [Revised: 12/20/2023] [Accepted: 12/24/2023] [Indexed: 01/12/2024] Open
Abstract
Cellular dysfunction during Parkinson's disease leads to neuroinflammation in various brain regions, inducing neuronal death and contributing to the progression of the disease. Different ion channels may influence the process of neurodegeneration. The peptides Ms 9a-1 and APHC3 can modulate the function of TRPA1 and TRPV1 channels, and we evaluated their cytoprotective effects in differentiated to dopaminergic neuron-like SH-SY5Y cells. We used the stable neuroblastoma cell lines SH-SY5Y, producing wild-type alpha-synuclein and its mutant A53T, which are prone to accumulation of thioflavin-S-positive aggregates. We analyzed the viability of cells, as well as the mRNA expression levels of TRPA1, TRPV1, ASIC1a channels, alpha-synuclein, and tyrosine hydroxylase after differentiation of these cell lines using RT-PCR. Overexpression of alpha-synuclein showed a neuroprotective effect and was accompanied by a reduction of tyrosine hydroxylase expression. A mutant alpha-synuclein A53T significantly increased the expression of the pro-apoptotic protein BAX and made cells more susceptible to apoptosis. Generally, overexpression of alpha-synuclein could be a model for the early stages of PD, while expression of mutant alpha-synuclein A53T mimics a genetic variant of PD. The peptides Ms 9a-1 and APHC3 significantly reduced the susceptibility to apoptosis of all cell lines but differentially influenced the expression of the genes of interest. Therefore, these modulators of TRPA1 and TRPV1 have the potential for the development of new therapeutic agents for neurodegenerative disease treatment.
Collapse
Affiliation(s)
- Yuliya S. Kolesova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (Y.S.K.); (E.E.M.); (A.M.M.)
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | - Yulia Y. Stroylova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia (V.I.M.)
| | - Ekaterina E. Maleeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (Y.S.K.); (E.E.M.); (A.M.M.)
| | - Anastasia M. Moysenovich
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (Y.S.K.); (E.E.M.); (A.M.M.)
- Department of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Denis V. Pozdyshev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia (V.I.M.)
| | - Vladimir I. Muronetz
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia (V.I.M.)
| | - Yaroslav A. Andreev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (Y.S.K.); (E.E.M.); (A.M.M.)
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| |
Collapse
|
21
|
Akan T, Aydın Y, Korkmaz OT, Ulupınar E, Saydam F. The Effects of Carvacrol on Transient Receptor Potential (TRP) Channels in an Animal Model of Parkinson's Disease. Neurotox Res 2023; 41:660-669. [PMID: 37452911 DOI: 10.1007/s12640-023-00660-5] [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: 03/20/2023] [Revised: 06/23/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023]
Abstract
In this study, we aimed to investigate the effects of carvacrol (CA), a widely used phytochemical having anti-oxidant and neuroprotective effects, on transient receptor potential (TRP) channels in an animal model of Parkinson's disease (PD). A total of 64 adult male Spraque-Dawley rats were divided into four groups: sham-operated, PD animal model (unilateral intrastriatal injections of 6-hydroxydopamine (6-OHDA), 6 µg/µl), PD + vehicle (dimethyl sulfoxide (DMSO)) treatment, and PD + CA treatment (10 mg/kg, every other day, for 14 days). Half of the brain samples of substantia nigra pars compacta (SNpc) and striatum (CPu) were collected for immunohistochemistry and the remaining half were used for molecular analyses. CA treatment significantly increased the density of dopaminergic neurons immunolabeled with tyrosine hydroxylase and transient receptor potential canonical 1 (TRPC1) channel in the SNpc of PD animals. In contrast, the density of astrocytes immunolabeled with glial fibrillary acetic acid and transient receptor potential ankyrin 1 (TRPA1) channel significantly decreased following CA treatment in the CPu of PD animals. RT-PCR and western blot analyses showed that 6-OHDA administration significantly reduced TRPA1 and TPRPC1 mRNA expression and protein levels in both SNpc and CPu. CA treatment significantly upregulated TRPA1 expression in PD group, while TRPC1 levels did not display an alteration. Based on this data it was concluded that CA treatment might protect the number of dopaminergic neurons by reducing the reactive astrogliosis and modulating the expression of TRP channels in both neurons and astrocytes in an animal model of PD.
Collapse
Affiliation(s)
- Tülay Akan
- Department of Physiology, Faculty of Medicine, Afyonkarahisar Health Sciences University, Zafer Sağlık Külliyesi B Blok, Dörtyol Mah, 2078 Sk, No. 3, 03030, Afyonkarahisar, Turkey.
| | - Yasemin Aydın
- Department of Physiology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Orhan Tansel Korkmaz
- Department of Physiology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Emel Ulupınar
- Department of Anatomy, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Faruk Saydam
- Department of Medical Biology, Faculty of Medicine, Recep Tayyip Erdogan University, Rize, Turkey
| |
Collapse
|
22
|
Thomasi B, Valdetaro L, Ricciardi MC, Gonçalves de Carvalho M, Fialho Tavares I, Tavares-Gomes AL. Enteric glia as a player of gut-brain interactions during Parkinson's disease. Front Neurosci 2023; 17:1281710. [PMID: 38027511 PMCID: PMC10644407 DOI: 10.3389/fnins.2023.1281710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
The enteric glia has been shown as a potential component of neuroimmune interactions that signal in the gut-brain axis during Parkinson's disease (PD). Enteric glia are a peripheral glial type found in the enteric nervous system (ENS) that, associated with enteric neurons, command various gastrointestinal (GI) functions. They are a unique cell type, with distinct phenotypes and distribution in the gut layers, which establish relevant neuroimmune modulation and regulate neuronal function. Comprehension of enteric glial roles during prodromal and symptomatic phases of PD should be a priority in neurogastroenterology research, as the reactive enteric glial profile, gastrointestinal dysfunction, and colonic inflammation have been verified during the prodromal phase of PD-a moment that may be interesting for interventions. In this review, we explore the mechanisms that should govern enteric glial signaling through the gut-brain axis to understand pathological events and verify the possible windows and pathways for therapeutic intervention. Enteric glia directly modulate several functional aspects of the intestine, such as motility, visceral sensory signaling, and immune polarization, key GI processes found deregulated in patients with PD. The search for glial biomarkers, the investigation of temporal-spatial events involving glial reactivity/signaling, and the proposal of enteric glia-based therapies are clearly demanded for innovative and intestine-related management of PD.
