1
|
Apweiler M, Saliba SW, Sun L, Streyczek J, Normann C, Hellwig S, Bräse S, Fiebich BL. Modulation of neuroinflammation and oxidative stress by targeting GPR55 - new approaches in the treatment of psychiatric disorders. Mol Psychiatry 2024; 29:3779-3788. [PMID: 38796643 PMCID: PMC11609097 DOI: 10.1038/s41380-024-02614-5] [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: 07/15/2023] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 05/28/2024]
Abstract
Pharmacological treatment of psychiatric disorders remains challenging in clinical, pharmacological, and scientific practice. Even if many different substances are established for treating different psychiatric conditions, subgroups of patients show only small or no response to the treatment. The neuroinflammatory hypothesis of the genesis of psychiatric disorders might explain underlying mechanisms in these non-responders. For that reason, recent research focus on neuroinflammatory processes and oxidative stress as possible causes of psychiatric disorders. G-protein coupled receptors (GPCRs) form the biggest superfamily of membrane-bound receptors and are already well known as pharmacological targets in various diseases. The G-protein coupled receptor 55 (GPR55), a receptor considered part of the endocannabinoid system, reveals promising modulation of neuroinflammatory and oxidative processes. Different agonists and antagonists reduce pro-inflammatory cytokine release, enhance the synthesis of anti-inflammatory mediators, and protect cells from oxidative damage. For this reason, GPR55 ligands might be promising compounds in treating subgroups of patients suffering from psychiatric disorders related to neuroinflammation or oxidative stress. New approaches in drug design might lead to new compounds targeting different pathomechanisms of those disorders in just one molecule.
Collapse
Affiliation(s)
- Matthias Apweiler
- Neuroimmunology and Neurochemistry Research Group, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, D-79104, Freiburg, Germany
- Department of Cardiology and Angiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, D-79106 Freiburg, Germany
| | - Soraya Wilke Saliba
- Neuroimmunology and Neurochemistry Research Group, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, D-79104, Freiburg, Germany
| | - Lu Sun
- Neuroimmunology and Neurochemistry Research Group, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, D-79104, Freiburg, Germany
| | - Jana Streyczek
- Neuroimmunology and Neurochemistry Research Group, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, D-79104, Freiburg, Germany
| | - Claus Normann
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, D-79104, Freiburg, Germany
| | - Sabine Hellwig
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, D-79104, Freiburg, Germany
| | - Stefan Bräse
- Institute of Organic Chemistry, Karlsruhe Institute of Technology (KIT), Kaiserstrasse 12, D-76131, Karlsruhe, Germany
- Institute of Biological and Chemical Systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Kaiserstrasse 12, D-76131, Karlsruhe, Germany
| | - Bernd L Fiebich
- Neuroimmunology and Neurochemistry Research Group, Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, D-79104, Freiburg, Germany.
| |
Collapse
|
2
|
Kim J, Choi C. Orphan GPCRs in Neurodegenerative Disorders: Integrating Structural Biology and Drug Discovery Approaches. Curr Issues Mol Biol 2024; 46:11646-11664. [PMID: 39451571 PMCID: PMC11505999 DOI: 10.3390/cimb46100691] [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: 09/30/2024] [Revised: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024] Open
Abstract
Neurodegenerative disorders, particularly Alzheimer's and Parkinson's diseases, continue to challenge modern medicine despite therapeutic advances. Orphan G-protein-coupled receptors (GPCRs) have emerged as promising targets in the central nervous system, offering new avenues for drug development. This review focuses on the structural biology of orphan GPCRs implicated in these disorders, providing a comprehensive analysis of their molecular architecture and functional mechanisms. We examine recent breakthroughs in structural determination techniques, such as cryo-electron microscopy and X-ray crystallography, which have elucidated the intricate conformations of these receptors. The review highlights how structural insights inform our understanding of orphan GPCR activation, ligand binding and signaling pathways. By integrating structural data with molecular pharmacology, we explore the potential of structure-guided approaches in developing targeted therapeutics toward orphan GPCRs. This structural-biology-centered perspective aims to deepen our comprehension of orphan GPCRs and guide future drug discovery efforts in neurodegenerative disorders.
Collapse
Affiliation(s)
- Jinuk Kim
- Department of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea;
| | | |
Collapse
|
3
|
Chen M, Xu X, Wang F, Xu X. Investigating causality and shared genetic architecture between body mass index and cognitive function: a genome-wide cross-trait analysis and bi-directional Mendelian randomization study. Front Aging Neurosci 2024; 16:1466799. [PMID: 39478699 PMCID: PMC11522962 DOI: 10.3389/fnagi.2024.1466799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/30/2024] [Indexed: 11/02/2024] Open
Abstract
Background and objectives Observational studies have established a connection between body mass index (BMI) and an increased risk of cognitive decline. However, a comprehensive investigation into the causal relationships between BMI and cognitive function across diverse age groups, as well as the genetic underpinnings of this relationship, has been notably lacking. This study aims to investigate causality and the shared genetic underpinnings of between BMI and cognitive function by conducting a thorough genome-wide analysis, thereby provide valuable insights for developing personalized intervention strategies to promote cognitive health. Methods Genetic associations between BMI and cognitive function were thoroughly investigated through covariate genetic analysis and chained imbalance score regression, utilizing data from genome-wide association studies (GWAS). Bi-directional Mendelian Randomization (MR) was employed to uncover associations and potential functional genes were further scrutinized through Cross-trait meta-analysis and Summary-data-based MR (SMR). Subsequently, a detailed examination of the expression profiles of the identified risk SNPs in tissues and cells was conducted. Results The study found a significant negative correlation between BMI and cognitive function (β = -0.16, P = 1.76E-05), suggesting a causal linkage where higher BMI values were predictive of cognitive impairment. We identified 5 genetic loci (rs6809216, rs7187776, rs11713193, rs13096480, and rs13107325) between BMI and cognitive function by cross-trait meta-analysis and 5 gene-tissue pairs were identified by SMR analysis. Moreover, two novel risk genes TUFM and MST1R were shared by both cross-trait analysis and SMR analysis, which had not been observed in previous studies. Furthermore, significant enrichment of single nucleotide polymorphisms (SNPs) at tissue- and cell-specific levels was identified for both BMI and cognitive function, predominantly within the brain. Conclusion This study uncovers a causal relationship between BMI and cognitive function, with the discovery of TUFM and MST1R as shared genetic factors associated with both conditions. This novel finding offers new insights into the development of preventative strategies for cognitive decline in obese individuals, and further enhances our understanding of the underlying pathophysiology of these conditions. Furthermore, these findings could serve as a guide for the development of innovative therapeutic approaches to address cognitive decline in obese individuals.
