101
|
Lee SYH, Yates NJ, Tye SJ. Inflammatory Mechanisms in Parkinson's Disease: From Pathogenesis to Targeted Therapies. Neuroscientist 2021; 28:485-506. [PMID: 33586516 DOI: 10.1177/1073858421992265] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Inflammation is a critical factor contributing to the progressive neurodegenerative process observed in Parkinson's disease (PD). Microglia, the immune cells of the central nervous system, are activated early in PD pathogenesis and can both trigger and propagate early disease processes via innate and adaptive immune mechanisms such as upregulated immune cells and antibody-mediated inflammation. Downstream cytokines and gene regulators such as microRNA (miRNA) coordinate later disease course and mediate disease progression. Biomarkers signifying the inflammatory and neurodegenerative processes at play within the central nervous system are of increasing interest to clinical teams. To be effective, such biomarkers must achieve the highest sensitivity and specificity for predicting PD risk, confirming diagnosis, or monitoring disease severity. The aim of this review was to summarize the current preclinical and clinical evidence that suggests that inflammatory processes contribute to the initiation and progression of neurodegenerative processes in PD. In this article, we further summarize the data about main inflammatory biomarkers described in PD to date and their potential for regulation as a novel target for disease-modifying pharmacological strategies.
Collapse
Affiliation(s)
- Stellina Y H Lee
- Queensland Brain Institute, The University of Queensland, Saint Lucia, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Saint Lucia, Queensland, Australia
| | - Nathanael J Yates
- Queensland Brain Institute, The University of Queensland, Saint Lucia, Queensland, Australia.,School of Human Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Susannah J Tye
- Queensland Brain Institute, The University of Queensland, Saint Lucia, Queensland, Australia.,Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA.,Department of Psychiatry, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
102
|
Yuda N, Tanaka M, Tokushima M, Abe F. Safety evaluation of high-dose intake of casein-derived peptide Met-Lys-Pro in healthy adults: A randomized, double-blind, placebo-controlled trial. Food Sci Nutr 2021; 9:662-671. [PMID: 33598151 PMCID: PMC7866589 DOI: 10.1002/fsn3.2028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/22/2020] [Accepted: 11/10/2020] [Indexed: 12/15/2022] Open
Abstract
Met-Lys-Pro (MKP) is a casein-derived angiotensin I-converting enzyme inhibitory peptide with the potential to cross the blood-brain barrier. It has shown preventive effects against high blood pressure (BP) and cognitive decline in animal models and human studies. MKP shows good water solubility and can be blended into a variety of foods. However, its ease of intake may contribute to the possibility of overdose. Therefore, we aimed to evaluate the safety of high-dose intake of MKP in healthy adults by conducting a randomized controlled trial with 30 subjects. Participants were randomly allocated to the MKP (n = 15) or placebo (n = 15) group. Over 4 weeks, the MKP group received test powder containing a daily dose of 1,000 μg of MKP, five times the minimum effective dose for cognitive improvement, whereas the placebo group received dextrin powder containing no detectable MKP. No clinical problems were observed in anthropometric and BP measurements or in blood and urine parameters. No adverse events owing to MKP intake were observed. These findings suggest that consumption of MKP is safe, and that it may be applicable as a safe preventive measure against hypertension and cognitive decline in future.
Collapse
Affiliation(s)
- Naoki Yuda
- Food Ingredients and Technology InstituteMorinaga Milk Industry Co., Ltd.KanagawaJapan
| | - Miyuki Tanaka
- Food Ingredients and Technology InstituteMorinaga Milk Industry Co., Ltd.KanagawaJapan
| | | | - Fumiaki Abe
- Food Ingredients and Technology InstituteMorinaga Milk Industry Co., Ltd.KanagawaJapan
| |
Collapse
|
103
|
Wijeratne T, Wijeratne C, Karimi L, Sales C, Crewther SG. Case Report: Posterior Reversible Leukoencephalopathy Syndrome (PRES) as a Biologically Predictable Neurological Association in Severe COVID-19. First Reported Case From Australia and Review of Internationally Published Cases. Front Neurol 2021; 11:600544. [PMID: 33584499 PMCID: PMC7875083 DOI: 10.3389/fneur.2020.600544] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 12/31/2020] [Indexed: 12/18/2022] Open
Abstract
Reports of different types of neurological manifestations of COVID-19 are rapidly increasing, including changes of posterior reversible leukoencephalopathy syndrome (PRES). Here we describe the first reported case of COVID-19 and PRES in Australia diagnosed on basis of MRI brain imaging and confirmed clinically by presence of confusion, delirium, headaches, also associated with hypertension and blood pressure variability and stable long-term kidney problems. He made full recovery as his blood pressure was controlled and clinical status was supported with appropriate supportive therapy. Although traditionally a rare condition, PRES is likely to be more common among patients with COVID-19 pathobiology there is Renin downregulation of ACE2 receptors, involvement of Renin-Angiotensin-Aldosterone system, endotheliitis, cytokine storm, and hyper-immune response. Thus we advocate clinical suspicion and early brain imaging with MRI brain among vulnerable patients with known co-morbidities, and diagnosed with COVID-19 given that hypertension and blood pressure variability are often exacerbated by acute SARS-CoV-2 immune reactions. Such acute hypertensive encephalopathy was able to be reversed with timely supportive therapy ensuring re-hydration and re-establishment of blood pressure control.
Collapse
Affiliation(s)
- Tissa Wijeratne
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia.,Department of Neurology, Western Health & University Melbourne, Australian Institute for Musculoskeletal Science (AIMSS), Western Centre for Health Research & Education (WHCRE), Sunshine Hospital, St Albans, VIC, Australia.,Department of Medicine, Faculty of Medicine, University of Rajarata, Anuradhapura, Sri Lanka
| | - Chanith Wijeratne
- Department of Neurology, Western Health & University Melbourne, Australian Institute for Musculoskeletal Science (AIMSS), Western Centre for Health Research & Education (WHCRE), Sunshine Hospital, St Albans, VIC, Australia.,Monash Medical School, Clayton, VIC, Australia
| | - Leila Karimi
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia.,Department of Neurology, Western Health & University Melbourne, Australian Institute for Musculoskeletal Science (AIMSS), Western Centre for Health Research & Education (WHCRE), Sunshine Hospital, St Albans, VIC, Australia
| | - Carmela Sales
- Department of Neurology, Western Health & University Melbourne, Australian Institute for Musculoskeletal Science (AIMSS), Western Centre for Health Research & Education (WHCRE), Sunshine Hospital, St Albans, VIC, Australia
| | - Sheila Gillard Crewther
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia.,Department of Neurology, Western Health & University Melbourne, Australian Institute for Musculoskeletal Science (AIMSS), Western Centre for Health Research & Education (WHCRE), Sunshine Hospital, St Albans, VIC, Australia
| |
Collapse
|
104
|
Miners S, Kehoe PG, Love S. Cognitive impact of COVID-19: looking beyond the short term. Alzheimers Res Ther 2020; 12:170. [PMID: 33380345 PMCID: PMC7772800 DOI: 10.1186/s13195-020-00744-w] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023]
Abstract
COVID-19 is primarily a respiratory disease but up to two thirds of hospitalised patients show evidence of central nervous system (CNS) damage, predominantly ischaemic, in some cases haemorrhagic and occasionally encephalitic. It is unclear how much of the ischaemic damage is mediated by direct or inflammatory effects of virus on the CNS vasculature and how much is secondary to extracranial cardiorespiratory disease. Limited data suggest that the causative SARS-CoV-2 virus may enter the CNS via the nasal mucosa and olfactory fibres, or by haematogenous spread, and is capable of infecting endothelial cells, pericytes and probably neurons. Extracranially, SARS-CoV-2 targets endothelial cells and pericytes, causing endothelial cell dysfunction, vascular leakage and immune activation, sometimes leading to disseminated intravascular coagulation. It remains to be confirmed whether endothelial cells and pericytes in the cerebral vasculature are similarly targeted. Several aspects of COVID-19 are likely to impact on cognition. Cerebral white matter is particularly vulnerable to ischaemic damage in COVID-19 and is also critically important for cognitive function. There is accumulating evidence that cerebral hypoperfusion accelerates amyloid-β (Aβ) accumulation and is linked to tau and TDP-43 pathology, and by inducing phosphorylation of α-synuclein at serine-129, ischaemia may also increase the risk of development of Lewy body disease. Current therapies for COVID-19 are understandably focused on supporting respiratory function, preventing thrombosis and reducing immune activation. Since angiotensin-converting enzyme (ACE)-2 is a receptor for SARS-CoV-2, and ACE inhibitors and angiotensin receptor blockers are predicted to increase ACE-2 expression, it was initially feared that their use might exacerbate COVID-19. Recent meta-analyses have instead suggested that these medications are protective. This is perhaps because SARS-CoV-2 entry may deplete ACE-2, tipping the balance towards angiotensin II-ACE-1-mediated classical RAS activation: exacerbating hypoperfusion and promoting inflammation. It may be relevant that APOE ε4 individuals, who seem to be at increased risk of COVID-19, also have lowest ACE-2 activity. COVID-19 is likely to leave an unexpected legacy of long-term neurological complications in a significant number of survivors. Cognitive follow-up of COVID-19 patients will be important, especially in patients who develop cerebrovascular and neurological complications during the acute illness.
Collapse
Affiliation(s)
- Scott Miners
- Dementia Research Group, Bristol Medical School (THS), University of Bristol, Learning & Research level 1, Southmead Hospital, Bristol, BS10 5NB, UK.
| | - Patrick G Kehoe
- Dementia Research Group, Bristol Medical School (THS), University of Bristol, Learning & Research level 1, Southmead Hospital, Bristol, BS10 5NB, UK
| | - Seth Love
- Dementia Research Group, Bristol Medical School (THS), University of Bristol, Learning & Research level 1, Southmead Hospital, Bristol, BS10 5NB, UK.
| |
Collapse
|
105
|
Ribeiro VT, de Souza LC, Simões E Silva AC. Renin-Angiotensin System and Alzheimer's Disease Pathophysiology: From the Potential Interactions to Therapeutic Perspectives. Protein Pept Lett 2020; 27:484-511. [PMID: 31886744 DOI: 10.2174/0929866527666191230103739] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 08/27/2019] [Accepted: 11/16/2019] [Indexed: 12/21/2022]
Abstract
New roles of the Renin-Angiotensin System (RAS), apart from fluid homeostasis and Blood Pressure (BP) regulation, are being progressively unveiled, since the discoveries of RAS alternative axes and local RAS in different tissues, including the brain. Brain RAS is reported to interact with pathophysiological mechanisms of many neurological and psychiatric diseases, including Alzheimer's Disease (AD). Even though AD is the most common cause of dementia worldwide, its pathophysiology is far from elucidated. Currently, no treatment can halt the disease course. Successive failures of amyloid-targeting drugs have challenged the amyloid hypothesis and increased the interest in the inflammatory and vascular aspects of AD. RAS compounds, both centrally and peripherally, potentially interact with neuroinflammation and cerebrovascular regulation. This narrative review discusses the AD pathophysiology and its possible interaction with RAS, looking forward to potential therapeutic approaches. RAS molecules affect BP, cerebral blood flow, neuroinflammation, and oxidative stress. Angiotensin (Ang) II, via angiotensin type 1 receptors may promote brain tissue damage, while Ang-(1-7) seems to elicit neuroprotection. Several studies dosed RAS molecules in AD patients' biological material, with heterogeneous results. The link between AD and clinical conditions related to classical RAS axis overactivation (hypertension, heart failure, and chronic kidney disease) supports the hypothesized role of this system in AD. Additionally, RAStargeting drugs as Angiotensin Converting Enzyme inhibitors (ACEis) and Angiotensin Receptor Blockers (ARBs) seem to exert beneficial effects on AD. Results of randomized controlled trials testing ACEi or ARBs in AD are awaited to elucidate whether AD-RAS interaction has implications on AD therapeutics.