Collapse
Affiliation(s)
- Beatriz Thomasi
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Luisa Valdetaro
- Department of Molecular Pathobiology, NYU College of Dentistry, New York, NY, United States
| | - Maria Carolina Ricciardi
- Neuroglial Interaction Lab, Neuroscience Program, Universidade Federal Fluminense, Niterói, Brazil
| | | | - Isabela Fialho Tavares
- Neuroglial Interaction Lab, Neurobiology Department, Universidade Federal Fluminense, Niterói, Brazil
| | - Ana Lucia Tavares-Gomes
- Neuroglial Interaction Lab, Neuroscience Program, Universidade Federal Fluminense, Niterói, Brazil
- Neuroglial Interaction Lab, Neurobiology Department, Universidade Federal Fluminense, Niterói, Brazil
| |
Collapse
|
23
|
Abdel-Salam OME, Mózsik G. Capsaicin, The Vanilloid Receptor TRPV1 Agonist in Neuroprotection: Mechanisms Involved and Significance. Neurochem Res 2023; 48:3296-3315. [PMID: 37493882 PMCID: PMC10514110 DOI: 10.1007/s11064-023-03983-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/09/2023] [Accepted: 07/04/2023] [Indexed: 07/27/2023]
Abstract
Hot peppers, also called chilli, chilli pepper, or paprika of the plant genus Capsicum (family Solanaceae), are one of the most used vegetables and spices worldwide. Capsaicin (8-methyl N-vanillyl-6-noneamide) is the main pungent principle of hot green and red peppers. By acting on the capsaicin receptor or transient receptor potential cation channel vanilloid subfamily member 1 (TRPV1), capsaicin selectively stimulates and in high doses defunctionalizes capsaicin-sensitive chemonociceptors with C and Aδ afferent fibers. This channel, which is involved in a wide range of neuronal processes, is expressed in peripheral and central branches of capsaicin-sensitive nociceptive neurons, sensory ganglia, the spinal cord, and different brain regions in neuronal cell bodies, dendrites, astrocytes, and pericytes. Several experimental and clinical studies provided evidence that capsaicin protected against ischaemic or excitotoxic cerebral neuronal injury and may lower the risk of cerebral stroke. By preventing neuronal death, memory impairment and inhibiting the amyloidogenic process, capsaicin may also be beneficial in neurodegenerative disorders such as Parkinson's or Alzheimer's diseases. Capsaicin given in systemic inflammation/sepsis exerted beneficial antioxidant and anti-inflammatory effects while defunctionalization of capsaicin-sensitive vagal afferents has been demonstrated to increase brain oxidative stress. Capsaicin may act in the periphery via the vagal sensory fibers expressing TRPV1 receptors to reduce immune oxidative and inflammatory signalling to the brain. Capsaicin given in small doses has also been reported to inhibit the experimentally-induced epileptic seizures. The aim of this review is to provide a concise account on the most recent findings related to this topic. We attempted to delineate such mechanisms by which capsaicin exerts its neuronal protective effects. We also aimed to provide the reader with the current knowledge on the mechanism of action of capsaicin on sensory receptors.
Collapse
Affiliation(s)
- Omar M E Abdel-Salam
- Department of Toxicology and Narcotics, Medical Research and Clinical Studies Institute, National Research Centre, Cairo, Egypt.
| | - Gyula Mózsik
- First Department of Medicine, Medical and Health Centre, University of Pécs, H-9724, Pecs, Hungary
| |
Collapse
|
24
|
Yaghoobi A, Seyedmirzaei H, Ala M. Genome- and Exome-Wide Association Studies Revealed Candidate Genes Associated with DaTscan Imaging Features. PARKINSON'S DISEASE 2023; 2023:2893662. [PMID: 37664790 PMCID: PMC10468272 DOI: 10.1155/2023/2893662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/02/2023] [Accepted: 08/16/2023] [Indexed: 09/05/2023]
Abstract
Introduction Despite remarkable progress in identifying Parkinson's disease (PD) genetic risk loci, the genetic basis of PD remains largely unknown. With the help of the endophenotype approach and using data from dopamine transporter single-photon emission computerized tomography (DaTscan), we identified potentially involved genes in PD. Method We conducted an imaging genetic study by performing exome-wide association study (EWAS) and genome-wide association study (GWAS) on the specific binding ratio (SBR) of six DaTscan anatomical areas between 489 and 559 subjects of Parkinson's progression markers initiative (PPMI) cohort and 83,623 and 36,845 single-nucleotide polymorphisms (SNPs)/insertion-deletion mutations (INDELs). We also investigated the association of cerebrospinal fluid (CSF) protein concentration of our significant genes with PD progression using PPMI CSF proteome data. Results Among 83,623 SNPs/INDELs in EWAS, one SNP (rs201465075) on 1 q32.1 locus was significantly (P value = 4.03 × 10-7) associated with left caudate DaTscan SBR, and 33 SNPs were suggestive. Among 36,845 SNPs in GWAS, one SNP (rs12450112) on 17 p.12 locus was significantly (P value = 1.34 × 10-6) associated with right anterior putamen DaTscan SBR, and 39 SNPs were suggestive among which 8 SNPs were intergenic. We found that rs201465075 and rs12450112 are most likely related to IGFN1 and MAP2K4 genes. The protein level of MAP2K4 in the CSF was significantly associated with PD progression in the PPMI cohort; however, proteomic data were not available for the IGFN1 gene. Conclusion We have shown that particular variants of IGFN1 and MAP2K4 genes may be associated with PD. Since DaTscan imaging could be positive in other Parkinsonian syndromes, caution should be taken when interpreting our results. Future experimental studies are also needed to verify these findings.
Collapse
Affiliation(s)
- Arash Yaghoobi
- Institute for Research in Fundamental Sciences (IPM), School of Biological Sciences, Tehran, Iran
| | - Homa Seyedmirzaei
- Interdisciplinary Neuroscience Research Program (INRP), Tehran University of Medical Sciences, Tehran, Iran
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Moein Ala
- Experimental Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Zhang M, Ma Y, Ye X, Zhang N, Pan L, Wang B. TRP (transient receptor potential) ion channel family: structures, biological functions and therapeutic interventions for diseases. Signal Transduct Target Ther 2023; 8:261. [PMID: 37402746 DOI: 10.1038/s41392-023-01464-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/26/2023] [Accepted: 04/25/2023] [Indexed: 07/06/2023] Open
Abstract
Transient receptor potential (TRP) channels are sensors for a variety of cellular and environmental signals. Mammals express a total of 28 different TRP channel proteins, which can be divided into seven subfamilies based on amino acid sequence homology: TRPA (Ankyrin), TRPC (Canonical), TRPM (Melastatin), TRPML (Mucolipin), TRPN (NO-mechano-potential, NOMP), TRPP (Polycystin), TRPV (Vanilloid). They are a class of ion channels found in numerous tissues and cell types and are permeable to a wide range of cations such as Ca2+, Mg2+, Na+, K+, and others. TRP channels are responsible for various sensory responses including heat, cold, pain, stress, vision and taste and can be activated by a number of stimuli. Their predominantly location on the cell surface, their interaction with numerous physiological signaling pathways, and the unique crystal structure of TRP channels make TRPs attractive drug targets and implicate them in the treatment of a wide range of diseases. Here, we review the history of TRP channel discovery, summarize the structures and functions of the TRP ion channel family, and highlight the current understanding of the role of TRP channels in the pathogenesis of human disease. Most importantly, we describe TRP channel-related drug discovery, therapeutic interventions for diseases and the limitations of targeting TRP channels in potential clinical applications.
Collapse
Affiliation(s)
- Miao Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- The Center for Microbes, Development and Health; Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yueming Ma
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xianglu Ye
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ning Zhang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Lei Pan
- The Center for Microbes, Development and Health; Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China.
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Bing Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, 201203, China.
| |
Collapse
|
26
|
Erin N, Szallasi A. Carcinogenesis and Metastasis: Focus on TRPV1-Positive Neurons and Immune Cells. Biomolecules 2023; 13:983. [PMID: 37371563 DOI: 10.3390/biom13060983] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/23/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Both sensory neurons and immune cells, albeit at markedly different levels, express the vanilloid (capsaicin) receptor, Transient Receptor Potential, Vanilloid-1 (TRPV1). Activation of TRPV1 channels in sensory afferent nerve fibers induces local effector functions by releasing neuropeptides (most notably, substance P) which, in turn, trigger neurogenic inflammation. There is good evidence that chronic activation or inactivation of this inflammatory pathway can modify tumor growth and metastasis. TRPV1 expression was also demonstrated in a variety of mammalian immune cells, including lymphocytes, dendritic cells, macrophages and neutrophils. Therefore, the effects of TRPV1 agonists and antagonists may vary depending on the prominent cell type(s) activated and/or inhibited. Therefore, a comprehensive understanding of TRPV1 activity on immune cells and nerve endings in distinct locations is necessary to predict the outcome of therapies targeting TRPV1 channels. Here, we review the neuro-immune modulation of cancer growth and metastasis, with focus on the consequences of TRPV1 activation in nerve fibers and immune cells. Lastly, the potential use of TRPV1 modulators in cancer therapy is discussed.