Collapse
Affiliation(s)
- Mingyi Chen
- Department of Neurology and Stroke Center, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Xiaoxin Xu
- Department of Neurology and Stroke Center, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Fang Wang
- Department of Neurology and Stroke Center, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Xiaohong Xu
- Department of Neurology and Stroke Center, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
4
|
Tyrakis P, Agridi C, Kourti M. A Comprehensive Exploration of the Multifaceted Neuroprotective Role of Cannabinoids in Alzheimer's Disease across a Decade of Research. Int J Mol Sci 2024; 25:8630. [PMID: 39201317 PMCID: PMC11354546 DOI: 10.3390/ijms25168630] [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: 05/20/2024] [Revised: 07/15/2024] [Accepted: 08/05/2024] [Indexed: 09/02/2024] Open
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disorder, manifests through dysregulation of brain function and subsequent loss of bodily control, attributed to β-amyloid plaque deposition and TAU protein hyperphosphorylation and aggregation, leading to neuronal death. Concurrently, similar cannabinoids to the ones derived from Cannabis sativa are present in the endocannabinoid system, acting through receptors CB1R and CB2R and other related receptors such as Trpv-1 and GPR-55, and are being extensively investigated for AD therapy. Given the limited efficacy and adverse effects of current available treatments, alternative approaches are crucial. Therefore, this review aims to identify effective natural and synthetic cannabinoids and elucidate their beneficial actions for AD treatment. PubMed and Scopus databases were queried (2014-2024) using keywords such as "Alzheimer's disease" and "cannabinoids". The majority of natural (Δ9-THC, CBD, AEA, etc.) and synthetic (JWH-133, WIN55,212-2, CP55-940, etc.) cannabinoids included showed promise in improving memory, cognition, and behavioral symptoms, potentially via pathways involving antioxidant effects of selective CB1R agonists (such as the BDNF/TrkB/Akt pathway) and immunomodulatory effects of selective CB2R agonists (TLR4/NF-κB p65 pathway). Combining anticholinesterase properties with a cannabinoid moiety may enhance therapeutic responses, addressing cholinergic deficits of AD brains. Thus, the positive outcomes of the vast majority of studies discussed support further advancing cannabinoids in clinical trials for AD treatment.
Collapse
Affiliation(s)
| | | | - Malamati Kourti
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia 1516, Cyprus; (P.T.); (C.A.)
| |
Collapse
|
5
|
Singh MK, Shin Y, Ju S, Han S, Kim SS, Kang I. Comprehensive Overview of Alzheimer's Disease: Etiological Insights and Degradation Strategies. Int J Mol Sci 2024; 25:6901. [PMID: 39000011 PMCID: PMC11241648 DOI: 10.3390/ijms25136901] [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: 05/21/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder and affects millions of individuals globally. AD is associated with cognitive decline and memory loss that worsens with aging. A statistical report using U.S. data on AD estimates that approximately 6.9 million individuals suffer from AD, a number projected to surge to 13.8 million by 2060. Thus, there is a critical imperative to pinpoint and address AD and its hallmark tau protein aggregation early to prevent and manage its debilitating effects. Amyloid-β and tau proteins are primarily associated with the formation of plaques and neurofibril tangles in the brain. Current research efforts focus on degrading amyloid-β and tau or inhibiting their synthesis, particularly targeting APP processing and tau hyperphosphorylation, aiming to develop effective clinical interventions. However, navigating this intricate landscape requires ongoing studies and clinical trials to develop treatments that truly make a difference. Genome-wide association studies (GWASs) across various cohorts identified 40 loci and over 300 genes associated with AD. Despite this wealth of genetic data, much remains to be understood about the functions of these genes and their role in the disease process, prompting continued investigation. By delving deeper into these genetic associations, novel targets such as kinases, proteases, cytokines, and degradation pathways, offer new directions for drug discovery and therapeutic intervention in AD. This review delves into the intricate biological pathways disrupted in AD and identifies how genetic variations within these pathways could serve as potential targets for drug discovery and treatment strategies. Through a comprehensive understanding of the molecular underpinnings of AD, researchers aim to pave the way for more effective therapies that can alleviate the burden of this devastating disease.
Collapse
Affiliation(s)
- Manish Kumar Singh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yoonhwa Shin
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Songhyun Ju
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung Soo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
6
|
Sharon N, Yarmolinsky L, Khalfin B, Fleisher-Berkovich S, Ben-Shabat S. Cannabinoids' Role in Modulating Central and Peripheral Immunity in Neurodegenerative Diseases. Int J Mol Sci 2024; 25:6402. [PMID: 38928109 PMCID: PMC11204381 DOI: 10.3390/ijms25126402] [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: 05/01/2024] [Revised: 05/30/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Cannabinoids (the endocannabinoids, the synthetic cannabinoids, and the phytocannabinoids) are well known for their various pharmacological properties, including neuroprotective and anti-inflammatory features, which are fundamentally important for the treatment of neurodegenerative diseases. The aging of the global population is causing an increase in these diseases that require the development of effective drugs to be even more urgent. Taking into account the unavailability of effective drugs for neurodegenerative diseases, it seems appropriate to consider the role of cannabinoids in the treatment of these diseases. To our knowledge, few reviews are devoted to cannabinoids' impact on modulating central and peripheral immunity in neurodegenerative diseases. The objective of this review is to provide the best possible information about the cannabinoid receptors and immuno-modulation features, peripheral immune modulation by cannabinoids, cannabinoid-based therapies for the treatment of neurological disorders, and the future development prospects of making cannabinoids versatile tools in the pursuit of effective drugs.
Collapse
Affiliation(s)
| | | | | | | | - Shimon Ben-Shabat
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (N.S.); (L.Y.); (B.K.); (S.F.-B.)
| |
Collapse
|
7
|
Sun T, Du YY, Zhang YQ, Tian QQ, Li X, Yu JY, Guo YY, Liu QQ, Yang L, Wu YM, Yang Q, Zhao MG. Activation of GPR55 Ameliorates Maternal Separation-Induced Learning and Memory Deficits by Augmenting 5-HT Synthesis in the Dorsal Raphe Nucleus of Juvenile Mice. ACS OMEGA 2024; 9:21838-21850. [PMID: 38799363 PMCID: PMC11112691 DOI: 10.1021/acsomega.3c08934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 04/16/2024] [Accepted: 04/24/2024] [Indexed: 05/29/2024]
Abstract
Maternal separation (MS) represents a profound early life stressor with enduring impacts on neuronal development and adult cognitive function in both humans and rodents. MS is associated with persistent dysregulations in neurotransmitter systems, including the serotonin (5-HT) pathway, which is pivotal for mood stabilization and stress-coping mechanisms. Although the novel cannabinoid receptor, GPR55, is recognized for its influence on learning and memory, its implications on the function and synaptic dynamics of 5-HT neurons within the dorsal raphe nucleus (DRN) remain to be elucidated. In this study, we sought to discern the repercussions of GPR55 activation on 5-HT synthesis within the DRN of adult C57BL/6J mice that experienced MS. Concurrently, we analyzed potential alterations in excitatory synaptic transmission, long-term synaptic plasticity, and relevant learning and memory outcomes. Our behavioral assessments indicated a marked amelioration in MS-induced learning and memory deficits following GPR55 activation. In conjunction with this, we noted a substantial decrease in 5-HT levels in the MS model, while GPR55 activation stimulated tryptophan hydroxylase 2 synthesis and fostered the release of 5-HT. Electrophysiological patch-clamp analyses highlighted the ability of GPR55 activation to alleviate MS-induced cognitive deficits by modulating the frequency and magnitude of miniature excitatory postsynaptic currents within the DRN. Notably, this cognitive enhancement was underpinned by the phosphorylation of both NMDA and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. In summary, our findings underscore the capacity of GPR55 to elevate 5-HT synthesis and modify synaptic transmissions within the DRN of juvenile mice, positing GPR55 as a promising therapeutic avenue for ameliorating MS-induced cognitive impairment.
