1
|
Ferreira Machado M, Muela HCS, Costa-Hong VA, Cristina Moraes N, Maia Memória C, Sanches Yassuda M, Bor-Seng-Shu E, Nitrini R, Aparecido Bortolotto L, de Carvalho Nogueira R. Angiotensin-converting enzyme inhibitors: a therapeutic option for controlling blood pressure associated with delayed cognitive processing speed. J Hum Hypertens 2024:10.1038/s41371-024-00965-8. [PMID: 39367178 DOI: 10.1038/s41371-024-00965-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
Antihypertensive treatment (AT) is essential for preventing hypertension-related cognitive decline. The goals of this observational study were to compare cognitive performance (CP) between non-hypertensive (NH) volunteers and hypertensive patients and to evaluate the correlation between CP and antihypertensive drugs (AHD). Three groups were constituted: NH (n = 30) [group 1], hypertensive with systolic blood pressure (SBP) < 140 mmHg and diastolic blood pressure (DBP) < 90 mmHg (n = 54) [group 2] and hypertensive with SBP ≥ 140 or DBP ≥ 90 (n = 31) [group 3]. To analyze the cognitive domains, a neuropsychological battery was applied and the raw performance values in these tests were transformed into z-scores. The domain was considered impaired if it presented a z-score below -1.5 SD. Compared to group 1, both groups of hypertensive were older (51 [ ± 12] years) and showed a worse CP in episodic memory (p = 0.014), language (p = 0.003) and processing speed (PS) [p = 0.05]. Angiotensin-converting enzyme inhibitors (ACEi) and angiotensin receptor blockers (ARB) were the most used AHD (46.3%, p = 0.01 [group 2] and 64.5%, p = 0.005 [group 3]) and showed correlations with PS. Linear regression models revealed a negative association of PS with the use of ACEi (β = -0.230, p = 0.004), but not with the use of ARB (β = 0.208, p = 0.008). The effect of AT on cognition appears to go beyond the search for lower blood pressure targets and also includes the mechanism of action of AHD on the brain, so that additional benefits may possibly be achieved with simple adaptations in the treatment regimen, particularly in patients without clinically manifest cognitive impairment.
Collapse
Affiliation(s)
- Michel Ferreira Machado
- Department of Neurology, Hospital das Clínicas, University of São Paulo Medical School, São Paulo, Brazil.
| | | | | | - Natalia Cristina Moraes
- Department of Neurology, Hospital das Clínicas, University of São Paulo Medical School, São Paulo, Brazil
| | - Claudia Maia Memória
- Department of Neurology, Hospital das Clínicas, University of São Paulo Medical School, São Paulo, Brazil
| | - Monica Sanches Yassuda
- Gerontology, School of Arts, Sciences and Humanities, University of São Paulo Medical School, São Paulo, Brazil
| | - Edson Bor-Seng-Shu
- Department of Neurology, Hospital das Clínicas, University of São Paulo Medical School, São Paulo, Brazil
| | - Ricardo Nitrini
- Department of Neurology, Hospital das Clínicas, University of São Paulo Medical School, São Paulo, Brazil
| | - Luiz Aparecido Bortolotto
- Hypertension Unit, Instituto do Coração (INCOR), University of São Paulo Medical School, São Paulo, Brazil
| | | |
Collapse
|
2
|
Gallardo-Ortíz IA, Oros-González A, Rodríguez-Manzo G, Garduño-Gutiérrez R, Aragón-Martínez A, Páez-Martínez N. Effect of exercise duration on toluene-induced locomotor sensitization in mice: a focus on the Renin Angiotensin System. Psychopharmacology (Berl) 2024; 241:2157-2170. [PMID: 38839630 DOI: 10.1007/s00213-024-06626-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 05/23/2024] [Indexed: 06/07/2024]
Abstract
RATIONALE Exercise attenuates addictive behavior; however, little is known about the contribution of exercise duration to this positive effect. The Renin Angiotensin System (RAS) has been implicated both in addictive responses and in the beneficial effects of exercise; though, its role in the advantageous effects of exercise on toluene-induced addictive responses has not been explored. OBJECTIVES To evaluate the impact of different exercise regimens in mitigating the expression of toluene-induced locomotor sensitization and to analyze changes in RAS elements' expression at the mesocorticolimbic system after repeated toluene exposure and following voluntary wheel running in toluene-sensitized animals. METHODS Toluene-induced addictive-like response was evaluated with a locomotor sensitization model in mice. Toluene-sensitized animals had access to running wheels 1, 2, 4 or 24 h/day for 4 weeks; thereafter, locomotor sensitization expression was evaluated after a toluene challenge. RAS elements (ACE and ACE2 enzymes; AT1, AT2 and Mas receptors) expression was determined by Western blot in the VTA, NAc and PFCx of toluene-sensitized mice with and without exercise. RESULTS Individual differences in toluene-induced locomotor sensitization development were observed. Access to wheel running 1 and 2 h/day reduced but 4 and 24 h/day completely blocked locomotor sensitization expression. Repeated toluene exposure changed RAS elements' expression in the VTA, NAc and PFCx, while exercise mainly modified ACE and AT1 in air-exposed and toluene-sensitized mice. CONCLUSIONS Inhalant-exposed animals show different sensitization phenotypes. Exercise duration determined its efficacy to attenuate the addictive-like response. Toluene exposure and exercise each modified RAS, the latter also modifying toluene-induced changes.
Collapse
Affiliation(s)
- Itzell A Gallardo-Ortíz
- Unidad de Biomedicina, Carrera de Enfermería, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de México, México
| | - Alain Oros-González
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México, México
| | - Gabriela Rodríguez-Manzo
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del IPN (Cinvestav), Sede Sur, Ciudad de México, México
| | - René Garduño-Gutiérrez
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del IPN (Cinvestav), Sede Sur, Ciudad de México, México
| | - Andrés Aragón-Martínez
- Unidad de Biomedicina, Carrera de Biología, Laboratorio de Gametos y Desarrollo Tecnológico, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Estado de México, México
| | - Nayeli Páez-Martínez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México, México.
- Laboratorio Integrativo para el Estudio de Sustancias Inhalables Adictivas, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría "Ramón de la Fuente Muñiz", Ciudad de México, México.
| |
Collapse
|
3
|
Tiwari P, Elgazzaz M, Lazartigues E, Hanif K. Effect of Diminazene Aceturate, an ACE2 activator, on platelet CD40L signaling induced glial activation in rat model of hypertension. Int Immunopharmacol 2024; 139:112654. [PMID: 38996777 DOI: 10.1016/j.intimp.2024.112654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 06/30/2024] [Accepted: 07/06/2024] [Indexed: 07/14/2024]
Abstract
Hypertension causes platelet activation and adhesion in the brain resulting in glial activation and neuroinflammation. Further, activation of Angiotensin-Converting Enzyme 2/Angiotensin (1-7)/Mas Receptor (ACE2/Ang (1-7)/MasR) axis of central Renin-Angiotensin System (RAS), is known to reduce glial activation and neuroinflammation, thereby exhibiting anti-hypertensive and anti-neuroinflammatory properties. Therefore, in the present study, the role of ACE2/Ang (1-7)/MasR axis was studied on platelet-induced glial activation and neuroinflammation using Diminazene Aceturate (DIZE), an ACE2 activator, in astrocytes and microglial cells as well as in rat model of hypertension. We found that the ACE2 activator DIZE, independently of its BP-lowering properties, efficiently prevented hypertension-induced glial activation, neuroinflammation, and platelet CD40-CD40L signaling via upregulation of ACE2/Ang (1-7)/MasR axis. Further, DIZE decreased platelet deposition in the brain by reducing the expression of adhesion molecules on the brain endothelium. Activation of ACE2 also reduced hypertension-induced endothelial dysfunction by increasing eNOS bioavailability. Interestingly, platelets isolated from hypertensive rats or activated with ADP had significantly increased sCD40L levels and induced significantly more glial activation than platelets from DIZE treated group. Therefore, injection of DIZE pre-treated ADP-activated platelets into normotensive rats strongly reduced glial activation compared to ADP-treated platelets. Moreover, CD40L-induced glial activation, CD40 expression, and NFкB-NLRP3 inflammatory signaling are reversed by DIZE. Furthermore, the beneficial effects of ACE2 activation, DIZE was found to be significantly blocked by MLN4760 (ACE2 inhibitor) as well as A779 (MasR antagonist) treatments. Hence, our study demonstrated that ACE2 activation reduced the platelet CD40-CD40L induced glial activation and neuroinflammation, hence imparted neuroprotection.