Collapse
Affiliation(s)
- Nuray Erin
- Department of Medical Pharmacology, School of Medicine, Akdeniz University, Antalya 07070, Turkey
- Immuno-Pharmacology and Immuno-Oncology Unit, School of Medicine, Akdeniz University, Antalya 07070, Turkey
| | - Arpad Szallasi
- Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary
| |
Collapse
|
27
|
Sardoiwala MN, Sood A, Biswal L, Roy Choudhury S, Karmakar S. Reconstituted Super Paramagnetic Protein "Magnetotransferrin" for Brain Targeting to Attenuate Parkinsonism. ACS APPLIED MATERIALS & INTERFACES 2023; 15:12708-12718. [PMID: 36857164 DOI: 10.1021/acsami.2c20990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Transferrin is an iron transporting protein consisting of bilobal protein shells (apotransferrin) with dual domains in each lobe, holding an interdomain iron binding cleft. This cleft is useful in synthesizing an iron oxide core inside the transferrin shell. In vitro reconstitution chemistry provides a nano-dimensional synthesis of the mineral core inside the protein shell. The present study demonstrates the synthesis of magnetotransferrin with reconstitution of apotransferrin to form iron oxide nanoparticles within the transferrin. Transmission electron microscopy investigations along with analysis of electronic diffraction patterns and magnetometry studies indicate entrapment of superparamagnetic iron (III) oxide nanoparticles. In vivo/ex vivo imaging of the brain and immunogold staining of brain sections further validate the brain targeting potential of "magnetotransferrin". The in vivo therapeutic potential of magneto transferrin has been demonstrated by induction of TRPV1 magnetic stimuli protein, having an important regulatory role in Parkinsonism management. In an exploration of neuroprotective mechanisms, deacetylation of H3K27 of synuclein has been revealed through the TRPV1-mediated HDAC3 activation in the treatment of Parkinsonism. Thus, this magnetic protein could be a potent candidate for brain targeting, bio-imaging, and therapy of neurological infirmities.
Collapse
Affiliation(s)
- Mohammed Nadim Sardoiwala
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, SAS Nagar 140306, Punjab, India
| | - Ankur Sood
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, SAS Nagar 140306, Punjab, India
| | - Liku Biswal
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, SAS Nagar 140306, Punjab, India
| | - Subhasree Roy Choudhury
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, SAS Nagar 140306, Punjab, India
| | - Surajit Karmakar
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, SAS Nagar 140306, Punjab, India
| |
Collapse
|
28
|
Wang C, Lu J, Sha X, Qiu Y, Chen H, Yu Z. TRPV1 regulates ApoE4-disrupted intracellular lipid homeostasis and decreases synaptic phagocytosis by microglia. Exp Mol Med 2023; 55:347-363. [PMID: 36720919 PMCID: PMC9981624 DOI: 10.1038/s12276-023-00935-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/29/2022] [Accepted: 12/06/2022] [Indexed: 02/02/2023] Open
Abstract
Although the ε4 allele of the apolipoprotein E (ApoE4) gene has been established as a genetic risk factor for many neurodegenerative diseases, including Alzheimer's disease, the mechanism of action remains poorly understood. Transient receptor potential vanilloid 1 (TRPV1) was reported to regulate autophagy to protect against foam cell formation in atherosclerosis. Here, we show that ApoE4 leads to lipid metabolism dysregulation in microglia, resulting in enhanced MHC-II-dependent antigen presentation and T-cell activation. Lipid accumulation and inflammatory reactions were accelerated in microglia isolated from TRPV1flox/flox; Cx3cr1cre-ApoE4 mice. We showed that metabolic boosting by treatment with the TRPV1 agonist capsaicin rescued lipid metabolic impairments in ApoE4 neurons and defects in autophagy caused by disruption of the AKT-mTOR pathway. TRPV1 activation with capsaicin reversed ApoE4-induced microglial immune dysfunction and neuronal autophagy impairment. Capsaicin rescued memory impairment, tau pathology, and neuronal autophagy in ApoE4 mice. Activation of TRPV1 decreased microglial phagocytosis of synapses in ApoE4 mice. TRPV1 gene deficiency exacerbated recognition memory impairment and tau pathology in ApoE4 mice. Our study suggests that TRPV1 regulation of lipid metabolism could be a therapeutic approach to alleviate the consequences of the ApoE4 allele.
Collapse
Affiliation(s)
- Chenfei Wang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jia Lu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xudong Sha
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yu Qiu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hongzhuan Chen
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Zhihua Yu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
29
|
Gerasimova T, Stepanenko E, Novosadova L, Arsenyeva E, Shimchenko D, Tarantul V, Grivennikov I, Nenasheva V, Novosadova E. Glial Cultures Differentiated from iPSCs of Patients with PARK2-Associated Parkinson's Disease Demonstrate a Pro-Inflammatory Shift and Reduced Response to TNFα Stimulation. Int J Mol Sci 2023; 24:ijms24032000. [PMID: 36768317 PMCID: PMC9916517 DOI: 10.3390/ijms24032000] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/13/2023] [Accepted: 01/15/2023] [Indexed: 01/20/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative diseases characterized by progressive loss of midbrain dopaminergic neurons in the substantia nigra. Mutations in the PARK2 gene are a frequent cause of familial forms of PD. Sustained chronic neuroinflammation in the central nervous system makes a significant contribution to neurodegeneration events. In response to inflammatory factors produced by activated microglia, astrocytes change their transcriptional programs and secretion profiles, thus acting as immunocompetent cells. Here, we investigated iPSC-derived glial cell cultures obtained from healthy donors (HD) and from PD patients with PARK2 mutations in resting state and upon stimulation by TNFα. The non-stimulated glia of PD patients demonstrated higher IL1B and IL6 expression levels and increased IL6 protein synthesis, while BDNF and GDNF expression was down-regulated when compared to that of the glial cells of HDs. In the presence of TNFα, all of the glial cultures displayed a multiplied expression of genes encoding inflammatory cytokines: TNFA, IL1B, and IL6, as well as IL6 protein synthesis, although PD glia responded to TNFα stimulation less strongly than HD glia. Our results demonstrated a pro-inflammatory shift, a suppression of the neuroprotective gene program, and some depletion of reactivity to TNFα in PARK2-deficient glia compared to glial cells of HDs.
Collapse
Affiliation(s)
- Tatiana Gerasimova
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
- Laboratory of Molecular Neurogenetics and Innate Immunity, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
- Correspondence:
| | - Ekaterina Stepanenko
- Laboratory of Molecular Neurogenetics and Innate Immunity, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
| | - Lyudmila Novosadova
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
- Laboratory of Molecular Neurogenetics and Innate Immunity, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
| | - Elena Arsenyeva
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
- Laboratory of Molecular Neurogenetics and Innate Immunity, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
| | - Darya Shimchenko
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
- Laboratory of Molecular Neurogenetics and Innate Immunity, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
| | - Vyacheslav Tarantul
- Laboratory of Molecular Neurogenetics and Innate Immunity, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
| | - Igor Grivennikov
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
- Laboratory of Molecular Neurogenetics and Innate Immunity, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
| | - Valentina Nenasheva
- Laboratory of Molecular Neurogenetics and Innate Immunity, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
| | - Ekaterina Novosadova
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
- Laboratory of Molecular Neurogenetics and Innate Immunity, Institute of Molecular Genetics of National Research Centre “Kurchatov Institute”, Moscow 123182, Russia
| |
Collapse
|
30
|
Cho E, Park J, Hwang EM, Kim HW, Park JY. 14-3-3γ haploinsufficiency leads to altered dopamine pathway and Parkinson's disease-like motor incoordination in mice. Mol Brain 2023; 16:2. [PMID: 36604743 PMCID: PMC9817279 DOI: 10.1186/s13041-022-00990-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 12/19/2022] [Indexed: 01/07/2023] Open
Abstract
The 14-3-3 protein family with seven isoforms found in mammals is widely expressed in the brain and plays various roles in cellular processes. Several studies have reported that 14-3-3γ, one of the 14-3-3 protein isoforms, is associated with neurological and psychiatric disorders, but the role of 14-3-3γ in the pathophysiology of brain diseases is unclear. Although studies have been conducted on the relationship between 14-3-3γ protein and Parkinson's disease (PD), a common neurodegenerative disorder with severe motor symptoms such as bradykinesia and rigidity, a direct connection remains to be elucidated. We recently showed that adult heterozygous 14-3-3γ knockout mice are hyperactive and exhibit anxiety-like behavior. In this study, we further characterized the molecular and behavioral changes in aged 14-3-3γ heterozygous mice to investigate the role of 14-3-3γ in the brain. We observed decreased dopamine levels and altered dopamine metabolism in the brains of these mice, including changes in the phosphorylation of proteins implicated in PD pathology. Furthermore, we confirmed that they displayed PD symptom-like behavioral deficits, such as impaired motor coordination and decreased ability to the nest-building activity. These findings suggest an association between 14-3-3γ dysfunction and PD pathophysiology.