Collapse
Affiliation(s)
- Ting Sun
- Precision
Pharmacy & Drug Development Center, Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical
University, Xi’an 710038, China
| | - Ya-Ya Du
- Precision
Pharmacy & Drug Development Center, Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical
University, Xi’an 710038, China
| | - Yong-Qiang Zhang
- Department
of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi’an 710032, China
| | - Qin-Qin Tian
- Department
of Chemistry, School of Pharmacy, Air Force
Medical University, Xi’an 710032, China
| | - Xi Li
- Precision
Pharmacy & Drug Development Center, Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical
University, Xi’an 710038, China
| | - Jiao-Yan Yu
- Precision
Pharmacy & Drug Development Center, Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical
University, Xi’an 710038, China
| | - Yan-Yan Guo
- Precision
Pharmacy & Drug Development Center, Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical
University, Xi’an 710038, China
| | - Qing-Qing Liu
- Precision
Pharmacy & Drug Development Center, Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical
University, Xi’an 710038, China
| | - Le Yang
- Precision
Pharmacy & Drug Development Center, Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical
University, Xi’an 710038, China
| | - Yu-Mei Wu
- Department
of Pharmacology, School of Pharmacy, Air
Force Medical University, Xi’an 710032, China
| | - Qi Yang
- Precision
Pharmacy & Drug Development Center, Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical
University, Xi’an 710038, China
| | - Ming-Gao Zhao
- Precision
Pharmacy & Drug Development Center, Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical
University, Xi’an 710038, China
| |
Collapse
|
8
|
Kadian M, Saini N, Khera A, Kumar A. Neuroprotective mechanism of trans,trans-Farnesol in an ICV-STZ-induced rat model of Alzheimer's pathology. Inflammopharmacology 2024; 32:1545-1573. [PMID: 38308793 DOI: 10.1007/s10787-023-01413-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 12/13/2023] [Indexed: 02/05/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is a prominent cause of dementia, resulting in neurodegeneration and memory impairment. This condition imposes a considerable public health burden on both patients and their families due to the patients' functional impairments as well as the psychological and financial constraints. It has been well demonstrated that its aetiology involves proteinopathy, mitochondriopathies, and enhanced reactive oxygen species (ROS) generation, which are some of the key features of AD brains that further result in oxidative stress, excitotoxicity, autophagy, and mitochondrial dysfunction. OBJECTIVE The current investigation was created with the aim of elucidating the neurological defence mechanism of trans,trans-Farnesol (TF) against intracerebroventricular-streptozotocin (ICV-STZ)-induced Alzheimer-like symptoms and related pathologies in rodents. MATERIALS AND METHODS The current investigation involved male SD rats receiving TF (25-100 mg/kg, per oral) consecutively for 21 days in ICV-STZ-treated animals. An in silico study was carried out to explore the possible interaction between TF and NADH dehydrogenase and succinate dehydrogenase. Further, various behavioural (Morris water maze and novel object recognition test), biochemical (oxidants and anti-oxidant markers), activities of mitochondrial enzyme complexes and acetylcholinesterase (AChE), pro-inflammatory (tumor necrosis factor-alpha; TNF-α) levels, and histopathological studies were evaluated in specific brain regions. RESULTS Rats administered ICV-STZ followed by treatment with TF (25, 50, and 100 mg/kg) for 21 days had significantly better mental performance (reduced escape latency to access platform, extended time spent in target quadrant, and improved differential index) in the Morris water maze test and new object recognition test models when compared to control (ICV-STZ)-treated groups. Further, TF treatment significantly restored redox proportion, anti-oxidant levels, regained mitochondrial capacities, attenuated altered AChE action, levels of TNF-α, and histopathological alterations in certain brain regions in comparison with control. In in silico analysis, TF caused greater interaction with NADH dehydrogenase and succinate dehydrogenase. CONCLUSION The current work demonstrates the neuroprotective ability of TF in an experimental model with AD-like pathologies. The study further suggests that the neuroprotective impacts of TF may be related to its effects on TNF-α levels, oxidative stress pathways, and mitochondrial complex capabilities.
Collapse
Affiliation(s)
- Monika Kadian
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Neetu Saini
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Alka Khera
- Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Anil Kumar
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
9
|
Zhou TY, Ma RX, Li J, Zou B, Yang H, Ma RY, Wu ZQ, Li J, Yao Y. Review of PINK1-Parkin-mediated mitochondrial autophagy in Alzheimer's disease. Eur J Pharmacol 2023; 959:176057. [PMID: 37751832 DOI: 10.1016/j.ejphar.2023.176057] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 09/14/2023] [Accepted: 09/14/2023] [Indexed: 09/28/2023]
Abstract
Mitochondrial autophagy plays an important role in maintaining the complexity of mitochondrial functions and removing damaged mitochondria, of which the PINK1-Parkin signal pathway is one of the most classical pathways. Thus, a comprehensive and in-depth interpretation of the PINK1-Parkin signal pathway might deepen our understanding on the impacts of mitochondrial autophagy. Alzheimer's disease (AD) is a classical example of neurodegenerative disease. Research on the pathogenesis and treatments of AD has been a focus of scientific research because of its complexity and the limitations of current drug therapies. It was reported that the pathogenesis of AD might be related to mitochondrial autophagy due to excessive deposition of Aβ protein and aggravation of the phosphorylation of Tau protein. Two key proteins in the PINK1-Parkin signaling pathway, PINK1 and Parkin, have important roles in the folding and accumulation of Aβ protein and the phosphorylation of Tau protein. In addition, the intermediate signal molecules in the PINK1-Parkin signal pathway also have certain effects on AD. In this paper, we first described the role of PINK1-Parkin signal pathway on mitochondrial autophagy, then discussed and analyzed the effect of the PINK1-Parkin signal pathway in AD and other metabolic diseases. Our aim was to provide a theoretical direction to further elucidate the pathogenesis of AD and highlight the key molecules related to AD that could be important targets used for AD drug development.
Collapse
Affiliation(s)
- Ting-Yuan Zhou
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Rui-Xia Ma
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Jia Li
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Bin Zou
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Hui Yang
- Research Center of Medical Science and Technology, Ningxia Medical University, Yinchuan, 750004, China
| | - Rui-Yin Ma
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Zi-Qi Wu
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Juan Li
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China; Ningxia Engineering and Technology Research Center for Modernization of Characteristic Chinese Medicine, and Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, 750004, China.
| | - Yao Yao
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
10
|
Choi K, Lee Y, Kim C. An In Silico Study for Expanding the Utility of Cannabidiol in Alzheimer's Disease Therapeutic Development. Int J Mol Sci 2023; 24:16013. [PMID: 37959001 PMCID: PMC10648567 DOI: 10.3390/ijms242116013] [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: 09/29/2023] [Revised: 10/18/2023] [Accepted: 10/20/2023] [Indexed: 11/15/2023] Open
Abstract
Cannabidiol (CBD), a major non-psychoactive component of the cannabis plant, has shown therapeutic potential in Alzheimer's disease (AD). In this study, we identified potential CBD targets associated with AD using a drug-target binding affinity prediction model and generated CBD analogs using a genetic algorithm combined with a molecular docking system. As a result, we identified six targets associated with AD: Endothelial NOS (ENOS), Myeloperoxidase (MPO), Apolipoprotein E (APOE), Amyloid-beta precursor protein (APP), Disintegrin and metalloproteinase domain-containing protein 10 (ADAM10), and Presenilin-1 (PSEN1). Furthermore, we generated CBD analogs for each target that optimize for all desired drug-likeness properties and physicochemical property filters, resulting in improved pIC50 values and docking scores compared to CBD. Molecular dynamics (MD) simulations were applied to analyze each target's CBD and highest-scoring CBD analogs. The MD simulations revealed that the complexes of ENOS, MPO, and ADAM10 with CBD exhibited high conformational stability, and the APP and PSEN1 complexes with CBD analogs demonstrated even higher conformational stability and lower interaction energy compared to APP and PSEN1 complexes with CBD. These findings demonstrated the capable binding of the six identified targets with CBD and the enhanced binding stability achieved with the developed CBD analogs for each target.