Collapse
Affiliation(s)
- Priya Tiwari
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mona Elgazzaz
- Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Eric Lazartigues
- Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Kashif Hanif
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
4
|
Smith HC, Yu Z, Iyer L, Marvar PJ. Sex-Dependent Effects of Angiotensin Type 2 Receptor-Expressing Medial Prefrontal Cortex Interneurons in Fear Extinction Learning. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:100340. [PMID: 39140003 PMCID: PMC11321323 DOI: 10.1016/j.bpsgos.2024.100340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/17/2024] [Accepted: 05/19/2024] [Indexed: 08/15/2024] Open
Abstract
Background The renin-angiotensin system has been identified as a potential therapeutic target for posttraumatic stress disorder, although its mechanisms are not well understood. Brain angiotensin type 2 receptors (AT2Rs) are a subtype of angiotensin II receptors located in stress and anxiety-related regions, including the medial prefrontal cortex (mPFC), but their function and mechanism in the mPFC remain unexplored. Therefore, we used a combination of imaging, cre/lox, and behavioral methods to investigate mPFC-AT2R-expressing neurons in fear and stess related behavior. Methods To characterize mPFC-AT2R-expressing neurons in the mPFC, AT2R-Cre/tdTomato male and female mice were used for immunohistochemistry. mPFC brain sections were stained with glutamatergic or interneuron markers, and density of AT2R+ cells and colocalization with each marker were quantified. To assess fear-related behaviors in AT2R-flox mice, we selectively deleted AT2R from mPFC neurons using a Cre-expressing adeno-associated virus. Mice then underwent Pavlovian auditory fear conditioning, elevated plus maze, and open field testing. Results Immunohistochemistry results revealed that AT2R was densely expressed throughout the mPFC and primarily expressed in somatostatin interneurons in a sex-dependent manner. Following fear conditioning, mPFC-AT2R Cre-lox deletion impaired extinction and increased exploratory behavior in female but not male mice, while locomotion was unaltered by mPFC-AT2R deletion in both sexes. Conclusions These results identify mPFC-AT2R+ neurons as a novel subgroup of somatostatin interneurons and reveal their role in regulating fear learning in a sex-dependent manner, potentially offering insights into novel therapeutic targets for posttraumatic stress disorder.
Collapse
Affiliation(s)
- Hannah C. Smith
- Department of Neuroscience, George Washington University, Washington, DC
| | - Zhe Yu
- Department of Pharmacology & Physiology, George Washington University, Washington, District of Columbia
| | - Laxmi Iyer
- Department of Pharmacology & Physiology, George Washington University, Washington, District of Columbia
| | - Paul J. Marvar
- Department of Neuroscience, George Washington University, Washington, DC
- Department of Pharmacology & Physiology, George Washington University, Washington, District of Columbia
- Department of Psychiatry and Behavioral Sciences, George Washington University, Washington, DC
| |
Collapse
|
5
|
Xu W, Langhans SA, Johnson DK, Stauff E, Kandula VVR, Kecskemethy HH, Averill LW, Yue X. Radiotracers for Molecular Imaging of Angiotensin-Converting Enzyme 2. Int J Mol Sci 2024; 25:9419. [PMID: 39273366 PMCID: PMC11395405 DOI: 10.3390/ijms25179419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/23/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
Angiotensin-converting enzymes (ACE) are well-known for their roles in both blood pressure regulation via the renin-angiotensin system as well as functions in fertility, immunity, hematopoiesis, and many others. The two main isoforms of ACE include ACE and ACE-2 (ACE2). Both isoforms have similar structures and mediate numerous effects on the cardiovascular system. Most remarkably, ACE2 serves as an entry receptor for SARS-CoV-2. Understanding the interaction between the virus and ACE2 is vital to combating the disease and preventing a similar pandemic in the future. Noninvasive imaging techniques such as positron emission tomography and single photon emission computed tomography could noninvasively and quantitatively assess in vivo ACE2 expression levels. ACE2-targeted imaging can be used as a valuable tool to better understand the mechanism of the infection process and the potential roles of ACE2 in homeostasis and related diseases. Together, this information can aid in the identification of potential therapeutic drugs for infectious diseases, cancer, and many ACE2-related diseases. The present review summarized the state-of-the-art radiotracers for ACE2 imaging, including their chemical design, pharmacological properties, radiochemistry, as well as preclinical and human molecular imaging findings. We also discussed the advantages and limitations of the currently developed ACE2-specific radiotracers.
Collapse
Affiliation(s)
- Wenqi Xu
- Department of Radiology, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
- Diagnostic & Research PET/MR Center, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
| | - Sigrid A Langhans
- Diagnostic & Research PET/MR Center, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
- Division of Neurology, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
| | - David K Johnson
- Computational Chemical Biology Core, Molecular Graphics and Modeling Laboratory, University of Kansas, Lawrence, KS 66047, USA
| | - Erik Stauff
- Department of Radiology, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
- Diagnostic & Research PET/MR Center, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
| | - Vinay V R Kandula
- Department of Radiology, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
| | - Heidi H Kecskemethy
- Department of Radiology, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
- Diagnostic & Research PET/MR Center, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
| | - Lauren W Averill
- Department of Radiology, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
- Diagnostic & Research PET/MR Center, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
| | - Xuyi Yue
- Department of Radiology, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
- Diagnostic & Research PET/MR Center, Nemours Children's Health, Delaware, Wilmington, DE 19803, USA
| |
Collapse
|
6
|
Liu T, Li J, Sun L, Zhu C, Wei J. The role of ACE2 in RAS axis on microglia activation in Parkinson's disease. Neuroscience 2024; 553:128-144. [PMID: 38986737 DOI: 10.1016/j.neuroscience.2024.06.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/19/2024] [Accepted: 06/22/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND The classic renin-angiotensin system (RAS) induces organ damage, while the ACE2/Ang-(1-7)/MasR axis opposes it. However, the role of ACE2 in the brain is unclear. We studied ACE2's role in the brain. METHOD We used male C57BL/6J (WT) mice, ACE2 knockout (KO) mice, and MPTP-induced mice. Behavioral tests confirmed successful modeling. We assessed the impact of ACE2 KO on the RAS axis and PD index, including ACE, ACE2, AT1, AT2, MasR, TH, α-syn, and Iba1. We investigated ACE2 and MasR's involvement in microglial activation via western blot and immunofluorescence. GSE10867 and GSE26532 datasets were used to analyze the effects of AT1 antagonists and in vitro PD models on microglia. RESULT Behavioral tests revealed that MPTP mice displayed motor deficits, depression, anxiety, and increased inflammatory markers in the SN and CPU, with reduced antioxidant capacity. ACE2 KO worsened these symptoms and exacerbated inflammation and oxidative stress. LPS-induced ACE2/MasR activation in BV2 cells demonstrated anti-inflammatory and neuroprotective effects by modulating microglial polarization. Antagonists inhibited microglial activation via inflammation and ROS processes. CONCLUSION The RAS axis regulates inflammation and oxidative stress to maintain CNS function, suggesting potential targets for neurologic disease treatment. Understanding microglial RAS activation can offer new therapeutic strategies.
