1
|
Xu TX, Jiang HY, Yang ZY. Renin-angiotensin system inhibitor use and risk of Parkinson's disease: a meta-analysis. Acta Neurol Belg 2025; 125:53-60. [PMID: 38669003 PMCID: PMC11876239 DOI: 10.1007/s13760-024-02560-7] [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: 12/26/2023] [Accepted: 04/07/2024] [Indexed: 03/05/2025]
Abstract
BACKGROUND Hypertension is a recognized risk factor for Parkinson's disease (PD). The renin-angiotensin system (RAS) inhibitors are widely used to treat hypertension. However, the association of RAS inhibitor use with PD has still been an area of controversy. METHODS Thus, we conducted a meta-analysis to investigate the relationship between RAS inhibitor use and PD. PUBMED and EMBASE databases were searched for articles published up to Oct 2023. All studies that examined the relationship between RAS inhibitor use and the incidence of PD were included. RESULTS Seven studies with total 3,495,218 individuals met our inclusion criteria for this meta-analysis. Overall, RAS inhibitor use was associated with a reduction in PD risk (OR = 0.88, 95%CI = 0.79-0.98) compared with the controls. When restricted the analysis to individuals with RAS inhibitor use indication, RAS inhibitor exposure was also associated with a decreased risk of PD (OR = 0.76, 95%CI = 0.62-0.92). Pooled results of cohort studies also did support a protective role of angiotensin converting enzyme inhibitors (ACEIs) (OR = 0.97, 95%CI = 0.89-1.07) users and angiotensin II receptor blockers (ARBs) (OR = 0.8, 95%CI = 0.63-1.02) in PD. CONCLUSION Overall, RAS inhibitor use as a class is associated with a reduction in PD risk. However, the findings of ACEIs and ARBs may be limited by small sample size. Future well-designed studies considering the classification by inhibitor type, duration, dose, or property of BBB penetration of RAS inhibitors are needed to clarify the contribution of these exposure parameters on the risk of PD.
Collapse
Affiliation(s)
- Tian-Xiang Xu
- Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Hai-Yin Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zeng-Yan Yang
- Guangxi University of Chinese Medicine, Nanning, Guangxi, China.
| |
Collapse
|
2
|
Tan S, Chi H, Wang P, Zhao R, Zhang Q, Gao Z, Xue H, Tang Q, Li G. Protein tyrosine phosphatase receptor type O serves as a key regulator of insulin resistance-induced α-synuclein aggregation in Parkinson's disease. Cell Mol Life Sci 2024; 81:403. [PMID: 39276174 PMCID: PMC11401831 DOI: 10.1007/s00018-024-05436-4] [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: 04/04/2024] [Revised: 08/05/2024] [Accepted: 09/02/2024] [Indexed: 09/16/2024]
Abstract
Insulin resistance (IR) was found to be a critical element in the pathogenesis of Parkinson's disease (PD), facilitating abnormal α-synuclein (α-Syn) aggregation in neurons and thus promoting PD development. However, how IR contributes to abnormal α-Syn aggregation remains ill-defined. Here, we analyzed six PD postmortem brain transcriptome datasets to reveal module genes implicated in IR-mediated α-Syn aggregation. In addition, we induced IR in cultured dopaminergic (DA) neurons overexpressing α-Syn to identify IR-modulated differentially expressed genes (DEGs). Integrated analysis of data from PD patients and cultured neurons revealed 226 genes involved in α-Syn aggregation under IR conditions, of which 53 exhibited differential expression between PD patients and controls. Subsequently, we conducted an integrated analysis of the 53 IR-modulated genes employing transcriptome data from PD patients with different Braak stages and DA neuron subclasses with varying α-Syn aggregation scores. Protein tyrosine phosphatase receptor type O (PTPRO) was identified to be closely associated with PD progression and α-Syn aggregation. Experimental validation in a cultured PD cell model confirmed that both mRNA and protein of PTPRO were reduced under IR conditions, and the downregulation of PTPRO significantly facilitated α-Syn aggregation and cell death. Collectively, our findings identified PTPRO as a key regulator in IR-mediated α-Syn aggregation and uncovered its prospective utility as a therapeutic target in PD patients with IR.
Collapse
Affiliation(s)
- Shichuan Tan
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, 250012, China
- Department of Emergency Neurosurgical Intensive Care Unit, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, China
| | - Huizhong Chi
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, 250012, China
| | - Pin Wang
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission (NHC) Key Laboratory of Otorhinolaryngology, Shandong University, Jinan, 250012, Shandong, China
| | - Rongrong Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, 250012, China
| | - Qinran Zhang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, China
- Department of Epidemiology and Health Statistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zijie Gao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, 250012, China
| | - Hao Xue
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, China
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, 250012, China
| | - Qilin Tang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, China.
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, 250012, China.
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, China.
- Shandong Key Laboratory of Brain Health and Function Remodeling, Jinan, 250012, China.
| |
Collapse
|
3
|
Liu T, Li J, Sun L, Zhu C, Wei J. The role of ACE2 in RAS axis on microglia activation in Parkinson's disease. Neuroscience 2024; 553:128-144. [PMID: 38986737 DOI: 10.1016/j.neuroscience.2024.06.024] [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: 03/26/2024] [Revised: 06/19/2024] [Accepted: 06/22/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND The classic renin-angiotensin system (RAS) induces organ damage, while the ACE2/Ang-(1-7)/MasR axis opposes it. However, the role of ACE2 in the brain is unclear. We studied ACE2's role in the brain. METHOD We used male C57BL/6J (WT) mice, ACE2 knockout (KO) mice, and MPTP-induced mice. Behavioral tests confirmed successful modeling. We assessed the impact of ACE2 KO on the RAS axis and PD index, including ACE, ACE2, AT1, AT2, MasR, TH, α-syn, and Iba1. We investigated ACE2 and MasR's involvement in microglial activation via western blot and immunofluorescence. GSE10867 and GSE26532 datasets were used to analyze the effects of AT1 antagonists and in vitro PD models on microglia. RESULT Behavioral tests revealed that MPTP mice displayed motor deficits, depression, anxiety, and increased inflammatory markers in the SN and CPU, with reduced antioxidant capacity. ACE2 KO worsened these symptoms and exacerbated inflammation and oxidative stress. LPS-induced ACE2/MasR activation in BV2 cells demonstrated anti-inflammatory and neuroprotective effects by modulating microglial polarization. Antagonists inhibited microglial activation via inflammation and ROS processes. CONCLUSION The RAS axis regulates inflammation and oxidative stress to maintain CNS function, suggesting potential targets for neurologic disease treatment. Understanding microglial RAS activation can offer new therapeutic strategies.
Collapse
Affiliation(s)
- Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China; Institute of Neurourology and Urodynamics, Huaihe Hospital of Henan University, Kaifeng 475000, China
| | - Jingwen Li
- Institute of Neurourology and Urodynamics, Huaihe Hospital of Henan University, Kaifeng 475000, China
| | - Lin Sun
- College of Chemistry and Molecular Sciences, Henan University, Kaifeng 475000, China.
| | - Chaoyang Zhu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Jianshe Wei
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China; Institute of Neurourology and Urodynamics, Huaihe Hospital of Henan University, Kaifeng 475000, China.
| |
Collapse
|
4
|
Park HY, Lee GS, Go J, Ryu YK, Lee CH, Moon JH, Kim KS. Angiotensin-converting enzyme inhibition prevents l-dopa-induced dyskinesia in a 6-ohda-induced mouse model of Parkinson's disease. Eur J Pharmacol 2024; 973:176573. [PMID: 38642669 DOI: 10.1016/j.ejphar.2024.176573] [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: 10/11/2023] [Revised: 03/11/2024] [Accepted: 04/09/2024] [Indexed: 04/22/2024]
Abstract
Parkinson's disease (PD) is characterised by severe movement defects and the degeneration of dopaminergic neurones in the midbrain. The symptoms of PD can be managed with dopamine replacement therapy using L-3, 4-dihydroxyphenylalanine (L-dopa), which is the gold standard therapy for PD. However, long-term treatment with L-dopa can lead to motor complications. The central renin-angiotensin system (RAS) is associated with the development of neurodegenerative diseases in the brain. However, the role of the RAS in dopamine replacement therapy for PD remains unclear. Here, we tested the co-treatment of the angiotensin-converting enzyme inhibitor (ACEI) with L-dopa altered L-dopa-induced dyskinesia (LID) in a 6-hydroxydopamine (6-OHDA)-lesioned mouse model of PD. Perindopril, captopril, and enalapril were used as ACEIs. The co-treatment of ACEI with L-dopa significantly decreased LID development in 6-OHDA-lesioned mice. In addition, the astrocyte and microglial transcripts involving Ccl2, C3, Cd44, and Iigp1 were reduced by co-treatment with ACEI and L-dopa in the 6-OHDA-lesioned striatum. In conclusion, co-treatment with ACEIs and L-dopa, such as perindopril, captopril, and enalapril, may mitigate the severity of L-DOPA-induced dyskinesia in a mouse model of PD.
Collapse
Affiliation(s)
- Hye-Yeon Park
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Ga Seul Lee
- Core Research Facility & Analysis Center, KRIBB, Daejeon 34141, Republic of Korea; College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - Jun Go
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Young-Kyoung Ryu
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea; KRIBB School, University of Science and Technology, Daejeon 34141, Republic of Korea
| | - Jeong Hee Moon
- Core Research Facility & Analysis Center, KRIBB, Daejeon 34141, Republic of Korea.
| | - Kyoung-Shim Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea; KRIBB School, University of Science and Technology, Daejeon 34141, Republic of Korea.
| |
Collapse
|
5
|
Schrag A, Kostev K. Association of antihypertensives and Parkinson's disease in a primary care population matched for underlying diagnosis. PLoS One 2024; 19:e0299985. [PMID: 38507360 PMCID: PMC10954140 DOI: 10.1371/journal.pone.0299985] [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: 10/23/2023] [Accepted: 02/19/2024] [Indexed: 03/22/2024] Open
Abstract
PURPOSE To examine the association of several antihypertensive medication classes with incidence of Parkinson's disease (PD), taking into account possible underlying conditions. METHODS In a case-control study based on a large primary care database and including 21,981 PD cases and 21,981 non-PD controls matched for age, sex, and possible treatment indications associations with different antihypertensive medication groups, including diuretics, betablockers, calcium channel blockers, angiotensin-converting enzyme inhibitors and angiotensin-II receptor-blockers and PD were examined. RESULTS Antihypertensive medications overall were associated with a lower risk of subsequent diagnosis of PD (OR: 0.94, 95% CI 0.90-0.97), with the negative association most significant for medications acting on the renin-angiotensin-aldosterone system. A positive association with diagnosis of PD was only seen for betablockers and restricted to those with relatively young age and not in those with longer treatment duration. CONCLUSION When taking into account underlying diagnoses, antihypertensive medications overall were associated with a reduced incidence of PD.