Collapse
Affiliation(s)
- Eunsil Cho
- grid.222754.40000 0001 0840 2678Department of Integrated Biomedical and Life Sciences, Korea University, Seoul, 02708 Korea ,grid.222754.40000 0001 0840 2678BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul, 02841 Korea
| | - Jinsil Park
- grid.263333.40000 0001 0727 6358College of Life Sciences, Sejong University, Seoul, 05006 Korea
| | - Eun Mi Hwang
- grid.35541.360000000121053345Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, 02792 Korea
| | - Hyung Wook Kim
- grid.263333.40000 0001 0727 6358College of Life Sciences, Sejong University, Seoul, 05006 Korea
| | - Jae-Yong Park
- grid.222754.40000 0001 0840 2678Department of Integrated Biomedical and Life Sciences, Korea University, Seoul, 02708 Korea ,grid.222754.40000 0001 0840 2678BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul, 02841 Korea ,ASTRION, Seoul, 02842 Korea
| |
Collapse
|
31
|
Cho E, Kim K, Kim H, Cho SR. Reelin protects against pathological α-synuclein accumulation and dopaminergic neurodegeneration after environmental enrichment in Parkinson's disease. Neurobiol Dis 2022; 175:105898. [DOI: 10.1016/j.nbd.2022.105898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/25/2022] [Accepted: 10/12/2022] [Indexed: 11/05/2022] Open
|
32
|
Luo J, Wu H, Li J, Xian W, Li W, Locascio JJ, Pei Z, Liu G. Joint Modeling Study Identifies Blood-Based Transcripts Link to Cognitive Decline in Parkinson's Disease. Mov Disord 2022; 37:2386-2395. [PMID: 36087011 DOI: 10.1002/mds.29213] [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: 02/17/2022] [Revised: 08/05/2022] [Accepted: 08/17/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Cognitive decline in Parkinson's disease (PD) is prevalent, insidious, and burdensome during the progression of the disease. OBJECTIVES We aimed to find transcriptome-wide biomarkers in blood to predict cognitive decline and identify patients at high risk with cognitive impairment in PD. METHODS We carried out joint modeling analysis to characterize transcriptome-wide longitudinal gene expression and its association with the progression of mild cognitive impairment (MCI) in PD patients. The average time-dependent area under the curves (AUCs) were used for evaluating the accuracy of the significant joint models. A cognitive survival score (CogSs) derived from joint model was leveraged to predict the occurrence of MCI. All predicting models were built in a discovery cohort with 272 patients and replicated in an independent cohort with 177 patients. RESULTS We identified five longitudinal varied expression of transcripts that were significantly associated with MCI progression in patients with PD. The most significant transcript IGLC1 joint model accurately predicted the progression of MCI in PD patients in the discovery and replication cohorts (average time-dependent AUCs >0.82). The CogSs derived from the optimal IGLC1 joint model had a high accuracy at early study stage in both cohorts (AUC ≥0.91). CONCLUSIONS Our transcriptome-wide joint modeling analysis uncovered five blood-based transcripts related to cognitive decline in PD. The joint models will serve as a useful resource for clinicians and researchers to screen PD patients with high risk of development of cognitive impairment and pave the path for Parkinson's personalized medicine. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Junfeng Luo
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Hao Wu
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Jinxia Li
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Wenbiao Xian
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weimin Li
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Joseph J Locascio
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Zhong Pei
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | |
Collapse
|
33
|
Signorelli L, Hescham SA, Pralle A, Gregurec D. Magnetic nanomaterials for wireless thermal and mechanical neuromodulation. iScience 2022; 25:105401. [PMID: 36388996 PMCID: PMC9641224 DOI: 10.1016/j.isci.2022.105401] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Magnetic fields are very attractive for non-invasive neuromodulation because they easily penetrate trough the skull and tissue. Cell specific neuromodulation requires the magnetic field energy to be converted by an actuator to a biologically relevant signal. Miniaturized actuators available today range from small, isotropic magnetic nanoparticles to larger, submicron anisotropic magnetic nanomaterials. Depending on the parameters of external magnetic fields and the properties of the nanoactuators, they create either a thermal or a mechanical stimulus. Ferromagnetic nanomaterials generate heat in response to high frequency alternating magnetic fields associated with dissipative losses. Anisotropic nanomaterials with large magnetic moments are capable of exerting forces at stationary or slowly varying magnetic fields. These tools allow exploiting thermosensitive or mechanosensitive neurons in circuit or cell specific tetherless neuromodulation schemes. This review will address assortment of available magnetic nanomaterial-based neuromodulation techniques that rely on application of external magnetic fields.
Collapse
Affiliation(s)
- Lorenzo Signorelli
- Department of Chemistry and Pharmacy, Chair of Aroma and Smell Research, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sarah- Anna Hescham
- Department of Neurosurgery, Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Arnd Pralle
- Department of Physics, University at Buffalo, Buffalo, NY, USA
| | - Danijela Gregurec
- Department of Chemistry and Pharmacy, Chair of Aroma and Smell Research, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
34
|
Synaptic Effects of Palmitoylethanolamide in Neurodegenerative Disorders. Biomolecules 2022; 12:biom12081161. [PMID: 36009055 PMCID: PMC9405819 DOI: 10.3390/biom12081161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/03/2022] [Accepted: 08/06/2022] [Indexed: 02/06/2023] Open
Abstract
Increasing evidence strongly supports the key role of neuroinflammation in the pathophysiology of neurodegenerative diseases, such as Alzheimer’s disease, frontotemporal dementia, and amyotrophic lateral sclerosis. Neuroinflammation may alter synaptic transmission contributing to the progression of neurodegeneration, as largely documented in animal models and in patients’ studies. In the last few years, palmitoylethanolamide (PEA), an endogenous lipid mediator, and its new composite, which is a formulation constituted of PEA and the well-recognized antioxidant flavonoid luteolin (Lut) subjected to an ultra-micronization process (co-ultraPEALut), has been identified as a potential therapeutic agent in different disorders by exerting potential beneficial effects on neurodegeneration and neuroinflammation by modulating synaptic transmission. In this review, we will show the potential therapeutic effects of PEA in animal models and in patients affected by neurodegenerative disorders.
Collapse
|
35
|
Lee WS, Kang JH, Lee JH, Kim YS, Kim JJ, Kim HS, Kim HW, Shin US, Yoon BE. Improved gliotransmission by increasing intracellular Ca 2+ via TRPV1 on multi-walled carbon nanotube platforms. J Nanobiotechnology 2022; 20:367. [PMID: 35953847 PMCID: PMC9367080 DOI: 10.1186/s12951-022-01551-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Background Astrocyte is a key regulator of neuronal activity and excitatory/inhibitory balance via gliotransmission. Recently, gliotransmission has been identified as a novel target for neurological diseases. However, using the properties of nanomaterials to modulate gliotransmission has not been uncovered. Results We prepared non-invasive CNT platforms for cells with different nanotopography and properties such as hydrophilicity and conductivity. Using CNT platforms, we investigated the effect of CNT on astrocyte functions participating in synaptic transmission by releasing gliotransmitters. Astrocytes on CNT platforms showed improved cell adhesion and proliferation with upregulated integrin and GFAP expression. In addition, intracellular GABA and glutamate in astrocytes were augmented on CNT platforms. We also demonstrated that gliotransmitters in brain slices were increased by ex vivo incubation with CNT. Additionally, intracellular resting Ca2+ level, which is important for gliotransmission, was also increased via TRPV1 on CNT platforms. Conclusion CNT can improve astrocyte function including adhesion, proliferation and gliotransmission by increasing resting Ca2+ level. Therefore, our study suggests that CNT would be utilized as a new therapeutic platform for central nervous system diseases by modulating gliotransmission. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01551-1.