Collapse
Affiliation(s)
- Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China; Institute of Neurourology and Urodynamics, Huaihe Hospital of Henan University, Kaifeng 475000, China
| | - Jingwen Li
- Institute of Neurourology and Urodynamics, Huaihe Hospital of Henan University, Kaifeng 475000, China
| | - Lin Sun
- College of Chemistry and Molecular Sciences, Henan University, Kaifeng 475000, China.
| | - Chaoyang Zhu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China
| | - Jianshe Wei
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng 475004, China; Institute of Neurourology and Urodynamics, Huaihe Hospital of Henan University, Kaifeng 475000, China.
| |
Collapse
|
7
|
Zika O, Appel J, Klinge C, Shkreli L, Browning M, Wiech K, Reinecke A. Reduction of Aversive Learning Rates in Pavlovian Conditioning by Angiotensin II Antagonist Losartan: A Randomized Controlled Trial. Biol Psychiatry 2024; 96:247-255. [PMID: 38309320 DOI: 10.1016/j.biopsych.2024.01.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 01/12/2024] [Accepted: 01/22/2024] [Indexed: 02/05/2024]
Abstract
BACKGROUND Angiotensin receptor blockade has been linked to aspects of aversive learning and memory formation and to the prevention of posttraumatic stress disorder symptom development. METHODS We investigated the influence of the angiotensin receptor blocker losartan on aversive Pavlovian conditioning using a probabilistic learning paradigm. In a double-blind, randomized, placebo-controlled design, we tested 45 (18 female) healthy volunteers during a baseline session, after application of losartan or placebo (drug session), and during a follow-up session. During each session, participants engaged in a task in which they had to predict the probability of an electrical stimulation on every trial while the true shock contingencies switched repeatedly between phases of high and low shock threat. Computational reinforcement learning models were used to investigate learning dynamics. RESULTS Acute administration of losartan significantly reduced participants' adjustment during both low-to-high and high-to-low threat changes. This was driven by reduced aversive learning rates in the losartan group during the drug session compared with baseline. The 50-mg drug dose did not induce reduction of blood pressure or change in reaction times, ruling out a general reduction in attention and engagement. Decreased adjustment of aversive expectations was maintained at a follow-up session 24 hours later. CONCLUSIONS This study shows that losartan acutely reduces Pavlovian learning in aversive environments, thereby highlighting a potential role of the renin-angiotensin system in anxiety development.
Collapse
Affiliation(s)
- Ondrej Zika
- Max Planck Institute for Human Development, Berlin, Germany
| | - Judith Appel
- Behavioural Science Institute, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Corinna Klinge
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Lorika Shkreli
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Michael Browning
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom; Oxford Health NHS Trust, Warneford Hospital, Oxford, United Kingdom
| | - Katja Wiech
- Wellcome Centre for Integrative Functional Neuroimaging, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Andrea Reinecke
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom; Oxford Health NHS Trust, Warneford Hospital, Oxford, United Kingdom.
| |
Collapse
|
8
|
Amado Costa L, Oliveira Amaral LB, Mourão FAG, Bader M, Santos RAS, Campagnole-Santos MJ, Kangussu LM. Anxiolytic effect of alamandine in male transgenic rats with low brain angiotensinogen is dependent on activation of MrgD receptors. Horm Behav 2024; 163:105551. [PMID: 38678724 DOI: 10.1016/j.yhbeh.2024.105551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/23/2024] [Accepted: 04/22/2024] [Indexed: 05/01/2024]
Abstract
Alamandine is a peptide hormone belonging to the renin-angiotensin system (RAS). It acts through the Mas-related G-protein coupled receptor type D, MrgD, which is expressed in different tissues, including the brain. In the present study, we hypothesize that a lack of alamandine, through MrgD, could cause the anxiety-like behavior in transgenic rats with low brain angiotensinogen [TGR(ASrAOGEN)680]. Adult male transgenic rats exhibited a significant increase in the latency to feeding time in the novelty suppressed feeding test and a decrease in the percentage of time and entries in the open arms in the elevated plus maze. These effects were reversed by intracerebroventricular infusion of alamandine. Pretreatment with D-Pro7-Ang-(1-7), a Mas and MrgD receptor antagonist, prevented the anxiolytic effects induced by this peptide. However, its effects were not altered by the selective Mas receptor antagonist, A779. In conclusion, our data indicates that alamandine, through MrgD, attenuates anxiety-like behavior in male TGR(ASrAOGEN)680, which reinforces the importance of the counter-regulatory RAS axis as promising target for the treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Laura Amado Costa
- Department of Morphology, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Laura B Oliveira Amaral
- Department of Morphology, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Flávio A G Mourão
- Department of Physiology and Biophysics, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Michael Bader
- National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar); Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Institute for Biology, University of Lübeck, Lübeck, Germany; Charité University Medicine Berlin, Berlin, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany
| | - Robson A S Santos
- Department of Physiology and Biophysics, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil; National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar)
| | - Maria José Campagnole-Santos
- Department of Physiology and Biophysics, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil; National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar)
| | - Lucas M Kangussu
- Department of Morphology, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil; National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar).
| |
Collapse
|
9
|
Barbaresi P, Fabri M, Lorenzi T, Sagrati A, Morroni M. Intrinsic organization of the corpus callosum. Front Physiol 2024; 15:1393000. [PMID: 39035452 PMCID: PMC11259024 DOI: 10.3389/fphys.2024.1393000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/16/2024] [Indexed: 07/23/2024] Open
Abstract
The corpus callosum-the largest commissural fiber system connecting the two cerebral hemispheres-is considered essential for bilateral sensory integration and higher cognitive functions. Most studies exploring the corpus callosum have examined either the anatomical, physiological, and neurochemical organization of callosal projections or the functional and/or behavioral aspects of the callosal connections after complete/partial callosotomy or callosal lesion. There are no works that address the intrinsic organization of the corpus callosum. We review the existing information on the activities that take place in the commissure in three sections: I) the topographical and neurochemical organization of the intracallosal fibers, II) the role of glia in the corpus callosum, and III) the role of the intracallosal neurons.