Collapse
Affiliation(s)
- Anette Schrag
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Karel Kostev
- University Clinic, Philipps-University, Marburg, Germany
- Epidemiology, IQVIA, Frankfurt, Germany
| |
Collapse
|
6
|
el Bouhaddani S, Höllerhage M, Uh HW, Moebius C, Bickle M, Höglinger G, Houwing-Duistermaat J. Statistical integration of multi-omics and drug screening data from cell lines. PLoS Comput Biol 2024; 20:e1011809. [PMID: 38295113 PMCID: PMC10878536 DOI: 10.1371/journal.pcbi.1011809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 02/20/2024] [Accepted: 01/08/2024] [Indexed: 02/02/2024] Open
Abstract
Data integration methods are used to obtain a unified summary of multiple datasets. For multi-modal data, we propose a computational workflow to jointly analyze datasets from cell lines. The workflow comprises a novel probabilistic data integration method, named POPLS-DA, for multi-omics data. The workflow is motivated by a study on synucleinopathies where transcriptomics, proteomics, and drug screening data are measured in affected LUHMES cell lines and controls. The aim is to highlight potentially druggable pathways and genes involved in synucleinopathies. First, POPLS-DA is used to prioritize genes and proteins that best distinguish cases and controls. For these genes, an integrated interaction network is constructed where the drug screen data is incorporated to highlight druggable genes and pathways in the network. Finally, functional enrichment analyses are performed to identify clusters of synaptic and lysosome-related genes and proteins targeted by the protective drugs. POPLS-DA is compared to other single- and multi-omics approaches. We found that HSPA5, a member of the heat shock protein 70 family, was one of the most targeted genes by the validated drugs, in particular by AT1-blockers. HSPA5 and AT1-blockers have been previously linked to α-synuclein pathology and Parkinson's disease, showing the relevance of our findings. Our computational workflow identified new directions for therapeutic targets for synucleinopathies. POPLS-DA provided a larger interpretable gene set than other single- and multi-omic approaches. An implementation based on R and markdown is freely available online.
Collapse
Affiliation(s)
| | | | - Hae-Won Uh
- Dept. Data science & Biostatistics, UMC Utrecht, Utrecht, Netherlands
| | - Claudia Moebius
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Marc Bickle
- Roche Institute for Translational Bioengineering, Basel, Switzerland
| | - Günter Höglinger
- Department of Neurology, Hannover Medical School, Hannover, Germany
- Department of Neurology, Ludwig-Maximilians-Universität, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Jeanine Houwing-Duistermaat
- Dept. Data science & Biostatistics, UMC Utrecht, Utrecht, Netherlands
- Dept. of Mathematics, Radboud University, Nijmegen, Netherlands
| |
Collapse
|
7
|
Sarpolaki MK, Vafaei A, Fattahi MR, Iranmehr A. Mini-Review: Role of Drugs Affecting Renin-Angiotensin System (RAS) in Traumatic Brain Injury (TBI): What We Know and What We Should Know. Korean J Neurotrauma 2023; 19:195-203. [PMID: 37431373 PMCID: PMC10329892 DOI: 10.13004/kjnt.2023.19.e26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 05/23/2023] [Accepted: 06/12/2023] [Indexed: 07/12/2023] Open
Abstract
Traumatic brain injuries (TBIs) are among the most important clinical and research areas in neurosurgery, owing to their devastating effects and high prevalence. Over the last few decades, there has been increasing research on the complex pathophysiology of TBI and secondary injuries following TBI. A growing body of evidence has shown that the renin-angiotensin system (RAS), a well-known cardiovascular regulatory pathway, plays a role in TBI pathophysiology. Acknowledging these complex and poorly understood pathways and their role in TBI could help design new clinical trials involving drugs that alter the RAS network, most notably angiotensin receptor blockers and angiotensin-converting enzyme inhibitors. This study aimed to briefly review the molecular, animal, and human studies on these drugs in TBI and provide a clear vision for researchers to fill knowledge gaps in the future.
Collapse
Affiliation(s)
- Mohammad Kazem Sarpolaki
- Neurological Surgery Department, Imam Khomeini Hospital Complex (IKHC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Ali Vafaei
- Experimental Medicine Research Center, Department of Pharmacology, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Reza Fattahi
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Arad Iranmehr
- Neurological Surgery Department, Imam Khomeini Hospital Complex (IKHC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
8
|
Rivas-Santisteban R, Lillo J, Raïch I, Muñoz A, Lillo A, Rodríguez-Pérez AI, Labandeira-García JL, Navarro G, Franco R. The cannabinoid CB 1 receptor interacts with the angiotensin AT 2 receptor. Overexpression of AT 2-CB 1 receptor heteromers in the striatum of 6-hydroxydopamine hemilesioned rats. Exp Neurol 2023; 362:114319. [PMID: 36632949 DOI: 10.1016/j.expneurol.2023.114319] [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: 10/02/2022] [Revised: 12/14/2022] [Accepted: 01/06/2023] [Indexed: 01/11/2023]
Abstract
It is of particular interest the potential of cannabinoid and angiotensin receptors as targets in the therapy of Parkinson's disease (PD). While endocannabinoids are neuromodulators that act through the CB1 and CB2 cannabinoid receptors, the renin angiotensin-system is relevant for regulation of the correct functioning of several brain circuits. Resonance energy transfer assays in a heterologous system showed that the CB1 receptor (CB1R) can directly interact with the angiotensin AT2 receptor (AT2R). Coactivation of the two receptors results in increased Gi-signaling. The AT2-CB1 receptor heteromer imprint consists of a blockade of AT2R-mediated signaling by rimonabant, a CB1R antagonist. Interestingly, the heteromer imprint, discovered in the heterologous system, was also found in primary striatal neurons thus demonstrating the expression of the heteromer in these cells. In situ proximity ligation assays confirmed the occurrence of AT2-CB1 receptor heteromers in striatal neurons. In addition, increased expression of the AT2-CB1 receptor heteromeric complexes was detected in the striatum of a rodent PD model consisting of rats hemilesioned using 6-hydroxydopamine. Expression of the heteromer was upregulated in the striatum of lesioned animals and, also, of lesioned animals that upon levodopa treatment became dyskinetic. In contrast, there was no upregulation in the striatum of lesioned rats that did not become dyskinetic upon chronic levodopa treatment. The results suggest that therapeutic developments focused on the CB1R should consider that this receptor can interact with the AT2R, which in the CNS is involved in mechanisms related to addictive behaviors and to neurodegenerative and neuroinflammatory diseases.
Collapse
Affiliation(s)
- Rafael Rivas-Santisteban
- Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain; CiberNed. Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
| | - Jaume Lillo
- Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain; CiberNed. Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
| | - Iu Raïch
- Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain; CiberNed. Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
| | - Ana Muñoz
- CiberNed. Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain; Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Alejandro Lillo
- Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain; CiberNed. Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
| | - Ana I Rodríguez-Pérez
- CiberNed. Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain; Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - José L Labandeira-García
- CiberNed. Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain; Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Gemma Navarro
- Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain; CiberNed. Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain; Neurosciences Institute, University of Barcelona (NeuroUB), Facultad de Psicología Campus de Mundet Paseo de la Vall d'Hebron, 171 08035 Barcelona, Spain.
| | - Rafael Franco
- CiberNed. Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain; Molecular Neurobiology laboratory, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain; School of Chemistry, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
9
|
Kulisevsky J, Martínez-Horta S, Campolongo A, Pascual-Sedano B, Marín-Lahoz J, Bejr-Kasem H, Labandeira-Garcia JL, Lanciego JL, Puig-Davi A, Horta-Barba A, Pagonabarraga J, Rodríguez-Antigüedad J. A randomized clinical trial of candesartan for cognitive impairment in Parkinson's disease. Parkinsonism Relat Disord 2023; 110:105367. [PMID: 36963339 DOI: 10.1016/j.parkreldis.2023.105367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/20/2023] [Accepted: 03/12/2023] [Indexed: 03/26/2023]
Affiliation(s)
- Jaime Kulisevsky
- Movement Disorders Unit, Neurology Department, Sant Pau Hospital, Barcelona, Spain; Universitat Autònoma de Barcelona (UAB), Medicine Department, Barcelona, Spain; Institut d'Investigacions Biomèdiques-Sant Pau (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Universitat Oberta de Catalunya (UOC), Spain.
| | - Saul Martínez-Horta
- Movement Disorders Unit, Neurology Department, Sant Pau Hospital, Barcelona, Spain; Universitat Autònoma de Barcelona (UAB), Medicine Department, Barcelona, Spain; Institut d'Investigacions Biomèdiques-Sant Pau (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Antonia Campolongo
- Movement Disorders Unit, Neurology Department, Sant Pau Hospital, Barcelona, Spain; Universitat Autònoma de Barcelona (UAB), Medicine Department, Barcelona, Spain; Institut d'Investigacions Biomèdiques-Sant Pau (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Universitat Oberta de Catalunya (UOC), Spain
| | - Berta Pascual-Sedano
- Movement Disorders Unit, Neurology Department, Sant Pau Hospital, Barcelona, Spain; Universitat Autònoma de Barcelona (UAB), Medicine Department, Barcelona, Spain; Institut d'Investigacions Biomèdiques-Sant Pau (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Universitat Oberta de Catalunya (UOC), Spain
| | - Juan Marín-Lahoz
- Movement Disorders Unit, Neurology Department, Sant Pau Hospital, Barcelona, Spain; Universitat Autònoma de Barcelona (UAB), Medicine Department, Barcelona, Spain; Institut d'Investigacions Biomèdiques-Sant Pau (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Helena Bejr-Kasem
- Movement Disorders Unit, Neurology Department, Sant Pau Hospital, Barcelona, Spain; Universitat Autònoma de Barcelona (UAB), Medicine Department, Barcelona, Spain; Institut d'Investigacions Biomèdiques-Sant Pau (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jose L Labandeira-Garcia
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas, Faculty of Medicine, University of Santiago de Compostela, Spain
| | - Jose L Lanciego
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Neurosciences Division, Centro de Investigación Médica Aplicada (CIMA), University of Navarra, Pamplona, Spain
| | - Arnau Puig-Davi
- Movement Disorders Unit, Neurology Department, Sant Pau Hospital, Barcelona, Spain; Universitat Autònoma de Barcelona (UAB), Medicine Department, Barcelona, Spain; Institut d'Investigacions Biomèdiques-Sant Pau (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Andrea Horta-Barba
- Movement Disorders Unit, Neurology Department, Sant Pau Hospital, Barcelona, Spain; Universitat Autònoma de Barcelona (UAB), Medicine Department, Barcelona, Spain; Institut d'Investigacions Biomèdiques-Sant Pau (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Javier Pagonabarraga
- Movement Disorders Unit, Neurology Department, Sant Pau Hospital, Barcelona, Spain; Universitat Autònoma de Barcelona (UAB), Medicine Department, Barcelona, Spain; Institut d'Investigacions Biomèdiques-Sant Pau (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jon Rodríguez-Antigüedad
- Movement Disorders Unit, Neurology Department, Sant Pau Hospital, Barcelona, Spain; Universitat Autònoma de Barcelona (UAB), Medicine Department, Barcelona, Spain; Institut d'Investigacions Biomèdiques-Sant Pau (IIB-Sant Pau), Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
10
|
Ahmad MH, Rizvi MA, Ali M, Mondal AC. Neurobiology of depression in Parkinson's disease: Insights into epidemiology, molecular mechanisms and treatment strategies. Ageing Res Rev 2023; 85:101840. [PMID: 36603690 DOI: 10.1016/j.arr.2022.101840] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 12/25/2022] [Accepted: 12/31/2022] [Indexed: 01/03/2023]
Abstract
Parkinson's disease (PD) is characterized mainly by motor dysfunctions due to the progressive loss of dopaminergic neurons. However, PD patients experience a multitude of debilitating non-motor symptoms, including depression, which may have deleteriously detrimental effects on life. Depression is multifactorial and exhibits a bimodal progression in PD, but its underlying molecular mechanisms are poorly understood. Studies demonstrating the pathophysiology of depression in PD and the specific treatment strategies for depression-like symptoms in PD patients are largely lacking, often underrated, under-recognized and, consequently, inadequately/under-treated. Nevertheless, reports suggest that the incidence of depression is approximately 20-30% of PD patients and may precede the onset of motor symptoms. Diagnosing depression in PD becomes difficult due to the clinical overlap in symptomatology between the two diseases, and the nigrostriatal dysfunction alone is insufficient to explain depressive symptoms in PD. Therefore, the current study provides an overview of the molecular mechanisms underlying the development of depression in PD and new insights into developing current antidepressant strategies to treat depression in PD. This review will identify and understand the molecular pathological mechanisms of depression in PD that will fundamentally help tailoring therapeutic interventions for depressive symptoms in PD.