Collapse
Affiliation(s)
- Victor Teatini Ribeiro
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Leonardo Cruz de Souza
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil.,Department of Internal Medicine, Service of Neurology, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Ana Cristina Simões E Silva
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
106
|
Haghighi MM, Kakhki EG, Sato C, Ghani M, Rogaeva E. The Intersection between COVID-19, the Gene Family of ACE2 and Alzheimer's Disease. Neurosci Insights 2020; 15:2633105520975743. [PMID: 33283188 PMCID: PMC7686598 DOI: 10.1177/2633105520975743] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 11/03/2020] [Indexed: 01/08/2023] Open
Abstract
We reviewed factors that might influence COVID-19 outcomes (eg, neurological symptoms), including the link to Alzheimer's disease. Since the virus triggers COVID-19 infection through binding to ACE2, we focused on the ACE2 gene family, including ACE. Both ACE2 and ACE are involved in the renin-angiotensin system (RAS). In general, ACE causes inflammation and vasoconstriction, while ACE2 leads to anti-inflammation activity and vasodilation. The disturbed balance between these counter-regulatory pathways could influence susceptibility to COVID-19. Notably, dysregulation of the RAS-equilibrium contributes to Alzheimer's disease. Differences in the incidence and symptoms of COVID-19 in diverse populations could be attributed to variability in the human genome. For example, ACE and ACE2 variations could modify the outcome of COVID-19 in different populations. It would be important to conduct genome-wide studies to detect variants influencing COVID-19 presentation, with a special focus on variants affecting immune-related pathways and expression of RAS-related genes.
Collapse
Affiliation(s)
- Mahdi Montazer Haghighi
- Tanz Centre for Research in
Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Erfan Ghani Kakhki
- Tanz Centre for Research in
Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- DisorDATA Analytics, Ottawa, ON,
Canada
| | - Christine Sato
- Tanz Centre for Research in
Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | | | - Ekaterina Rogaeva
- Tanz Centre for Research in
Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Division of Neurology, Department of
Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|
107
|
Panariello F, Cellini L, Speciani M, De Ronchi D, Atti AR. How Does SARS-CoV-2 Affect the Central Nervous System? A Working Hypothesis. Front Psychiatry 2020; 11:582345. [PMID: 33304284 PMCID: PMC7701095 DOI: 10.3389/fpsyt.2020.582345] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 10/07/2020] [Indexed: 12/15/2022] Open
Abstract
Interstitial pneumonia was the first manifestation to be recognized as caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2); however, in just a few weeks, it became clear that the coronavirus disease-2019 (COVID-19) overrun tissues and more body organs than just the lungs, so much so that it could be considered a systemic pathology. Several studies reported the involvement of the conjunctiva, the gut, the heart and its pace, and vascular injuries such as thromboembolic complications and Kawasaki disease in children and toddlers were also described. More recently, it was reported that in a sample of 214 SARS-CoV-2 positive patients, 36.4% complained of neurological symptoms ranging from non-specific manifestations (dizziness, headache, and seizures), to more specific symptoms such hyposmia or hypogeusia, and stroke. Older individuals, especially males with comorbidities, appear to be at the highest risk of developing such severe complications related to the Central Nervous System (CNS) involvement. Neuropsychiatric manifestations in COVID-19 appear to develop in patients with and without pre-existing neurological disorders. Growing evidence suggests that SARS-CoV-2 binds to the human Angiotensin-Converting Enzyme 2 (ACE2) for the attachment and entrance inside host cells. By describing ACE2 and the whole Renin Angiotensin Aldosterone System (RAAS) we may better understand whether specific cell types may be affected by SARS-CoV-2 and whether their functioning can be disrupted in case of an infection. Since clear evidences of neurological interest have already been shown, by clarifying the topographical distribution and density of ACE2, we will be able to speculate how SARS-CoV-2 may affect the CNS and what is the pathogenetic mechanism by which it contributes to the specific clinical manifestations of the disease. Based on such evidences, we finally hypothesize the process of SARS-CoV-2 invasion of the CNS and provide a possible explanation for the onset or the exacerbation of some common neuropsychiatric disorders in the elderly including cognitive impairment and Alzheimer disease.
Collapse
Affiliation(s)
- Fabio Panariello
- Department of Mental Health, Local Health Authorities, Bologna, Italy
| | - Lorenzo Cellini
- Department of Biomedical and Neuromotor Sciences, Psychiatry, Bologna University, Bologna, Italy
| | - Maurizio Speciani
- Department of Biomedical and Neuromotor Sciences, Psychiatry, Bologna University, Bologna, Italy
| | - Diana De Ronchi
- Department of Biomedical and Neuromotor Sciences, Psychiatry, Bologna University, Bologna, Italy
| | - Anna Rita Atti
- Department of Biomedical and Neuromotor Sciences, Psychiatry, Bologna University, Bologna, Italy
| |
Collapse
|
108
|
Li X, Xuan W, Chen D, Gao H, Wang G, Guo Q, Wang Y, Song H, Cai B. Research Progress of Alzheimer's Disease Therapeutic Drugs: Based on Renin-Angiotensin System Axis. J Alzheimers Dis 2020; 78:1315-1338. [PMID: 33164932 DOI: 10.3233/jad-200770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
It is widely recognized that Alzheimer's disease (AD) has a complicate link to renin-angiotensin system (RAS). It is known that cerebrovascular disease has some connections with AD, but most of the studies are still conducted in parallel or independently. Although previous research came up with large number of hypotheses about the pathogenesis of AD, it does not include the mechanism of RAS-related regulation of AD. It has been found that many components of RAS have been changed in AD. For example, the multifunctional and high-efficiency vasoconstrictor Ang II and Ang III with similar effects are changed under the action of other RAS signal peptides; these signal peptides are believed to help improve nerve injury and cognitive function. These changes may lead to neuropathological changes of AD, and progressive defects of cognitive function, which are association with some hypotheses of AD. The role of RAS in AD gradually attracts our attention, and RAS deserved to be considered carefully in the pathogenesis of AD. This review discusses the mechanisms of RAS participating in the three current hypotheses of AD: neuroinflammation, oxidative stress and amyloid-β protein (Aβ) hypothesis, as well as the drugs that regulate RAS systems already in clinical or in clinical trials. It further demonstrates the importance of RAS in the pathogenesis of AD, not only because of its multiple aspects of participation, which may be accidental, but also because of the availability of RAS drugs, which can be reused as therapies of AD.
Collapse
Affiliation(s)
- Xinquan Li
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Weiting Xuan
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Dabao Chen
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Huawu Gao
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Guangyun Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Qiaoru Guo
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the Fifth Affiliated Hospital and School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yan Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Hang Song
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Biao Cai
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| |
Collapse
|
109
|
Bouças AP, Rheinheimer J, Lagopoulos J. Why Severe COVID-19 Patients Are at Greater Risk of Developing Depression: A Molecular Perspective. Neuroscientist 2020; 28:11-19. [PMID: 33135582 DOI: 10.1177/1073858420967892] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The prevailing evidence suggests that patients with severe COVID-19 seem to have an overreaction of the immune system demonstrating exacerbated levels of inflammation caused by a "cytokine storm." At this early stage, the mechanisms underpinning COVID-19 are still subject to intense scrutiny and the long-term mental health consequences as a result of the disease are unknown. Here we discuss the hypothesis that patients who survive severe COVID-19 and who experience significant activation of the immune system, are at greater risk of developing depression. We posit that a phenomenon known as cytokine storm dramatically activates the enzyme indoleamine 2,3-dioxygenase (IDO-1), resulting in the increase in kynurenine metabolites. Kynurenine is metabolized by IDO-1 in the brain, producing chemokines, in which a prolonged exposure may result long-term brain impairment. In this article, we also propose the possibility that a SARS-CoV-2 neuroinvasion increases the local levels of angiotensin II by angiotensin-converting enzyme 2 down-regulation. Thereby, angiotensin II could increase kynurenine metabolites producing pro-oxidative and pro-inflammatory effects, resulting in impairment of cognitive function, enhanced oxidative stress and decreased brain-derived neurotrophic factor. It is our premise that patients who experience such a cytokine storm may be at increased risk of long-term mental illness, such as depression.
Collapse
Affiliation(s)
- Ana P Bouças
- Sunshine Coast Mind and Neuroscience Thompson Institute, University of the Sunshine Coast, Sunshine Coast, Queensland, Australia
| | - Jakeline Rheinheimer
- Postgraduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Jim Lagopoulos
- Sunshine Coast Mind and Neuroscience Thompson Institute, University of the Sunshine Coast, Sunshine Coast, Queensland, Australia
| |
Collapse
|
110
|
Linial M, Stern A, Weinstock M. Effect of ladostigil treatment of aging rats on gene expression in four brain areas associated with regulation of memory. Neuropharmacology 2020; 177:108229. [PMID: 32738309 DOI: 10.1016/j.neuropharm.2020.108229] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/17/2020] [Accepted: 07/01/2020] [Indexed: 02/09/2023]
Abstract
Episodic and spatial memory decline in aging and are controlled by the hippocampus, perirhinal, frontal and parietal cortices and the connections between them. Ladostigil, a drug with antioxidant and anti-inflammatory activity, was shown to prevent the loss of episodic and spatial memory in aging rats. To better understand the molecular effects of aging and ladostigil on these brain regions we characterized the changes in gene expression using RNA-sequencing technology in rats aged 6 and 22 months. We found that the changes induced by aging and chronic ladostigil treatment were brain region specific. In the hippocampus, frontal and perirhinal cortex, ladostigil decreased the overexpression of genes regulating calcium homeostasis, ion channels and those adversely affecting synaptic function. In the parietal cortex, ladostigil increased the expression of several genes that provide neurotrophic support, while reducing that of pro-apoptotic genes and those encoding pro-inflammatory cytokines and their receptors. Ladostigil also decreased the expression of axonal growth inhibitors and those impairing mitochondrial function. Together, these actions could explain the protection by ladostigil against age-related memory decline.
Collapse
Affiliation(s)
- Michal Linial
- Department of Biological Chemistry, Life Science Institute, Israel; The Rachel and Selim Benin School of Computer Science and Engineering, Israel
| | - Amos Stern
- Department of Biological Chemistry, Life Science Institute, Israel
| | - Marta Weinstock
- Institute of Drug Research, School of Pharmacy, The Hebrew University of Jerusalem, Israel.
| |
Collapse
|
111
|
Labandeira-Garcia JL, Valenzuela R, Costa-Besada MA, Villar-Cheda B, Rodriguez-Perez AI. The intracellular renin-angiotensin system: Friend or foe. Some light from the dopaminergic neurons. Prog Neurobiol 2020; 199:101919. [PMID: 33039415 PMCID: PMC7543790 DOI: 10.1016/j.pneurobio.2020.101919] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 08/20/2020] [Accepted: 10/04/2020] [Indexed: 12/11/2022]
Abstract
The renin-angiotensin system (RAS) is one of the oldest hormone systems in vertebrate phylogeny. RAS was initially related to regulation of blood pressure and sodium and water homeostasis. However, local or paracrine RAS were later identified in many tissues, including brain, and play a major role in their physiology and pathophysiology. In addition, a major component, ACE2, is the entry receptor for SARS-CoV-2. Overactivation of tissue RAS leads several oxidative stress and inflammatory processes involved in aging-related degenerative changes. In addition, a third level of RAS, the intracellular or intracrine RAS (iRAS), with still unclear functions, has been observed. The possible interaction between the intracellular and extracellular RAS, and particularly the possible deleterious or beneficial effects of the iRAS activation are controversial. The dopaminergic system is particularly interesting to investigate the RAS as important functional interactions between dopamine and RAS have been observed in the brain and several peripheral tissues. Our recent observations in mitochondria and nucleus of dopaminergic neurons may clarify the role of the iRAS. This may be important for the developing of new therapeutic strategies, since the effects on both extracellular and intracellular RAS must be taken into account, and perhaps better understanding of COVID-19 cell mechanisms.
Collapse
Affiliation(s)
- Jose L Labandeira-Garcia
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CiberNed), Madrid, Spain.
| | - Rita Valenzuela
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CiberNed), Madrid, Spain
| | - Maria A Costa-Besada
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CiberNed), Madrid, Spain
| | - Begoña Villar-Cheda
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CiberNed), Madrid, Spain
| | - Ana I Rodriguez-Perez
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CiberNed), Madrid, Spain
| |
Collapse
|
112
|
Cosarderelioglu C, Nidadavolu LS, George CJ, Oh ES, Bennett DA, Walston JD, Abadir PM. Brain Renin-Angiotensin System at the Intersect of Physical and Cognitive Frailty. Front Neurosci 2020; 14:586314. [PMID: 33117127 PMCID: PMC7561440 DOI: 10.3389/fnins.2020.586314] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 08/25/2020] [Indexed: 12/15/2022] Open
Abstract
The renin–angiotensin system (RAS) was initially considered to be part of the endocrine system regulating water and electrolyte balance, systemic vascular resistance, blood pressure, and cardiovascular homeostasis. It was later discovered that intracrine and local forms of RAS exist in the brain apart from the endocrine RAS. This brain-specific RAS plays essential roles in brain homeostasis by acting mainly through four angiotensin receptor subtypes; AT1R, AT2R, MasR, and AT4R. These receptors have opposing effects; AT1R promotes vasoconstriction, proliferation, inflammation, and oxidative stress while AT2R and MasR counteract the effects of AT1R. AT4R is critical for dopamine and acetylcholine release and mediates learning and memory consolidation. Consequently, aging-associated dysregulation of the angiotensin receptor subtypes may lead to adverse clinical outcomes such as Alzheimer’s disease and frailty via excessive oxidative stress, neuroinflammation, endothelial dysfunction, microglial polarization, and alterations in neurotransmitter secretion. In this article, we review the brain RAS from this standpoint. After discussing the functions of individual brain RAS components and their intracellular and intracranial locations, we focus on the relationships among brain RAS, aging, frailty, and specific neurodegenerative diseases, such as Alzheimer’s disease, Parkinson’s disease, and vascular cognitive impairment, through oxidative stress, neuroinflammation, and vascular dysfunction. Finally, we discuss the effects of RAS-modulating drugs on the brain RAS and their use in novel treatment approaches.