Collapse
Affiliation(s)
- Paolo Barbaresi
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Marche Polytechnic University, Ancona, Italy
| | - Mara Fabri
- Department of Life and Environmental Sciences, Marche Polytechnic University, Ancona, Italy
| | - Teresa Lorenzi
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Marche Polytechnic University, Ancona, Italy
| | - Andrea Sagrati
- Department of Life and Environmental Sciences, Marche Polytechnic University, Ancona, Italy
| | - Manrico Morroni
- Electron Microscopy Unit, Azienda Ospedaliero-Universitaria, Ancona, Italy
| |
Collapse
|
10
|
Zuo HJ, Ren XQ, Shi JS, Shi HL, Guo K, Wang PX, Zhao M, Li JJ. Gastrodin regulates the expression of renin-angiotensin system-SIRT3 and proinflammatory mediators in reactive astrocytes via activated microglia. Eur J Neurosci 2024; 60:3677-3693. [PMID: 38711280 DOI: 10.1111/ejn.16371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 03/19/2024] [Accepted: 04/11/2024] [Indexed: 05/08/2024]
Abstract
Gastrodin, an anti-inflammatory herbal agent, is known to suppress microglia activation. Here, we investigated whether it would exert a similar effect in reactive astrocytes and whether it might act through the renin-angiotensin system (RAS) and sirtuin 3 (SIRT3). Angiotensinogen (ATO), angiotensin-converting enzyme (ACE), angiotensin II type 1 (AT1) and type 2 (AT2) receptor and SIRT3 expression was detected in TNC-1 astrocytes treated with BV-2 microglia conditioned medium (CM) with or without gastrodin and lipopolysaccharide (LPS) pre-treatment by RT-PCR, immunofluorescence and western blotting analysis. Expression of C3 (A1 astrocyte marker), S100A10 (A2 astrocyte marker), proinflammatory cytokines and neurotrophic factors was then evaluated. The results showed a significant increase of ATO, ACE, AT1, SIRT3, C3, proinflammatory cytokines and neurotrophic factors expression in TNC-1 astrocytes incubated in CM + LPS when compared with cells incubated in the CM, but AT2 and S100A10 expression was reduced. TNC-1 astrocytes responded vigorously to BV-2 CM treated with gastrodin + LPS as compared with the control. This was evident by the decreased expression of the abovementioned protein markers, except for AT2 and S100A10. Interestingly, SIRT3, IGF-1 and BDNF expression was enhanced, suggesting that gastrodin inhibited the expression of RAS and proinflammatory mediators but promoted the expression of neurotrophic factors. And gastrodin regulated the phenotypic changes of astrocytes through AT1. Additionally, azilsartan (a specific inhibitor of AT1) inhibited the expression of C3 and S100A10, which remained unaffected in gastrodin and azilsartan combination treatment. These findings provide evidence that gastrodin may have a therapeutic effect via regulating RAS-SIRT3.
Collapse
Affiliation(s)
- Han-Jun Zuo
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Xue-Qi Ren
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Jin-Sha Shi
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Hao-Long Shi
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Kun Guo
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Peng-Xiang Wang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Min Zhao
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Juan-Juan Li
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, China
| |
Collapse
|
11
|
Etebar N, Naderpour S, Akbari S, Zali A, Akhlaghdoust M, Daghighi SM, Baghani M, Sefat F, Hamidi SH, Rahimzadegan M. Impacts of SARS-CoV-2 on brain renin angiotensin system related signaling and its subsequent complications on brain: A theoretical perspective. J Chem Neuroanat 2024; 138:102423. [PMID: 38705215 DOI: 10.1016/j.jchemneu.2024.102423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/08/2024] [Accepted: 04/18/2024] [Indexed: 05/07/2024]
Abstract
Cellular ACE2 (cACE2), a vital component of the renin-angiotensin system (RAS), possesses catalytic activity to maintain AngII and Ang 1-7 balance, which is necessary to prevent harmful effects of AngII/AT2R and promote protective pathways of Ang (1-7)/MasR and Ang (1-7)/AT2R. Hemostasis of the brain-RAS is essential for maintaining normal central nervous system (CNS) function. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a viral disease that causes multi-organ dysfunction. SARS-CoV-2 mainly uses cACE2 to enter the cells and cause its downregulation. This, in turn, prevents the conversion of Ang II to Ang (1-7) and disrupts the normal balance of brain-RAS. Brain-RAS disturbances give rise to one of the pathological pathways in which SARS-CoV-2 suppresses neuroprotective pathways and induces inflammatory cytokines and reactive oxygen species. Finally, these impairments lead to neuroinflammation, neuronal injury, and neurological complications. In conclusion, the influence of RAS on various processes within the brain has significant implications for the neurological manifestations associated with COVID-19. These effects include sensory disturbances, such as olfactory and gustatory dysfunctions, as well as cerebrovascular and brain stem-related disorders, all of which are intertwined with disruptions in the RAS homeostasis of the brain.
Collapse
Affiliation(s)
- Negar Etebar
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Faculty of Pharmacy - Eastern Mediterranean University Famagusta, North Cyprus via Mersin 10, Turkey
| | - Saghi Naderpour
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Faculty of Pharmacy - Eastern Mediterranean University Famagusta, North Cyprus via Mersin 10, Turkey
| | - Setareh Akbari
- Neuroscience and Research Committee, School of Advanced Technology in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Zali
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Meisam Akhlaghdoust
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran; USERN Office, Functional Neurosurgery Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mojtaba Daghighi
- Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Matin Baghani
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farshid Sefat
- Department of Biomedical Engineering, School of Engineering, University of Bradford, Bradford, UK
| | - Seyed Hootan Hamidi
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Acharya BM Reddy College of Pharmacy, Rajiv Gandhi University of Health Sciences, Bangalore, India
| | - Milad Rahimzadegan
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Olschewski DN, Nazarzadeh N, Lange F, Koenig AM, Kulka C, Abraham JA, Blaschke SJ, Merkel R, Hoffmann B, Fink GR, Schroeter M, Rueger MA, Vay SU. The angiotensin II receptors type 1 and 2 modulate astrocytes and their crosstalk with microglia and neurons in an in vitro model of ischemic stroke. BMC Neurosci 2024; 25:29. [PMID: 38926677 PMCID: PMC11202395 DOI: 10.1186/s12868-024-00876-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Astrocytes are the most abundant cell type of the central nervous system and are fundamentally involved in homeostasis, neuroprotection, and synaptic plasticity. This regulatory function of astrocytes on their neighboring cells in the healthy brain is subject of current research. In the ischemic brain we assume disease specific differences in astrocytic acting. The renin-angiotensin-aldosterone system regulates arterial blood pressure through endothelial cells and perivascular musculature. Moreover, astrocytes express angiotensin II type 1 and 2 receptors. However, their role in astrocytic function has not yet been fully elucidated. We hypothesized that the angiotensin II receptors impact astrocyte function as revealed in an in vitro system mimicking cerebral ischemia. Astrocytes derived from neonatal wistar rats were exposed to telmisartan (angiotensin II type 1 receptor-blocker) or PD123319 (angiotensin II type 2 receptor-blocker) under normal conditions (control) or deprivation from oxygen and glucose. Conditioned medium (CM) of astrocytes was harvested to elucidate astrocyte-mediated indirect effects on microglia and cortical neurons. RESULT The blockade of angiotensin II type 1 receptor by telmisartan increased the survival of astrocytes during ischemic conditions in vitro without affecting their proliferation rate or disturbing their expression of S100A10, a marker of activation. The inhibition of the angiotensin II type 2 receptor pathway by PD123319 resulted in both increased expression of S100A10 and proliferation rate. The CM of telmisartan-treated astrocytes reduced the expression of pro-inflammatory mediators with simultaneous increase of anti-inflammatory markers in microglia. Increased neuronal activity was observed after treatment of neurons with CM of telmisartan- as well as PD123319-stimulated astrocytes. CONCLUSION Data show that angiotensin II receptors have functional relevance for astrocytes that differs in healthy and ischemic conditions and effects surrounding microglia and neuronal activity via secretory signals. Above that, this work emphasizes the strong interference of the different cells in the CNS and that targeting astrocytes might serve as a therapeutic strategy to influence the acting of glia-neuronal network in de- and regenerative context.