Collapse
Affiliation(s)
- Kyudam Choi
- Heerae Co., Ltd., Seoul 06253, Republic of Korea;
| | - Yurim Lee
- Department of Software, Sejong University, Seoul 05006, Republic of Korea;
| | - Cheongwon Kim
- Department of Software, Sejong University, Seoul 05006, Republic of Korea;
| |
Collapse
|
11
|
Wang X, Zhang H, Liu Y, Xu Y, Yang B, Li H, Chen L. An overview on synthetic and biological activities of cannabidiol (CBD) and its derivatives. Bioorg Chem 2023; 140:106810. [PMID: 37659147 DOI: 10.1016/j.bioorg.2023.106810] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/17/2023] [Accepted: 08/22/2023] [Indexed: 09/04/2023]
Abstract
(-)-Cannabidiol is a class of non-psychoactive plant cannabinoids derived from cannabis plants. Currently, Epidiolex (Cannabidiol) has been approved by the FDA for the treatment of two rare and severe forms of epilepsy related diseases, namely Lennox-Gastaut syndrome (LGS) and Dravet (DS). In addition, Cannabidiol and its structural analogues have received increasing attention due to their potential therapeutic effects such as neuroprotection, anti-epilepsy, anti-inflammation, anti-anxiety, and anti-cancer. Based on literature review, no comprehensive reviews on the synthesis of Cannabidiol and its derivatives have been found in recent years. Therefore, this article summarizes the published synthesis methods of Cannabidiol and the synthesis routes of Cannabidiol derivatives, and introduces the biological activities of some Cannabidiol analogues that have been studied extensively and have significant activities.
Collapse
Affiliation(s)
- Xiuli Wang
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Huanbang Zhang
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yan Liu
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Heilongjiang 150006, China
| | - Yang Xu
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Bingyou Yang
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Heilongjiang 150006, China.
| | - Hua Li
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China; Institute of Structural Pharmacology & TCM Chemical Biology, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China.
| | - Lixia Chen
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
12
|
Lingli C, Hongmei N, Penghuan J, Hongli Z, Yuye L, Rui W, Fei R, Zhihong Y, Dongfang H, Yaming G. Inhibition of RhoA/ROCK signalling pathway activity improves neural damage and cognitive deficits in the fluorosis model. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 266:115554. [PMID: 37806133 DOI: 10.1016/j.ecoenv.2023.115554] [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: 06/17/2023] [Revised: 09/28/2023] [Accepted: 10/05/2023] [Indexed: 10/10/2023]
Abstract
Excessive fluoride intake poses health risks to humans and animals. Many studies have indicated that fluoride exposure can damage the cytoskeleton and synapses, which has negative effects on the intellectual development of humans and animals. Our previous study suggested that the RhoA/ROCK signalling pathway is activated by NaF exposure in HT-22 cells and plays a vital role in cytoskeletal assembly and synaptogenesis. However, the mechanism underlying RhoA/ROCK-mediated cytoskeletal injury induced by fluoride remains unclear. In this study, Neuro-2A cells and ICR mice were used to investigate the effects of RhoA/ROCK activation inhibition on NaF-induced synaptic dysfunction and cognitive impairment. We detected the expression of GAP, RhoA, ROCK1/2, and (p)-MLC in vivo and in vitro model. The results showed that NaF exposure activated the RhoA/ROCK/MLC signalling pathway. We measured the effects of RhoA/ROCK inhibition on synaptic injury and intellectual impairment induced by NaF exposure. In vitro, Y-27632 suppressed activated RhoA/ROCK, attenuated morphological and ultrastructural damage, and decreased the survival rate and synapse-functional protein expression caused by NaF. In vivo, the results showed that the RhoA/ROCK/MLC pathway was inhibited by fasudil and improved pathological damage in the hippocampus, cognitive impairment, and decreased expression of neurofunctional proteins induced by NaF. Overall, these results suggest that fasudil and Y-27632 can reverse neurotoxicity caused by fluoride exposure. Furthermore, inhibition of RhoA/ROCK may be a future treatment for CNS injury, and more detailed studies on other neurodegenerative disease models are required to confirm its effectiveness.
Collapse
Affiliation(s)
- Chen Lingli
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan Provence, 453003, China; Postdoctoral Research and Development Base, Henan Institute of Science and Technology, Xinxiang, Henan Provence, 453003, China
| | - Ning Hongmei
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan Provence, 453003, China
| | - Jia Penghuan
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan Provence, 453003, China
| | - Zhang Hongli
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan Provence, 453003, China
| | - Liu Yuye
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan Provence, 453003, China
| | - Wang Rui
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan Provence, 453003, China
| | - Ren Fei
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan Provence, 453003, China
| | - Yin Zhihong
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan Provence, 453003, China
| | - Hu Dongfang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan Provence, 453003, China
| | - Ge Yaming
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan Provence, 453003, China.
| |
Collapse
|
13
|
Medina-Vera D, Zhao H, Bereczki E, Rosell-Valle C, Shimozawa M, Chen G, de Fonseca FR, Nilsson P, Tambaro S. The Expression of the Endocannabinoid Receptors CB2 and GPR55 Is Highly Increased during the Progression of Alzheimer's Disease in AppNL-G-F Knock-In Mice. BIOLOGY 2023; 12:805. [PMID: 37372090 DOI: 10.3390/biology12060805] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023]
Abstract
BACKGROUND The endocannabinoid system (ECS) and associated lipid transmitter-based signaling systems play an important role in modulating brain neuroinflammation. ECS is affected in neurodegenerative disorders, such as Alzheimer's disease (AD). Here we have evaluated the non-psychotropic endocannabinoid receptor type 2 (CB2) and lysophosphatidylinositol G-protein-coupled receptor 55 (GPR55) localization and expression during Aβ-pathology progression. METHODS Hippocampal gene expression of CB2 and GPR55 was explored by qPCR analysis, and brain distribution was evaluated by immunofluorescence in the wild type (WT) and APP knock-in AppNL-G-F AD mouse model. Furthermore, the effects of Aβ42 on CB2 and GPR55 expression were assessed in primary cell cultures. RESULTS CB2 and GPR55 mRNA levels were significantly upregulated in AppNL-G-F mice at 6 and 12 months of age, compared to WT. CB2 was highly expressed in the microglia and astrocytes surrounding the Aβ plaques. Differently, GPR55 staining was mainly detected in neurons and microglia but not in astrocytes. In vitro, Aβ42 treatment enhanced CB2 receptor expression mainly in astrocytes and microglia cells, whereas GPR55 expression was enhanced primarily in neurons. CONCLUSIONS These data show that Aβ pathology progression, particularly Aβ42, plays a crucial role in increasing the expression of CB2 and GPR55 receptors, supporting CB2 and GPR55 implications in AD.