Collapse
Affiliation(s)
- Caglar Cosarderelioglu
- Division of Geriatrics, Department of Internal Medicine, Ankara University School of Medicine, Ankara, Turkey.,Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lolita S Nidadavolu
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Claudene J George
- Division of Geriatrics, Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, United States
| | - Esther S Oh
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, United States
| | - Jeremy D Walston
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Peter M Abadir
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
113
|
Campos J, Pacheco R. Involvement of dopaminergic signaling in the cross talk between the renin-angiotensin system and inflammation. Semin Immunopathol 2020; 42:681-696. [PMID: 32997225 PMCID: PMC7526080 DOI: 10.1007/s00281-020-00819-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 09/25/2020] [Indexed: 12/11/2022]
Abstract
The renin-angiotensin system (RAS) is a fundamental regulator of blood pressure and has emerged as an important player in the control of inflammatory processes. Accordingly, imbalance on RAS components either systemically or locally might trigger the development of inflammatory disorders by affecting immune cells. At the same time, alterations in the dopaminergic system have been consistently involved in the physiopathology of inflammatory disorders. Accordingly, the interaction between the RAS and the dopaminergic system has been studied in the context of inflammation of the central nervous system (CNS), kidney, and intestine, where they exert antagonistic actions in the regulation of the immune system. In this review, we summarized, integrated, and discussed the cross talk of the dopaminergic system and the RAS in the regulation of inflammatory pathologies, including neurodegenerative disorders, such as Parkinson’s disease. We analyzed the molecular mechanisms underlying the interaction between both systems in the CNS and in systemic pathologies. Moreover, we also analyzed the impact of the commensal microbiota in the regulation of RAS and dopaminergic system and how it is involved in inflammatory disorders. Furthermore, we summarized the therapeutic approaches that have yielded positive results in preclinical or clinical studies regarding the use of drugs targeting the RAS and dopaminergic system for the treatment of inflammatory conditions. Further understanding of the molecular and cellular regulation of the RAS-dopaminergic cross talk should allow the formulation of new therapies consisting of novel drugs and/or repurposing already existing drugs, alone or in combination, for the treatment of inflammatory disorders.
Collapse
Affiliation(s)
- Javier Campos
- Laboratorio de Neuroinmunología, Fundación Ciencia & Vida, Av. Zañartu 1482, 7780272 Ñuñoa, Santiago, Chile
| | - Rodrigo Pacheco
- Laboratorio de Neuroinmunología, Fundación Ciencia & Vida, Av. Zañartu 1482, 7780272 Ñuñoa, Santiago, Chile. .,Universidad San Sebastián, 7510156 Providencia, Santiago, Chile.
| |
Collapse
|
114
|
Vadhan JD, Speth RC. The role of the brain renin-angiotensin system (RAS) in mild traumatic brain injury (TBI). Pharmacol Ther 2020; 218:107684. [PMID: 32956721 DOI: 10.1016/j.pharmthera.2020.107684] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2020] [Indexed: 02/07/2023]
Abstract
There is considerable interest in traumatic brain injury (TBI) induced by repeated concussions suffered by athletes in sports, military personnel from combat-and non-combat related activities, and civilian populations who suffer head injuries from accidents and domestic violence. Although the renin-angiotensin system (RAS) is primarily a systemic cardiovascular regulatory system that, when dysregulated, causes hypertension and cardiovascular pathology, the brain contains a local RAS that plays a critical role in the pathophysiology of several neurodegenerative diseases. This local RAS includes receptors for angiotensin (Ang) II within the brain parenchyma, as well as on circumventricular organs outside the blood-brain-barrier. The brain RAS acts primarily via the type 1 Ang II receptor (AT1R), exacerbating insults and pathology. With TBI, the brain RAS may contribute to permanent brain damage, especially when a second TBI occurs before the brain recovers from an initial injury. Agents are needed that minimize the extent of injury from an acute TBI, reducing TBI-mediated permanent brain damage. This review discusses how activation of the brain RAS following TBI contributes to this damage, and how drugs that counteract activation of the AT1R including AT1R blockers (ARBs), renin inhibitors, angiotensin-converting enzyme (ACE) inhibitors, and agonists at type 2 Ang II receptors (AT2) and at Ang (1-7) receptors (Mas) can potentially ameliorate TBI-induced brain damage.
Collapse
Affiliation(s)
- Jason D Vadhan
- College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States of America
| | - Robert C Speth
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, United States of America; School of Medicine, Georgetown University, Washington, DC, United States of America.
| |
Collapse
|
115
|
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
|
116
|
Tada AM, Hamezah HS, Yanagisawa D, Morikawa S, Tooyama I. Neuroprotective Effects of Casein-Derived Peptide Met-Lys-Pro (MKP) in a Hypertensive Model. Front Neurosci 2020; 14:845. [PMID: 32922259 PMCID: PMC7457086 DOI: 10.3389/fnins.2020.00845] [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: 04/27/2020] [Accepted: 07/20/2020] [Indexed: 01/08/2023] Open
Abstract
We have previously reported that casein hydrolysate, CH-3, from bovine milk and casein-derived tripeptide Met-Lys-Pro (MKP) has ACE inhibitory activity and reduces blood pressure. In this study, we investigated the therapeutic effects of MKP in a hypertensive rat model (7-week-old male SHRSP/Izm rats). For long term evaluation, rats were fed either a diet containing CH-3 or normal diet. The survival rate of SHRSP rats was significantly improved by intake of CH-3 for 181 days. For short term evaluation, rats were orally administered synthetic tripeptide MKP or distilled water for 4 weeks. MRI study demonstrated that hemorrhagic lesions were observed in two of five rats in the control group, while no hemorrhagic lesions were observed in the MKP group. Volumetric analysis using MRI revealed that MKP administration inhibited atrophy of diencephalic regions. Histological examinations revealed that hemorrhage areas and astrogliosis in the hippocampus and cerebral cortex were lower in the MKP group than in the control group. Gene expression analysis indicated that MKP administration reduced expression of genes related to cerebral circulation insufficiency such as immune responses (Cd74 and Prkcd), response to hypoxia (Ddit4, Apold1, and Prkcd), reactive oxygen species metabolic process (Ddit4 and Pdk4), and apoptotic process (Ddit4, Prkcd, and Sgk1), suggesting that MKP administration prevented cerebral ischemia associated with hypertension. In addition, some genes encoding responses to hormone stimulus (Fos, Dusp1, and Sik1) were also downregulated. Serum aldosterone and corticosterone levels were also significantly decreased following MKP administration. The present study indicates that MKP shows neuroprotective effects in SHRSP rats by regulating cerebral circulation insufficiency and corticoid levels. MKP administration may therefore be a potential therapeutic strategy for hypertensive brain diseases such as cerebrovascular disease.
Collapse
Affiliation(s)
- Asuka Matsuzaki Tada
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Japan.,Functional Food Ingredients Group, Food Ingredients and Technology Institute, R&D Division, Morinaga Milk Industry Co., Ltd., Zama, Japan
| | | | - Daijiro Yanagisawa
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Japan
| | - Shigehiro Morikawa
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Japan
| | - Ikuo Tooyama
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
117
|
Abo-Youssef AM, Khallaf WA, Khattab MM, Messiha BA. The anti-Alzheimer effect of telmisartan in a hyperglycemic ovariectomized rat model; role of central angiotensin and estrogen receptors. Food Chem Toxicol 2020; 142:111441. [DOI: 10.1016/j.fct.2020.111441] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/14/2020] [Accepted: 05/17/2020] [Indexed: 12/19/2022]
|
118
|
Royea J, Hamel E. Brain angiotensin II and angiotensin IV receptors as potential Alzheimer's disease therapeutic targets. GeroScience 2020; 42:1237-1256. [PMID: 32700176 DOI: 10.1007/s11357-020-00231-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that is multifactorial in nature. Yet, despite being the most common form of dementia in the elderly, AD's primary cause remains unknown. As such, there is currently little to offer AD patients as the vast majority of recently tested therapies have either failed in well-controlled clinical trials or inadequately treat AD. Recently, emerging preclinical and clinical evidence has associated the brain renin angiotensin system (RAS) to AD pathology. Accordingly, various components of the brain RAS were shown to be altered in AD patients and mouse models, including the angiotensin II type 1 (AT1R), angiotensin IV receptor (AT4R), and Mas receptors. Collectively, the changes observed within the RAS have been proposed to contribute to many of the neuropathological hallmarks of AD, including the neuronal, cognitive, and vascular dysfunctions. Accumulating evidence has additionally identified antihypertensive medications targeting the RAS, particularly angiotensin receptor blockers (ARBs) and angiotensin-converting enzyme inhibitors (ACEIs), to delay AD onset and progression. In this review, we will discuss the emergence of the RAS's involvement in AD and highlight putative mechanisms of action underlying ARB's beneficial effects that may explain their ability to modify the risk of developing AD or AD progression. The RAS may provide novel molecular targets for recovering memory pathways, cerebrovascular function, and other pathological landmarks of AD.
Collapse
Affiliation(s)
- Jessika Royea
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, Montréal, QC, H3A 2B4, Canada
| | - Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, Montréal, QC, H3A 2B4, Canada.
| |
Collapse
|
119
|
Franco R, Rivas-Santisteban R, Serrano-Marín J, Rodríguez-Pérez AI, Labandeira-García JL, Navarro G. SARS-CoV-2 as a Factor to Disbalance the Renin–Angiotensin System: A Suspect in the Case of Exacerbated IL-6 Production. THE JOURNAL OF IMMUNOLOGY 2020; 205:1198-1206. [DOI: 10.4049/jimmunol.2000642] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023]
|
120
|
Yao YY, Ling EA, Lu D. Microglia mediated neuroinflammation - signaling regulation and therapeutic considerations with special reference to some natural compounds. Histol Histopathol 2020; 35:1229-1250. [PMID: 32662061 DOI: 10.14670/hh-18-239] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Neuroinflammation plays a central role in multiple neurodegenerative diseases and neurological disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), cerebral ischemic injury etc. In this connection, microglia, the key players in the central nervous system, mediate the inflammatory response process. In brain injuries, activated microglia can clear the cellular debris and invading pathogens and release neurotrophic factors; however, prolonged microglia activation may cause neuronal death through excessive release of inflammatory mediators. Therefore, it is of paramount importance to understand the underlying molecular mechanisms of microglia activation to design an effective therapeutic strategy to alleviate neuronal injury. Recent studies have shown that some natural compounds and herbal extracts possess anti-inflammatory properties that may suppress microglial activation and ameliorate neuroinflammation and hence are neuroprotective. In this review, we will update some of the common signaling pathways that regulate microglia activation. Among the various signaling pathways, the Notch-1, mitogen-activated protein kinases (MAPKs), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) have been reported to exacerbate microglia mediated neuroinflammation that is implicated in different neuropathological diseases. The search for natural compounds or agents, specifically those derived from natural herbal extracts such as Gastrodin, scutellarin, RG1 etc. has been the focus of many of our recent studies because they have been found to regulate microglia activation. The pharmacological effects of these agents and their potential mechanisms for regulating microglia activation are systematically reviewed here for a fuller understanding of their biochemical action and therapeutic potential for treatment of microglia mediated neuropathological diseases.