Collapse
Affiliation(s)
- Daniel Navin Olschewski
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany.
| | - Nilufar Nazarzadeh
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Felix Lange
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Anna Maria Koenig
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Christina Kulka
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Jella-Andrea Abraham
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
- Department of Mechanobiology, Institute of Biological Information Processing (IBI-2), Research Centre Juelich, Juelich, Germany
| | - Stefan Johannes Blaschke
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany
| | - Rudolf Merkel
- Department of Mechanobiology, Institute of Biological Information Processing (IBI-2), Research Centre Juelich, Juelich, Germany
| | - Bernd Hoffmann
- Department of Mechanobiology, Institute of Biological Information Processing (IBI-2), Research Centre Juelich, Juelich, Germany
| | - Gereon Rudolf Fink
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany
| | - Michael Schroeter
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany
| | - Maria Adele Rueger
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany
| | - Sabine Ulrike Vay
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
13
|
Hong J, Li Y, Chen L, Han D, Li Y, Mi X, Liu K, Wang Q, Song Y, Liu T, Yang N, Liu Y, Li Z, Guo X. A53T α-synuclein mutation increases susceptibility to postoperative delayed neurocognitive recovery via hippocampal Ang-(1-7)/MasR axis. Biochem Pharmacol 2024; 224:116261. [PMID: 38705534 DOI: 10.1016/j.bcp.2024.116261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/28/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Delayed neurocognitive recovery (dNCR) is a common complication in geriatric surgical patients. The impact of anesthesia and surgery on patients with neurodegenerative diseases, such as Parkinson's disease (PD) or prion disease, has not yet been reported. In this study, we aimed to determine the association between a pre-existing A53T genetic background, which involves a PD-related point mutation, and the development of postoperative dNCR. We observed that partial hepatectomy induced hippocampus-dependent cognitive deficits in 5-month-old A53T transgenic mice, a model of early-stage PD without cognitive deficits, unlike in age-matched wild-type (WT) mice. We respectively examined molecular changes at 6 h, 1 day, and 2 days after partial hepatectomy and observed that cognitive changes were accompanied by weakened angiotensin-(1-7)/Mas receptor [Ang-(1-7)/MasR] axis, increased alpha-synuclein (α-syn) expression and phosphorylation, decreased methylated protein phosphatase-2A (Me-PP2A), and prompted microglia M1 polarization and neuronal apoptosis in the hippocampus at 1 day after surgery. Nevertheless, no changes in blood-brain barrier (BBB) integrity or plasma α-syn levels in either A53T or WT mice. Furthermore, intranasal administration of selective MasR agonist AVE 0991, reversed the mentioned cognitive deficits in A53T mice, enhanced MasR expression, reduced α-syn accumulation and phosphorylation, and attenuated microglia activation and apoptotic response. Our findings suggest that individuals with the A53T genetic background may be more susceptible to developing postoperative dNCR. This susceptibility could be linked to central α-syn accumulation mediated by the weakened Ang-(1-7)/MasR/methyl-PP2A signaling pathway in the hippocampus following surgery, independent of plasma α-syn level and BBB.
Collapse
Affiliation(s)
- Jingshu Hong
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Yue Li
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Lei Chen
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Dengyang Han
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Yitong Li
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Xinning Mi
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Kaixi Liu
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Qian Wang
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Yanan Song
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China; Beijing Center of Quality Control and Improvement on Clinical Anesthesia, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Taotao Liu
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Ning Yang
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Yajie Liu
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Zhengqian Li
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China; Beijing Center of Quality Control and Improvement on Clinical Anesthesia, No. 49, North Garden Street, Haidian District, Beijing 100191, China; Anesthesia and Perioperative Medicine Branch of China International Exchange and Promotive Association for Medical and Health Care (CPAM), No. 49, North Garden Street, Haidian District, Beijing 100191, China.
| | - Xiangyang Guo
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China; Beijing Center of Quality Control and Improvement on Clinical Anesthesia, No. 49, North Garden Street, Haidian District, Beijing 100191, China; Anesthesia and Perioperative Medicine Branch of China International Exchange and Promotive Association for Medical and Health Care (CPAM), No. 49, North Garden Street, Haidian District, Beijing 100191, China.
| |
Collapse
|
14
|
Moore GJ, Ridway H, Gadanec LK, Apostolopoulos V, Zulli A, Swiderski J, Kelaidonis K, Vidali VP, Matsoukas MT, Chasapis CT, Matsoukas JM. Structural Features Influencing the Bioactive Conformation of Angiotensin II and Angiotensin A: Relationship between Receptor Desensitization, Addiction, and the Blood-Brain Barrier. Int J Mol Sci 2024; 25:5779. [PMID: 38891966 PMCID: PMC11171751 DOI: 10.3390/ijms25115779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/04/2024] [Accepted: 05/08/2024] [Indexed: 06/21/2024] Open
Abstract
The N-terminal portion of the octapeptide angiotensin II (DRVYIHPF; AngII), a vasopressor peptide that favorably binds to, and activates, AngII type 1 receptor (AT1R), has an important role in maintaining bioactive conformation. It involves all three charged groups, namely (i) the N-terminal amino group cation, (ii) the Asp sidechain anion and (iii) the Arg guanidino cation. Neutralization of any one of these three charged groups results in a substantial reduction (<5%) in bioactivity, implicating a specialized function for this cluster. In contrast, angiotensin A (ARVYIHPF; AngA) has reduced bioactivity at AT1R; however, replacement of Asp in AngII with sarcosine (N-methyl-glycine) not only restores bioactivity but increases the activity of agonist, antagonist, and inverse agonist analogues. A bend produced at the N-terminus by the introduction of the secondary amino acid sarcosine is thought to realign the functional groups that chaperone the C-terminal portion of AngII, allowing transfer of the negative charge originating at the C-terminus to be transferred to the Tyr hydroxyl-forming tyrosinate anion, which is required to activate the receptor and desensitizes the receptor (tachyphylaxis). Peptide (sarilesin) and nonpeptide (sartans) moieties, which are long-acting inverse agonists, appear to desensitize the receptor by a mechanism analogous to tachyphylaxis. Sartans/bisartans were found to bind to alpha adrenergic receptors resulting in structure-dependent desensitization or resensitization. These considerations have provided information on the mechanisms of receptor desensitization/tolerance and insights into possible avenues for treating addiction. In this regard sartans, which appear to cross the blood-brain barrier more readily than bisartans, are the preferred drug candidates.
Collapse
Affiliation(s)
- Graham J. Moore
- Pepmetics Inc., 772 Murphy Place, Victoria, BC V8Y 3H4, Canada;
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Harry Ridway
- Institute for Sustainable Industries and Liveable Cities, Victoria University, Melbourne, VIC 8001, Australia;
| | - Laura Kate Gadanec
- Institute for Health and Sport, Immunology and Translational Research, Victoria University, Melbourne, VIC 3030, Australia; (L.K.G.); (V.A.); (A.Z.); (J.S.)
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Immunology and Translational Research, Victoria University, Melbourne, VIC 3030, Australia; (L.K.G.); (V.A.); (A.Z.); (J.S.)