Collapse
Affiliation(s)
- Dina Medina-Vera
- Instituto de Investigación Biomédica de Málaga-IBIMA, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Facultad de Ciencias, Universidad de Málaga, 29010 Málaga, Spain
| | - Hongjing Zhao
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, China
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, 17164 Solna, Sweden
| | - Erika Bereczki
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, 17164 Solna, Sweden
| | - Cristina Rosell-Valle
- Instituto de Investigación Biomédica de Málaga-IBIMA, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Makoto Shimozawa
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, 17164 Solna, Sweden
| | - Gefei Chen
- Department of Biosciences and Nutrition, Karolinska Institutet, 14152 Huddinge, Sweden
| | - Fernando Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga-IBIMA, Unidad de Gestión Clínica de Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, 17164 Solna, Sweden
| | - Simone Tambaro
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, 17164 Solna, Sweden
| |
Collapse
|
14
|
Huat TJ, Onraet T, Camats-Perna J, Newcombe EA, Ngo KC, Sue AN, Mirzaei M, LaFerla FM, Medeiros R. Deletion of MyD88 in astrocytes prevents β-amyloid-induced neuropathology in mice. Glia 2023; 71:431-449. [PMID: 36271704 PMCID: PMC9970273 DOI: 10.1002/glia.24285] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 10/04/2022] [Accepted: 10/07/2022] [Indexed: 11/07/2022]
Abstract
As the understanding of immune responses in Alzheimer's disease (AD) is in its early phases, there remains an urgency to identify the cellular and molecular processes driving chronic inflammation. In AD, a subpopulation of astrocytes acquires a neurotoxic phenotype which prompts them to lose typical physiological features. While the underlying molecular mechanisms are still unknown, evidence suggests that myeloid differentiation primary response 88 (MyD88) adaptor protein may play a role in coordinating these cells' immune responses in AD. Herein, we combined studies in human postmortem samples with a conditional genetic knockout mouse model to investigate the link between MyD88 and astrocytes in AD. In silico analyses of bulk and cell-specific transcriptomic data from human postmortem brains demonstrated an upregulation of MyD88 expression in astrocytes in AD versus non-AD individuals. Proteomic studies revealed an increase in glial fibrillary acidic protein in multiple brain regions of AD subjects. These studies also showed that although overall MyD88 steady-state levels were unaffected by AD, this protein was enriched in astrocytes near amyloid plaques and neurofibrillary tangles. Functional studies in mice indicated that the deletion of astrocytic MyD88 protected animals from the acute synaptic toxicity and cognitive impairment caused by the intracerebroventricular administration of β-amyloid (Aβ). Lastly, unbiased proteomic analysis revealed that loss of astrocytic MyD88 resulted in altered astrocyte reactivity, lower levels of immune-related proteins, and higher expression of synaptic-related proteins in response to Aβ. Our studies provide evidence of the pivotal role played by MyD88 in the regulation of astrocytes response to AD.
Collapse
Affiliation(s)
- Tee Jong Huat
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland. Brisbane, QLD, Australia
- Centre for Stem Cell Ageing and Regenerative Engineering, The University of Queensland. Brisbane, QLD, Australia
| | - Tessa Onraet
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland. Brisbane, QLD, Australia
| | - Judith Camats-Perna
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland. Brisbane, QLD, Australia
| | - Estella A. Newcombe
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland. Brisbane, QLD, Australia
| | - Kim C. Ngo
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine. Irvine, CA, USA
| | - Ashley N. Sue
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine. Irvine, CA, USA
| | - Mehdi Mirzaei
- Clinical Medicine Department, Faculty of Medicine, Health and Human Sciences, Macquarie University. Sydney, NSW, Australia
| | - Frank M. LaFerla
- Department of Neurobiology and Behavior, University of California, Irvine. Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine. Irvine, CA, USA
| | - Rodrigo Medeiros
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland. Brisbane, QLD, Australia
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine. Irvine, CA, USA
- Correspondence: Rodrigo Medeiros, University of California, Irvine, 3400A Biological Sciences III, Irvine, CA 92697-4545.
| |
Collapse
|
15
|
Anderson LL, Bahceci DA, Hawkins NA, Everett-Morgan D, Banister SD, Kearney JA, Arnold JC. Heterozygous deletion of Gpr55 does not affect a hyperthermia-induced seizure, spontaneous seizures or survival in the Scn1a+/- mouse model of Dravet syndrome. PLoS One 2023; 18:e0280842. [PMID: 36701411 PMCID: PMC9879440 DOI: 10.1371/journal.pone.0280842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
A purified preparation of cannabidiol (CBD), a cannabis constituent, has been approved for the treatment of intractable childhood epilepsies such as Dravet syndrome. Extensive pharmacological characterization of CBD shows activity at numerous molecular targets but its anticonvulsant mechanism(s) of action is yet to be delineated. Many suggest that the anticonvulsant action of CBD is the result of G protein-coupled receptor 55 (GPR55) inhibition. Here we assessed whether Gpr55 contributes to the strain-dependent seizure phenotypes of the Scn1a+/- mouse model of Dravet syndrome. The Scn1a+/- mice on a 129S6/SvEvTac (129) genetic background have no overt phenotype, while those on a [129 x C57BL/6J] F1 background exhibit a severe phenotype that includes hyperthermia-induced seizures, spontaneous seizures and reduced survival. We observed greater Gpr55 transcript expression in the cortex and hippocampus of mice on the seizure-susceptible F1 background compared to those on the seizure-resistant 129 genetic background, suggesting that Gpr55 might be a genetic modifier of Scn1a+/- mice. We examined the effect of heterozygous genetic deletion of Gpr55 and pharmacological inhibition of GPR55 on the seizure phenotypes of F1.Scn1a+/- mice. Heterozygous Gpr55 deletion and inhibition of GPR55 with CID2921524 did not affect the temperature threshold of a thermally-induced seizure in F1.Scn1a+/- mice. Neither was there an effect of heterozygous Gpr55 deletion observed on spontaneous seizure frequency or survival of F1.Scn1a+/- mice. Our results suggest that GPR55 antagonism may not be a suitable anticonvulsant target for Dravet syndrome drug development programs, although future research is needed to provide more definitive conclusions.
Collapse
Affiliation(s)
- Lyndsey L. Anderson
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, Discipline of Pharmacology, Sydney Pharmacy School, The University of Sydney, Sydney, NSW, Australia
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
| | - Dilara A. Bahceci
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, Discipline of Pharmacology, Sydney Pharmacy School, The University of Sydney, Sydney, NSW, Australia
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
| | - Nicole A. Hawkins
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Evanston, IL, United States of America
| | - Declan Everett-Morgan
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
| | - Samuel D. Banister
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
- Faculty of Science, School of Chemistry, The University of Sydney, Sydney, NSW, Australia
| | - Jennifer A. Kearney
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Evanston, IL, United States of America
| | - Jonathon C. Arnold
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
- Faculty of Medicine and Health, Discipline of Pharmacology, Sydney Pharmacy School, The University of Sydney, Sydney, NSW, Australia
- Lambert Initiative for Cannabinoid Therapeutics, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
16
|
Liang W, Xie Z, Liao D, Li Y, Li Z, Zhao Y, Li X, Dong M. Inhibiting microRNA-142-5p improves learning and memory in Alzheimer's disease rats via targeted regulation of the PTPN1-mediated Akt pathway. Brain Res Bull 2023; 192:107-114. [PMID: 35219754 DOI: 10.1016/j.brainresbull.2022.02.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 12/03/2021] [Accepted: 02/22/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVE MicroRNAs (miRNAs) have been recognized as possible biomarkers for Alzheimer's disease (AD). MiR-142-5p has been reported to be abnormally expressed in brain tissues. However, the role of miR-142-5p in AD pathogenesis keeps unclear. This study aimed to investigate the effect of miR-142-5p on the learning and memory of AD rats via regulation of protein tyrosine phosphatase nonreceptor type 1 (PTPN1)-mediated protein kinase B (Akt) pathway. METHODS The AD model was established by injection of Aβ1-42 oligomer once into the lateral ventricle of rats, and the spatial learning and memory ability of rats was measured. AD rats were injected with miR-142-5p or PTPN1 vectors to explore their functions in inflammation, Aβ, p-tau protein, apoptosis in brain tissues, and the effects on Akt pathway. The targeting relationship between miR-142-5p and PTPN1 was detected. RESULTS Overexpressed miR-142-5p, down-regulated PTPN1 and inactivated Akt pathway were exhibited in AD. MiR-142-5p targeted PTPN1 to mediate Akt pathway. Reduced miR-142-5p and elevated PTPN1 improved the behavior of AD rats. MiR-142-5p targeted PTPN1 to effectively inhibit Aβ formation and abnormal phosphorylation of p-tau protein, suppress the inflammation in the brain tissues of AD rat, and improve the survival rate of brain tissue cells. MiR-142-5p regulated PTPN1 to activate the Akt pathway, further inhibiting the apoptosis of brain neurons in AD rats. CONCLUSION Down-regulating miR-142-5p targets PTPN1 to activate Akt pathway, thus improving the learning and memory of AD rats and playing an anti-AD role.