Collapse
Affiliation(s)
- Won-Seok Lee
- Department of Molecular Biology, Dankook University, Cheonan, 31116, Republic of Korea.,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea.,Mechanobiology Dental Medicine Research Center, Cheonan, 31116, Republic of Korea
| | - Ji-Hye Kang
- Department of Nanobiomedical Science, BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea.,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
| | - Jung-Hwan Lee
- Department of Nanobiomedical Science, BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea.,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea.,Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea.,Dental Medicine Innovation Centre, UCL Eastman-Korea, Dankook University, Cheonan, 31116, Republic of Korea.,Mechanobiology Dental Medicine Research Center, Cheonan, 31116, Republic of Korea
| | - Yoo Sung Kim
- Department of Molecular Biology, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jongmin Joseph Kim
- Department of Molecular Biology, Dankook University, Cheonan, 31116, Republic of Korea
| | - Han-Sem Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
| | - Hae-Won Kim
- Department of Nanobiomedical Science, BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea.,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea.,Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea.,Dental Medicine Innovation Centre, UCL Eastman-Korea, Dankook University, Cheonan, 31116, Republic of Korea.,Mechanobiology Dental Medicine Research Center, Cheonan, 31116, Republic of Korea
| | - Ueon Sang Shin
- Department of Nanobiomedical Science, BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea. .,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea.
| | - Bo-Eun Yoon
- Department of Molecular Biology, Dankook University, Cheonan, 31116, Republic of Korea. .,Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea. .,Mechanobiology Dental Medicine Research Center, Cheonan, 31116, Republic of Korea.
| |
Collapse
|
36
|
Lu J, Wang C, Cheng X, Wang R, Yan X, He P, Chen H, Yu Z. A breakdown in microglial metabolic reprogramming causes internalization dysfunction of α-synuclein in a mouse model of Parkinson’s disease. J Neuroinflammation 2022; 19:113. [PMID: 35599331 PMCID: PMC9124408 DOI: 10.1186/s12974-022-02484-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 05/15/2022] [Indexed: 12/11/2022] Open
Abstract
Background The α-synuclein released by neurons activates microglia, which then engulfs α-synuclein for degradation via autophagy. Reactive microglia are a major pathological feature of Parkinson’s disease (PD), although the exact role of microglia in the pathogenesis of PD remains unclear. Transient receptor potential vanilloid type 1 (TRPV1) channels are nonselective cation channel protein that have been proposed as neuroprotective targets in neurodegenerative diseases. Methods Using metabolic profiling, microglia energy metabolism was measured including oxidative phosphorylation and aerobic glycolysis. The mRFP-GFP-tagged LC3 reporter was introduced to characterize the role of TRPV1 in microglial autophagy. α-synuclein preformed fibril (PFF) TRPV1flox/flox; Cx3cr1Cre mouse model of sporadic PD were employed to study the capacity of TRPV1 activation to attenuate neurodegeneration process. Results We found that acute exposure to PFF caused microglial activation as a result of metabolic reprogramming from oxidative phosphorylation to aerobic glycolysis via the AKT–mTOR–HIF-1α pathway. Activated microglia eventually reached a state of chronic PFF-tolerance, accompanied by broad defects in energy metabolism. We showed that metabolic boosting by treatment with the TRPV1 agonist capsaicin rescued metabolic impairments in PFF-tolerant microglia and also defects in mitophagy caused by disruption of the AKT–mTOR–HIF-1α pathway. Capsaicin attenuated phosphorylation of α-synuclein in primary neurons by boosting phagocytosis in PFF-tolerant microglia in vitro. Finally, we found that behavioral deficits and loss of dopaminergic neurons were accelerated in the PFF TRPV1flox/flox; Cx3cr1Cre mouse model of sporadic PD. We identified defects in energy metabolism, mitophagy and phagocytosis of PFF in microglia from the substantia nigra pars compacta of TRPV1flox/flox; Cx3cr1Cre mice. Conclusion The findings suggest that modulating microglial metabolism might be a new therapeutic strategy for PD. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02484-0.
Collapse
|
37
|
Meza RC, Ancatén-González C, Chiu CQ, Chávez AE. Transient Receptor Potential Vanilloid 1 Function at Central Synapses in Health and Disease. Front Cell Neurosci 2022; 16:864828. [PMID: 35518644 PMCID: PMC9062234 DOI: 10.3389/fncel.2022.864828] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/29/2022] [Indexed: 12/11/2022] Open
Abstract
The transient receptor potential vanilloid 1 (TRPV1), a ligand-gated nonselective cation channel, is well known for mediating heat and pain sensation in the periphery. Increasing evidence suggests that TRPV1 is also expressed at various central synapses, where it plays a role in different types of activity-dependent synaptic changes. Although its precise localizations remain a matter of debate, TRPV1 has been shown to modulate both neurotransmitter release at presynaptic terminals and synaptic efficacy in postsynaptic compartments. In addition to being required in these forms of synaptic plasticity, TRPV1 can also modify the inducibility of other types of plasticity. Here, we highlight current evidence of the potential roles for TRPV1 in regulating synaptic function in various brain regions, with an emphasis on principal mechanisms underlying TRPV1-mediated synaptic plasticity and metaplasticity. Finally, we discuss the putative contributions of TRPV1 in diverse brain disorders in order to expedite the development of next-generation therapeutic treatments.
Collapse
Affiliation(s)
- Rodrigo C Meza
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Carlos Ancatén-González
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile.,Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
| | - Chiayu Q Chiu
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Andrés E Chávez
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
38
|
Zou L, Xu K, Tian H, Fang Y. Remote neural regulation mediated by nanomaterials. NANOTECHNOLOGY 2022; 33:272002. [PMID: 35442216 DOI: 10.1088/1361-6528/ac62b1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/29/2022] [Indexed: 06/14/2023]
Abstract
Neural regulation techniques play an essential role in the functional dissection of neural circuits and also the treatment of neurological diseases. Recently, a series of nanomaterials, including upconversion nanoparticles (UCNPs), magnetic nanoparticles (MNPs), and silicon nanomaterials (SNMs) that are responsive to remote optical or magnetic stimulation, have been applied as transducers to facilitate localized control of neural activities. In this review, we summarize the latest advances in nanomaterial-mediated neural regulation, especially in a remote and minimally invasive manner. We first give an overview of existing neural stimulation techniques, including electrical stimulation, transcranial magnetic stimulation, chemogenetics, and optogenetics, with an emphasis on their current limitations. Then we focus on recent developments in nanomaterial-mediated neural regulation, including UCNP-mediated fiberless optogenetics, MNP-mediated magnetic neural regulation, and SNM-mediated non-genetic neural regulation. Finally, we discuss the possibilities and challenges for nanomaterial-mediated neural regulation.