- Immunology Program, Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
| | - Anthony Zulli
- Institute for Health and Sport, Immunology and Translational Research, Victoria University, Melbourne, VIC 3030, Australia; (L.K.G.); (V.A.); (A.Z.); (J.S.)
| | - Jordan Swiderski
- Institute for Health and Sport, Immunology and Translational Research, Victoria University, Melbourne, VIC 3030, Australia; (L.K.G.); (V.A.); (A.Z.); (J.S.)
| | | | - Veroniki P. Vidali
- Institute of Nanoscience and Nanotechnology, National Centre for Scientific Research “Demokritos”, 15341 Athens, Greece;
| | | | - Christos T. Chasapis
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece;
| | - John M. Matsoukas
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
- Institute for Health and Sport, Immunology and Translational Research, Victoria University, Melbourne, VIC 3030, Australia; (L.K.G.); (V.A.); (A.Z.); (J.S.)
- NewDrug/NeoFar PC, Patras Science Park, 26504 Patras, Greece;
- Department of Chemistry, University of Patras, 26504 Patras, Greece
| |
Collapse
|
15
|
Liu T, Wu H, Sun L, Wei J. Role of Inflammation in the Development of COVID-19 to Parkinson's Disease. J Inflamm Res 2024; 17:3259-3282. [PMID: 38800597 PMCID: PMC11127656 DOI: 10.2147/jir.s460161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/16/2024] [Indexed: 05/29/2024] Open
Abstract
Background The coronavirus disease 2019 (COVID-19) can lead to neurological symptoms such as headaches, confusion, seizures, hearing loss, and loss of smell. The link between COVID-19 and Parkinson's disease (PD) is being investigated, but more research is needed for a definitive connection. Methods Datasets GSE22491 and GSE164805 were selected to screen differentially expressed gene (DEG), and immune infiltration and gene set enrichment analysis (GSEA) of the DEG were performed. WGCNA analyzed the DEG and selected the intersection genes. Potential biological functions and signaling pathways were determined, and diagnostic genes were further screened using gene expression and receiver operating characteristic (ROC) curves. Screening and molecular docking of ibuprofen as a therapeutic target. The effectiveness of ibuprofen was verified by constructing a PD model in vitro, and constructing "COVID19-PD" signaling pathway, and exploring the role of angiotensin-converting enzyme 2 (ACE2) in PD. Results A total of 13 DEG were screened from the GSE36980 and GSE5281 datasets. Kyoto encyclopedia of genes and genomes (KEGG) analysis showed that the DEG were mainly associated with the hypoxia-inducible factor (HIF-1), epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor resistance, etc. After analysis, it is found that ibuprofen alleviates PD symptoms by inhibiting the expression of nuclear factor kappa-B (NF-κB), interleukin-1β (IL-1β), IL-6, and tumor necrosis factor-α (TNF-α). Based on signal pathway construction, the importance of ACE2 in COVID-19-induced PD has been identified. ACE2 is found to have widespread distribution in the brain. In the 1-methyl-4-phenyl-1,2,3,6-te-trahydropyridine (MPTP)-induced ACE2-null PD mice model, more severe motor and non-motor symptoms, increased NF-κB p65 and α-synuclein (α-syn) expression with significant aggregation, decreased tyrosine hydroxylase (TH), severe neuronal loss, and neurodegenerative disorders. Conclusion Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection increases the risk of PD through an inflammatory environment and downregulation of ACE2, providing evidence for the molecular mechanism and targeted therapy associated with COVID-19 and PD.
Collapse
Affiliation(s)
- Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Institute of Neurourology and Urodynamics, Huaihe Hospital of Henan University, Kaifeng, 475004, People’s Republic of China
| | - Haojie Wu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Institute of Neurourology and Urodynamics, Huaihe Hospital of Henan University, Kaifeng, 475004, People’s Republic of China
| | - Lin Sun
- College of Chemistry and Molecular Sciences, Henan University, Kaifeng, 475004, People’s Republic of China
| | - Jianshe Wei
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Institute of Neurourology and Urodynamics, Huaihe Hospital of Henan University, Kaifeng, 475004, People’s Republic of China
| |
Collapse
|
16
|
Khan N, Kurnik-Łucka M, Latacz G, Gil K. Systematic-Narrative Hybrid Literature Review: Crosstalk between Gastrointestinal Renin-Angiotensin and Dopaminergic Systems in the Regulation of Intestinal Permeability by Tight Junctions. Int J Mol Sci 2024; 25:5566. [PMID: 38791603 PMCID: PMC11122119 DOI: 10.3390/ijms25105566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/12/2024] [Accepted: 05/18/2024] [Indexed: 05/26/2024] Open
Abstract
In the first part of this article, the role of intestinal epithelial tight junctions (TJs), together with gastrointestinal dopaminergic and renin-angiotensin systems, are narratively reviewed to provide sufficient background. In the second part, the current experimental data on the interplay between gastrointestinal (GI) dopaminergic and renin-angiotensin systems in the regulation of intestinal epithelial permeability are reviewed in a systematic manner using the PRISMA methodology. Experimental data confirmed the copresence of DOPA decarboxylase (DDC) and angiotensin converting enzyme 2 (ACE2) in human and rodent enterocytes. The intestinal barrier structure and integrity can be altered by angiotensin (1-7) and dopamine (DA). Both renin-angiotensin and dopaminergic systems influence intestinal Na+/K+-ATPase activity, thus maintaining electrolyte and nutritional homeostasis. The colocalization of B0AT1 and ACE2 indicates the direct role of the renin-angiotensin system in amino acid absorption. Yet, more studies are needed to thoroughly define the structural and functional interaction between TJ-associated proteins and GI renin-angiotensin and dopaminergic systems.
Collapse
Affiliation(s)
- Nadia Khan
- Faculty of Medicine, Department of Pathophysiology, Jagiellonian University Medical College, Czysta 18, 31-121 Krakow, Poland
- Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 31-008 Krakow, Poland
| | - Magdalena Kurnik-Łucka
- Faculty of Medicine, Department of Pathophysiology, Jagiellonian University Medical College, Czysta 18, 31-121 Krakow, Poland
| | - Gniewomir Latacz
- Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 31-008 Krakow, Poland
| | - Krzysztof Gil
- Faculty of Medicine, Department of Pathophysiology, Jagiellonian University Medical College, Czysta 18, 31-121 Krakow, Poland
| |
Collapse
|
17
|
Rodríguez-Pérez AI, Garrido-Gil P, García-Garrote M, Muñoz A, Parga JA, Labandeira-García JL, Rodríguez-Pallares J. Non-HLA angiotensin-type-1 receptor autoantibodies mediate the long-term loss of grafted neurons in Parkinson's disease models. Stem Cell Res Ther 2024; 15:138. [PMID: 38735991 PMCID: PMC11089721 DOI: 10.1186/s13287-024-03751-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 05/02/2024] [Indexed: 05/14/2024] Open
Abstract
BACKGROUND Clinical trials have provided evidence that transplants of dopaminergic precursors, which may be replaced by new in vitro stem cell sources, can integrate into the host tissue, and alleviate motor symptoms in Parkinson´s disease (PD). In some patients, deterioration of graft function occurred several months after observing a graft-derived functional improvement. Rejection of peripheral organs was initially related to HLA-specific antibodies. However, the role of non-HLA antibodies is now considered also relevant for rejection. Angiotensin-II type-1 receptor autoantibodies (AT1-AA) act as agonists of the AT1 receptors. AT1-AA are the non-HLA antibodies most widely associated with graft dysfunction or rejection after transplantation of different solid organs and hematopoietic stem cells. However, it is not known about the presence and possible functional effects of AT1-AA in dopaminergic grafts, and the effects of treatment with AT1 receptor blockers (ARBs) such as candesartan on graft survival. METHODS In a 6-hydroxydopamine PD rat model, we studied the short-term (10 days)- and long-term (3 months) effects of chronic treatment with the ARB candesartan on survival of grafted dopaminergic neurons and microglial graft infiltration, as well as the effects of dopaminergic denervation and grafting on serum and CSF AT1-AA levels. The expression of AT1 receptors in grafted neurons was determined by laser capture microdissection. RESULTS At the early period post-grafting, the number of grafted dopaminergic neurons that survived was not significantly different between treated and untreated hosts (i.e., control rats and rats treated with candesartan), probably because, just after grafting, other deleterious factors are predominant for dopaminergic cell death, such as mechanical trauma, lack of growth factors/nutrients and ischemia. However, several months post-grafting, we observed a significantly higher number of surviving dopaminergic neurons and a higher density of striatal dopaminergic terminals in the candesartan-treated group. For several months, grafted rats showed blood and cerebrospinal fluid levels of AT1-AA higher than normal controls, and also higher AT1-AA levels than non-grafted parkinsonian rats. CONCLUSIONS The results suggest the use of ARBs such as candesartan in PD patients, particularly before and after dopaminergic grafts, and the need to monitor AT1-AA levels in PD patients, particularly in those candidates for dopaminergic grafting.