Collapse
Affiliation(s)
- Mir Hilal Ahmad
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India; Genome Biology Lab, Department of Biosciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Moshahid Alam Rizvi
- Genome Biology Lab, Department of Biosciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Mansoor Ali
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Amal Chandra Mondal
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
11
|
Franco R, Serrano-Marín J. Can chronic therapeutic drug use by the elderly affect Alzheimer’s disease risk and rate of progression? EXPLORATION OF NEUROPROTECTIVE THERAPY 2023; 3:8-23. [DOI: 10.37349/ent.2023.00035] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/31/2023] [Indexed: 01/03/2025]
Abstract
There is no approved drug capable of halting the progression of the most prevalent neurodegenerative disorders, namely Alzheimer’s disease (AD) and Parkinson’s disease (PD). Current therapeutic strategies focus mainly on the inhibition of the formation of protein aggregates and their deposition in the central nervous system. However, after almost a hundred years, proper management of the disease is still lacking. The fact of not finding effective management tools in the various clinical trials already carried out suggests that new hypotheses and strategies should be explored. Although vast resources have been allocated to the investigation of protein aggregates and the pathophysiology is now better understood, clues to the actual etiology are lacking. It is well known that brain homeostasis is of paramount importance for the survival of neurons. Drugs that target the periphery are often not subject to evaluation for their potential effect on the central nervous system. While acute treatments may be irrelevant, pills used for chronic conditions can be detrimental to neurons, especially in terms of progressive damage leading to a long-term decline in neuronal survival. Due to the lack of advances in the search for a curative treatment for neurodegenerative diseases, and the lack of new hypotheses about their etiology, a novel hypothesis is here proposed. It consists of assuming that the effects of the drugs most commonly used by the elderly, such as antihypertensive, hypoglycemic, and hypocholesterolemic, could have a negative impact on neuronal survival.
Collapse
Affiliation(s)
- Rafael Franco
- CiberNed, Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, 28029 Madrid, Spain; Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain; School of Chemistry, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Joan Serrano-Marín
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
12
|
Yang J, Gao Y, Duan Q, Qiu Y, Feng S, Zhan C, Huang Y, Zhang Y, Ma G, Nie K, Wang L. Renin-angiotensin system blockers affect cognitive decline in Parkinson's disease: The PPMI dataset. Parkinsonism Relat Disord 2022; 105:90-95. [PMID: 36395543 DOI: 10.1016/j.parkreldis.2022.10.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/13/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE To explore the potential clinical effects of renin-angiotensin system blocker (RASB, angiotensin II receptor blockers (ARBs) and angiotensin-converting enzyme inhibitors (ACEIs)) in patients from the Parkinson's Progress Marker Initiative (PPMI) study database. METHODS One hundred and seven untreated, newly diagnosed PD patients with hypertension, from the PPMI were included. We measured cognitive performance, biomarkers in CSF, and magnetic resonance imaging (MRI) during the five follow-up years for patients exposed or not to renal-angiotensin system blockers. Sixteen PD patients with hypertension underwent [18F]florbetaben positron emission tomography (PET) scanning. SUVRs of region of interest (ROI) were calculated and compared within different groups. RESULT Treatment with ARBs but not ACEIs improved global cognitive function evaluated by MoCA score in PD patients with hypertension compared to other hypertensive medicines up to 5 years follow up. Specifically, ARBs improved visuospatial, memory, executive abilities, processing speed attention test scores in PD. There was no significant impact of ARBs on α-syn, tau, Aβ in CSF. RASBs reduced [18F] florbetaben uptake in cortex and subcortex nuclei in the brain. CONCLUSIONS These results show potential protective effect with ARBs in cognitive impairment of parkinson's disease with hypertension.
Collapse
Affiliation(s)
- Jianhua Yang
- The Second School of Clinical Medicine, Southern Medical University, No.1023, South Shatai Road, Baiyun District, Guangzhou, 510515, China; Department of Neurology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Neuroscience Institute, No.106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, China; Department of Neurology, Ganzhou People's Hospital, Ganzhou, 341000, China
| | - Yuyuan Gao
- Department of Neurology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Neuroscience Institute, No.106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, China
| | - Qingrui Duan
- Department of Neurology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Neuroscience Institute, No.106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, China
| | - Yihui Qiu
- Department of Neurology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Neuroscience Institute, No.106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, China
| | - Shujun Feng
- Department of Neurology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Neuroscience Institute, No.106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, China
| | - Cuijing Zhan
- The Second School of Clinical Medicine, Southern Medical University, No.1023, South Shatai Road, Baiyun District, Guangzhou, 510515, China; Department of Neurology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Neuroscience Institute, No.106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, China
| | - Yin Huang
- The Second School of Clinical Medicine, Southern Medical University, No.1023, South Shatai Road, Baiyun District, Guangzhou, 510515, China; Department of Neurology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Neuroscience Institute, No.106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, China
| | - Yuhu Zhang
- Department of Neurology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Neuroscience Institute, No.106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, China
| | - Guixian Ma
- Department of Neurology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Neuroscience Institute, No.106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, China
| | - Kun Nie
- Department of Neurology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Neuroscience Institute, No.106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, China.
| | - Lijuan Wang
- The Second School of Clinical Medicine, Southern Medical University, No.1023, South Shatai Road, Baiyun District, Guangzhou, 510515, China; Department of Neurology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Neuroscience Institute, No.106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, China.
| |
Collapse
|
13
|
Zhang L, Li Q. Neuroprotective effects of tanshinone IIA in experimental model of Parkinson disease in rats. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.104269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
14
|
Caniceiro AB, Bueschbell B, Schiedel AC, Moreira IS. Class A and C GPCR Dimers in Neurodegenerative Diseases. Curr Neuropharmacol 2022; 20:2081-2141. [PMID: 35339177 PMCID: PMC9886835 DOI: 10.2174/1570159x20666220327221830] [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: 09/14/2021] [Revised: 02/21/2022] [Accepted: 03/23/2022] [Indexed: 11/22/2022] Open
Abstract
Neurodegenerative diseases affect over 30 million people worldwide with an ascending trend. Most individuals suffering from these irreversible brain damages belong to the elderly population, with onset between 50 and 60 years. Although the pathophysiology of such diseases is partially known, it remains unclear upon which point a disease turns degenerative. Moreover, current therapeutics can treat some of the symptoms but often have severe side effects and become less effective in long-term treatment. For many neurodegenerative diseases, the involvement of G proteincoupled receptors (GPCRs), which are key players of neuronal transmission and plasticity, has become clearer and holds great promise in elucidating their biological mechanism. With this review, we introduce and summarize class A and class C GPCRs, known to form heterodimers or oligomers to increase their signalling repertoire. Additionally, the examples discussed here were shown to display relevant alterations in brain signalling and had already been associated with the pathophysiology of certain neurodegenerative diseases. Lastly, we classified the heterodimers into two categories of crosstalk, positive or negative, for which there is known evidence.
Collapse
Affiliation(s)
- Ana B. Caniceiro
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; ,These authors contributed equally to this work.
| | - Beatriz Bueschbell
- PhD Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão, 3030-789 Coimbra, Portugal; ,These authors contributed equally to this work.
| | - Anke C. Schiedel
- Department of Pharmaceutical & Medicinal Chemistry, Pharmaceutical Institute, University of Bonn, D-53121 Bonn, Germany;
| | - Irina S. Moreira
- University of Coimbra, Department of Life Sciences, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal; ,Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, 3004-504 Coimbra, Portugal,Address correspondence to this author at the Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, 3004-504 Coimbra, Portugal; E-mail:
| |
Collapse
|
15
|
Onaolapo AY, Ojo FO, Olofinnade AT, Falade J, Lawal IA, Onaolapo OJ. Microbiome-Based Therapies in Parkinson's Disease: Can Tuning the Microbiota Become a Viable Therapeutic Strategy? CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 22:CNSNDDT-EPUB-126136. [PMID: 36056826 DOI: 10.2174/1871527321666220903114559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/20/2022] [Accepted: 07/16/2022] [Indexed: 06/15/2023]
Abstract
Progressive neurodegenerative disorders such as Parkinson's disease (PD) have continued to baffle medical science, despite strides in the understanding of their pathology. The inability of currently available therapies to halt disease progression is a testament to an incomplete understanding of pathways crucial to disease initiation, progression and management. Science has continued to link the activities and equilibrium of the gut microbiome to the health and proper functioning of brain neurons. They also continue to stir interest in the potential applications of technologies that may shift the balance of the gut microbiome towards achieving a favourable outcome in PD management. There have been suggestions that an improved understanding of the roles of the gut microbiota is likely to lead to the emergence of an era where their manipulation becomes a recognized strategy for PD management. This review examines the current state of our journey in the quest to understand how the gut microbiota can influence several aspects of PD. We highlight the relationship between the gut microbiome/microbiota and PD pathogenesis, as well as preclinical and clinical evidence evaluating the effect of postbiotics, probiotics and prebiotics in PD management. This is with a view to ascertaining if we are at the threshold of discovering the application of a usable tool in our quest for disease modifying therapies in PD.
Collapse
Affiliation(s)
- Adejoke Y Onaolapo
- Behavioural Neuroscience/Neurobiology Unit, Department of Anatomy, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Folusho O Ojo
- Department of Anatomy, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Anthony T Olofinnade
- Department of Pharmacology, Therapeutics and Toxicology, Faculty of Basic Clinical Sciences, College of Medicine, Lagos State University, Lagos State
| | - Joshua Falade
- Department of Mental Health, Afe-Babalola University Ado-Ekiti Ekiti State Nigeria
| | - Ismail A Lawal
- Department of Anatomy, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
- Department of Anatomy, Faculty of Health Sciences. Alhikmah University Ilorin, Kwara State, Nigeria
| | - Olakunle J Onaolapo
- Behavioural Neuroscience/Neuropharmacology Unit, Department of Pharmacology and Therapeutics, Faculty of Basic Clinical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| |
Collapse
|
16
|
Association of Angiotensin Receptor Blockers with Incident Parkinson Disease in Patients with Hypertension: A Retrospective Cohort Study. Am J Med 2022; 135:1001-1007. [PMID: 35580718 DOI: 10.1016/j.amjmed.2022.04.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 04/18/2022] [Accepted: 04/27/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Angiotensin receptor blockers (ARBs), which are commonly used antihypertensives, have been proposed to lower the risk of Parkinson disease by reducing oxidative stress based on animal and in vitro studies. Thus, this study aimed to test this association in patients with newly diagnosed hypertension. METHODS This retrospective cohort study enrolled 107,207 patients with newly diagnosed hypertension between 2001 and 2013. The hazard ratios for Parkinson disease were calculated for ARB treatment compared with those who never used ARBs and among the 5 subgroups receiving different cumulative ARB dosages. RESULTS We identified 527 (1.1%) Parkinson disease cases among patients with ARB treatment in a median observation period of 8.4 years compared to the 1,255 (2.2%) Parkinson disease cases among those without ARB treatment in a median observation period of 6.8 years. Overall, risk for developing Parkinson disease was statistically lower in the ARB-treated group with a hazard ratio of 0.56 (95% confidence interval: 0.51-0.63) than those without ARB. CONCLUSIONS ARB treatment was associated with a statistically important reduction of Parkinson disease risk in patients with newly diagnosed hypertension. Therefore, ARB may constitute an effective neuroprotective strategy to lower Parkinson disease risk in such patients.