Collapse
Affiliation(s)
- Weiwei Liang
- Department of General Practice, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Zhuojun Xie
- Department of The Third outpatient, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Dong Liao
- Department of Ultrasound, Yunnan Geriatric Hospital, Kunming, Yunnan, China
| | - Ying Li
- Department of Rehabilitation Medicine, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Zhengyu Li
- Department of Rehabilitation Medicine, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Yuanru Zhao
- Department of Laboratory Medicine, Yunnan Xinkunhua Hospital, Kunming, Yunnan, China
| | - Xiaobo Li
- Department of Geriatric Medicine, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China.
| | - Manli Dong
- Department of Rehabilitation Medicine, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China.
| |
Collapse
|
17
|
Kurano M, Saito Y, Uranbileg B, Saigusa D, Kano K, Aoki J, Yatomi Y. Modulations of bioactive lipids and their receptors in postmortem Alzheimer's disease brains. Front Aging Neurosci 2022; 14:1066578. [PMID: 36570536 PMCID: PMC9780287 DOI: 10.3389/fnagi.2022.1066578] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022] Open
Abstract
Background Analyses of brain samples from Alzheimer's disease (AD) patients may be expected to help us improve our understanding of the pathogenesis of AD. Bioactive lipids, including sphingolipids, glycerophospholipids, and eicosanoids/related mediators have been demonstrated to exert potent physiological actions and to be involved in the pathogenesis of various human diseases. In this cross-sectional study, we attempted to elucidate the associations of these bioactive lipids with the pathogenesis/pathology of AD through postmortem studies of human brains. Methods We measured the levels of glycerophospholipids, sphingolipids, and eicosanoids/related mediators in the brains of patients with AD (AD brains), patients with Cerad score B (Cerad-b brains), and control subjects (control brains), using a liquid chromatography-mass spectrometry method; we also measured the mRNA levels of specific receptors for these bioactive lipids in the same brain specimens. Results The levels of several species of sphingomyelins and ceramides were higher in the Cerad-b and AD brains. Levels of several species of lysophosphatidic acids (LPAs), lysophosphatidylcholine, lysophosphatidylserine, lysophosphatidylethanolamine (LPE), lysophosphatidylinositol, phosphatidylcholine, phosphatidylserine (PS), phosphatidylethanolamine (PE), phosphatidylinositol, and phosphatidylglycerol were especially high in the Cerad-b brains, while those of lysophosphatidylglycerol (LPG) were especially high in the AD brains. Several eicosanoids, including metabolites of prostaglandin E2, oxylipins, metabolites of epoxide, and metabolites of DHA and EPA, such as resolvins, were also modulated in the AD brains. Among the lipid mediators, the levels of S1P2, S1P5, LPA1, LPA2, LPA6, P2Y10, GPR174, EP1, DP1, DP2, IP, FP, and TXA2r were lower in the AD and/or Cerad-b brains. The brain levels of ceramides, LPC, LPI, PE, and PS showed strong positive correlations with the Aβ contents, while those of LPG showed rather strong positive correlations with the presence of senile plaques and neurofibrillary tangles. A discriminant analysis revealed that LPG is especially important for AD and the LPE/PE axis is important for Cerad-b. Conclusions Comprehensive lipidomics, together with the measurement of lipid receptor expression levels provided novel evidence for the associations of bioactive lipids with AD, which is expected to facilitate future translational research and reverse translational research.
Collapse
Affiliation(s)
- Makoto Kurano
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan,*Correspondence: Makoto Kurano,
| | - Yuko Saito
- Tokyo Metropolitan Geriatric Hospital, Institute of Gerontology, Tokyo, Japan
| | - Baasanjav Uranbileg
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Daisuke Saigusa
- Laboratory of Biomedical and Analytical Sciences, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
| | - Kuniyuki Kano
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Junken Aoki
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
18
|
Qian XH, Liu XL, Chen G, Chen SD, Tang HD. Injection of amyloid-β to lateral ventricle induces gut microbiota dysbiosis in association with inhibition of cholinergic anti-inflammatory pathways in Alzheimer's disease. J Neuroinflammation 2022; 19:236. [PMID: 36171620 PMCID: PMC9520842 DOI: 10.1186/s12974-022-02599-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 09/18/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common neurodegenerative disease and its pathogenesis is still unclear. There is dysbiosis of gut microbiota in AD patients. More importantly, dysbiosis of the gut microbiota has been observed not only in AD patients, but also in patients with mild cognitive impairment (MCI). However, the mechanism of gut microbiota dysbiosis in AD is poorly understood. Cholinergic anti-inflammatory pathway is an important pathway for the central nervous system (CNS) regulation of peripheral immune homeostasis, especially in the gut. Therefore, we speculated that dysfunction of cholinergic anti-inflammatory pathway is a potential pathway for dysbiosis of the gut microbiota in AD. METHODS In this study, we constructed AD model mice by injecting Aβ1-42 into the lateral ventricle, and detected the cognitive level of mice by the Morris water maze test. In addition, 16S rDNA high-throughput analysis was used to detect the gut microbiota abundance of each group at baseline, 2 weeks and 4 weeks after surgery. Furthermore, immunofluorescence and western blot were used to detect alteration of intestinal structure of mice, cholinergic anti-inflammatory pathway, and APP process of brain and colon in each group. RESULTS Aβ1-42 i.c.v induced cognitive impairment and neuron damage in the brain of mice. At the same time, Aβ1-42 i.c.v induced alteration of gut microbiota at 4 weeks after surgery, while there was no difference at the baseline and 2 weeks after surgery. In addition, changes in colon structure and increased levels of pro-inflammatory factors were detected in Aβ1-42 treatment group, accompanied by inhibition of cholinergic anti-inflammatory pathways. Amyloidogenic pathways in both the brain and colon were accelerated in Aβ1-42 treatment group. CONCLUSIONS The present findings suggested that Aβ in the CNS can induce gut microbiota dysbiosis, alter intestinal structure and accelerate the amyloidogenic pathways, which were related to inhibiting cholinergic anti-inflammatory pathways.
Collapse
Affiliation(s)
- Xiao-Hang Qian
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiao-Li Liu
- Department of Neurology, Shanghai Fengxian District Central Hospital, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai, 201406, China
| | - Guang Chen
- The Second Hospital of Anhui Medical University, Anhui, 230601, China
| | - Sheng-di Chen
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Hui-Dong Tang
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Medical Center on Aging of Ruijin Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
19
|
Medicinal Herbs and Their Derived Ingredients Protect against Cognitive Decline in In Vivo Models of Alzheimer’s Disease. Int J Mol Sci 2022; 23:ijms231911311. [PMID: 36232612 PMCID: PMC9569503 DOI: 10.3390/ijms231911311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/24/2022] Open
Abstract
Alzheimer’s disease (AD) has pathological hallmarks including amyloid beta (Aβ) plaque formation. Currently approved single-target drugs cannot effectively ameliorate AD. Medicinal herbs and their derived ingredients (MHDIs) have multitarget and multichannel properties, engendering exceptional AD treatment outcomes. This review delineates how in in vivo models MHDIs suppress Aβ deposition by downregulating β- and γ-secretase activities; inhibit oxidative stress by enhancing the antioxidant activities and reducing lipid peroxidation; prevent tau hyperphosphorylation by upregulating protein phosphatase 2A expression and downregulating glycogen synthase kinase-3β expression; reduce inflammatory mediators partly by upregulating brain-derived neurotrophic factor/extracellular signal-regulated protein kinase 1/2-mediated signaling and downregulating p38 mitogen-activated protein kinase (p38 MAPK)/c-Jun N-terminal kinase (JNK)-mediated signaling; attenuate synaptic dysfunction by increasing presynaptic protein, postsynaptic protein, and acetylcholine levels and preventing acetylcholinesterase activity; and protect against neuronal apoptosis mainly by upregulating Akt/cyclic AMP response element-binding protein/B-cell lymphoma 2 (Bcl-2)-mediated anti-apoptotic signaling and downregulating p38 MAPK/JNK/Bcl-2-associated x protein (Bax)/caspase-3-, Bax/apoptosis-inducing factor-, C/EBP homologous protein/glucose-regulated protein 78-, and autophagy-mediated apoptotic signaling. Therefore, MHDIs listed in this review protect against Aβ-induced cognitive decline by inhibiting Aβ accumulation, oxidative stress, tau hyperphosphorylation, inflammation, synaptic damage, and neuronal apoptosis in the cortex and hippocampus during the early and late AD phases.