Collapse
Affiliation(s)
- Ana I Rodríguez-Pérez
- Research Center for Molecular Medicine and Chronic Diseases (CiMUS), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Pablo Garrido-Gil
- Research Center for Molecular Medicine and Chronic Diseases (CiMUS), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Maria García-Garrote
- Research Center for Molecular Medicine and Chronic Diseases (CiMUS), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Ana Muñoz
- Research Center for Molecular Medicine and Chronic Diseases (CiMUS), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Juan A Parga
- Research Center for Molecular Medicine and Chronic Diseases (CiMUS), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jose Luis Labandeira-García
- Research Center for Molecular Medicine and Chronic Diseases (CiMUS), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, 15782, Spain.
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| | - Jannette Rodríguez-Pallares
- Research Center for Molecular Medicine and Chronic Diseases (CiMUS), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, 15782, Spain.
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| |
Collapse
|
18
|
Labandeira-Garcia JL, Labandeira CM, Guerra MJ, Rodriguez-Perez AI. The role of the brain renin-angiotensin system in Parkinson´s disease. Transl Neurodegener 2024; 13:22. [PMID: 38622720 PMCID: PMC11017622 DOI: 10.1186/s40035-024-00410-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/22/2024] [Indexed: 04/17/2024] Open
Abstract
The renin-angiotensin system (RAS) was classically considered a circulating hormonal system that regulates blood pressure. However, different tissues and organs, including the brain, have a local paracrine RAS. Mutual regulation between the dopaminergic system and RAS has been observed in several tissues. Dysregulation of these interactions leads to renal and cardiovascular diseases, as well as progression of dopaminergic neuron degeneration in a major brain center of dopamine/angiotensin interaction such as the nigrostriatal system. A decrease in the dopaminergic function induces upregulation of the angiotensin type-1 (AT1) receptor activity, leading to recovery of dopamine levels. However, AT1 receptor overactivity in dopaminergic neurons and microglial cells upregulates the cellular NADPH-oxidase-superoxide axis and Ca2+ release, which mediate several key events in oxidative stress, neuroinflammation, and α-synuclein aggregation, involved in Parkinson's disease (PD) pathogenesis. An intraneuronal antioxidative/anti-inflammatory RAS counteracts the effects of the pro-oxidative AT1 receptor overactivity. Consistent with this, an imbalance in RAS activity towards the pro-oxidative/pro-inflammatory AT1 receptor axis has been observed in the substantia nigra and striatum of several animal models of high vulnerability to dopaminergic degeneration. Interestingly, autoantibodies against angiotensin-converting enzyme 2 and AT1 receptors are increased in PD models and PD patients and contribute to blood-brain barrier (BBB) dysregulation and nigrostriatal pro-inflammatory RAS upregulation. Therapeutic strategies addressed to the modulation of brain RAS, by AT1 receptor blockers (ARBs) and/or activation of the antioxidative axis (AT2, Mas receptors), may be neuroprotective for individuals with a high risk of developing PD or in prodromal stages of PD to reduce progression of the disease.
Collapse
Affiliation(s)
- Jose Luis Labandeira-Garcia
- Cellular and Molecular Neurobiology of Parkinson´S Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain.
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| | | | - Maria J Guerra
- Cellular and Molecular Neurobiology of Parkinson´S Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Ana I Rodriguez-Perez
- Cellular and Molecular Neurobiology of Parkinson´S Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain.
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| |
Collapse
|
19
|
Gising J, Honarnejad S, Bras M, Baillie GL, McElroy SP, Jones PS, Morrison A, Beveridge J, Hallberg M, Larhed M. The Discovery of New Inhibitors of Insulin-Regulated Aminopeptidase by a High-Throughput Screening of 400,000 Drug-like Compounds. Int J Mol Sci 2024; 25:4084. [PMID: 38612894 PMCID: PMC11012289 DOI: 10.3390/ijms25074084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/25/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
With the ambition to identify novel chemical starting points that can be further optimized into small drug-like inhibitors of insulin-regulated aminopeptidase (IRAP) and serve as potential future cognitive enhancers in the clinic, we conducted an ultra-high-throughput screening campaign of a chemically diverse compound library of approximately 400,000 drug-like small molecules. Three biochemical and one biophysical assays were developed to enable large-scale screening and hit triaging. The screening funnel, designed to be compatible with high-density microplates, was established with two enzyme inhibition assays employing either fluorescent or absorbance readouts. As IRAP is a zinc-dependent enzyme, the remaining active compounds were further evaluated in the primary assay, albeit with the addition of zinc ions. Rescreening with zinc confirmed the inhibitory activity for most compounds, emphasizing a zinc-independent mechanism of action. Additionally, target engagement was confirmed using a complementary biophysical thermal shift assay where compounds causing positive/negative thermal shifts were considered genuine binders. Triaging based on biochemical activity, target engagement, and drug-likeness resulted in the selection of 50 qualified hits, of which the IC50 of 32 compounds was below 3.5 µM. Despite hydroxamic acid dominance, diverse chemotypes with biochemical activity and target engagement were discovered, including non-hydroxamic acid compounds. The most potent compound (QHL1) was resynthesized with a confirmed inhibitory IC50 of 320 nM. Amongst these compounds, 20 new compound structure classes were identified, providing many new starting points for the development of unique IRAP inhibitors. Detailed characterization and optimization of lead compounds, considering both hydroxamic acids and other diverse structures, are in progress for further exploration.