Collapse
Affiliation(s)
- Yue-Yi Yao
- Technology Transfer Center, Kunming Medical University, Kunming, China
| | - Eng-Ang Ling
- Department of Anatomy, Young Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Di Lu
- Technology Transfer Center, Kunming Medical University, Kunming, China.
| |
Collapse
|
121
|
Assis AD, Mascarenhas FNADP, Araújo FDA, Santos RAS, Zanon RG. Angiotensin-(1-7) receptor Mas antagonist (A779) influenced gliosis and reduced synaptic density in the spinal cord after peripheral axotomy. Peptides 2020; 129:170329. [PMID: 32437718 DOI: 10.1016/j.peptides.2020.170329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 12/20/2022]
Abstract
The peptide angiotensin-(1-7) [Ang (1-7)] and its receptor Mas are involved in controlling arterial pressure and display actions on the nervous system. In a previous study, our laboratory showed that A779 [(peptidyl antagonist of the Ang-(1-7)] treatment had a negative effect following a lesion of the sciatic nerve, possibly by delaying the responses of Schwann cells, resulting in a decreased axonal organization along with a slowed functional return. In the present work, we investigated the central cellular changes after sciatic nerve injury in rodents treated with A779 after two weeks. In the lumbar spinal cords, where the neuronal bodies that make up the sciatic are, the treatment with A779 showed reduced reactivity of astrocytes (p = 0.004, Mann-Whitney U test) and less synaptic density (p = 0.004, Mann-Whitney U test) after injury. Also, the treatment upregulated microglia activity in both sides (p = 0.004, Mann-Whitney U test), ipsilateral and contralateral to the lesion, of the spinal cord. In addition, the Mas expression in spine neurons was increased in response to axotomy especially after two weeks (p = 0.03, Mann-Whitney U test) following the nerve lesion in comparison to earlier stages after injury. Therefore, we can conclude that Ang-(1-7)/Mas axis plays a role during spinal cord recovery after peripheral nerve injury.
Collapse
Affiliation(s)
- Alex Dias Assis
- Department of Human Anatomy, Institute of Biomedical Sciences, Federal University of Uberlandia (UFU), Uberlandia, MG, Brazil
| | | | - Fernanda de Assis Araújo
- Department of Physiology, Institute of Biomedical Sciences, Federal University of Uberlandia (UFU), Uberlandia, MG, Brazil
| | | | - Renata Graciele Zanon
- Department of Human Anatomy, Institute of Biomedical Sciences, Federal University of Uberlandia (UFU), Uberlandia, MG, Brazil.
| |
Collapse
|
122
|
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
|
123
|
Wang H, Tang X, Fan H, Luo Y, Song Y, Xu Y, Chen Y. Potential mechanisms of hemorrhagic stroke in elderly COVID-19 patients. Aging (Albany NY) 2020; 12:10022-10034. [PMID: 32527987 PMCID: PMC7346040 DOI: 10.18632/aging.103335] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 05/14/2020] [Indexed: 04/11/2023]
Abstract
The novel severe acute respiratory syndrome coronavirus 2 is the causative agent of coronavirus disease 2019, a new human infectious disease. While fever, cough, and respiratory distress are typical first symptoms, a fraction of those affected present instead with neurological symptoms suggestive of central nervous system compromise. This review summarizes the potential contribution of coronavirus disease 2019 to hemorrhagic stroke in the elderly and proposes possible mechanisms. Reports show that the most affected patients have underlying chronic diseases such as hypertension and diabetes, which are two key risk factors for hemorrhagic stroke. Angiotensin-converting enzyme 2 is the main host cell surface receptor interacting with the severe acute respiratory syndrome coronavirus 2 spike glycoprotein to allow viral entry and infection. We speculate that ensuing downregulation of angiotensin-converting enzyme 2 expression may compound the risk conferred by pre-existing comorbidities and critically influence the pathogenesis of hemorrhagic stroke by elevating blood pressure and impairing cerebrovascular endothelial function. Additionally, both age- and/or disease-related immune dysfunction and enhanced catecholamine release secondary to anxiety and stress may also aggravate central nervous system symptoms of severe acute respiratory syndrome coronavirus 2 infection. Thus, assessment of systemic inflammatory biomarkers and tight control of hemodynamic parameters upon admission are crucial to minimize mortality and morbidity in coronavirus disease 2019 patients with central nervous system symptoms suggestive of incipient stroke.
Collapse
Affiliation(s)
- Haili Wang
- Department of Neurology, Clinical Medical College, Yangzhou University, Yangzhou 225000, Jiangsu, China
- Department of Neurology, Clinical Medical College of Yangzhou, Dalian Medical University, Yangzhou 225000, Jiangsu, China
| | - Xiaojia Tang
- Department of Neurology, Clinical Medical College, Yangzhou University, Yangzhou 225000, Jiangsu, China
- Department of Neurology, Clinical Medical College of Yangzhou, Dalian Medical University, Yangzhou 225000, Jiangsu, China
| | - Hongyang Fan
- Department of Neurology, Clinical Medical College, Yangzhou University, Yangzhou 225000, Jiangsu, China
| | - Yuhan Luo
- Department of Neurology, Clinical Medical College, Yangzhou University, Yangzhou 225000, Jiangsu, China
- Department of Neurology, Clinical Medical College of Yangzhou, Dalian Medical University, Yangzhou 225000, Jiangsu, China
| | - Yuxia Song
- Department of Neurology, Clinical Medical College, Yangzhou University, Yangzhou 225000, Jiangsu, China
- Department of Neurology, Clinical Medical College of Yangzhou, Dalian Medical University, Yangzhou 225000, Jiangsu, China
| | - Yao Xu
- Department of Neurology, Clinical Medical College, Yangzhou University, Yangzhou 225000, Jiangsu, China
| | - Yingzhu Chen
- Department of Neurology, Clinical Medical College, Yangzhou University, Yangzhou 225000, Jiangsu, China
| |
Collapse
|
124
|
Yuda N, Tanaka M, Yamauchi K, Abe F, Kakiuchi I, Kiyosawa K, Miyasaka M, Sakane N, Nakamura M. Effect of the Casein-Derived Peptide Met-Lys-Pro on Cognitive Function in Community-Dwelling Adults Without Dementia: A Randomized, Double-Blind, Placebo-Controlled Trial. Clin Interv Aging 2020; 15:743-754. [PMID: 32546992 PMCID: PMC7266326 DOI: 10.2147/cia.s253116] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background Preventative measures have recently been taken to reduce the incidence of Alzheimer’s disease worldwide. We previously showed that Met-Lys-Pro (MKP), a casein-derived angiotensin-converting enzyme inhibitory peptide with the potential to cross the blood–brain barrier, attenuated cognitive decline in a mouse model of Alzheimer’s disease. However, the effect of MKP on cognitive function improvement in humans remains unknown. This exploratory study sought to investigate whether MKP intake could improve cognitive function in adults without dementia. Methods A total of 268 community-dwelling adults without dementia participated in this 24-week randomized controlled trial. Participants were randomly allocated to the MKP (n = 134) or placebo (n = 134) group. The MKP group received four tablets daily, each containing 50 μg MKP, while the placebo group received four dextrin tablets containing no detectable MKP for 24 weeks. Scores on the Japanese version of the cognitive subscale of the Alzheimer’s Disease Assessment Scale (ADAS-cog) were used as the primary outcome to compare cognitive function between the MKP and placebo groups. The study products were also evaluated for safety. Results The intention-to-treat analysis showed that there was no significant difference between the groups in terms of the ADAS-cog total score. Orientation, as measured by the respective ADAS-cog subscale, was significantly improved compared to placebo at 24 weeks post-MKP administration (P = 0.022). No serious adverse events due to MKP intake were observed. Conclusion To the best of our knowledge, this is the first study to report the effects of MKP on human cognition. These preliminary results suggested the safety of daily MKP intake and its potential to improve orientation in adults without dementia. Further clinical studies are needed to confirm the present findings and the benefits of MKP on cognitive function.
Collapse
Affiliation(s)
- Naoki Yuda
- Food Ingredients and Technology Institute, Morinaga Milk Industry Co., Ltd., Zama, Kanagawa, Japan
| | - Miyuki Tanaka
- Food Ingredients and Technology Institute, Morinaga Milk Industry Co., Ltd., Zama, Kanagawa, Japan
| | - Koji Yamauchi
- Food Ingredients and Technology Institute, Morinaga Milk Industry Co., Ltd., Zama, Kanagawa, Japan
| | - Fumiaki Abe
- Food Ingredients and Technology Institute, Morinaga Milk Industry Co., Ltd., Zama, Kanagawa, Japan
| | - Izumi Kakiuchi
- Department of Nursing, Matsumoto Junior College, Matsumoto, Nagano, Japan
| | - Kyoko Kiyosawa
- Department of Nursing, Matsumoto Junior College, Matsumoto, Nagano, Japan
| | - Mitsunaga Miyasaka
- Department of Nursing, Matsumoto Junior College, Matsumoto, Nagano, Japan
| | - Naoki Sakane
- Division of Preventive Medicine, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | | |
Collapse
|
125
|
Cao DY, Saito S, Veiras LC, Okwan-Duodu D, Bernstein EA, Giani JF, Bernstein KE, Khan Z. Role of angiotensin-converting enzyme in myeloid cell immune responses. Cell Mol Biol Lett 2020; 25:31. [PMID: 32508938 PMCID: PMC7249647 DOI: 10.1186/s11658-020-00225-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 05/05/2020] [Indexed: 02/07/2023] Open
Abstract
Angiotensin-converting enzyme (ACE), a dicarboxypeptidase, plays a major role in the regulation of blood pressure by cleaving angiotensin I into angiotensin II (Ang II), a potent vasoconstrictor. Because of its wide substrate specificity and tissue distribution, ACE affects many diverse biological processes. In inflammatory diseases, including granuloma, atherosclerosis, chronic kidney disease and bacterial infection, ACE expression gets upregulated in immune cells, especially in myeloid cells. With increasing evidences connecting ACE functions to the pathogenesis of these acquired diseases, it is suggested that ACE plays a vital role in immune functions. Recent studies with mouse models of bacterial infection and tumor suggest that ACE plays an important role in the immune responses of myeloid cells. Inhibition of ACE suppresses neutrophil immune response to bacterial infection. In contrast, ACE overexpression in myeloid cells strongly induced bacterial and tumor resistance in mice. A detailed biochemical understanding of how ACE activates myeloid cells and which ACE peptide(s) (substrate or product) mediate these effects could lead to the development of novel therapies for boosting immunity against a variety of stimuli, including bacterial infection and tumor.
Collapse
Affiliation(s)
- Duo-Yao Cao
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - Suguru Saito
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - Luciana C Veiras
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - Derick Okwan-Duodu
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA.,Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Davis Res. Bldg., Rm. 2014, 8700 Beverly Blvd, Los Angeles, CA 90048 USA
| | - Ellen A Bernstein
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - Jorge F Giani
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA.,Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Davis Res. Bldg., Rm. 2014, 8700 Beverly Blvd, Los Angeles, CA 90048 USA
| | - Kenneth E Bernstein
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA.,Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Davis Res. Bldg., Rm. 2014, 8700 Beverly Blvd, Los Angeles, CA 90048 USA
| | - Zakir Khan
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA.,Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Davis Res. Bldg., Rm. 2014, 8700 Beverly Blvd, Los Angeles, CA 90048 USA
| |
Collapse
|
126
|
Dupont AG, Légat L. GABA is a mediator of brain AT 1 and AT 2 receptor-mediated blood pressure responses. Hypertens Res 2020; 43:995-1005. [PMID: 32451494 DOI: 10.1038/s41440-020-0470-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/24/2020] [Accepted: 04/01/2020] [Indexed: 02/07/2023]
Abstract
The nucleus tractus solitarius (NTS), paraventricular nucleus (PVN), and rostral ventrolateral medulla (RVLM) are the most targeted regions of central blood pressure control studies. Glutamate and gamma-aminobutyric acid (GABA) interact within these brain regions to modulate blood pressure. The brain renin-angiotensin system also participates in central blood pressure control. Angiotensin II increases blood pressure through the stimulation of angiotensin II type 1 (AT1) receptors within the PVN and RVLM and attenuates baroreceptor sensitivity, resulting in elevated blood pressure within the NTS. Angiotensin II type 2 (AT2) receptors in cardiovascular control centers in the brain also appear to be involved in blood pressure control and counteract AT1 receptor-mediated effects. The current review is focused on the interaction of GABA with AT1 and AT2 receptors in the control of blood pressure within the RVLM, PVN and NTS. Within the NTS, GABA is released from local GABAergic interneurons that are stimulated by local AT1 receptors and mediates a hypertensive response. In contrast, the local increase in GABA levels observed after AT2 receptor stimulation within the RVLM, likely from GABAergic nerve endings originating in the caudal ventrolateral medulla, is important in the mediation of the hypotensive response. Preliminary results suggest that the hypertensive response to AT1 receptor stimulation within the RVLM is associated with a reduction in GABA release. The current experimental evidence therefore indicates that GABA is an important mediator of brainstem responses to AT1 and AT2 receptor stimulation and that increased GABA release may play a role in hypertensive and hypotensive responses, depending on the site of action.