Collapse
|
20
|
Lee DH, Lee JY, Hong DY, Lee EC, Park SW, Jo YN, Park YJ, Cho JY, Cho YJ, Chae SH, Lee MR, Oh JS. ROCK and PDE-5 Inhibitors for the Treatment of Dementia: Literature Review and Meta-Analysis. Biomedicines 2022; 10:biomedicines10061348. [PMID: 35740369 PMCID: PMC9219677 DOI: 10.3390/biomedicines10061348] [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: 05/06/2022] [Revised: 06/02/2022] [Accepted: 06/05/2022] [Indexed: 12/14/2022] Open
Abstract
Dementia is a disease in which memory, thought, and behavior-related disorders progress gradually due to brain damage caused by injury or disease. It is mainly caused by Alzheimer’s disease or vascular dementia and several other risk factors, including genetic factors. It is difficult to treat as its incidence continues to increase worldwide. Many studies have been performed concerning the treatment of this condition. Rho-associated kinase (ROCK) and phosphodiesterase-5 (PDE-5) are attracting attention as pharmacological treatments to improve the symptoms. This review discusses how ROCK and PDE-5 affect Alzheimer’s disease, vascular restructuring, and exacerbation of neuroinflammation, and how their inhibition helps improve cognitive function. In addition, the results of the animal behavior analysis experiments utilizing the Morris water maze were compared through meta-analysis to analyze the effects of ROCK inhibitors and PDE-5 inhibitors on cognitive function. According to the selection criteria, 997 publications on ROCK and 1772 publications on PDE-5 were screened, and conclusions were drawn through meta-analysis. Both inhibitors showed good improvement in cognitive function tests, and what is expected of the synergy effect of the two drugs was confirmed in this review.
Collapse
Affiliation(s)
- Dong-Hun Lee
- Department of Neurosurgery, College of Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan 31151, Korea; (D.-H.L.); (J.Y.L.); (D.-Y.H.); (E.C.L.); (S.-W.P.)
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea
| | - Ji Young Lee
- Department of Neurosurgery, College of Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan 31151, Korea; (D.-H.L.); (J.Y.L.); (D.-Y.H.); (E.C.L.); (S.-W.P.)
| | - Dong-Yong Hong
- Department of Neurosurgery, College of Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan 31151, Korea; (D.-H.L.); (J.Y.L.); (D.-Y.H.); (E.C.L.); (S.-W.P.)
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea
| | - Eun Chae Lee
- Department of Neurosurgery, College of Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan 31151, Korea; (D.-H.L.); (J.Y.L.); (D.-Y.H.); (E.C.L.); (S.-W.P.)
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea
| | - Sang-Won Park
- Department of Neurosurgery, College of Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan 31151, Korea; (D.-H.L.); (J.Y.L.); (D.-Y.H.); (E.C.L.); (S.-W.P.)
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea
| | - Yu Na Jo
- Department of Medicine, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea; (Y.N.J.); (Y.J.P.); (J.Y.C.); (Y.J.C.); (S.H.C.)
| | - Yu Jin Park
- Department of Medicine, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea; (Y.N.J.); (Y.J.P.); (J.Y.C.); (Y.J.C.); (S.H.C.)
| | - Jae Young Cho
- Department of Medicine, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea; (Y.N.J.); (Y.J.P.); (J.Y.C.); (Y.J.C.); (S.H.C.)
| | - Yoo Jin Cho
- Department of Medicine, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea; (Y.N.J.); (Y.J.P.); (J.Y.C.); (Y.J.C.); (S.H.C.)
| | - Su Hyun Chae
- Department of Medicine, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea; (Y.N.J.); (Y.J.P.); (J.Y.C.); (Y.J.C.); (S.H.C.)
| | - Man Ryul Lee
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea
- Correspondence: (M.R.L.); (J.S.O.)
| | - Jae Sang Oh
- Department of Neurosurgery, College of Medicine, Soonchunhyang University, Cheonan Hospital, Cheonan 31151, Korea; (D.-H.L.); (J.Y.L.); (D.-Y.H.); (E.C.L.); (S.-W.P.)
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soon Chun Hyang University, Cheonan 31151, Korea
- Correspondence: (M.R.L.); (J.S.O.)
| |
Collapse
|
21
|
Pharmacological Activation of GPR55 Improved Cognitive Impairment Induced by Lipopolysaccharide in Mice. J Mol Neurosci 2022; 72:1656-1669. [PMID: 35596056 DOI: 10.1007/s12031-022-02020-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 05/01/2022] [Indexed: 10/18/2022]
Abstract
Our previous research found that activation of GPR55 can alleviate cognitive impairment induced by amyloid-beta 1-42 (Aβ1-42) and streptozotocin in mice, but the role of GPR55 in the pathogenesis of cognitive impairment remains unknown. Here, we used a lipopolysaccharide (LPS) mouse model to further investigate the role and mechanism of O-1602, a GPR55 agonist, on cognitive dysfunction. ICR mice were treated with an intracerebroventricular (i.c.v.) injection of LPS, followed by cognitive function tests. The expression of GPR55, NF-κB p65, caspase-3, Bax, and Bcl-2 in the hippocampus was examined by Western blotting. Inflammatory cytokines and microglia were detected by ELISA kit and immunohistochemical analyses, respectively. The levels of MDA, GSH, SOD, and CAT were examined by assay kits. Furthermore, TUNEL-staining was used to detect neuronal apoptosis. Our results showed that i.c.v. injection of LPS in mice exhibited impaired performance in the behavior tests, which were ameliorated by O-1602 treatment (2.0 or 4.0 μg/mouse, i.c.v.). Importantly, we found that O-1602 treatment reversed GPR55 downregulation, decreased the expression of NF-κB p65, suppressed the accumulation of proinflammatory cytokines and microglia activation, increased the anti-inflammatory cytokines, and reduced the levels of MDA, increased the levels of GSH, SOD, and CAT in the hippocampus. In addition, O-1602 treatment also significantly reduced Bax and increased Bcl-2 expression as well as decreased caspase-3 activity and TUNEL-positive cells in the hippocampus. These observations indicate that O-1602 may ameliorate LPS-induced cognition deficits via inhibiting neuroinflammation, oxidative stress, and apoptosis mediated by the NF-κB pathway in mice.