Collapse
|
17
|
Ishola I, Afolayan O, Badru A, Olubodun-Obadun T, John N, Adeyemi O. Angiotensin converting enzyme inhibitor captopril prevents neuronal overexpression of amyloid-beta and alpha-synuclein in Drosophila melanogaster genetic models of neurodegenerative diseases. Niger J Physiol Sci 2022; 37:21-28. [PMID: 35947848 DOI: 10.54548/njps.v37i1.3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 07/09/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Parkinson disease (PD) and Alzheimer's disease (AD) are progressive neurodegenerative disorders characterized by loss of selective neurons in discreet part of the brain. The peptide angiotensin II (Ang II) plays significant role in hippocampal and striatal neurons degeneration through the generation of reactive oxygen species. Blockade of the angiotensin converting enzyme or ATI receptors provides protection in animal models of neurodegenerative diseases. In the present study, the neuroprotective effect of captopril was investigated in Drosophila melanogaster model using the UAS-GAL4 system to express the synuclein and Aβ42 peptide in the flies' neurons. METHODS The disease causing human Aβ42 peptide or α-syn was expressed pan-neuronally (elav-GAL4) or dopamine neuron (DDC-GAL4) using the UAS-GAL4 system. Flies were either grown in food media with or without captopril (1, 5, or 10µM). This was followed by fecundity, larva motility, negative geotaxis assay (climbing) and lifespan as a measure of neurodegeneration. RESULTS Elav-Gal4<Aβ or DDC-GAL4<α-syn flies displayed significant decrease in larva motility when compared with normal control (w1118) which was reversed by the supplementation of the media with captopril (5 or 10 mM) indicative of neuroprotection. Interestingly, supplementation of flies' media with captopril improved climbing activity in Elav-Gal4<Aβ or DDC-GAL4<α-syn flies when compared with vehicle treated only. Moreover, flies grown on captopril caused no significant change in lifespan. Conclusion: Findings from this study confirmed the neuroprotective action of captopril in genetic or familial forms of neurodegeneration.
Collapse
|
18
|
GLP-1 Receptor Agonists in Neurodegeneration: Neurovascular Unit in the Spotlight. Cells 2022; 11:cells11132023. [PMID: 35805109 PMCID: PMC9265397 DOI: 10.3390/cells11132023] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 02/07/2023] Open
Abstract
Defects in brain energy metabolism and proteopathic stress are implicated in age-related degenerative neuronopathies, exemplified by Alzheimer’s disease (AD) and Parkinson’s disease (PD). As the currently available drug regimens largely aim to mitigate cognitive decline and/or motor symptoms, there is a dire need for mechanism-based therapies that can be used to improve neuronal function and potentially slow down the underlying disease processes. In this context, a new class of pharmacological agents that achieve improved glycaemic control via the glucagon-like peptide 1 (GLP-1) receptor has attracted significant attention as putative neuroprotective agents. The experimental evidence supporting their potential therapeutic value, mainly derived from cellular and animal models of AD and PD, has been discussed in several research reports and review opinions recently. In this review article, we discuss the pathological relevance of derangements in the neurovascular unit and the significance of neuron–glia metabolic coupling in AD and PD. With this context, we also discuss some unresolved questions with regard to the potential benefits of GLP-1 agonists on the neurovascular unit (NVU), and provide examples of novel experimental paradigms that could be useful in improving our understanding regarding the neuroprotective mode of action associated with these agents.
Collapse
|
19
|
Grotemeyer A, McFleder RL, Wu J, Wischhusen J, Ip CW. Neuroinflammation in Parkinson's Disease - Putative Pathomechanisms and Targets for Disease-Modification. Front Immunol 2022; 13:878771. [PMID: 35663989 PMCID: PMC9158130 DOI: 10.3389/fimmu.2022.878771] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/20/2022] [Indexed: 12/15/2022] Open
Abstract
Parkinson’s disease (PD) is a progressive and debilitating chronic disease that affects more than six million people worldwide, with rising prevalence. The hallmarks of PD are motor deficits, the spreading of pathological α-synuclein clusters in the central nervous system, and neuroinflammatory processes. PD is treated symptomatically, as no causally-acting drug or procedure has been successfully established for clinical use. Various pathways contributing to dopaminergic neuron loss in PD have been investigated and described to interact with the innate and adaptive immune system. We discuss the possible contribution of interconnected pathways related to the immune response, focusing on the pathophysiology and neurodegeneration of PD. In addition, we provide an overview of clinical trials targeting neuroinflammation in PD.
Collapse
Affiliation(s)
| | | | - Jingjing Wu
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Jörg Wischhusen
- Section for Experimental Tumor Immunology, Department of Obstetrics and Gynecology, University Hospital of Würzburg, Würzburg, Germany
| | - Chi Wang Ip
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
20
|
Mogi M. Aldosterone breakthrough from a pharmacological perspective. Hypertens Res 2022; 45:967-975. [PMID: 35422512 DOI: 10.1038/s41440-022-00913-4] [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: 01/24/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 01/13/2023]
Abstract
Aldosterone (Aldo) breakthrough is a well-known phenomenon that occurs in patients with long-term renin-angiotensin aldosterone system (RAAS) blockade using inhibitors of renin or angiotensin converting enzyme or angiotensin II type 1 receptor blockers. The blockade of the mineralocorticoid receptor (MR), an Aldo binding receptor, is effective in managing patients with resistant hypertension, defined as uncontrollable blood pressure despite the concurrent use of three antihypertensive drugs. In other words, MR inhibitors are not used as first-line antihypertensive drugs in most guidelines for hypertension management. Aldo breakthrough puts hypertensive patients at higher risk of cardiovascular disease and worsens future outcomes. This review discusses Aldo secretion and the mechanism of Aldo breakthrough, dependent or independent of the RAAS, with consideration of the pharmacological aspects of this phenomenon, as well as hypothetical views.
Collapse
Affiliation(s)
- Masaki Mogi
- Department of Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Tohon, Ehime, 791-0295, Japan.
| |
Collapse
|
21
|
Jo Y, Kim S, Ye BS, Lee E, Yu YM. Protective Effect of Renin-Angiotensin System Inhibitors on Parkinson's Disease: A Nationwide Cohort Study. Front Pharmacol 2022; 13:837890. [PMID: 35308220 PMCID: PMC8927987 DOI: 10.3389/fphar.2022.837890] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Renin-angiotensin system (RAS) inhibitors have been suggested as protective agents in Parkinson's disease (PD). However, epidemiological evidence on the association between RAS inhibitors and the development of PD is inconsistent. Objectives: To investigate the effect of RAS inhibitors on PD risk in patients with ischemic heart disease (IHD) by type and cumulative duration of RAS inhibitors and their degree of blood-brain barrier (BBB) penetration ability. Methods: This was a propensity score-matched retrospective cohort study using 2008-2019 healthcare claims data from the Korean Health Insurance Review and Assessment database. The association between RAS inhibitor use and PD in patients with IHD was evaluated using multivariate Cox proportional hazard regression analysis. The risks are presented as adjusted hazard ratios (aHRs) and 95% confidence intervals (CIs). Results: Over a 10-year follow-up, 1,086 of 62,228 IHD patients developed PD. The Cox regression model showed that the use of RAS inhibitors was significantly associated with a lower risk of PD (aHR = 0.75; 95% CI 0.66-0.85) than the non-use of RAS inhibitors. Specifically, this reduced risk of PD only remained with the use of BBB-crossing angiotensin II receptor blockers (ARBs) (aHR = 0.62; 95% CI = 0.53-0.74), and this association was more definite with an increasing cumulative duration. A significantly reduced risk of PD was not observed with the use of BBB-crossing angiotensin-converting enzyme inhibitors. Conclusions: The use of ARBs with BBB-penetrating properties and a high cumulative duration significantly reduces the risk of PD in IHD patients. This protective effect could provide insight into disease-modifying drug candidates for PD.
Collapse
Affiliation(s)
- Youngkwon Jo
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, South Korea
| | - Seungyeon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea.,Transdisciplinary Department of Medicine and Advanced Technology, Seoul National University Hospital, Seoul, South Korea
| | - Byoung Seok Ye
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Euni Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | - Yun Mi Yu
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, South Korea.,Department of Pharmaceutical Medicine and Regulatory Sciences, Colleges of Medicine and Pharmacy, Yonsei University, Incheon, South Korea
| |
Collapse
|
22
|
Shared Molecular Targets in Parkinson’s Disease and Arterial Hypertension: A Systematic Review. Biomedicines 2022; 10:biomedicines10030653. [PMID: 35327454 PMCID: PMC8945026 DOI: 10.3390/biomedicines10030653] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/04/2022] [Accepted: 03/07/2022] [Indexed: 12/10/2022] Open
Abstract
(1) Background: Parkinson’s disease and arterial hypertension are likely to coexist in the elderly, with possible bidirectional interactions. We aimed to assess the role of antihypertensive agents in PD emergence and/or progression. (2) We performed a systematic search on the PubMed database. Studies enrolling patients with Parkinson’s disease who underwent treatment with drugs pertaining to one of the major antihypertensive drug classes (β-blockers, diuretics, angiotensin-converting enzyme inhibitors, angiotensin receptor blockers and calcium-channel blockers) prior to or after the diagnosis of parkinsonism were scrutinized. We divided the outcome into two categories: neuroprotective and disease-modifying effect. (3) We included 20 studies in the qualitative synthesis, out of which the majority were observational studies, with only one randomized controlled trial. There are conflicting results regarding the effect of antihypertensive drugs on Parkinson’s disease pathogenesis, mainly because of heterogeneous protocols and population. (4) Conclusions: There is low quality evidence that antihypertensive agents might be potential therapeutic targets in Parkinson’s disease, but this hypothesis needs further testing.
Collapse
|
23
|
Maeker E, Maeker-Poquet B. Syndromes extrapyramidaux induits par les médicaments. NPG NEUROLOGIE - PSYCHIATRIE - GÉRIATRIE 2022; 22:25-34. [DOI: 10.1016/j.npg.2021.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
24
|
Gao Q, Chen R, Wu L, Huang Q, Wang XX, Tian YY, Zhang YD. Angiotensin-(1-7) reduces α-synuclein aggregation by enhancing autophagic activity in Parkinson's disease. Neural Regen Res 2021; 17:1138-1145. [PMID: 34558543 PMCID: PMC8552854 DOI: 10.4103/1673-5374.324854] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Abnormal accumulation of α-synuclein contributes to the formation of Lewy bodies in the substantia nigra, which is considered the typical pathological hallmark of Parkinson's disease. Recent research indicates that angiotensin-(1-7) plays a crucial role in several neurodegenerative disorders, including Parkinson's disease, but the underlying mechanisms remain elusive. In this study, we used intraperitoneal administration of rotenone to male Sprague-Dawley rats for 4 weeks to establish a Parkinson's disease model. We investigated whether angiotensin-(1-7) is neuroprotective in this model by continuous administration of angiotensin-(1-7) into the right substantia nigra for 4 weeks. We found that angiotensin-(1-7) infusion relieved characteristic parkinsonian behaviors and reduced α-synuclein aggregation in the substantia nigra. Primary dopaminergic neurons were extracted from newborn Sprague-Dawley rat substantia nigras and treated with rotenone, angiotensin-(1-7), and/or the Mas receptor blocker A-779 for 24 hours. After binding to the Mas receptor, angiotensin-(1-7) attenuated apoptosis and α-synuclein aggregation in rotenone-treated cells. Primary dopaminergic neurons were also treated with angiotensin-(1-7) and/or the autophagy inhibitor 3-methyladenine for 24 hours. Angiotensin-(1-7) increased α-synuclein removal and increased the autophagy of rotenone-treated cells. We conclude that angiotensin-(1-7) reduces α-synuclein aggregation by alleviating autophagy dysfunction in Parkinson's disease. Therefore, the angiotensin-(1-7)/Mas receptor axis plays an important role in the pathogenesis of Parkinson's disease and angiotensin-(1-7) has potential therapeutic value for Parkinson's disease. All experiments were approved by the Biological Research Ethics Committee of Nanjing First Hospital (approval No. DWSY-2000932) in January 2020.