Collapse
Affiliation(s)
- Alain G Dupont
- Department of Pharmacology, Research Group Experimental Pharmacology (EFAR), Center for Neurosciences, (C4N) Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium
| | - Laura Légat
- Department of Pharmacology, Research Group Experimental Pharmacology (EFAR), Center for Neurosciences, (C4N) Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, 1090, Belgium.
| |
Collapse
|
127
|
Mocayar Marón FJ, Ferder L, Reiter RJ, Manucha W. Daily and seasonal mitochondrial protection: Unraveling common possible mechanisms involving vitamin D and melatonin. J Steroid Biochem Mol Biol 2020; 199:105595. [PMID: 31954766 DOI: 10.1016/j.jsbmb.2020.105595] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/09/2020] [Accepted: 01/16/2020] [Indexed: 12/11/2022]
Abstract
From an evolutionary point of view, vitamin D and melatonin appeared very early and share functions related to defense mechanisms. In the current clinical setting, vitamin D is exclusively associated with phosphocalcic metabolism. Meanwhile, melatonin has chronobiological effects and influences the sleep-wake cycle. Scientific evidence, however, has identified new actions of both molecules in different physiological and pathological settings. The biosynthetic pathways of vitamin D and melatonin are inversely related relative to sun exposure. A deficiency of these molecules has been associated with the pathogenesis of cardiovascular diseases, including arterial hypertension, neurodegenerative diseases, sleep disorders, kidney diseases, cancer, psychiatric disorders, bone diseases, metabolic syndrome, and diabetes, among others. During aging, the intake and cutaneous synthesis of vitamin D, as well as the endogenous synthesis of melatonin are remarkably depleted, therefore, producing a state characterized by an increase of oxidative stress, inflammation, and mitochondrial dysfunction. Both molecules are involved in the homeostatic functioning of the mitochondria. Given the presence of specific receptors in the organelle, the antagonism of the renin-angiotensin-aldosterone system (RAAS), the decrease of reactive species of oxygen (ROS), in conjunction with modifications in autophagy and apoptosis, anti-inflammatory properties inter alia, mitochondria emerge as the final common target for melatonin and vitamin D. The primary purpose of this review is to elucidate the common molecular mechanisms by which vitamin D and melatonin might share a synergistic effect in the protection of proper mitochondrial functioning.
Collapse
Affiliation(s)
- Feres José Mocayar Marón
- Área de Farmacología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Argentina; Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Mendoza, Argentina
| | - León Ferder
- Department of Pediatrics, Nephrology Division, Miller School of Medicine, University of Miami, FL, USA
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science at San Antonio, San Antonio, TX, USA
| | - Walter Manucha
- Área de Farmacología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Argentina; Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Mendoza, Argentina.
| |
Collapse
|
128
|
Mogi M. Effect of renin-angiotensin system on senescence. Geriatr Gerontol Int 2020; 20:520-525. [PMID: 32346971 DOI: 10.1111/ggi.13927] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/24/2020] [Accepted: 03/04/2020] [Indexed: 12/11/2022]
Abstract
The renin-angiotensin system (RAS) plays crucial roles in the control of blood pressure and sodium homeostasis. Moreover, RAS also acts as a key player in cell and organ senescence, mainly by activation of the classical axis of angiotensin (Ang) converting enzyme (ACE)/Ang II/Ang II type 1 receptor via overproduction of reactive oxygen species. Overactivation of the classical RAS axis induces organ dysfunction in the vasculature, brain, kidney and skeletal muscle, resulting in atherosclerosis, stroke, chronic kidney disease and sarcopenia. Moreover, RAS has been shown to regulate lifespan, using gene-modification models. Recently, mice lacking the Ang II type 1 receptor were shown to exhibit an increase in lifespan compared with control mice. Here, the effect of RAS on age-related tissue dysfunction in several organs is reviewed, including not only the classical axis but also protective functions of RAS such as the ACE2/Ang (1-7)/Mas axis. Geriatr Gerontol Int 2020; ••: ••-••.
Collapse
Affiliation(s)
- Masaki Mogi
- Department of Pharmacology, Ehime University Graduate School of Medicine, Ehime, Japan
| |
Collapse
|
129
|
Association between Use of Angiotensin-converting Enzyme Inhibitors or Angiotensin Receptor Blockers and Postoperative Delirium. Anesthesiology 2020; 133:119-132. [DOI: 10.1097/aln.0000000000003329] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Background
Angiotensin-converting enzyme inhibitors and angiotensin receptor blockers improve cognitive function. The authors therefore tested the primary hypothesis that preoperative use of angiotensin inhibitors is associated with less delirium in critical care patients. Post hoc, the association between postoperative use of angiotensin system inhibitors and delirium was assessed.
Methods
The authors conducted a single-site cohort study of adults admitted to Cleveland Clinic critical care units after noncardiac procedures between 2013 and 2018 who had at least one Confusion Assessment Method delirium assessment. Patients with preexisting dementia, Alzheimer’s disease or other cognitive decline, and patients who had neurosurgical procedures were excluded. For the primary analysis, the confounder-adjusted association between preoperative angiotensin inhibitor use and the incidence of postoperative delirium was assessed. Post hoc, the confounder-adjusted association between postoperative angiotensin system inhibitor use and the incidence of delirium was assessed.
Results
The incidence of delirium was 39% (551 of 1,396) among patients who were treated preoperatively with angiotensin system inhibitors and 39% (1,344 of 3,468) in patients who were not. The adjusted odds ratio of experiencing delirium during critical care was 0.98 (95% CI, 0.86 to 1.10; P = 0.700) for preoperative use of angiotensin system inhibitors versus control. Delirium was observed in 23% (100 of 440) of patients who used angiotensin system inhibitors postoperatively before intensive care discharge, and in 41% (1,795 of 4,424) of patients who did not (unadjusted P < 0.001). The confounder-adjusted odds ratio for experiencing delirium in patients who used angiotensin system inhibitors postoperatively was 0.55 (95% CI, 0.43 to 0.72; P < 0.001).
Conclusions
Preoperative use of angiotensin system inhibitors is not associated with reduced postoperative delirium. In contrast, treatment during intensive care was associated with lower odds of delirium. Randomized trials of postoperative angiotensin-converting enzymes inhibitors and angiotensin receptor blockers seem justified.
Editor’s Perspective
What We Already Know about This Topic
What This Article Tells Us That Is New
Collapse
|
130
|
Synthesis and Evaluation of [ 18F]FEtLos and [ 18F]AMBF 3Los as Novel 18F-Labelled Losartan Derivatives for Molecular Imaging of Angiotensin II Type 1 Receptors. Molecules 2020; 25:molecules25081872. [PMID: 32325695 PMCID: PMC7221519 DOI: 10.3390/molecules25081872] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/18/2020] [Accepted: 01/23/2020] [Indexed: 01/13/2023] Open
Abstract
Losartan is widely used in clinics to treat cardiovascular related diseases by selectively blocking the angiotensin II type 1 receptors (AT1Rs), which regulate the renin-angiotensin system (RAS). Therefore, monitoring the physiological and pathological biodistribution of AT1R using positron emission tomography (PET) might be a valuable tool to assess the functionality of RAS. Herein, we describe the synthesis and characterization of two novel losartan derivatives PET tracers, [18F]fluoroethyl-losartan ([18F]FEtLos) and [18F]ammoniomethyltrifluoroborate-losartan ([18F]AMBF3Los). [18F]FEtLos was radiolabeled by 18F-fluoroalkylation of losartan potassium using the prosthetic group 2-[18F]fluoroethyl tosylate; whereas [18F]AMBF3Los was prepared following an one-step 18F-19F isotopic exchange reaction, in an overall yield of 2.7 ± 0.9% and 11 ± 4%, respectively, with high radiochemical purity (>95%). Binding competition assays in AT1R-expressing membranes showed that AMBF3Los presented an almost equivalent binding affinity (Ki 7.9 nM) as the cold reference Losartan (Ki 1.5 nM), unlike FEtLos (Ki 2000 nM). In vitro and in vivo assays showed that [18F]AMBF3Los displayed a good binding affinity for AT1R-overexpressing CHO cells and was able to specifically bind to renal AT1R. Hence, our data demonstrate [18F]AMBF3Los as a new tool for PET imaging of AT1R with possible applications for the diagnosis of cardiovascular, inflammatory and cancer diseases.
Collapse
|
131
|
Kuter KZ, Cenci MA, Carta AR. The role of glia in Parkinson's disease: Emerging concepts and therapeutic applications. PROGRESS IN BRAIN RESEARCH 2020; 252:131-168. [PMID: 32247363 DOI: 10.1016/bs.pbr.2020.02.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Originally believed to primarily affect neurons, Parkinson's disease (PD) has recently been recognized to also affect the functions and integrity of microglia and astroglia, two cell categories of fundamental importance to brain tissue homeostasis, defense, and repair. Both a loss of glial supportive-defensive functions and a toxic gain of glial functions are implicated in the neurodegenerative process. Moreover, the chronic treatment with L-DOPA may cause maladaptive glial plasticity favoring a development of therapy complications. This chapter focuses on the pathophysiology of PD from a glial point of view, presenting this rapidly growing field from the first discoveries made to the most recent developments. We report and compare histopathological and molecular findings from experimental models of PD and human studies. We moreover discuss the important role played by astrocytes in compensatory adaptations taking place during presymptomatic disease stages. We finally describe examples of potential therapeutic applications stemming from an increased understanding of the important roles of glia in PD.
Collapse
Affiliation(s)
- Katarzyna Z Kuter
- Department of Neuropsychopharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland.
| | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Anna R Carta
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Cagliari, Italy.
| |
Collapse
|
132
|
Zhang Z, Zheng X, Zhang X, Zhang Y, Huang B, Luo T. Aging alters Hv1-mediated microglial polarization and enhances neuroinflammation after peripheral surgery. CNS Neurosci Ther 2020; 26:374-384. [PMID: 31774629 PMCID: PMC7053237 DOI: 10.1111/cns.13271] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/30/2019] [Accepted: 11/01/2019] [Indexed: 02/05/2023] Open
Abstract
Perioperative neurocognitive disorders have been widely recognized as common adverse events after surgical intervention. Aging is one of the most important independent risk factors for worsened cognitive outcome, and this deterioration is linked to exacerbated microglia-mediated neuroinflammation in the aged brain. Under pathological stimulation, microglia are capable of polarizing toward proinflammatory M1 and anti-inflammatory M2 phenotypes. In the present study, we examined how aging affects microglial responses and neuroinflammation following peripheral surgery. Adult (2-3 months) and aged (18 months old) male C57/BL6 mice were subjected to tibial fracture or sham surgery. Aged mice exhibited higher level of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) in the hippocampus. The expression of synaptic protein synaptophysin (SYP) was also markedly reduced in the aged brain after the surgery. Both adult and aged mice showed significant increases in M1 microglial polarization (CD16/32). In contrast, tibial fracture surgery induced a decreased M2 microglial polarization (CD206, Ym1/2, Arg1) in aged brain but enhanced M2 microglial polarization in adult brain. Aged mice have upregulated voltage-gated proton channel (Hv1) and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase subunit expression compared with adult mice. The percentage of CD16/32-positive M1 microglia colabeling with Hv1 was higher in aged mice after tibial fracture surgery. Thus, Hv1/NADPH oxidase upregulation in the aged brain may shift the dynamic equilibrium of microglial activation toward M1 polarization and exaggerate postoperative neuroinflammatory responses after peripheral surgical intervention.