Collapse
Affiliation(s)
- Liang Zou
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Ke Xu
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Huihui Tian
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
| | - Ying Fang
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, People's Republic of China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| |
Collapse
|
39
|
Wang S, Wang B, Shang D, Zhang K, Yan X, Zhang X. Ion Channel Dysfunction in Astrocytes in Neurodegenerative Diseases. Front Physiol 2022; 13:814285. [PMID: 35222082 PMCID: PMC8864228 DOI: 10.3389/fphys.2022.814285] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/03/2022] [Indexed: 11/13/2022] Open
Abstract
Astrocytes play an important role in the central nervous system (CNS). Ion channels in these cells not only function in ion transport, and maintain water/ion metabolism homeostasis, but also participate in physiological processes of neurons and glial cells by regulating signaling pathways. Increasing evidence indicates the ion channel proteins of astrocytes, such as aquaporins (AQPs), transient receptor potential (TRP) channels, adenosine triphosphate (ATP)-sensitive potassium (K-ATP) channels, and P2X7 receptors (P2X7R), are strongly associated with oxidative stress, neuroinflammation and characteristic proteins in neurodegenerative disorders, including Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD) and amyotrophic lateral sclerosis (ALS). Since ion channel protein dysfunction is a significant pathological feature of astrocytes in neurodegenerative diseases, we discuss these critical proteins and their signaling pathways in order to understand the underlying molecular mechanisms, which may yield new therapeutic targets for neurodegenerative disorders.
Collapse
Affiliation(s)
- Sijian Wang
- Center of Implant Dentistry, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Biyao Wang
- The VIP Department, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Dehao Shang
- Center of Implant Dentistry, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Kaige Zhang
- Center of Implant Dentistry, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Xu Yan
- The VIP Department, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Xinwen Zhang
- Center of Implant Dentistry, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| |
Collapse
|
40
|
Hong AR, Jang JG, Chung YC, Won SY, Jin BK. Interleukin 13 on Microglia is Neurotoxic in Lipopolysaccharide-injected Striatum in vivo. Exp Neurobiol 2022; 31:42-53. [PMID: 35256543 PMCID: PMC8907255 DOI: 10.5607/en21032] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 02/09/2022] [Accepted: 02/16/2022] [Indexed: 11/19/2022] Open
Abstract
To explore the potential function of interleukin-13 (IL-13), lipopolysaccharide (LPS) or PBS as a control was unilaterally microinjected into striatum of rat brain. Seven days after LPS injection, there was a significant loss of neurons and microglial activation in the striatum, visualized by immunohistochemical staining against neuronal nuclei (NeuN) and the OX-42 (complement receptor type 3, CR3), respectively. In parallel, IL-13 immunoreactivity was increased as early as 3 days and sustained up to 7 days post LPS injection, compared to PBS-injected control and detected exclusively within microglia. Moreover, GFAP immunostaining and blood brain barrier (BBB) permeability evaluation showed the loss of astrocytes and disruption of BBB, respectively. By contrast, treatment with IL-13 neutralizing antibody (IL-13NA) protects NeuN+ neurons against LPS-induced neurotoxicity in vivo . Accompanying neuroprotection, IL-13NA reduced loss of GFAP+ astrocytes and damage of BBB in LPS-injected striatum. Intriguingly, treatment with IL-13NA produced neurotrophic factors (NTFs) on survived astrocytes in LPS-injected rat striatum. Taken together, the present study suggests that LPS induces expression of IL-13 on microglia, which contributes to neurodegeneration via damage on astrocytes and BBB disruption in the striatum in vivo.
Collapse
Affiliation(s)
- Ah Reum Hong
- Department of Neuroscience, Graduate School, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Jae Geun Jang
- Department of Neuroscience, Graduate School, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Young Cheul Chung
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Korea
| | - So-Yoon Won
- Department of Biochemistry & Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Byung Kwan Jin
- Department of Neuroscience, Graduate School, School of Medicine, Kyung Hee University, Seoul 02447, Korea.,Department of Biochemistry & Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
41
|
Morales P, Muller C, Jagerovic N, Reggio PH. Targeting CB2 and TRPV1: Computational Approaches for the Identification of Dual Modulators. Front Mol Biosci 2022; 9:841190. [PMID: 35281260 PMCID: PMC8914543 DOI: 10.3389/fmolb.2022.841190] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/03/2022] [Indexed: 11/13/2022] Open
Abstract
Both metabotropic (CBRs) and ionotropic cannabinoid receptors (ICRs) have implications in a range of neurological disorders. The metabotropic canonical CBRs CB1 and CB2 are highly implicated in these pathological events. However, selective targeting at CB2 versus CB1 offers optimized pharmacology due to the absence of psychoactive outcomes. The ICR transient receptor potential vanilloid type 1 (TRPV1) has also been reported to play a role in CNS disorders. Thus, activation of both targets, CB2 and TRPV1, offers a promising polypharmacological strategy for the treatment of neurological events including analgesia and neuroprotection. This brief research report aims to identify chemotypes with a potential dual CB2/TRPV1 profile. For this purpose, we have rationalized key structural features for activation and performed virtual screening at both targets using curated chemical libraries.
Collapse
Affiliation(s)
- Paula Morales
- Medicinal Chemistry Institute, Spanish National Research Council, Madrid, Spain
| | - Chanté Muller
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC, United States
| | - Nadine Jagerovic
- Medicinal Chemistry Institute, Spanish National Research Council, Madrid, Spain
| | - Patricia H. Reggio
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC, United States
| |
Collapse
|
42
|
Iglesias LP, Aguiar DC, Moreira FA. TRPV1 blockers as potential new treatments for psychiatric disorders. Behav Pharmacol 2022; 33:2-14. [PMID: 33136616 DOI: 10.1097/fbp.0000000000000603] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The transient receptor potential vanilloid-1 channel (TRPV1) is responsible for decoding physical and chemical stimuli. TRPV1 is activated by capsaicin (a compound from chili peppers), heat (above 43°C) and acid environment, playing a major role in pain, inflammation and body temperature. Molecular and histological studies have suggested TRPV1 expression in specific brain regions, where it can be activated primarily by the endocannabinoid anandamide, fostering studies on its potential role in psychiatric disorders. TRPV1 blockers are effective in various animal models predictive of anxiolytic and antipanic activities, in addition to reducing conditioned fear. In models of antidepressant activity, these compounds reduce behavioral despair and promote active stress-coping behavior. TRPV1 blockers also reduce the effects of certain drugs of abuse and revert behavioral changes in animal models of neurodevelopmental disorders. The main limiting factor in developing TRPV1 blockers as therapeutic agents concerns their effects on body temperature, particularly hyperthermia. New compounds, which block specific states of the channel, could represent an alternative. Moreover, compounds blocking both TRPV1 and the anandamide-hydrolyzing enzyme, fatty acid amide hydrolase (FAAH), termed dual TRPV1/FAAH blockers, have been investigated with promising results. Overall, preclinical studies yield favorable results with TRPV1 blockers in animal models of psychiatric disorders.
Collapse
Affiliation(s)
- Lia P Iglesias
- Department of Pharmacology, Graduate School of Neuroscience
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gera, Brazil
| | - Daniele C Aguiar
- Department of Pharmacology, Graduate School of Neuroscience
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gera, Brazil
| | - Fabrício A Moreira
- Department of Pharmacology, Graduate School of Neuroscience
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gera, Brazil
| |
Collapse
|
43
|
Wang C, Huang W, Lu J, Chen H, Yu Z. TRPV1-Mediated Microglial Autophagy Attenuates Alzheimer’s Disease-Associated Pathology and Cognitive Decline. Front Pharmacol 2022; 12:763866. [PMID: 35115924 PMCID: PMC8804218 DOI: 10.3389/fphar.2021.763866] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 12/06/2021] [Indexed: 01/21/2023] Open
Abstract
Autophagy is a major regulator of the ageing process of the central nervous system and neurodegeneration. Autophagy dysfunction has been implicated in the pathogenesis of Alzheimer’s disease (AD). TRPV1 was reported to regulate autophagy to protect against foam cell formation and reduce the release of inflammatory factors in atherosclerosis. In this study, pharmacological activation of TRPV1 with the TRPV1 agonist capsaicin induced autophagy in a TRPV1-dependent manner in both primary microglia and BV2 cells. TRPV1-mediated autophagy regulated glycolysis and oxidative phosphorylation by controlling the expression of genes required for aerobic glycolysis and mitochondrial respiration in primary microglia. TRPV1 agonist capsaicin decreased amyloid and phosphorylated tau pathology and reversed memory deficits by promoting microglia activation, metabolism, and autophagy in 3xTg mice. These results indicate that TRPV1 was a potential therapeutic target for AD, which suggests that capsaicin should be further assessed as a possible treatment for AD.