Collapse
|
22
|
Calvillo-Robledo A, Cervantes-Villagrana RD, Morales P, Marichal-Cancino BA. The oncogenic lysophosphatidylinositol (LPI)/GPR55 signaling. Life Sci 2022; 301:120596. [PMID: 35500681 DOI: 10.1016/j.lfs.2022.120596] [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: 02/26/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 10/18/2022]
Abstract
GPR55 is a class A orphan G protein-coupled receptor that has drawn important therapeutic attention in the last decade because of its role in pathophysiological processes including vascular functions, metabolic dysfunction, neurodegenerative disorders, or bone turnover among others. Several cannabinoids of phytogenic, endogenous, and synthetic nature have shown to modulate this receptor leading to propose it as a member of the endocannabinoid system. The putative endogenous GPR55 ligand is L-α-lysophosphatidylinositol (LPI) and it has been associated with several processes that control cell survival and tumor progression. The relevance of GPR55 in cancer is currently being extensively studied in vitro and in vivo using diverse cancer models. The LPI/GPR55 axis has been reported to participate in pro-oncogenic processes including cellular proliferation, differentiation, migration, invasion, and metastasis being altered in several cancer cells via G12/13 and Gq signaling. Moreover, GRP55 and its bioactive lipid have been proposed as potential biomarkers for cancer diagnosis. Indeed, GPR55 overexpression or high expression has been shown to correlate with cancer aggressiveness in specific tumors including acute myeloid leukemia, uveal melanoma, low grade glioma and renal cancer. This review aims to analyze and summarize current evidence on the cancerogenic role of the LPI/GPR55 axis providing a critical view of the therapeutic prospects of this promising target.
Collapse
Affiliation(s)
- Argelia Calvillo-Robledo
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Ciudad Universitaria, 20131 Aguascalientes, Ags., Mexico
| | | | - Paula Morales
- Instituto de Química Médica, CSIC, 28006 Madrid, Spain
| | - Bruno A Marichal-Cancino
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Ciudad Universitaria, 20131 Aguascalientes, Ags., Mexico.
| |
Collapse
|
23
|
Burgaz S, García C, Gonzalo-Consuegra C, Gómez-Almería M, Ruiz-Pino F, Unciti JD, Gómez-Cañas M, Alcalde J, Morales P, Jagerovic N, Rodríguez-Cueto C, de Lago E, Muñoz E, Fernández-Ruiz J. Preclinical Investigation in Neuroprotective Effects of the GPR55 Ligand VCE-006.1 in Experimental Models of Parkinson's Disease and Amyotrophic Lateral Sclerosis. Molecules 2021; 26:molecules26247643. [PMID: 34946726 PMCID: PMC8708356 DOI: 10.3390/molecules26247643] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 01/18/2023] Open
Abstract
Cannabinoids act as pleiotropic compounds exerting, among others, a broad-spectrum of neuroprotective effects. These effects have been investigated in the last years in different preclinical models of neurodegeneration, with the cannabinoid type-1 (CB1) and type-2 (CB2) receptors concentrating an important part of this research. However, the issue has also been extended to additional targets that are also active for cannabinoids, such as the orphan G-protein receptor 55 (GPR55). In the present study, we investigated the neuroprotective potential of VCE-006.1, a chromenopyrazole derivative with biased orthosteric and positive allosteric modulator activity at GPR55, in murine models of two neurodegenerative diseases. First, we proved that VCE-006.1 alone could induce ERK1/2 activation and calcium mobilization, as well as increase cAMP response but only in the presence of lysophosphatidyl inositol. Next, we investigated this compound administered chronically in two neurotoxin-based models of Parkinson's disease (PD), as well as in some cell-based models. VCE-006.1 was active in reversing the motor defects caused by 6-hydroxydopamine (6-OHDA) in the pole and the cylinder rearing tests, as well as the losses in tyrosine hydroxylase-containing neurons and the elevated glial reactivity detected in the substantia nigra. Similar cytoprotective effects were found in vitro in SH-SY5Y cells exposed to 6-OHDA. We also investigated VCE-006.1 in LPS-lesioned mice with similar beneficial effects, except against glial reactivity and associated inflammatory events, which remained unaltered, a fact confirmed in BV2 cells treated with LPS and VCE-006.1. We also analyzed GPR55 in these in vivo models with no changes in its gene expression, although GPR55 was down-regulated in BV2 cells treated with LPS, which may explain the lack of efficacy of VCE-006.1 in such an assay. Furthermore, we investigated VCE-006.1 in two genetic models of amyotrophic lateral sclerosis (ALS), mutant SOD1, or TDP-43 transgenic mice. Neither the neurological decline nor the deteriorated rotarod performance were prevented with this compound, and the same happened with the elevated microglial and astroglial reactivities, albeit modest spinal motor neuron preservation was achieved in both models. We also analyzed GPR55 in these in vivo models and found no changes in both TDP-43 transgenic and mSOD1 mice. Therefore, our findings support the view that targeting the GPR55 may afford neuroprotection in experimental PD, but not in ALS, thus stressing the specificities for the development of cannabinoid-based therapies in the different neurodegenerative disorders.
Collapse
Affiliation(s)
- Sonia Burgaz
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain; (S.B.); (C.G.); (C.G.-C.); (M.G.-A.); (M.G.-C.); (J.A.); (C.R.-C.); (E.d.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28040 Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28040 Madrid, Spain
| | - Concepción García
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain; (S.B.); (C.G.); (C.G.-C.); (M.G.-A.); (M.G.-C.); (J.A.); (C.R.-C.); (E.d.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28040 Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28040 Madrid, Spain
| | - Claudia Gonzalo-Consuegra
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain; (S.B.); (C.G.); (C.G.-C.); (M.G.-A.); (M.G.-C.); (J.A.); (C.R.-C.); (E.d.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28040 Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28040 Madrid, Spain
| | - Marta Gómez-Almería
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain; (S.B.); (C.G.); (C.G.-C.); (M.G.-A.); (M.G.-C.); (J.A.); (C.R.-C.); (E.d.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28040 Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28040 Madrid, Spain
| | - Francisco Ruiz-Pino
- Emerald Health Biotechnology España, 14014 Córdoba, Spain; (F.R.-P.); (J.D.U.); (E.M.)
| | - Juan Diego Unciti
- Emerald Health Biotechnology España, 14014 Córdoba, Spain; (F.R.-P.); (J.D.U.); (E.M.)
| | - María Gómez-Cañas
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain; (S.B.); (C.G.); (C.G.-C.); (M.G.-A.); (M.G.-C.); (J.A.); (C.R.-C.); (E.d.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28040 Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28040 Madrid, Spain
| | - Juan Alcalde
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain; (S.B.); (C.G.); (C.G.-C.); (M.G.-A.); (M.G.-C.); (J.A.); (C.R.-C.); (E.d.L.)
| | - Paula Morales
- Instituto de Química Médica, CSIC, 28006 Madrid, Spain; (P.M.); (N.J.)
| | - Nadine Jagerovic
- Instituto de Química Médica, CSIC, 28006 Madrid, Spain; (P.M.); (N.J.)
| | - Carmen Rodríguez-Cueto
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain; (S.B.); (C.G.); (C.G.-C.); (M.G.-A.); (M.G.-C.); (J.A.); (C.R.-C.); (E.d.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28040 Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28040 Madrid, Spain
| | - Eva de Lago
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain; (S.B.); (C.G.); (C.G.-C.); (M.G.-A.); (M.G.-C.); (J.A.); (C.R.-C.); (E.d.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28040 Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28040 Madrid, Spain
| | - Eduardo Muñoz
- Emerald Health Biotechnology España, 14014 Córdoba, Spain; (F.R.-P.); (J.D.U.); (E.M.)
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), 14004 Córdoba, Spain
- Department of Cellular Biology, Physiology and Immunology, University of Córdoba, 14071 Córdoba, Spain
- Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
| | - Javier Fernández-Ruiz
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, 28040 Madrid, Spain; (S.B.); (C.G.); (C.G.-C.); (M.G.-A.); (M.G.-C.); (J.A.); (C.R.-C.); (E.d.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28040 Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28040 Madrid, Spain
- Correspondence: ; Tel.: +34–913941450
| |
Collapse
|