Collapse
Affiliation(s)
- Zhi‐jing Zhang
- Department of AnesthesiologyPeking University Shenzhen HospitalShenzhenChina
- Shantou University Medical CollegeShantouChina
| | - Xin‐xun Zheng
- Department of AnesthesiologyPeking University Shenzhen HospitalShenzhenChina
| | - Xin‐yun Zhang
- Department of AnesthesiologyPeking University Shenzhen HospitalShenzhenChina
- Shantou University Medical CollegeShantouChina
| | - Yi Zhang
- Department of AnesthesiologyPeking University Shenzhen HospitalShenzhenChina
- Anhui Medical UniversityHefeiChina
| | - Bao‐yi Huang
- Department of AnesthesiologyPeking University Shenzhen HospitalShenzhenChina
- Shantou University Medical CollegeShantouChina
| | - Tao Luo
- Department of AnesthesiologyPeking University Shenzhen HospitalShenzhenChina
| |
Collapse
|
133
|
Evans CE, Miners JS, Piva G, Willis CL, Heard DM, Kidd EJ, Good MA, Kehoe PG. ACE2 activation protects against cognitive decline and reduces amyloid pathology in the Tg2576 mouse model of Alzheimer's disease. Acta Neuropathol 2020; 139:485-502. [PMID: 31982938 PMCID: PMC7035243 DOI: 10.1007/s00401-019-02098-6] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/16/2019] [Accepted: 11/08/2019] [Indexed: 02/07/2023]
Abstract
Mid-life hypertension and cerebrovascular dysfunction are associated with increased risk of later life dementia, including Alzheimer’s disease (AD). The classical renin–angiotensin system (cRAS), a physiological regulator of blood pressure, functions independently within the brain and is overactive in AD. cRAS-targeting anti-hypertensive drugs are associated with reduced incidence of AD, delayed onset of cognitive decline, and reduced levels of Aβ and tau in both animal models and human pathological studies. cRAS activity is moderated by a downstream regulatory RAS pathway (rRAS), which is underactive in AD and is strongly associated with pathological hallmarks in human AD, and cognitive decline in animal models of CNS disease. We now show that enhancement of brain ACE2 activity, a major effector of rRAS, by intraperitoneal administration of diminazene aceturate (DIZE), an established activator of ACE2, lowered hippocampal Aβ and restored cognition in mid-aged (13–14-month-old) symptomatic Tg2576 mice. We confirmed that the protective effects of DIZE were directly mediated through ACE2 and were associated with reduced hippocampal soluble Aβ42 and IL1-β levels. DIZE restored hippocampal MasR levels in conjunction with increased NMDA NR2B and downstream ERK signalling expression in hippocampal synaptosomes from Tg2576 mice. Chronic (10 weeks) administration of DIZE to pre-symptomatic 9–10-month-old Tg2576 mice, and acute (10 days) treatment in cognitively impaired 12–13-month-old mice, prevented the development of cognitive impairment. Together these data demonstrate that ACE2 enhancement protects against and reverses amyloid-related hippocampal pathology and cognitive impairment in a preclinical model of AD.
Collapse
|
134
|
Role of brain renin angiotensin system in neurodegeneration: An update. Saudi J Biol Sci 2020; 27:905-912. [PMID: 32127770 PMCID: PMC7042626 DOI: 10.1016/j.sjbs.2020.01.026] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 01/20/2020] [Accepted: 01/22/2020] [Indexed: 01/12/2023] Open
Abstract
Renin angiotensin system (RAS) is an endocrine system widely known for its physiological roles in electrolyte homeostasis, body fluid volume regulation and cardiovascular control in peripheral circulation. However, brain RAS is an independent form of RAS expressed locally in the brain, which is known to be involved in brain functions and disorders. There is strong evidence for a major involvement of excessive brain angiotensin converting enzyme (ACE)/Angiotensin II (Ang II)/Angiotensin type-1 receptor (AT-1R) axis in increased activation of oxidative stress, apoptosis and neuroinflammation causing neurodegeneration in several brain disorders. Numerous studies have demonstrated strong neuroprotective effects by blocking AT1R in these brain disorders. Additionally, the angiotensin converting enzyme 2 (ACE2)/Angiotensin (1–7)/Mas receptor (MASR), is another axis of brain RAS which counteracts the damaging effects of ACE/Ang II/AT1R axis on neurons in the brain. Thus, angiotensin II receptor blockers (ARBs) and activation of ACE2/Angiotensin (1–7)/MASR axis may serve as an exciting and novel method for neuroprotection in several neurodegenerative diseases. Here in this review article, we discuss the expression of RAS in the brain and highlight how altered RAS level may cause neurodegeneration. Understanding the pathophysiology of RAS and their links to neurodegeneration has enormous potential to identify potentially effective pharmacological tools to treat neurodegenerative diseases in the brain.
Collapse
|
135
|
Chen YJ, Qian ZM, Sheng Y, Zheng J, Liu Y. Angiotensin II down-regulates transferrin receptor 1 and ferroportin 1 expression in Neuro-2a cells via activation of type-1 receptor. Neurosci Lett 2019; 716:134684. [PMID: 31830506 DOI: 10.1016/j.neulet.2019.134684] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/30/2019] [Accepted: 12/09/2019] [Indexed: 12/14/2022]
Abstract
Angiotensin II (ANGII) modulates expression of iron intake and export proteins in cultured neurons. However, the relevant mechanisms have not been fully elucidated. Here, we investigated the effects of ANGII and/or candesartan, a ANGII-Type-1 Receptor (AT1R) antagonist, and PD123319, a ANGII-Type-2 Receptor (AT2R) antagonist on expression of transferrin receptor 1 (TfR1), ferroportin 1 (Fpn1)and ferritin as well as iron regulatory proteins (IRPs), hepcidin and nuclear factor E2-related factor 2 (Nrf2) in Neuro-2a cells. We demonstrated that ANGII induces a significant reduction in expression of TfR1, Fpn1, IRP2 proteins and Nrf2 mRNA and an increase in ferritin protein and hepcidin mRNA, while candesartan, but not PD123319, significantly attenuated or reversed all these ANGII-induced changes in Neuro-2a cells. These findings imply that ANGII down-regulates TfR1 expression likely via the AT1R/IRP2 pathway, and Fpn1 expression via ATR1/hepcidin and AT1R/ Nrf2 pathways.
Collapse
Affiliation(s)
- Yun-Jin Chen
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, 201203, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Zhong-Ming Qian
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, 201203, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China; Institute of Translational & Precision Medicine, Nantong University, Nantong, JS, 226019, China.
| | - Yuan Sheng
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, 201203, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jie Zheng
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, 201203, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yong Liu
- Department of Neurology, The Second Affiliated (Xinqiao) Hospital, The Army (Third Military) Medical University, Chongqing, China.
| |
Collapse
|
136
|
Garcia-Garrote M, Perez-Villalba A, Garrido-Gil P, Belenguer G, Parga JA, Perez-Sanchez F, Labandeira-Garcia JL, Fariñas I, Rodriguez-Pallares J. Interaction between Angiotensin Type 1, Type 2, and Mas Receptors to Regulate Adult Neurogenesis in the Brain Ventricular-Subventricular Zone. Cells 2019; 8:E1551. [PMID: 31801296 PMCID: PMC6952803 DOI: 10.3390/cells8121551] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/21/2019] [Accepted: 11/28/2019] [Indexed: 12/30/2022] Open
Abstract
The renin-angiotensin system (RAS), and particularly its angiotensin type-2 receptors (AT2), have been classically involved in processes of cell proliferation and maturation during development. However, the potential role of RAS in adult neurogenesis in the ventricular-subventricular zone (V-SVZ) and its aging-related alterations have not been investigated. In the present study, we analyzed the role of major RAS receptors on neurogenesis in the V-SVZ of adult mice and rats. In mice, we showed that the increase in proliferation of cells in this neurogenic niche was induced by activation of AT2 receptors but depended partially on the AT2-dependent antagonism of AT1 receptor expression, which restricted proliferation. Furthermore, we observed a functional dependence of AT2 receptor actions on Mas receptors. In rats, where the levels of the AT1 relative to those of AT2 receptor are much lower, pharmacological inhibition of the AT1 receptor alone was sufficient in increasing AT2 receptor levels and proliferation in the V-SVZ. Our data revealed that interactions between RAS receptors play a major role in the regulation of V-SVZ neurogenesis, particularly in proliferation, generation of neuroblasts, and migration to the olfactory bulb, both in young and aged brains, and suggest potential beneficial effects of RAS modulators on neurogenesis.
Collapse
MESH Headings
- Age Factors
- Angiotensin II/metabolism
- Animals
- Immunohistochemistry
- Lateral Ventricles/metabolism
- Male
- Mice
- Mice, Knockout
- Models, Biological
- Neural Stem Cells/metabolism
- Neurogenesis/genetics
- Protein Binding
- Rats
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 2/genetics
- Receptor, Angiotensin, Type 2/metabolism
Collapse
Affiliation(s)
- Maria Garcia-Garrote
- Laboratorio de Neurobiología Celular y Molecular de la Enfermedad de Parkinson, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Dpto. Ciencias Morfolóxicas, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Ana Perez-Villalba
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Faculty of Psychology, Universidad Católica de Valencia, Valencia, 46100 Burjassot, Spain
| | - Pablo Garrido-Gil
- Laboratorio de Neurobiología Celular y Molecular de la Enfermedad de Parkinson, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Dpto. Ciencias Morfolóxicas, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - German Belenguer
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Departamento de Biología Celular, Biología Funcional y Antropología Física and Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain
| | - Juan A Parga
- Laboratorio de Neurobiología Celular y Molecular de la Enfermedad de Parkinson, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Dpto. Ciencias Morfolóxicas, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Francisco Perez-Sanchez
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Departamento de Biología Celular, Biología Funcional y Antropología Física and Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain
| | - Jose Luis Labandeira-Garcia
- Laboratorio de Neurobiología Celular y Molecular de la Enfermedad de Parkinson, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Dpto. Ciencias Morfolóxicas, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Isabel Fariñas
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Departamento de Biología Celular, Biología Funcional y Antropología Física and Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain
| | - Jannette Rodriguez-Pallares
- Laboratorio de Neurobiología Celular y Molecular de la Enfermedad de Parkinson, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Dpto. Ciencias Morfolóxicas, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| |
Collapse
|
137
|
Jermakowicz WJ, Carballosa-Gautam MM, Vitores AA, Hentall ID. Brainstem-Evoked Transcription of Defensive Genes After Spinal Cord Injury. Front Cell Neurosci 2019; 13:510. [PMID: 31803022 PMCID: PMC6877476 DOI: 10.3389/fncel.2019.00510] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 10/29/2019] [Indexed: 12/13/2022] Open
Abstract
The spinal cord after injury shows altered transcription in numerous genes. We tested in a pilot study whether the nucleus raphé magnus, a descending serotonergic brainstem region whose stimulation improves recovery after incomplete spinal cord injury (SCI), can influence these transcriptional changes. Rats received 2 h of low-frequency electrical stimulation in the raphé magnus 3 days after an impact contusion at segment T8. Comparison groups lacked injuries or activated stimulators or both. Immediately following stimulation, spinal cords were extracted, their RNA transcriptome sequenced, and differential gene expression quantified. Confirming many previous studies, injury primarily increased inflammatory and immune transcripts and decreased those related to lipid and cholesterol synthesis and neuronal signaling. Stimulation plus injury, contrasted with injury alone, caused significant changes in 43 transcripts (39 increases, 4 decreases), all protein-coding. Injury itself decreased only four of these 43 transcripts, all reversed by stimulation, and increased none of them. The non-specific 5-HT7 receptor antagonist pimozide reversed 25 of the 43 changes. Stimulation in intact rats principally caused decreases in transcripts related to oxidative phosphorylation, none of which were altered by stimulation in injury. Gene ontology (biological process) annotations comparing stimulation with either no stimulation or pimozide treatment in injured rats highlighted defense responses to lipopolysaccharides and microorganisms, and also erythrocyte development and oxygen transport (possibly yielding cellular oxidant detoxification). Connectivity maps of human orthologous genes generated in the CLUE database of perturbagen-response transcriptional signatures showed that drug classes whose effects in injured rats most closely resembled stimulation without pimozide include peroxisome proliferator-activated receptor agonists and angiotensin receptor blockers, which are reportedly beneficial in SCI. Thus the initial transcriptional response of the injured spinal cord to raphé magnus stimulation is upregulation of genes that in various ways are mostly protective, some probably located in recently arrived myeloid cells.
Collapse
Affiliation(s)
- Walter J Jermakowicz
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, Miami, FL, United States
| | - Melissa M Carballosa-Gautam
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, Miami, FL, United States
| | - Alberto A Vitores
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, Miami, FL, United States
| | - Ian D Hentall
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami, Miami, FL, United States
| |
Collapse
|
138
|
Sharifi F, Reisi P, Malek M. Angiotensin 1 receptor antagonist attenuates acute kidney injury-induced cognitive impairment and synaptic plasticity via modulating hippocampal oxidative stress. Life Sci 2019; 234:116775. [DOI: 10.1016/j.lfs.2019.116775] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/12/2019] [Accepted: 08/15/2019] [Indexed: 01/13/2023]
|
139
|
Muñoz A, Corrêa CL, Lopez-Lopez A, Costa-Besada MA, Diaz-Ruiz C, Labandeira-Garcia JL. Physical Exercise Improves Aging-Related Changes in Angiotensin, IGF-1, SIRT1, SIRT3, and VEGF in the Substantia Nigra. J Gerontol A Biol Sci Med Sci 2019; 73:1594-1601. [PMID: 29659739 DOI: 10.1093/gerona/gly072] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Indexed: 01/04/2023] Open
Abstract
Dysregulation of tissue renin-angiotensin system (RAS) is involved in oxidative and inflammatory processes observed in major aging-related diseases, including neurodegenerative diseases such as Parkinson's disease (PD). Physical exercise has beneficial effects against aging-related changes, dopaminergic neuron vulnerability, and PD progression. The present study indicates that sedentary aged rats have an increase in activity of the nigral angiotensin (Ang) II/Ang type 1 receptor (AT1) axis (ie, the pro-oxidative pro-inflammatory arm), and a decrease in the activity of the RAS protective arm (ie, Ang II/AT2 and Ang 1-7/Mas receptor axis) in comparison with young rats. In addition, sedentary aged rats showed a decrease in levels of nigral IGF-1, SIRT1, SIRT3, and VEGF. Treadmill running induced a significant increase in levels of IGF-1, SIRT1, SIRT3, and VEGF, as well as an increase in expression of the protective Ang 1-7/Mas axis and inhibition of the Ang II/AT1 axis. The exercise-induced increase in IGF-1 and sirtuins may mediate the effects of exercise on the nigral RAS. However, exercise may induce the increase in VEGF and modulation of RAS activity by different pathways. Exercise, via RAS, contributes to inhibition of the pro-oxidative and proinflammatory state that increase dopaminergic neuron vulnerability and risk of PD with aging.