Collapse
Affiliation(s)
- Chenfei Wang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Huang
- Cardiology Department, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jia Lu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongzhuan Chen
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Zhihua Yu, ; Hongzhuan Chen,
| | - Zhihua Yu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Zhihua Yu, ; Hongzhuan Chen,
| |
Collapse
|
44
|
Jang J, Hong A, Chung Y, Jin B. Interleukin-4 Aggravates LPS-Induced Striatal Neurodegeneration In Vivo via Oxidative Stress and Polarization of Microglia/Macrophages. Int J Mol Sci 2022; 23:ijms23010571. [PMID: 35008995 PMCID: PMC8745503 DOI: 10.3390/ijms23010571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 12/10/2022] Open
Abstract
The present study investigated the effects of interleukin (IL)-4 on striatal neurons in lipopolysaccharide (LPS)-injected rat striatum in vivo. Either LPS or PBS as a control was unilaterally injected into the striatum, and brain tissues were processed for immunohistochemical and Nissl staining or for hydroethidine histochemistry at the indicated time points after LPS injection. Analysis by NeuN and Nissl immunohistochemical staining showed a significant loss of striatal neurons at 1, 3, and 7 days post LPS. In parallel, IL-4 immunoreactivity was upregulated as early as 1 day, reached a peak at 3 days, and was sustained up to 7 days post LPS. Increased levels of IL-4 immunoreactivity were exclusively detected in microglia/macrophages, but not in neurons nor astrocytes. The neutralizing antibody (NA) for IL-4 significantly protects striatal neurons against LPS-induced neurotoxicity in vivo. Accompanying neuroprotection, IL-4NA inhibited activation of microglia/macrophages, production of reactive oxygen species (ROS), ROS-derived oxidative damage and nitrosative stress, and produced polarization of microglia/macrophages shifted from M1 to M2. These results suggest that endogenous IL-4 expressed in LPS-activated microglia/macrophages contributes to striatal neurodegeneration in which oxidative/nitrosative stress and M1/M2 polarization are implicated.
Collapse
Affiliation(s)
- Jaegeun Jang
- Department of Neuroscience, Graduate School of Medicine, Kyung Hee University, Seoul 02447, Korea; (J.J.); (A.H.)
| | - Ahreum Hong
- Department of Neuroscience, Graduate School of Medicine, Kyung Hee University, Seoul 02447, Korea; (J.J.); (A.H.)
| | - Youngcheul Chung
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Korea
- Correspondence: (Y.C.); (B.J.); Tel.: +82-42-610-8383 (Y.C.); +82-2-961-9288 (B.J.); Fax: +82-2-969-4570 (Y.C.); +82-42-610-8148 (B.J.)
| | - Byungkwan Jin
- Department of Neuroscience, Graduate School of Medicine, Kyung Hee University, Seoul 02447, Korea; (J.J.); (A.H.)
- Department of Biochemistry & Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (Y.C.); (B.J.); Tel.: +82-42-610-8383 (Y.C.); +82-2-961-9288 (B.J.); Fax: +82-2-969-4570 (Y.C.); +82-42-610-8148 (B.J.)
| |
Collapse
|
45
|
Long W, Johnson J, Kalyaanamoorthy S, Light P. TRPV1 channels as a newly identified target for vitamin D. Channels (Austin) 2021; 15:360-374. [PMID: 33825665 PMCID: PMC8032246 DOI: 10.1080/19336950.2021.1905248] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/15/2021] [Accepted: 03/15/2021] [Indexed: 11/23/2022] Open
Abstract
Vitamin D is known to elicit many biological effects in diverse tissue types and is thought to act almost exclusively upon its canonical receptor within the nucleus, leading to gene transcriptional changes and the subsequent cellular response. However, not all the observed effects of vitamin D can be attributed to this sole mechanism, and other cellular targets likely exist but remain to be identified. Our recent discovery that vitamin D is a partial agonist of the Transient Receptor Potential Vanilloid family 1 (TRPV1) channel may provide new insights as to how this important vitamin exerts its biological effects either independently or in addition to the nuclear vitamin D receptor. In this review, we discuss the literature surrounding this apparent discrepancy in vitamin D signaling and compare vitamin D with known TRPV1 ligands with respect to their binding to TRPV1. Furthermore, we provide evidence supporting the notion that this novel vitamin D/TRPV1 axis may explain some of the beneficial actions of this vitamin in disease states where TRPV1 expression and vitamin D deficiency are known to overlap. Finally, we discuss whether vitamin D may also act on other members of the TRP family of ion channels.
Collapse
Affiliation(s)
- Wentong Long
- Department of Pharmacology and the Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Janyne Johnson
- Department of Pharmacology and the Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | | | - Peter Light
- Department of Pharmacology and the Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
46
|
Soti M, Ranjbar H, Kohlmeier KA, Shabani M. Parkinson's disease related alterations in cannabinoid transmission. Brain Res Bull 2021; 178:82-96. [PMID: 34808322 DOI: 10.1016/j.brainresbull.2021.11.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/29/2021] [Accepted: 11/15/2021] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is characterized by the progressive loss of dopaminergic (DAergic) neurons of the substantia nigra pars compacta (SNc) by neurodegeneration. Recent findings in animal models of PD propose tonic inhibition of the remaining DA neurons through GABA release from reactive glial cells. Movement dysfunctions could be ameliorated by promotion of activity in dormant DA cells. The endocannabinoid system (ECS) is extensively present in basal ganglia (BG) and is known as an indirect modulator of DAergic neurotransmission, thus drugs designed to target this system have shown promising therapeutic potential in PD patients. Interestingly, down/up-regulation of cannabinoid receptors (CBRs) varies across the different stages of PD, suggesting that some of the motor/ non-motor deficits may be related to changes in CBRs. Determination of the profile of changes of these receptors across the different stages of PD as well as their neural distribution within the BG could improve understanding of PD and identify pathways important in disease pathobiology. In this review, we focus on temporal and spatial alterations of CBRs during PD in the BG. At present, as inconclusive, but suggestive results have been obtained, future investigations should be conducted to extend preclinical studies examining CBRs changes within each stage in controlled clinical trials in order to determine the potential of targeting CBRs in management of PD.
Collapse
Affiliation(s)
- Monavareh Soti
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| | - Hoda Ranjbar
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| | - Kristi A Kohlmeier
- Department of Drug Design and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Mohammad Shabani
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
47
|
Zhang C, Zhao M, Wang B, Su Z, Guo B, Qin L, Zhang W, Zheng R. The Nrf2-NLRP3-caspase-1 axis mediates the neuroprotective effects of Celastrol in Parkinson's disease. Redox Biol 2021; 47:102134. [PMID: 34600334 PMCID: PMC8487081 DOI: 10.1016/j.redox.2021.102134] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/10/2021] [Accepted: 09/12/2021] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is a chronic neurodegenerative disorder that is characterized by motor symptoms as a result of a loss of dopaminergic neurons in the substantia nigra pars compacta (SNc), accompanied by chronic neuroinflammation, oxidative stress, formation of α-synuclein aggregates. Celastrol, a potent anti-inflammatory and anti-oxidative pentacyclic triterpene, has emerged as a neuroprotective agent. However, the mechanisms by which celastrol is neuroprotective in PD remain elusive. Here we show that celastrol protects against dopamine neuron loss, mitigates neuroinflammation, and relieves motor deficits in MPTP-induced PD mouse model and AAV-mediated human α-synuclein overexpression PD model. Whole-genome deep sequencing analysis revealed that Nrf2, NLRP3 and caspase-1 in SNc may be associated with the neuroprotective actions of celastrol in PD. By using multiple genetically modified mice (Nrf2-KO, NLRP3-KO and Caspase-1-KO), we identified that celastrol inhibits NLRP3 inflammasome activation, relieves motor deficits and nigrostriatal dopaminergic degeneration through Nrf2-NLRP3-caspase-1 pathway. Taken together, these findings suggest that Nrf2-NLRP3-caspase-1 axis may serve as a key target of celastrol in PD treatment, and highlight the favorable properties of celastrol for neuroprotection, making celastrol as a promising disease-modifying agent for PD.