Collapse
Affiliation(s)
- Qing Gao
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Rui Chen
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Liang Wu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Qing Huang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xi-Xi Wang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - You-Yong Tian
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ying-Dong Zhang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
25
|
Sfera A, Osorio C, Maguire G, Rahman L, Afzaal J, Cummings M, Maldonado JC. COVID-19, ferrosenescence and neurodegeneration, a mini-review. Prog Neuropsychopharmacol Biol Psychiatry 2021; 109:110230. [PMID: 33373681 PMCID: PMC7832711 DOI: 10.1016/j.pnpbp.2020.110230] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/16/2020] [Accepted: 12/18/2020] [Indexed: 02/06/2023]
Abstract
Exacerbation of cognitive, motor and nonmotor symptoms have been described in critically ill COVID-19 patients, indicating that, like prior pandemics, neurodegenerative sequelae may mark the aftermath of this viral infection. Moreover, SARS-CoV-2, the causative agent of COVID-19 disease, was associated with hyperferritinemia and unfavorable prognosis in older individuals, suggesting virus-induced ferrosenescence. We have previously defined ferrosenescence as an iron-associated disruption of both the human genome and its repair mechanisms, leading to premature cellular senescence and neurodegeneration. As viruses replicate more efficiently in iron-rich senescent cells, they may have developed the ability to induce this phenotype in host tissues, predisposing to both immune dysfunction and neurodegenerative disorders. In this mini-review, we summarize what is known about the SARS-CoV-2-induced cellular senescence and iron dysmetabolism. We also take a closer look at immunotherapy with natural killer cells, angiotensin II receptor blockers ("sartans"), iron chelators and dipeptidyl peptidase 4 inhibitors ("gliptins") as adjunct treatments for both COVID-19 and its neurodegenerative complications.
Collapse
Affiliation(s)
- Adonis Sfera
- Patton State Hospital, California, United States of America.
| | | | - Gerald Maguire
- University of California, Riverside, United States of America
| | - Leah Rahman
- Patton State Hospital, California, United States of America
| | - Jafri Afzaal
- Patton State Hospital, California, United States of America
| | | | | |
Collapse
|
26
|
Kobiec T, Otero-Losada M, Chevalier G, Udovin L, Bordet S, Menéndez-Maissonave C, Capani F, Pérez-Lloret S. The Renin-Angiotensin System Modulates Dopaminergic Neurotransmission: A New Player on the Scene. Front Synaptic Neurosci 2021; 13:638519. [PMID: 33967734 PMCID: PMC8100578 DOI: 10.3389/fnsyn.2021.638519] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/11/2021] [Indexed: 11/13/2022] Open
Abstract
Parkinson's disease (PD) is an extrapyramidal disorder characterized by neuronal degeneration in several regions of the peripheral and central nervous systems. It is the second most frequent neurodegenerative disease after Alzheimer's. It has become a major health problem, affecting 1% of the world population over 60 years old and 3% of people beyond 80 years. The main histological findings are intracellular Lewy bodies composed of misfolded α-synuclein protein aggregates and loss of dopaminergic neurons in the central nervous system. Neuroinflammation, apoptosis, mitochondrial dysfunction, altered calcium homeostasis, abnormal protein degradation, and synaptic pathobiology have been put forward as mechanisms leading to cell death, α-synuclein deposition, or both. A progressive loss of dopaminergic neurons in the substantia nigra late in the neurodegeneration leads to developing motor symptoms like bradykinesia, tremor, and rigidity. The renin-angiotensin system (RAS), which is involved in regulating blood pressure and body fluid balance, also plays other important functions in the brain. The RAS is involved in the autocrine and paracrine regulation of the nigrostriatal dopaminergic synapses. Dopamine depletion, as in PD, increases angiotensin II expression, which stimulates or inhibits dopamine synthesis and is released via AT1 or AT2 receptors. Furthermore, angiotensin II AT1 receptors inhibit D1 receptor activation allosterically. Therefore, the RAS may have an important modulating role in the flow of information from the brain cortex to the basal ganglia. High angiotensin II levels might even aggravate neurodegeneration, activating the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex, which leads to increased reactive oxygen species production.
Collapse
Affiliation(s)
- Tamara Kobiec
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía, Facultad de Psicología y Psicopedagogía, Universidad Católica Argentina, Buenos Aires, Argentina
| | - Matilde Otero-Losada
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Guenson Chevalier
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Lucas Udovin
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Sofía Bordet
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía, Facultad de Psicología y Psicopedagogía, Universidad Católica Argentina, Buenos Aires, Argentina
| | - Camila Menéndez-Maissonave
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía, Facultad de Psicología y Psicopedagogía, Universidad Católica Argentina, Buenos Aires, Argentina
| | - Francisco Capani
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía, Facultad de Psicología y Psicopedagogía, Universidad Católica Argentina, Buenos Aires, Argentina
- Facultad de Psicología y Psicopedagogía, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
- Departamento de Biología, Universidad Argentina John F. Kennedy, Buenos Aires, Argentina
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago de Chile, Chile
| | - Santiago Pérez-Lloret
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Facultad de Medicina, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
- Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
27
|
Angiotensin-II Modulates GABAergic Neurotransmission in the Mouse Substantia Nigra. eNeuro 2021; 8:ENEURO.0090-21.2021. [PMID: 33771900 PMCID: PMC8174047 DOI: 10.1523/eneuro.0090-21.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 03/22/2021] [Indexed: 11/26/2022] Open
Abstract
GABAergic projections neurons of the substantia nigra reticulata (SNr), through an extensive network of dendritic arbors and axon collaterals, provide robust inhibitory input to neighboring dopaminergic neurons in the substantia nigra compacta (SNc). Angiotensin-II (Ang-II) receptor signaling increases SNc dopaminergic neuronal sensitivity to insult, thus rendering these cells susceptible to dysfunction and destruction. However, the mechanisms by which Ang-II regulates SNc dopaminergic neuronal activity are unclear. Given the complex relationship between SN dopaminergic and GABAergic neurons, we hypothesized that Ang-II could regulate SNc dopaminergic neuronal activity directly and indirectly by modulating SNr GABAergic neurotransmission. Here, using transgenic mice, slice electrophysiology, and optogenetics, we provide evidence of an AT1 receptor-mediated signaling mechanism in SNr GABAergic neurons where Ang-II suppresses electrically-evoked neuronal output by facilitating postsynaptic GABAA receptors (GABAARs) and prolonging the action potential (AP) duration. Unexpectedly, Ang-II had no discernable effects on the electrical properties of SNc dopaminergic neurons. Also, and indicating a nonlinear relationship between electrical activity and neuronal output, following phasic photoactivation of SNr GABAergic neurons, Ang-II paradoxically enhanced the feedforward inhibitory input to SNc dopaminergic neurons. In sum, our observations describe an increasingly complex and heterogeneous response of the SN to Ang-II by revealing cell-specific responses and nonlinear effects on intranigral GABAergic neurotransmission. Our data further implicate the renin-angiotensin-system (RAS) as a functionally relevant neuromodulator in the substantia nigra, thus underscoring a need for additional inquiry.
Collapse
|
28
|
Rivas-Santisteban R, Lillo J, Muñoz A, Rodríguez-Pérez AI, Labandeira-García JL, Navarro G, Franco R. Novel Interactions Involving the Mas Receptor Show Potential of the Renin-Angiotensin system in the Regulation of Microglia Activation: Altered Expression in Parkinsonism and Dyskinesia. Neurotherapeutics 2021; 18:998-1016. [PMID: 33474655 PMCID: PMC7817140 DOI: 10.1007/s13311-020-00986-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2020] [Indexed: 12/30/2022] Open
Abstract
The renin-angiotensin system (RAS) not only plays an important role in controlling blood pressure but also participates in almost every process to maintain homeostasis in mammals. Interest has recently increased because SARS viruses use one RAS component (ACE2) as a target-cell receptor. The occurrence of RAS in the basal ganglia suggests that the system may be targeted to improve the therapy of neurodegenerative diseases. RAS-related data led to the hypothesis that RAS receptors may interact with each other. The aim of this paper was to find heteromers formed by Mas and angiotensin receptors and to address their functionality in neurons and microglia. Novel interactions were discovered by using resonance energy transfer techniques. The functionality of individual and interacting receptors was assayed by measuring levels of the second messengers cAMP and Ca2+ in transfected human embryonic kidney cells (HEK-293T) and primary cultures of striatal cells. Receptor complex expression was assayed by in situ proximity ligation assay. Functionality and expression were assayed in parallel in primary cultures of microglia treated or not with lipopolysaccharide and interferon-γ (IFN-γ). The proximity ligation assay was used to assess heteromer expression in parkinsonian and dyskinetic conditions. Complexes formed by Mas and the angiotensin AT1 or AT2 receptors were identified in both a heterologous expression system and in neural primary cultures. In the heterologous system, we showed that the three receptors-MasR, AT1R, and AT2R-can interact to form heterotrimers. The expression of receptor dimers (AT1R-MasR or AT2R-MasR) was higher in microglia than in neurons and was differentially affected upon microglial activation with lipopolysaccharide and IFN-γ. In all cases, agonist-induced signaling was reduced upon coactivation, and in some cases just by coexpression. Also, the blockade of signaling of two receptors in a complex by the action of a given (selective) receptor antagonist (cross-antagonism) was often observed. Differential expression of the complexes was observed in the striatum under parkinsonian conditions and especially in animals rendered dyskinetic by levodopa treatment. The negative modulation of calcium mobilization (mediated by AT1R activation), the multiplicity of possibilities on RAS affecting the MAPK pathway, and the disbalanced expression of heteromers in dyskinesia yield new insight into the operation of the RAS system, how it becomes unbalanced, and how a disbalanced RAS can be rebalanced. Furthermore, RAS components in activated microglia warrant attention in drug-development approaches to address neurodegeneration.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Animals
- Corpus Striatum/drug effects
- Corpus Striatum/metabolism
- Dyskinesia, Drug-Induced/metabolism
- HEK293 Cells
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Microglia/drug effects
- Microglia/metabolism
- Oxidopamine/toxicity
- Parkinsonian Disorders/chemically induced
- Parkinsonian Disorders/metabolism
- Proto-Oncogene Mas/agonists
- Proto-Oncogene Mas/metabolism
- Rats
- Rats, Wistar
- Receptor, Angiotensin, Type 1/agonists
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 2/agonists
- Receptor, Angiotensin, Type 2/metabolism
- Renin-Angiotensin System/drug effects
- Renin-Angiotensin System/physiology
Collapse
Affiliation(s)
- Rafael Rivas-Santisteban
- Department Biochemistry and Molecular Biomedicine, School of Biology, University of Barcelona, Diagonal 643, Barcelona, Catalonia, 08028, Spain
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIberNed), Instituto de Salud Carlos III, Valderrebollo 5, Madrid, Madrid, 28031, Spain
| | - Jaume Lillo
- Department Biochemistry and Molecular Biomedicine, School of Biology, University of Barcelona, Diagonal 643, Barcelona, Catalonia, 08028, Spain
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIberNed), Instituto de Salud Carlos III, Valderrebollo 5, Madrid, Madrid, 28031, Spain
| | - Ana Muñoz
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIberNed), Instituto de Salud Carlos III, Valderrebollo 5, Madrid, Madrid, 28031, Spain
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Ana I Rodríguez-Pérez
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIberNed), Instituto de Salud Carlos III, Valderrebollo 5, Madrid, Madrid, 28031, Spain
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - José Luís Labandeira-García
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIberNed), Instituto de Salud Carlos III, Valderrebollo 5, Madrid, Madrid, 28031, Spain
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Gemma Navarro
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIberNed), Instituto de Salud Carlos III, Valderrebollo 5, Madrid, Madrid, 28031, Spain
- Department of Biochemistry and Physiology, School of Pharmacy and Food Science, University of Barcelona, Barcelona, Catalonia, 08028, Spain
| | - Rafael Franco
- Department Biochemistry and Molecular Biomedicine, School of Biology, University of Barcelona, Diagonal 643, Barcelona, Catalonia, 08028, Spain.