Collapse
Affiliation(s)
- Ana Muñoz
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Clynton L Corrêa
- Faculty of Medicine, Master Program of Physical Education - Universidade Federal do Rio de Janeiro, Brazil
| | - Andrea Lopez-Lopez
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Maria A Costa-Besada
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Carmen Diaz-Ruiz
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jose L Labandeira-Garcia
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, University of Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| |
Collapse
|
140
|
Cui C, Xu P, Li G, Qiao Y, Han W, Geng C, Liao D, Yang M, Chen D, Jiang P. Vitamin D receptor activation regulates microglia polarization and oxidative stress in spontaneously hypertensive rats and angiotensin II-exposed microglial cells: Role of renin-angiotensin system. Redox Biol 2019; 26:101295. [PMID: 31421410 PMCID: PMC6831892 DOI: 10.1016/j.redox.2019.101295] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/02/2019] [Accepted: 08/07/2019] [Indexed: 01/19/2023] Open
Abstract
Hypertension is one of the major predisposing factors for neurodegenerative disease characterized with activated renin-angiotensin system (RAS) in both periphery and brain. Vitamin D (VitD) is recently recognized as a pleiotropic hormone with strong neuroprotective properties. While multiple lines of evidence suggest that VitD can act on RAS, the evidence concerning the crosstalk between VitD and RAS in the brain is limited. Therefore, this study aims to evaluate whether VitD can modulate brain RAS to trigger neuroprotective actions in the brain of spontaneously hypertensive rats (SHR). Our data showed that calcitriol treatment induced VDR expression and inhibited neural death in the prefrontal cortex of SHR. Sustained calcitriol administration also inhibited microglia M1 polarization, but enhanced M2 polarization, accompanied with decreased expression of proinflammatory cytokines. We then further explored the potential mechanisms and showed that SHR exhibited overactivated classical RAS with increased expression of angiotensin II (Ang II) receptor type 1 (AT1), angiotensin converting enzyme (ACE) and Ang II production, whereas the counteracting arm of traditional RAS, ACE2/Ang(1-7)/MasR, was impaired in the SHR brain. Calcitriol nonsignificantly suppressed AT1 and ACE but markedly reduced Ang II formation. Intriguingly, calcitriol exerted pronouncedly impact on ACE2/Ang(1-7)/MasR axis with enhanced expression of ACE2, MasR and Ang(1-7) generation. Meanwhile, calcitriol ameliorated the overactivation of NADPH-oxidase (Nox), the downstream of RAS, in SHR, and also mitigated oxidative stress. In microglial (BV2) cells, we further found that calcitriol induced ACE2 and MasR with no significant impact on ACE and AT1. In accordance, calcitriol also attenuated Ang II-induced Nox activation and ROS production, and shifted the microglia polarization from M1 to M2 phenotype. However, co-treatment with A779, a specific MasR antagonist, abrogated the antioxidant and neuroimmune modulating actions of VitD. These findings strongly indicate the involvement of ACE2/Ang(1-7)/MasR pathway in the neuroprotective mechanisms of VitD in the hypertensive brain.
Collapse
Affiliation(s)
- Changmeng Cui
- Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Pengfei Xu
- Jining First People's Hospital, Jining Medical University, Jining, China
| | - Gongying Li
- Department of Mental Health, Jining Medical University, Jining, China
| | - Yi Qiao
- Department of Public Health, Jining Medical University, Jining, China
| | - Wenxiu Han
- Jining First People's Hospital, Jining Medical University, Jining, China
| | - Chunmei Geng
- Jining First People's Hospital, Jining Medical University, Jining, China
| | - Dehua Liao
- Hunan Cancer Hospital, Central South University, Changsha, China
| | - Mengqi Yang
- Jining First People's Hospital, Jining Medical University, Jining, China
| | - Dan Chen
- Jining First People's Hospital, Jining Medical University, Jining, China
| | - Pei Jiang
- Jining First People's Hospital, Jining Medical University, Jining, China; Department of Precision Medicine, Tengzhou Central People's Hospital, Tengzhou, China.
| |
Collapse
|
141
|
Lund B, Stone R, Levy A, Lee S, Amundson E, Kashani N, Rodgers K, Kelland E. Reduced disease severity following therapeutic treatment with angiotensin 1–7 in a mouse model of multiple sclerosis. Neurobiol Dis 2019; 127:87-100. [DOI: 10.1016/j.nbd.2019.02.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 02/20/2019] [Indexed: 12/19/2022] Open
|
142
|
Yao K, Zu HB. Microglial polarization: novel therapeutic mechanism against Alzheimer's disease. Inflammopharmacology 2019; 28:95-110. [PMID: 31264132 DOI: 10.1007/s10787-019-00613-5] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 06/16/2019] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease that results in progressive dementia, and exhibits high disability and fatality rates. Recent evidence has demonstrated that neuroinflammation is critical in the pathophysiological processes of AD, which is characterized by the activation of microglia and astrocytes. Under different stimuli, microglia are usually activated into two polarized states, termed the classical 'M1' phenotype and the alternative 'M2' phenotype. M1 microglia are considered to promote inflammatory injury in AD; in contrast, M2 microglia exert neuroprotective effects. Imbalanced microglial polarization, in the form of excessive activation of M1 microglia and dysfunction of M2 microglia, markedly promotes the development of AD. Furthermore, an increasing number of studies have shown that the transition of microglia from the M1 to M2 phenotype could potently alleviate pathological damage in AD. Hence, this article reviews the current knowledge regarding the role of microglial M1/M2 polarization in the pathophysiology of AD. In addition, we summarize several approaches that protect against AD by altering the polarization states of microglia. This review aims to contribute to a better understanding of the pathogenesis of AD and, moreover, to explore the potential of novel drugs for the treatment of AD in the future.
Collapse
Affiliation(s)
- Kai Yao
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No. 1508 Longhang Road, Jinshan District, Shanghai, 201508, China
| | - Heng-Bing Zu
- Department of Neurology, Jinshan Hospital Affiliated to Fudan University, No. 1508 Longhang Road, Jinshan District, Shanghai, 201508, China.
| |
Collapse
|
143
|
Costa-Ferreira W, Morais-Silva G, Gomes-de-Souza L, Marin MT, Crestani CC. The AT1 Receptor Antagonist Losartan Does Not Affect Depressive-Like State and Memory Impairment Evoked by Chronic Stressors in Rats. Front Pharmacol 2019; 10:705. [PMID: 31293424 PMCID: PMC6598205 DOI: 10.3389/fphar.2019.00705] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/31/2019] [Indexed: 12/15/2022] Open
Abstract
The present study investigated the effect of the treatment with the angiotensin II type 1 receptor (AT1) antagonist losartan in the depressive-like state and memory impairment evoked by exposure to either homotypic (i.e., repeated exposure to the same type of stressor) or heterotypic (i.e., exposure to different aversive stimuli) chronic stressors in rats. For this, male Wistar rats were subjected to a 10 days regimen of repeated restraint stress (RRS, homotypic stressor) or chronic variable stress (CVS, heterotypic stressor) while being concurrently treated daily with losartan (30 mg/kg/day, p.o.). Depressive-like state was evaluated by analysis of the alterations considered as markers of depression (decreased sucrose preference and body weight and coat state deterioration), whereas cognitive non-emotional performance was tested using the novel object recognition (NOR) test. Locomotor activity was also evaluated in the open field test. Both RRS and CVS impaired sucrose preference and caused coat state deterioration, whereas only CVS impaired body weight gain. Besides, RRS impaired short-term memory (but not long-term memory) in the NOR test. Neither depressive-like state nor memory impairment evoked by the chronic stressors was affected by the treatment with losartan. Nevertheless, CVS increased the locomotion, which was inhibited by losartan. Taken together, these results provide evidence that the chronic treatment with losartan does not affect the depressive-like state and memory impairment evoked by either homotypic or heterotypic chronic stress regimens in rats.
Collapse
Affiliation(s)
- Willian Costa-Ferreira
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil.,Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, Brazil
| | - Gessynger Morais-Silva
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil.,Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, Brazil
| | - Lucas Gomes-de-Souza
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil.,Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, Brazil
| | - Marcelo T Marin
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil.,Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, Brazil
| | - Carlos C Crestani
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil.,Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, Brazil
| |
Collapse
|
144
|
Guo Y, Hong W, Wang X, Zhang P, Körner H, Tu J, Wei W. MicroRNAs in Microglia: How do MicroRNAs Affect Activation, Inflammation, Polarization of Microglia and Mediate the Interaction Between Microglia and Glioma? Front Mol Neurosci 2019; 12:125. [PMID: 31133802 PMCID: PMC6522842 DOI: 10.3389/fnmol.2019.00125] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/26/2019] [Indexed: 12/31/2022] Open
Abstract
The essential roles of microglia in maintaining homeostasis in the healthy brain and contributing to neuropathology are well documented. Emerging evidence suggests that epigenetic modulation regulates microglial behavior in both physiological and pathological conditions. MicroRNAs (miRNAs) are short, non-coding epigenetic regulators that repress target gene expression mostly via binding to 3'-untranslated region (3'-UTR) of mRNA in a Dicer-dependent manner. Dysregulation of certain miRNAs can contribute to microglial hyper-activation, persistent neuroinflammation, and abnormal macrophage polarization in the brain. These abnormal conditions can support the pathogenesis of neurological disorders such as glioma, Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), stroke, ischemia, and spinal cord injury (SCI). However, the roles of miRNAs in microglia in health and neurological disease have not been systematically summarized. This review will first report the role of Dicer, a key endoribonulease that is responsible for most miRNA biogenesis in microglia. Second, we will focus on recent research about the function of miRNAs in activation, inflammation and polarization of microglia, respectively. In addition, potential crosstalk between microglia and glioma cells via miRNAs will be discussed in this part. Finally, the role of two essential miRNAs, miR-124, and miR-155, in microglia will be highlighted.
Collapse
Affiliation(s)
- Yawei Guo
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| | - Wenming Hong
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xinming Wang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| | - Pengying Zhang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| | - Heinrich Körner
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| | - Jiajie Tu
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| | - Wei Wei
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| |
Collapse
|
145
|
Timaru-Kast R, Gotthardt P, Luh C, Huang C, Hummel R, Schäfer MKE, Thal SC. Angiotensin II Receptor 1 Blockage Limits Brain Damage and Improves Functional Outcome After Brain Injury in Aged Animals Despite Age-Dependent Reduction in AT1 Expression. Front Aging Neurosci 2019; 11:63. [PMID: 31105549 PMCID: PMC6499023 DOI: 10.3389/fnagi.2019.00063] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/06/2019] [Indexed: 11/13/2022] Open
Abstract
Traumatic brain injury (TBI) is a frequent pathology associated with poor neurological outcome in the aged population. We recently observed accelerated cerebral inflammation in aged mice in response to TBI. Candesartan is a potent specific inhibitor of angiotensin II receptor type 1 (AT1) which limits cerebral inflammation and brain damage in juvenile animals after experimental TBI. In the present study, we show significantly lower posttraumatic AT1 mRNA levels in aged (21 months) compared to young (2 months) mice. Despite low cerebral At1 expression, pharmacologic blockade by treatment with candesartan [daily, beginning 30 min after experimental TBI by controlled cortical impact (CCI)] was highly effective in both young and aged animals and reduced histological brain damage by -20% after 5 days. In young mice, neurological improvement was enhanced by AT1 inhibition 5 days after CCI. In older animals, candesartan treatment reduced functional impairment already on day 3 after TBI and post-traumatic body weight (BW) loss was attenuated. Candesartan reduced microglia activation (-40%) in young and aged animals, and neutrophil infiltration (-40% to 50%) in aged mice, whereas T-cell infiltration was not changed in either age group. In young animals, markers of anti-inflammatory microglia M2a polarization [arginase 1 (Arg1), chitinase3-like 3 (Ym1)] were increased by candesartan at days 1 and 5 after insult. In older mice 5 days after insult, expression of Arg1 was significantly higher independently of the treatment, whereas Ym1 gene expression was further enhanced by AT1 inhibition. Despite age-dependent posttraumatic differences in At1 expression levels, inhibition of AT1 was highly effective in a posttreatment paradigm. Targeting inflammation with candesartan is, therefore, a promising therapeutic strategy to limit secondary brain damage independent of the age.