Collapse
Affiliation(s)
- Chenyu Zhang
- Department of Anatomy, Histology and Embryology, Health Science Center, Peking University, Beijing, China
| | - Miao Zhao
- Department of Anatomy, Histology and Embryology, Health Science Center, Peking University, Beijing, China
| | - Bingwei Wang
- Department of Anatomy, Histology and Embryology, Health Science Center, Peking University, Beijing, China
| | - Zhijie Su
- Department of Anatomy, Histology and Embryology, Health Science Center, Peking University, Beijing, China
| | - Bingbing Guo
- Department of Anatomy, Histology and Embryology, Health Science Center, Peking University, Beijing, China
| | - Lihua Qin
- Department of Anatomy, Histology and Embryology, Health Science Center, Peking University, Beijing, China
| | - Weiguang Zhang
- Department of Anatomy, Histology and Embryology, Health Science Center, Peking University, Beijing, China
| | - Ruimao Zheng
- Department of Anatomy, Histology and Embryology, Health Science Center, Peking University, Beijing, China; Neuroscience Research Institute, Peking University, Beijing, China; Key Laboratory for Neuroscience of Ministry of Education, Peking University, Beijing, China; Key Laboratory for Neuroscience of National Health Commission, Peking University, Beijing, China.
| |
Collapse
|
48
|
Hescham SA, Chiang PH, Gregurec D, Moon J, Christiansen MG, Jahanshahi A, Liu H, Rosenfeld D, Pralle A, Anikeeva P, Temel Y. Magnetothermal nanoparticle technology alleviates parkinsonian-like symptoms in mice. Nat Commun 2021; 12:5569. [PMID: 34552093 PMCID: PMC8458499 DOI: 10.1038/s41467-021-25837-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 09/01/2021] [Indexed: 12/02/2022] Open
Abstract
Deep brain stimulation (DBS) has long been used to alleviate symptoms in patients suffering from psychiatric and neurological disorders through stereotactically implanted electrodes that deliver current to subcortical structures via wired pacemakers. The application of DBS to modulate neural circuits is, however, hampered by its mechanical invasiveness and the use of chronically implanted leads, which poses a risk for hardware failure, hemorrhage, and infection. Here, we demonstrate that a wireless magnetothermal approach to DBS (mDBS) can provide similar therapeutic benefits in two mouse models of Parkinson's disease, the bilateral 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and in the unilateral 6-hydroxydopamine (6-OHDA) model. We show magnetothermal neuromodulation in untethered moving mice through the activation of the heat-sensitive capsaicin receptor (transient receptor potential cation channel subfamily V member 1, TRPV1) by synthetic magnetic nanoparticles. When exposed to an alternating magnetic field, the nanoparticles dissipate heat, which triggers reversible firing of TRPV1-expressing neurons. We found that mDBS in the subthalamic nucleus (STN) enables remote modulation of motor behavior in healthy mice. Moreover, mDBS of the STN reversed the motor deficits in a mild and severe parkinsonian model. Consequently, this approach is able to activate deep-brain circuits without the need for permanently implanted hardware and connectors.
Collapse
Affiliation(s)
- Sarah-Anna Hescham
- Department of Neurosurgery, Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands.
| | - Po-Han Chiang
- Research Laboratory of Electronics and McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute of Biomedical Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan, ROC
| | - Danijela Gregurec
- Research Laboratory of Electronics and McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry and Pharmacy, Chair of Aroma and Smell Research, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Junsang Moon
- Research Laboratory of Electronics and McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Ali Jahanshahi
- Department of Neurosurgery, Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Huajie Liu
- Department of Neurosurgery, Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Dekel Rosenfeld
- Research Laboratory of Electronics and McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Arnd Pralle
- Department of Physics, University at Buffalo, Buffalo, NY, USA
| | - Polina Anikeeva
- Research Laboratory of Electronics and McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yasin Temel
- Department of Neurosurgery, Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
49
|
Ling ZM, Wang Q, Ma Y, Xue P, Gu Y, Cao MH, Wei ZY. Astrocyte Pannexin 1 Suppresses LPS-Induced Inflammatory Responses to Protect Neuronal SH-SY5Y Cells. Front Cell Neurosci 2021; 15:710820. [PMID: 34475813 PMCID: PMC8406772 DOI: 10.3389/fncel.2021.710820] [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: 05/17/2021] [Accepted: 07/16/2021] [Indexed: 01/05/2023] Open
Abstract
Reactive astrogliosis is a key hallmark of inflammatory responses in the pathogenesis of brain injury, including Parkinson’s disease (PD), but its role and regulatory mechanisms are not fully understood. Pannexin 1 (Panx 1) is a membrane channel that mediates substance release in many neurodegenerative diseases. However, the role of astrocyte Panx 1 in the regulation of PD-like neuroinflammation remains elusive. Here, we characterized the expression of Panx 1 in isolated primary astrocytes and a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD model. The functions of Panx 1 in inflammatory cytokines expression and the viability of neuronal SH-SY5Y cells were examined in cultured cells treated with lipopolysaccharide (LPS) and 1-methyl-4-phenylpyridinium (MPP+). We found that Panx 1 expression was significantly increased under both LPS- and MPP+-treated conditions. Panx 1 downregulation suppressed LPS-induced pro-inflammatory cytokine expression but did not significantly affect MPP+-induced astrocyte apoptosis or inflammatory cytokine expression through treatment with the Panx 1 inhibitor carbenoxolone (CBX) and Panx 1 siRNA. Moreover, silencing Panx 1 in reactive astrocytes had a potentially protective effect on the viability of neuronal SH-SY5Y cells. Therefore, we propose that Panx 1 may serve as a key regulator in reactive astrocytes to intervene in the inflammatory response and maintain neuronal viability in the context of PD-like conditions.
Collapse
Affiliation(s)
- Zhuo-Min Ling
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Medical School of Nantong University, Nantong, China
| | - Qian Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yu Ma
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Peng Xue
- Medical School of Nantong University, Nantong, China
| | - Yun Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Mao-Hong Cao
- Medical School of Nantong University, Nantong, China.,Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China
| | - Zhong-Ya Wei
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
50
|
Neuroprotective and Symptomatic Effects of Cannabidiol in an Animal Model of Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22168920. [PMID: 34445626 PMCID: PMC8396349 DOI: 10.3390/ijms22168920] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 08/14/2021] [Accepted: 08/16/2021] [Indexed: 12/27/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the loss of dopaminergic neurons in the Substantia Nigra pars compacta, leading to classical PD motor symptoms. Current therapies are purely symptomatic and do not modify disease progression. Cannabidiol (CBD), one of the main phytocannabinoids identified in Cannabis Sativa, which exhibits a large spectrum of therapeutic properties, including anti-inflammatory and antioxidant effects, suggesting its potential as disease-modifying agent for PD. The aim of this study was to evaluate the effects of chronic treatment with CBD (10 mg/kg, i.p.) on PD-associated neurodegenerative and neuroinflammatory processes, and motor deficits in the 6-hydroxydopamine model. Moreover, we investigated the potential mechanisms by which CBD exerted its effects in this model. CBD-treated animals showed a reduction of nigrostriatal degeneration accompanied by a damping of the neuroinflammatory response and an improvement of motor performance. In particular, CBD exhibits a preferential action on astrocytes and activates the astrocytic transient receptor potential vanilloid 1 (TRPV1), thus, enhancing the endogenous neuroprotective response of ciliary neurotrophic factor (CNTF). These results overall support the potential therapeutic utility of CBD in PD, as both neuroprotective and symptomatic agent.
Collapse
|