| |
Collapse
|
29
|
Udovin L, Otero-Losada M, Bordet S, Chevalier G, Quarracino C, Capani F, Pérez-Lloret S. Effects of angiotensin type 1 receptor antagonists on Parkinson's disease progression: An exploratory study in the PPMI database. Parkinsonism Relat Disord 2021; 86:34-37. [PMID: 33823471 DOI: 10.1016/j.parkreldis.2021.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 02/10/2021] [Accepted: 03/10/2021] [Indexed: 10/21/2022]
Abstract
INTRODUCTION We explored the potential clinical effects of angiotensin-II AT1 receptor blockers (ARBs) and angiotensin-converting enzyme inhibitors (ACEIs) in patients from the Parkinson's Progress Marker Initiative (PPMI) study database. METHODS We included 423 newly diagnosed PD patients, free from antiparkinsonian treatment, from the PPMI. We compared the proportion of patients starting on l-DOPA during the first year of follow-up, and the changes in MDS-UPDRS total score and sub-scores during the first five follow-up years for patients exposed or not to ARBs or ACEIs. RESULTS Treatment with ARBs did not affect the proportion of patients on l-DOPA during the first year (adjusted OR, 95% CI = 0.26, 0.03-2.18, N.S.) while reduced MDS-UPDRS total score (0.85, 0.76-0.95, p < 0.01). Patients treated with ACEIs experienced no changes in either measure. CONCLUSIONS These results show potential signals for a beneficial effect with ARBs. Further clinical trials are warranted.
Collapse
Affiliation(s)
- Lucas Udovin
- Centro de Altos Estudios en Ciencias Humanas y de la Salud- Universidad Abierta Interamericana. Consejo Nacional de Investigaciones Científicas y Técnicas. CAECIHS-UAI. CONICET, Buenos Aires, Argentina
| | - Matilde Otero-Losada
- Centro de Altos Estudios en Ciencias Humanas y de la Salud- Universidad Abierta Interamericana. Consejo Nacional de Investigaciones Científicas y Técnicas. CAECIHS-UAI. CONICET, Buenos Aires, Argentina
| | - Sofia Bordet
- Centro de Altos Estudios en Ciencias Humanas y de la Salud- Universidad Abierta Interamericana. Consejo Nacional de Investigaciones Científicas y Técnicas. CAECIHS-UAI. CONICET, Buenos Aires, Argentina; Centro de Investigaciones en Psicología y Psicopedagogía (CIPP), Facultad de Psicología y Psicopedagogía, Pontificia Universidad Católica Argentina (UCA), Buenos Aires, Argentina
| | - Guenson Chevalier
- Centro de Altos Estudios en Ciencias Humanas y de la Salud- Universidad Abierta Interamericana. Consejo Nacional de Investigaciones Científicas y Técnicas. CAECIHS-UAI. CONICET, Buenos Aires, Argentina
| | - Cecilia Quarracino
- Centro de Altos Estudios en Ciencias Humanas y de la Salud- Universidad Abierta Interamericana. Consejo Nacional de Investigaciones Científicas y Técnicas. CAECIHS-UAI. CONICET, Buenos Aires, Argentina
| | - Francisco Capani
- Centro de Altos Estudios en Ciencias Humanas y de la Salud- Universidad Abierta Interamericana. Consejo Nacional de Investigaciones Científicas y Técnicas. CAECIHS-UAI. CONICET, Buenos Aires, Argentina; Instituto Universitario de Ciencias de la Salud, Fundación H.A Barceló, Buenos Aires, Argentina; Departamento de Biología, Universidad John F. Kennedy, Buenos Aires, Argentina; Facultad de Medicina, Universidad Autónoma de Chile, Santiago, Chile
| | - Santiago Pérez-Lloret
- Centro de Altos Estudios en Ciencias Humanas y de la Salud- Universidad Abierta Interamericana. Consejo Nacional de Investigaciones Científicas y Técnicas. CAECIHS-UAI. CONICET, Buenos Aires, Argentina; Facultad de Ciencias Médicas, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina; Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires. Buenos Aires, Argentina.
| |
Collapse
|
30
|
Pierzchlińska A, Kwaśniak-Butowska M, Sławek J, Droździk M, Białecka M. Arterial Blood Pressure Variability and Other Vascular Factors Contribution to the Cognitive Decline in Parkinson's Disease. Molecules 2021; 26:molecules26061523. [PMID: 33802165 PMCID: PMC8001922 DOI: 10.3390/molecules26061523] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 11/24/2022] Open
Abstract
Dementia is one of the most disabling non-motor symptoms in Parkinson’s disease (PD). Unlike in Alzheimer’s disease, the vascular pathology in PD is less documented. Due to the uncertain role of commonly investigated metabolic or vascular factors, e.g., hypertension or diabetes, other factors corresponding to PD dementia have been proposed. Associated dysautonomia and dopaminergic treatment seem to have an impact on diurnal blood pressure (BP) variability, which may presumably contribute to white matter hyperintensities (WMH) development and cognitive decline. We aim to review possible vascular and metabolic factors: Renin-angiotensin-aldosterone system, vascular endothelial growth factor (VEGF), hyperhomocysteinemia (HHcy), as well as the dopaminergic treatment, in the etiopathogenesis of PD dementia. Additionally, we focus on the role of polymorphisms within the genes for catechol-O-methyltransferase (COMT), apolipoprotein E (APOE), vascular endothelial growth factor (VEGF), and for renin-angiotensin-aldosterone system components, and their contribution to cognitive decline in PD. Determining vascular risk factors and their contribution to the cognitive impairment in PD may result in screening, as well as preventive measures.
Collapse
Affiliation(s)
- Anna Pierzchlińska
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University, Aleja Powstańców Wlkp 72, 70-111 Szczecin, Poland;
- Correspondence: (A.P.); (M.D.)
| | - Magdalena Kwaśniak-Butowska
- Division of Neurological and Psychiatric Nursing, Medical University of Gdansk, Aleja Jana Pawła II 50, 80-462 Gdansk, Poland; (M.K.-B.); (J.S.)
- Department of Neurology, St Adalbert Hospital, Aleja Jana Pawła II 50, 80-462 Gdansk, Poland
| | - Jarosław Sławek
- Division of Neurological and Psychiatric Nursing, Medical University of Gdansk, Aleja Jana Pawła II 50, 80-462 Gdansk, Poland; (M.K.-B.); (J.S.)
- Department of Neurology, St Adalbert Hospital, Aleja Jana Pawła II 50, 80-462 Gdansk, Poland
| | - Marek Droździk
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, Aleja Powstańców Wlkp 72, 70-111 Szczecin, Poland
- Correspondence: (A.P.); (M.D.)
| | - Monika Białecka
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University, Aleja Powstańców Wlkp 72, 70-111 Szczecin, Poland;
| |
Collapse
|
31
|
Occhieppo VB, Basmadjian OM, Bregonzio C. Brain angiotensin II in dopaminergic imbalance-derived pathologies: neuroinflammation and vascular responses. Neural Regen Res 2021; 16:504-505. [PMID: 32985475 PMCID: PMC7996039 DOI: 10.4103/1673-5374.293144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Affiliation(s)
- Victoria Belén Occhieppo
- Instituto de Farmacología Experimental Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Osvaldo Martin Basmadjian
- Instituto de Farmacología Experimental Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Claudia Bregonzio
- Instituto de Farmacología Experimental Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
32
|
Receptors | Angiotensin Receptors. ENCYCLOPEDIA OF BIOLOGICAL CHEMISTRY III 2021. [PMCID: PMC8326513 DOI: 10.1016/b978-0-12-819460-7.00096-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The renin-angiotensin-aldosterone system (RAS) is a vital hormone-receptor system that regulates cardiovascular and renal functions. In this article, we discuss exciting new findings in the RAS field. Recently solved active state crystal structures of Angiotensin II type 1 (AT1R) and type 2 receptor (AT2R) helped in understanding receptor activation mechanisms in detail. Also, considerable attention is given to the developments in characterizing the counter-regulatory RAS axis due to current hope for harnessing this axis for the development of protective therapies against various cardiovascular diseases. We describe the RAS component, angiotensin-converting enzyme 2 (ACE2) functioning as cellular entry receptor for the causative agent of COVID-19 pandemic, SARS-CoV-2. Altogether, these discoveries paved the way for developing novel therapies targeting different components of the RAS in the future.
Collapse
|
33
|
McFall A, Nicklin SA, Work LM. The counter regulatory axis of the renin angiotensin system in the brain and ischaemic stroke: Insight from preclinical stroke studies and therapeutic potential. Cell Signal 2020; 76:109809. [PMID: 33059037 PMCID: PMC7550360 DOI: 10.1016/j.cellsig.2020.109809] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 01/01/2023]
Abstract
Stroke is the 2nd leading cause of death worldwide and the leading cause of physical disability and cognitive issues. Although we have made progress in certain aspects of stroke treatment, the consequences remain substantial and new treatments are needed. Hypertension has long been recognised as a major risk factor for stroke, both haemorrhagic and ischaemic. The renin angiotensin system (RAS) plays a key role in blood pressure regulation and this, plus local expression and signalling of RAS in the brain, both support the potential for targeting this axis therapeutically in the setting of stroke. While historically, focus has been on suppressing classical RAS signalling through the angiotensin type 1 receptor (AT1R), the identification of a counter-regulatory axis of the RAS signalling via the angiotensin type 2 receptor (AT2R) and Mas receptor has renewed interest in targeting the RAS. This review describes RAS signalling in the brain and the potential of targeting the Mas receptor and AT2R in preclinical models of ischaemic stroke. The animal and experimental models, and the route and timing of intervention, are considered from a translational perspective.
Collapse
Affiliation(s)
- Aisling McFall
- Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Stuart A Nicklin
- Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Lorraine M Work
- Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
34
|
Visanji NP, Madan P, Lacoste AMB, Buleje I, Han Y, Spangler S, Kalia LV, Hensley Alford S, Marras C. Using artificial intelligence to identify anti-hypertensives as possible disease modifying agents in Parkinson's disease. Pharmacoepidemiol Drug Saf 2020; 30:201-209. [PMID: 33219601 DOI: 10.1002/pds.5176] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 08/25/2020] [Accepted: 10/21/2020] [Indexed: 11/10/2022]
Abstract
PURPOSE Drug repurposing is an effective means of increasing treatment options for diseases, however identifying candidate molecules for the indication of interest from the thousands of approved drugs is challenging. We have performed a computational analysis of published literature to rank existing drugs according to predicted ability to reduce alpha synuclein (aSyn) oligomerization and analyzed real-world data to investigate the association between exposure to highly ranked drugs and PD. METHODS Using IBM Watson for Drug Discoveryâ (WDD) we identified several antihypertensive drugs that may reduce aSyn oligomerization. Using IBM MarketScanâ Research Databases we constructed a cohort of individuals with incident hypertension. We conducted univariate and multivariate Cox proportional hazard analyses (HR) with exposure as a time-dependent covariate. Diuretics were used as the referent group. Age at hypertension diagnosis, sex, and several comorbidities were included in multivariate analyses. RESULTS Multivariate results revealed inverse associations for time to PD diagnosis with exposure to the combination of the combination of angiotensin receptor II blockers (ARBs) and dihydropyridine calcium channel blockers (DHP-CCB) (HR = 0.55, p < 0.01) and angiotensin converting enzyme inhibitors (ACEi) and diuretics (HR = 0.60, p-value <0.01). Increased risk was observed with exposure to alpha-blockers alone (HR = 1.81, p < 0.001) and the combination of alpha-blockers and CCB (HR = 3.17, p < 0.05). CONCLUSIONS We present evidence that a computational approach can efficiently identify leads for disease-modifying drugs. We have identified the combination of ARBs and DHP-CCBs as of particular interest in PD.