Collapse
Affiliation(s)
- Ralph Timaru-Kast
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Philipp Gotthardt
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Clara Luh
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Changsheng Huang
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Regina Hummel
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Michael K E Schäfer
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany.,Center for Molecular Surgical Research, University Medical Center of the Johannes Gutenberg University, Mainz, Germany.,Focus Program Translational Neurosciences, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Serge C Thal
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany.,Center for Molecular Surgical Research, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
146
|
Lewitt MS, Boyd GW. The Role of Insulin-Like Growth Factors and Insulin-Like Growth Factor-Binding Proteins in the Nervous System. BIOCHEMISTRY INSIGHTS 2019; 12:1178626419842176. [PMID: 31024217 PMCID: PMC6472167 DOI: 10.1177/1178626419842176] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 03/15/2019] [Indexed: 01/23/2023]
Abstract
The insulin-like growth factors (IGF-I and IGF-II) and their receptors are widely expressed in nervous tissue from early embryonic life. They also cross the blood brain barriers by active transport, and their regulation as endocrine factors therefore differs from other tissues. In brain, IGFs have paracrine and autocrine actions that are modulated by IGF-binding proteins and interact with other growth factor signalling pathways. The IGF system has roles in nervous system development and maintenance. There is substantial evidence for a specific role for this system in some neurodegenerative diseases, and neuroprotective actions make this system an attractive target for new therapeutic approaches. In developing new therapies, interaction with IGF-binding proteins and other growth factor signalling pathways should be considered. This evidence is reviewed, gaps in knowledge are highlighted, and recommendations are made for future research.
Collapse
Affiliation(s)
- Moira S Lewitt
- School of Health & Life Sciences, University of the West of Scotland, Paisley, UK
| | - Gary W Boyd
- School of Health & Life Sciences, University of the West of Scotland, Paisley, UK
| |
Collapse
|
147
|
Quitterer U, AbdAlla S. Improvements of symptoms of Alzheimer`s disease by inhibition of the angiotensin system. Pharmacol Res 2019; 154:104230. [PMID: 30991105 DOI: 10.1016/j.phrs.2019.04.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/10/2019] [Accepted: 04/11/2019] [Indexed: 01/30/2023]
Abstract
With ageing of the global society, the frequency of ageing-related neurodegenerative diseases such as Alzheimer`s disease (AD) is on the rise worldwide. Currently, there is no cure for AD, and the four drugs approved for AD only have very small effects on AD symptoms. Consequently, there are enormous efforts worldwide to identify new targets for treatment of AD. Approaches that interfere with classical neuropathologic features of AD, such as extracellular senile plaques formed of aggregated amyloid-beta (Abeta), and intracellular neurofibrillary tangles of hyperphosphorylated tau have not been successful so far. In search for a treatment approach of AD, we found that inhibition of the angiotensin-converting enzyme (ACE) by a centrally acting ACE inhibitor retards symptoms of neurodegeneration, Abeta plaque formation and tau hyperphosphorylation in experimental models of AD. Our approach is currently being investigated in a clinical setting. Initial evidence with AD patients shows that a brain-penetrating ACE inhibitor counteracts the process of neurodegeneration and dementia. Moreover, centrally acting ACE inhibitors given in addition to the standard therapy, cholinesterase inhibition, can improve cognitive function of AD patients for several months. This is one of the most promising results for AD treatment since more than a decade.
Collapse
Affiliation(s)
- Ursula Quitterer
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland; Institute of Pharmacology and Toxicology, Department of Medicine, University of Zurich, Winterthurerstrasse 190, CH-8057, Zürich, Switzerland.
| | - Said AbdAlla
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| |
Collapse
|
148
|
Gong X, Hu H, Qiao Y, Xu P, Yang M, Dang R, Han W, Guo Y, Chen D, Jiang P. The Involvement of Renin-Angiotensin System in Lipopolysaccharide-Induced Behavioral Changes, Neuroinflammation, and Disturbed Insulin Signaling. Front Pharmacol 2019; 10:318. [PMID: 31001119 PMCID: PMC6454872 DOI: 10.3389/fphar.2019.00318] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 03/15/2019] [Indexed: 12/12/2022] Open
Abstract
Brain insulin signaling is accounted for the development of a variety of neuropsychiatric disorders, such as anxiety and depression, whereas both inflammation and the activated renin-angiotensin system (RAS) are two major contributors to insulin resistance. Intriguingly, inflammation and RAS can activate each other, forming a positive feedback loop that would result in exacerbated unwanted tissue damage. To further examine the interrelationship among insulin signaling, neuroinflammation and RAS in the brain, the effect of repeated lipopolysaccharide (LPS) exposure and co-treatment with the angiotensin II (Ang II) receptor type 1 (AT1) blocker, candesartan (Cand), on anxiety and depression-like behaviors, RAS, neuroinflammation and insulin signaling was explored. Our results demonstrated that prolonged LPS challenge successfully induced the rats into anxiety and depression-like state, accompanied with significant neural apoptosis and neuroinflammation. LPS also activated RAS as evidenced by the enhanced angiotensin converting enzyme (ACE) expression, Ang II generation and AT1 expression. However, blocking the activated RAS with Cand co-treatment conferred neurobehavioral protective properties. The AT1 blocker markedly ameliorated the microglial activation, the enhanced gene expression of the proinflammatory cytokines and the overactivated NF-κB signaling. In addition, Cand also mitigated the LPS-induced disturbance of insulin signaling with the normalized phosphorylation of serine 307 and tyrosine 896 of insulin receptor substrate-1 (IRS-1). Collectively, the present study, for the first time, provided the direct evidence indicating that the inflammatory condition may interact with RAS to impede brain insulin pathway, resulting in neurobehavioral damage, and inhibiting RAS seems to be a promising strategy to block the cross-talk and cut off the vicious cycle between RAS and immune system.
Collapse
Affiliation(s)
- Xiaoxue Gong
- Institute of Clinical Pharmacy and Pharmacology, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Hui Hu
- Department of Cardiology, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Yi Qiao
- Department of Public Health, Jining Medical University, Jining, China
| | - Pengfei Xu
- Institute of Clinical Pharmacy and Pharmacology, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Mengqi Yang
- Institute of Clinical Pharmacy and Pharmacology, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Ruili Dang
- Institute of Clinical Pharmacy and Pharmacology, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Wenxiu Han
- Institute of Clinical Pharmacy and Pharmacology, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Yujin Guo
- Institute of Clinical Pharmacy and Pharmacology, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Dan Chen
- Institute of Clinical Pharmacy and Pharmacology, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Pei Jiang
- Institute of Clinical Pharmacy and Pharmacology, Jining First People's Hospital, Jining Medical University, Jining, China
| |
Collapse
|
149
|
Bennett JP, Keeney PM, Brohawn DG. RNA Sequencing Reveals Small and Variable Contributions of Infectious Agents to Transcriptomes of Postmortem Nervous Tissues From Amyotrophic Lateral Sclerosis, Alzheimer's Disease and Parkinson's Disease Subjects, and Increased Expression of Genes From Disease-Activated Microglia. Front Neurosci 2019; 13:235. [PMID: 30983949 PMCID: PMC6447612 DOI: 10.3389/fnins.2019.00235] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 02/27/2019] [Indexed: 12/11/2022] Open
Abstract
Nervous tissues from both humans with neurodegenerative diseases (NDD) and animals with genetic models of human NDD, such as rare monogenic causes of Amyotrophic Lateral Sclerosis (ALS), Alzheimer's disease (AD), and Parkinson's disease (PD), show activated microglia, suggesting a potential causal role for inflammation in pathogenesis of NDD. We performed paired-end (PE) RNA sequencing (RNA seq) of total RNA's extracted from frozen sections of cervical spinal cords from ALS and CTL subjects, frontal cortical gray matter ribbons of AD and CTL subjects, and ventral midbrains of PD and CTL subjects. Trimmed PE reads were aligned against the hg38 human transcriptome using Tophat2/Bowtie2 (ALS) or HISAT2 (AD and PD) and quantitated with Cufflinks. PE reads were also aligned using Bowtie2 against genomes from representative species of Toxoplasma gondii and Trichinella sp. T6 (parasitic infectious agents), Babesia microti and Borrelia burgdorferi (tick-vector borne agents), and Treponema denticola and Porphyromonas gingivalis, agents causing chronic gingivitis. Primary aligned reads of each agent in each tissue sample were quantitated with SAMtools. We found small percentages (<0.1%) of transcriptomes aligned with B. microti, B. burgdorferi, T. denticola, and P. gingivalis genomes and larger percentages aligned with T. gondii (0.1-0.2%) and Trichinella sp. T6 (1.0-1.1%) genomes. In AD specimens, but in no others, primary aligned transcriptome percentages, although small, approached significance for being greater in AD compared to CTL samples for B. burgdorferi (p = 0.067) and P. gingivalis (p = 0.068). Genes' expressions in postmortem tissues of AD and ALS but not PD revealed significant changes among disease-associated microglial (DAM) genes. Infectious agents' transcripts can be detected in RNA seq reads of both NDD and CTL tissues and vary from agent to agent. Expressions of Stage 1 and Stage 2 DAM genes significantly changed, suggesting the presence of Stages 1 and 2 DAM in our NDD tissue samples.
Collapse
Affiliation(s)
- James P Bennett
- Neurodegeneration Therapeutics, Inc., Charlottesville, VA, United States.,Parkinson's and Movement Disorders Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Paula M Keeney
- Neurodegeneration Therapeutics, Inc., Charlottesville, VA, United States.,Parkinson's and Movement Disorders Center, Virginia Commonwealth University, Richmond, VA, United States
| | - David G Brohawn
- Parkinson's and Movement Disorders Center, Virginia Commonwealth University, Richmond, VA, United States.,Department of Medical Genetics, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
150
|
Mocayar Marón FJ, Ferder L, Saraví FD, Manucha W. Hypertension linked to allostatic load: from psychosocial stress to inflammation and mitochondrial dysfunction. Stress 2019; 22:169-181. [PMID: 30547701 DOI: 10.1080/10253890.2018.1542683] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Although a large number of available treatments and strategies, the prevalence of cardiovascular diseases continues to grow worldwide. Emerging evidence supports the notion of counteracting stress as a critical component of a comprehensive therapeutic strategy for cardiovascular disease. Indeed, an unhealthy lifestyle is a burden to biological variables such as plasma glucose, lipid profile, and blood pressure control. Recent findings identify allostatic load as a new paradigm for an integrated understanding of the importance of psychosocial stress and its impact on the development and maintenance of cardiovascular disease. Allostasis complement homeostasis and integrates behavioral and physiological mechanisms by which genes, early experiences, environment, lifestyle, diet, sleep, and physical exercise can modulate and adapt biological responses at the cellular level. For example, variability is a physiological characteristic of blood pressure necessary for survival and the allostatic load in hypertension can contribute to its related cardiovascular morbidity and mortality. Therefore, the current review will focus on the mechanisms that link hypertension to allostatic load, which includes psychosocial stress, inflammation, and mitochondrial dysfunction. We will describe and discuss new insights on neuroendocrine-immune effects linked to allostatic load and its impact on the cellular and molecular responses; the links between allostatic load, inflammation, and endothelial dysfunction; the epidemiological evidence supporting the pathophysiological origins of hypertension; and the biological embedding of allostatic load and hypertension with an emphasis on mitochondrial dysfunction.
Collapse
Affiliation(s)
- Feres José Mocayar Marón
- a Área de Química Biológica, Departamento de Morfofisiología, Facultad de Ciencias Médicas , Universidad Nacional de Cuyo , Mendoza , Argentina
| | - León Ferder
- b Department of Pediatrics , Nephrology Division, Miller School of Medicine, University of Miami , FL , USA
| | - Fernando Daniel Saraví
- c Instituto de Fisiología, Departamento de Morfofisiología, Facultad de Ciencias Médicas , Universidad Nacional de Cuyo , Mendoza , Argentina
| | - Walter Manucha
- d Área de Farmacología, Departamento de Patología, Facultad de Ciencias Médicas , Universidad Nacional de Cuyo , Mendoza , Argentina
- e Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET) , Mendoza , Argentina
| |
Collapse
|