Collapse
Affiliation(s)
- Naomi P Visanji
- Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, Ontario, Canada
| | | | | | - Italo Buleje
- Foundational Innovation, Health Care and Life Sciences, IBM Cambridge Research Center, Cambridge, Massachusetts, USA
| | - Yanyan Han
- IBM Almaden Research Center, San Jose, California, USA
| | | | - Lorraine V Kalia
- Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, Ontario, Canada
| | | | - Connie Marras
- Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, Ontario, Canada
| |
Collapse
|
35
|
Rivas-Santisteban R, Rodriguez-Perez AI, Muñoz A, Reyes-Resina I, Labandeira-García JL, Navarro G, Franco R. Angiotensin AT 1 and AT 2 receptor heteromer expression in the hemilesioned rat model of Parkinson's disease that increases with levodopa-induced dyskinesia. J Neuroinflammation 2020; 17:243. [PMID: 32807174 PMCID: PMC7430099 DOI: 10.1186/s12974-020-01908-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/21/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND/AIMS The renin-angiotensin system (RAS) is altered in Parkinson's disease (PD), a disease due to substantia nigra neurodegeneration and whose dopamine-replacement therapy, using the precursor levodopa, leads to dyskinesias as the main side effect. Angiotensin AT1 and AT2 receptors, mainly known for their role in regulating water homeostasis and blood pressure and able to form heterodimers (AT1/2Hets), are present in the central nervous system. We assessed the functionality and expression of AT1/2Hets in Parkinson disease (PD). METHODS Immunocytochemistry was used to analyze the colocalization between angiotensin receptors; bioluminescence resonance energy transfer was used to detect AT1/2Hets. Calcium and cAMP determination, MAPK activation, and label-free assays were performed to characterize signaling in homologous and heterologous systems. Proximity ligation assays were used to quantify receptor expression in mouse primary cultures and in rat striatal sections. RESULTS We confirmed that AT1 and AT2 receptors form AT1/2Hets that are expressed in cells of the central nervous system. AT1/2Hets are novel functional units with particular signaling properties. Importantly, the coactivation of the two receptors in the heteromer reduces the signaling output of angiotensin. Remarkably, AT1/2Hets that are expressed in both striatal neurons and microglia make possible that candesartan, the antagonist of AT1, increases the effect of AT2 receptor agonists. In addition, the level of striatal expression increased in the unilateral 6-OH-dopamine lesioned rat PD model and was markedly higher in parkinsonian-like animals that did not become dyskinetic upon levodopa chronic administration if compared with expression in those that became dyskinetic. CONCLUSION The results indicate that boosting the action of neuroprotective AT2 receptors using an AT1 receptor antagonist constitutes a promising therapeutic strategy in PD.
Collapse
Affiliation(s)
- Rafael Rivas-Santisteban
- Department of Biochemistry and Molecular Biomedicine, School of Biology, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación en Red, enfermedades Neurodegenerativas, CiberNed, Instituto de Salud Carlos III, Madrid, Spain
| | - Ana I Rodriguez-Perez
- Centro de Investigación en Red, enfermedades Neurodegenerativas, CiberNed, Instituto de Salud Carlos III, Madrid, Spain.,Laboratory of Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ana Muñoz
- Centro de Investigación en Red, enfermedades Neurodegenerativas, CiberNed, Instituto de Salud Carlos III, Madrid, Spain.,Laboratory of Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Irene Reyes-Resina
- Department of Biochemistry and Molecular Biomedicine, School of Biology, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación en Red, enfermedades Neurodegenerativas, CiberNed, Instituto de Salud Carlos III, Madrid, Spain.,Current adress: RG Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
| | - José Luis Labandeira-García
- Centro de Investigación en Red, enfermedades Neurodegenerativas, CiberNed, Instituto de Salud Carlos III, Madrid, Spain.,Laboratory of Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Gemma Navarro
- Centro de Investigación en Red, enfermedades Neurodegenerativas, CiberNed, Instituto de Salud Carlos III, Madrid, Spain. .,Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, Barcelona, Spain.
| | - Rafael Franco
- Centro de Investigación en Red, enfermedades Neurodegenerativas, CiberNed, Instituto de Salud Carlos III, Madrid, Spain. .,School of Chemistry, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
36
|
Garcia-Rosa S, Carvalho BS, Guest PC, Steiner J, Martins-de-Souza D. Blood plasma proteomic modulation induced by olanzapine and risperidone in schizophrenia patients. J Proteomics 2020; 224:103813. [DOI: 10.1016/j.jprot.2020.103813] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/27/2020] [Accepted: 05/05/2020] [Indexed: 12/29/2022]
|
37
|
Marras C, Mestre T, McDermott MP. Huntington's Disease and Hypertension: Sorting Out Mixed Messages. Mov Disord 2020; 35:915-917. [PMID: 32562461 DOI: 10.1002/mds.28076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 03/30/2020] [Indexed: 11/08/2022] Open
Affiliation(s)
- Connie Marras
- The Edmond J. Safra Program in Parkinson's Disease, Toronto Western Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Tiago Mestre
- Parkinson's Disease and Movement Disorders Clinic, Division of Neurology, Department of Medicine, The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Michael P McDermott
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, New York, USA
| |
Collapse
|
38
|
Sfera A, Osorio C, Jafri N, Diaz EL, Campo Maldonado JE. Intoxication With Endogenous Angiotensin II: A COVID-19 Hypothesis. Front Immunol 2020; 11:1472. [PMID: 32655579 PMCID: PMC7325923 DOI: 10.3389/fimmu.2020.01472] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/05/2020] [Indexed: 12/13/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 has spread rapidly around the globe. However, despite its high pathogenicity and transmissibility, the severity of the associated disease, COVID-19, varies widely. While the prognosis is favorable in most patients, critical illness, manifested by respiratory distress, thromboembolism, shock, and multi-organ failure, has been reported in about 5% of cases. Several studies have associated poor COVID-19 outcomes with the exhaustion of natural killer cells and cytotoxic T cells, lymphopenia, and elevated serum levels of D-dimer. In this article, we propose a common pathophysiological denominator for these negative prognostic markers, endogenous, angiotensin II toxicity. We hypothesize that, like in avian influenza, the outlook of COVID-19 is negatively correlated with the intracellular accumulation of angiotensin II promoted by the viral blockade of its degrading enzyme receptors. In this model, upregulated angiotensin II causes premature vascular senescence, leading to dysfunctional coagulation, and immunity. We further hypothesize that angiotensin II blockers and immune checkpoint inhibitors may be salutary for COVID-19 patients with critical illness by reversing both the clotting and immune defects (Graphical Abstract).
Collapse
Affiliation(s)
- Adonis Sfera
- Patton State Hospital, San Bernardino, CA, United States
| | - Carolina Osorio
- Department of Psychiatry, Loma Linda University, Loma Linda, CA, United States
| | - Nyla Jafri
- Patton State Hospital, San Bernardino, CA, United States
| | - Eddie Lee Diaz
- Patton State Hospital, San Bernardino, CA, United States
| | - Jose E Campo Maldonado
- Department of Medicine, The University of Texas Rio Grande Valley, Edinburg, TX, United States
| |
Collapse
|
39
|
Youssef MM, Abd El-Latif HA, El-Yamany MF, Georgy GS. Aliskiren and captopril improve cognitive deficits in poorly controlled STZ-induced diabetic rats via amelioration of the hippocampal P-ERK, GSK3β, P-GSK3β pathway. Toxicol Appl Pharmacol 2020; 394:114954. [PMID: 32171570 DOI: 10.1016/j.taap.2020.114954] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/08/2020] [Accepted: 03/10/2020] [Indexed: 12/22/2022]
Abstract
Learning and memory deficits are obvious symptoms that develop over time in patients with poorly controlled diabetes. Hyperactivity of the renin-angiotensin system (RAS) is directly associated with β-cell dysfunction and diabetic complications, including cognitive impairment. Here, we investigated the protective and molecular effects of two RAS modifiers, aliskiren; renin inhibitor and captopril; angiotensin converting enzyme inhibitor, on cognitive deficits in the rat hippocampus. Injection of low dose streptozotocin for 4 days resulted in type 1 diabetes. Then, poorly controlled diabetes was mimicked with ineffective daily doses of insulin for 4 weeks. The hyperglycaemia and pancreatic atrophy caused memory disturbance that were identifiable in behavioural tests, hippocampal neurodegeneration, and the following significant changes in the hippocampus, increases in the inflammatory marker interleukin 1β, cholinesterase, the oxidative stress marker malondialdehyde and protein expression of phosphorylated extracellular-signal-regulated kinase and glycogen synthase kinase-3 beta versus decrease in the antioxidant reduced glutathione and protein expression of phosphorylated glycogen synthase kinase-3 beta. Blocking RAS with either drugs along with insulin amended all previously mentioned parameters. Aliskiren stabilized the blood glucose level and restored normal pancreatic integrity and hippocampal malondialdehyde level. Aliskiren showed superior protection against the hippocampal degeneration displayed in the earlier behavioural modification in the passive avoidance test, and the aliskiren group outperformed the control group in the novel object recognition test. We therefore conclude that aliskiren and captopril reversed the diabetic state and cognitive deficits in rats with poorly controlled STZ-induced diabetes through reducing oxidative stress and inflammation and modulating protein expression.
Collapse
Affiliation(s)
- Madonna M Youssef
- Department of Pharmacology, National organization for drug control and research (NODCAR), Giza, Egypt.
| | - H A Abd El-Latif
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini St, Cairo 11562, Egypt
| | - M F El-Yamany
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini St, Cairo 11562, Egypt
| | - Gehan S Georgy
- Department of Pharmacology, National organization for drug control and research (NODCAR), Giza, Egypt
| |
Collapse
|
40
|
Buda V, Baul B, Andor M, Man DE, Ledeţi A, Vlase G, Vlase T, Danciu C, Matusz P, Peter F, Ledeţi I. Solid State Stability and Kinetics of Degradation for Candesartan-Pure Compound and Pharmaceutical Formulation. Pharmaceutics 2020; 12:pharmaceutics12020086. [PMID: 31972960 PMCID: PMC7076474 DOI: 10.3390/pharmaceutics12020086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/11/2020] [Accepted: 01/15/2020] [Indexed: 12/28/2022] Open
Abstract
The aim of this work was to assess the impact of an excipient in a pharmaceutical formulation containing candesartan cilexetil over the decomposition of the active pharmaceutical ingredient and to comparatively investigate the kinetics of degradation during thermolysis in an oxidative atmosphere under controlled thermal stress. To achieve this, the samples were chosen as follows: pure candesartan cilexetil and a commercial tablet of 32 mg strength. As a first investigational tool, Universal attenuated total reflection Fourier transform infrared (UATR-FTIR) spectroscopy was chosen in order to confirm the purity and identity of the samples, as well as to check if any interactions took place in the tablet between candesartan cilexetil and excipients under ambient conditions. Later on, samples were investigated by thermal analysis, and the elucidation of the decomposition mechanism was achieved solely after performing an in-depth kinetic study, namely the use of the modified non-parametric kinetics (NPK) method, since other kinetic methods (American Society for Testing and Materials—ASTM E698, Friedman and Flynn–Wall–Ozawa) led to inadvertencies. The NPK method suggested that candesartan cilexetil and the tablet were degraded by the contribution of two steps, the main being represented by chemical degradation and the secondary being a physical transformation. The excipients chosen in the formulation seemed to have a stabilizing effect on the decomposition of the candesartan cilexetil that was incorporated into the tablet, relative to pure active pharmaceutical ingredient (API), since the apparent activation energy for the decomposition of the tablet was 192.5 kJ/mol, in comparison to 154.5 kJ/mol for the pure API.
Collapse
Affiliation(s)
- Valentina Buda
- Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (V.B.); (C.D.)
| | - Bianca Baul
- Faculty of Industrial Chemistry and Environmental Engineering, Politehnica University of Timișoara, Vasile Parvan Street 6, 300223 Timisoara, Romania (F.P.)
| | - Minodora Andor
- Faculty of Medicine, “Victor Babeş” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (M.A.); (D.E.M.); (P.M.)
| | - Dana Emilia Man
- Faculty of Medicine, “Victor Babeş” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (M.A.); (D.E.M.); (P.M.)
| | - Adriana Ledeţi
- Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (V.B.); (C.D.)
- Correspondence: (A.L.); (I.L.); Tel.: +40-256-204-476 (A.L. & I.L.)
| | - Gabriela Vlase
- Research Centre for Thermal Analysis in Environmental Problems, West University of Timişoara, 300115 Timisoara, Romania; (G.V.); (T.V.)
| | - Titus Vlase
- Research Centre for Thermal Analysis in Environmental Problems, West University of Timişoara, 300115 Timisoara, Romania; (G.V.); (T.V.)
| | - Corina Danciu
- Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (V.B.); (C.D.)
| | - Petru Matusz
- Faculty of Medicine, “Victor Babeş” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (M.A.); (D.E.M.); (P.M.)
| | - Francisc Peter
- Faculty of Industrial Chemistry and Environmental Engineering, Politehnica University of Timișoara, Vasile Parvan Street 6, 300223 Timisoara, Romania (F.P.)
| | - Ionuţ Ledeţi
- Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (V.B.); (C.D.)
- Faculty of Industrial Chemistry and Environmental Engineering, Politehnica University of Timișoara, Vasile Parvan Street 6, 300223 Timisoara, Romania (F.P.)
- Correspondence: (A.L.); (I.L.); Tel.: +40-256-204-476 (A.L. & I.L.)
| |
Collapse
|
41
|
Oh SJ, Fan X. The Possible Role of the Angiotensin System in the Pathophysiology of Schizophrenia: Implications for Pharmacotherapy. CNS Drugs 2019; 33:539-547. [PMID: 30993607 DOI: 10.1007/s40263-019-00632-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A growing body of literature has elucidated the involvement of the central renin-angiotensin system (RAS) in various neuropsychiatric diseases. While consensus on the exact mechanism of the central RAS in schizophrenia pathophysiology does not currently exist, increasing evidence reveals promise in harnessing the therapeutic potential of RAS modulation in the treatment of schizophrenia. In this review, we examine how the central RAS affects inflammation, glutamate, dopamine, gamma-aminobutyric acid (GABA), and peroxisome proliferator-activated receptor (PPAR)-γ, all of which are associated with schizophrenia etiology. In addition, a recent study has demonstrated the therapeutic potential of RAS modulators, especially angiotensin II type 1 receptor blockers (ARBs), as adjunctive therapy to the currently available antipsychotic medications for schizophrenia treatment. With a greater understanding of how RAS inhibition directly modulates neurotransmitter balance in the brain, it is possible that compounds with RAS-inhibiting properties could be used to optimize physiological levels of glutamate, dopamine, and GABA, and the balance among the three neurotransmitters, analogously to how antipsychotic medications mediate the dopaminergic pathways. It can be hoped that a novel approach based on this concept, such as adjunctive telmisartan therapy, may offer practical interventional strategies to address currently unmet therapeutic needs in patients with schizophrenia, especially those with treatment-resistant schizophrenia.
Collapse
Affiliation(s)
| | - Xiaoduo Fan
- Psychotic Disorders Program, UMASS Memorial Medical Center, Biotech One, Suite 100, 365 Plantation Street, Worcester, MA, 01605, USA.
| |
Collapse
|
42
|
Xu X, Pan J, Li X, Cui Y, Mao Z, Wu B, Xu H, Zhou W, Liu Y. Inhibition of Methamphetamine Self-Administration and Reinstatement by Central Blockade of Angiotensin II Receptor in Rats. J Pharmacol Exp Ther 2019; 369:244-258. [PMID: 30867225 DOI: 10.1124/jpet.118.255729] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 03/04/2019] [Indexed: 02/06/2023] Open
Abstract
The molecular mechanism and treatment of methamphetamine (METH) use disorder remain unclear. The current study aimed to investigate the role of central angiotensin II receptor (ATR) in drug taking and seeking behavior associated with METH use disorder. The effect of an ATR type 1 (AT1R) antagonist, candesartan cilexetil, on the reinforcing and motivational effects of METH was first assessed using the animal model of METH self-administration (SA) and reinstatement. The levels of dopamine D2 receptor (D2R) and AT1R were subsequently examined. Furthermore, the present study determined the expression of microRNAs (miRNAs) by comparing METH SA, METH-yoked, and Saline-yoked groups. The target miRNAs were further overexpressed in the nucleus accumbens (NAc) via a lentivirus vector to investigate the effects of target miRNAs on METH SA maintained under a fixed ratio 1, progressive ratio, and cue/drug reinstatement of METH SA. The potential role of the AT1R-PLCβ-CREB signaling pathway was finally investigated. The results suggest that AT1R blockade effectively reduced METH SA and reinstatement, in conjunction with the counter-regulation of D2R and AT1R. A total of 17 miRNAs targeting Ang II in NAc were found to be associated with the voluntary intake of METH. Furthermore, overexpression of specific miR-219a-5p targeting AT1R-regulated METH SA and reinstatement. The AT1R-PLCβ-CREB signaling pathway was found to be associated with the effect of AT1R on the drug-taking and drug-seeking behavior involving METH use disorder.
Collapse
Affiliation(s)
- Xing Xu
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Jian Pan
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Xingxing Li
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Yan Cui
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Zijuan Mao
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Boliang Wu
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Huachong Xu
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Wenhua Zhou
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Yu Liu
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| |
Collapse
|
43
|
Wang J, Fu D, Senouthai S, Jiang Y, Hu R, You Y. Identification of the Transcriptional Networks and the Involvement in Angiotensin II-Induced Injury after CRISPR/Cas9-Mediated Knockdown of Cyr61 in HEK293T Cells. Mediators Inflamm 2019; 2019:8697257. [PMID: 31148949 PMCID: PMC6501185 DOI: 10.1155/2019/8697257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 02/14/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The transcriptional networks of Cyr61 and its function in cell injury are poorly understood. The present study depicted the lncRNA and mRNA profiles and the involvement in angiotensin II-induced injury after Cyr61 knockdown mediated by CRISPR/Cas9 in HEK293T cells. METHODS HEK293T cells were cultured, and Cyr61 knockdown was achieved by transfection of the CRISPR/Cas9 KO plasmid. lncRNA and mRNA microarrays were used to identify differentially expressed genes (DEGs). Gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed to determine biofunctions and signaling pathways. RT-PCR was used to validate the microarray results. Cells were divided into four groups: control, Cyr61 knockdown, angiotensin II (Ang II) without Cyr61 knockdown, and Ang II with Cyr61 knockdown. CCK8, western blotting, and flow cytometry analysis were carried out to dissect cellular function. RESULTS A total of 23184 lncRNAs and 28264 mRNAs were normalized. 26 lncRNAs and 212 mRNAs were upregulated, and 74 lncRNAs and 233 mRNAs were downregulated after Cyr61 knockdown. Analysis of cellular components, molecular functions, biological processes, and regulatory pathways associated with the differentially expressed mRNAs revealed downstream mechanisms of the Cyr61 gene. The differentially expressed genes were affected for small cell lung cancer, axon guidance, Fc gamma R-mediated phagocytosis, MAPK signaling pathway, focal adhesion, insulin resistance, and metabolic pathways. In addition, Cyr61 expression was increased in accordance with induction of cell cycle arrest and apoptosis and inhibition of cell proliferation induced by Ang II. Knockdown of Cyr61 in HEK293T cells promoted cell cycle procession, decreased apoptosis, and promoted cell proliferation. CONCLUSIONS The Cyr61 gene is involved in Ang II-induced injury in HEK293T cells. Functional mechanisms of the differentially expressed lncRNAs and mRNAs as well as identification of metabolic pathways will provide new therapeutic targets for Cyr61-realated diseases.
Collapse
Affiliation(s)
- Junjie Wang
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Zhuang Autonomous Region, China
| | - Dongdong Fu
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Zhuang Autonomous Region, China
| | - Soulixay Senouthai
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Zhuang Autonomous Region, China
| | - Yan Jiang
- Department of Clinical Laboratories, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Zhuang Autonomous Region, China
| | - Rentong Hu
- Science Lab Center, Youjiang Medical University for Nationalities, Baise, Guangxi Zhuang Autonomous Region, China
| | - Yanwu You
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
44
|
Tang Y, Yang K, Zhao J, Liang X, Wang J. Evidence of Repurposing Drugs and Identifying Contraindications from Real World Study in Parkinson's Disease. ACS Chem Neurosci 2019; 10:954-963. [PMID: 30702853 DOI: 10.1021/acschemneuro.8b00456] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
There is great unmet need in discovering novel treatment for Parkinson's disease (PD) and identifying the new agents potentially causing drug-induced parkinsonism. New indications and contraindications of drugs are typically approved following rigorous randomized controlled trial (RCT) evaluation. However, RCTs have their inherent limitations, since they are usually conducted in ideal conditions, with high cost and limited follow-up periods. In the past decade, large cohort studies with long follow-up outcome data was derived from a PD database in a real-world setting. Studies based on real world data (RWD) can help to augment and extrapolate data obtained in RCTs and provide information about the safety and effectiveness of a medication in heterogeneous, large populations. In the present review, we focus on the published real world studies designed to develop new treatment strategies for repurposing drugs and identifying contraindications for PD. We also outline the challenges and limitations in these studies. Subsequently we introduce PaWei app platform, which hopefully can facilitate PD management and address real-world problems associated with PD. Better understanding of RWD collection and analysis is needed if RWD is to achieve its full potential.
Collapse
Affiliation(s)
- Yilin Tang
- Department of Neurology and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ke Yang
- Department of Neurology and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jue Zhao
- Department of Neurology and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xiaoniu Liang
- Department of Neurology and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jian Wang
- Department of Neurology and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
45
|
The depressor axis of the renin–angiotensin system and brain disorders: a translational approach. Clin Sci (Lond) 2018; 132:1021-1038. [DOI: 10.1042/cs20180189] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/19/2018] [Accepted: 04/23/2018] [Indexed: 02/07/2023]
Abstract
All the components of the classic renin–angiotensin system (RAS) have been identified in the brain. Today, the RAS is considered to be composed mainly of two axes: the pressor axis, represented by angiotensin (Ang) II/angiotensin-converting enzyme/AT1 receptors, and the depressor and protective one, represented by Ang-(1–7)/ angiotensin-converting enzyme 2/Mas receptors. Although the RAS exerts a pivotal role on electrolyte homeostasis and blood pressure regulation, their components are also implicated in higher brain functions, including cognition, memory, anxiety and depression, and several neurological disorders. Overactivity of the pressor axis of the RAS has been implicated in stroke and several brain disorders, such as cognitive impairment, dementia, and Alzheimer or Parkinson’s disease. The present review is focused on the role of the protective axis of the RAS in brain disorders beyond its effects on blood pressure regulation. Furthermore, the use of drugs targeting centrally RAS and its beneficial effects on brain disorders are also discussed.
Collapse
|