Collapse
Affiliation(s)
- Johan Gising
- The Beijer Laboratory, Science for Life Laboratory, Department of Medicinal Chemistry, Biomedical Centre, Uppsala University, P.O. Box 574, SE-751 23 Uppsala, Sweden; (J.B.); (M.L.)
| | - Saman Honarnejad
- Pivot Park Screening Centre, Kloosterstraat 9, 5349 AB Oss, The Netherlands; (S.H.); (M.B.)
| | - Maaike Bras
- Pivot Park Screening Centre, Kloosterstraat 9, 5349 AB Oss, The Netherlands; (S.H.); (M.B.)
| | - Gemma L. Baillie
- BioAscent Discovery Ltd., Bo‘Ness Road, Newhouse, Motherwell ML1 5UH, UK; (G.L.B.); (S.P.M.); (P.S.J.); (A.M.)
| | - Stuart P. McElroy
- BioAscent Discovery Ltd., Bo‘Ness Road, Newhouse, Motherwell ML1 5UH, UK; (G.L.B.); (S.P.M.); (P.S.J.); (A.M.)
| | - Philip S. Jones
- BioAscent Discovery Ltd., Bo‘Ness Road, Newhouse, Motherwell ML1 5UH, UK; (G.L.B.); (S.P.M.); (P.S.J.); (A.M.)
| | - Angus Morrison
- BioAscent Discovery Ltd., Bo‘Ness Road, Newhouse, Motherwell ML1 5UH, UK; (G.L.B.); (S.P.M.); (P.S.J.); (A.M.)
| | - Julia Beveridge
- The Beijer Laboratory, Science for Life Laboratory, Department of Medicinal Chemistry, Biomedical Centre, Uppsala University, P.O. Box 574, SE-751 23 Uppsala, Sweden; (J.B.); (M.L.)
| | - Mathias Hallberg
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Neuropharmacology and Addiction Research, Biomedical Centre, Uppsala University, P.O. Box 591, SE-751 24 Uppsala, Sweden;
| | - Mats Larhed
- The Beijer Laboratory, Science for Life Laboratory, Department of Medicinal Chemistry, Biomedical Centre, Uppsala University, P.O. Box 574, SE-751 23 Uppsala, Sweden; (J.B.); (M.L.)
| |
Collapse
|
20
|
Lozinski M, Lumbers ER, Bowden NA, Martin JH, Fay MF, Pringle KG, Tooney PA. Upregulation of the Renin-Angiotensin System Is Associated with Patient Survival and the Tumour Microenvironment in Glioblastoma. Cells 2024; 13:634. [PMID: 38607073 PMCID: PMC11012120 DOI: 10.3390/cells13070634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024] Open
Abstract
Glioblastoma is a highly aggressive disease with poor survival outcomes. An emerging body of literature links the role of the renin-angiotensin system (RAS), well-known for its function in the cardiovascular system, to the progression of cancers. We studied the expression of RAS-related genes (ATP6AP2, AGTR1, AGTR2, ACE, AGT, and REN) in The Cancer Genome Atlas (TCGA) glioblastoma cohort, their relationship to patient survival, and association with tumour microenvironment pathways. The expression of RAS genes was then examined in 12 patient-derived glioblastoma cell lines treated with chemoradiation. In cases of glioblastoma within the TCGA, ATP6AP2, AGTR1, ACE, and AGT had consistent expressions across samples, while AGTR2 and REN were lowly expressed. High expression of AGTR1 was independently associated with lower progression-free survival (PFS) (p = 0.01) and had a non-significant trend for overall survival (OS) after multivariate analysis (p = 0.095). The combined expression of RAS receptors (ATP6AP2, AGTR1, and AGTR2) was positively associated with gene pathways involved in hypoxia, microvasculature, stem cell plasticity, and the molecular characterisation of glioblastoma subtypes. In patient-derived glioblastoma cell lines, ATP6AP2 and AGTR1 were upregulated after chemoradiotherapy and correlated with an increase in HIF1A expression. This data suggests the RAS is correlated with changes in the tumour microenvironment and associated with glioblastoma survival outcomes.
Collapse
Affiliation(s)
- Mathew Lozinski
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308, Australia; (M.L.); (N.A.B.); (J.H.M.); (M.F.F.)
- Mark Hughes Foundation Centre for Brain Cancer Research, University of Newcastle, Callaghan, NSW 2308, Australia
- Drug Repurposing and Medicines Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Eugenie R. Lumbers
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308, Australia; (E.R.L.); (K.G.P.)
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Nikola A. Bowden
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308, Australia; (M.L.); (N.A.B.); (J.H.M.); (M.F.F.)
- Drug Repurposing and Medicines Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Jennifer H. Martin
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308, Australia; (M.L.); (N.A.B.); (J.H.M.); (M.F.F.)
- Drug Repurposing and Medicines Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Michael F. Fay
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308, Australia; (M.L.); (N.A.B.); (J.H.M.); (M.F.F.)
- Mark Hughes Foundation Centre for Brain Cancer Research, University of Newcastle, Callaghan, NSW 2308, Australia
- Drug Repurposing and Medicines Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
- GenesisCare, Gateshead, NSW 2290, Australia
| | - Kirsty G. Pringle
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308, Australia; (E.R.L.); (K.G.P.)
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Paul A. Tooney
- Mark Hughes Foundation Centre for Brain Cancer Research, University of Newcastle, Callaghan, NSW 2308, Australia
- Drug Repurposing and Medicines Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW 2308, Australia; (E.R.L.); (K.G.P.)
| |
Collapse
|
21
|
Carroll KR, Mizrachi M, Simmons S, Toz B, Kowal C, Wingard J, Tehrani N, Zarfeshani A, Kello N, El Khoury L, Weissman-Tsukamoto R, Levin JZ, Volpe BT, Diamond B. Lupus autoantibodies initiate neuroinflammation sustained by continuous HMGB1:RAGE signaling and reversed by increased LAIR-1 expression. Nat Immunol 2024; 25:671-681. [PMID: 38448779 PMCID: PMC11141703 DOI: 10.1038/s41590-024-01772-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 01/25/2024] [Indexed: 03/08/2024]
Abstract
Cognitive impairment is a frequent manifestation of neuropsychiatric systemic lupus erythematosus, present in up to 80% of patients and leading to a diminished quality of life. In the present study, we used a model of lupus-like cognitive impairment that is initiated when antibodies that crossreact with excitatory neuronal receptors penetrate the hippocampus, causing immediate, self-limited, excitotoxic death of hippocampal neurons, which is then followed by a significant loss of dendritic complexity in surviving neurons. This injury creates a maladaptive equilibrium that is sustained in mice for at least 1 year. We identified a feedforward loop of microglial activation and microglia-dependent synapse elimination dependent on neuronal secretion of high mobility group box 1 protein (HMGB1) which binds the receptor for advanced glycation end products (RAGE) and leads to microglial secretion of C1q, upregulation of interleukin-10 with consequent downregulation of leukocyte-associated immunoglobulin-like receptor 1 (LAIR-1), an inhibitory receptor for C1q. Treatment with a centrally acting angiotensin-converting enzyme inhibitor or with an angiotensin-receptor blocker restored a healthy equilibrium, microglial quiescence and intact spatial memory.
Collapse
Affiliation(s)
- Kaitlin R Carroll
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Mark Mizrachi
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Sean Simmons
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bahtiyar Toz
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Czeslawa Kowal
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Jeffrey Wingard
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Nazila Tehrani
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Aida Zarfeshani
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | | | | | | | - Joshua Z Levin
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bruce T Volpe
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Betty Diamond
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, USA.
| |
Collapse
|
22
|
Rao A, Bhat SA, Shibata T, Giani JF, Rader F, Bernstein KE, Khan Z. Diverse biological functions of the renin-angiotensin system. Med Res Rev 2024; 44:587-605. [PMID: 37947345 DOI: 10.1002/med.21996] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 08/30/2023] [Accepted: 10/29/2023] [Indexed: 11/12/2023]
Abstract
The renin-angiotensin system (RAS) has been widely known as a circulating endocrine system involved in the control of blood pressure. However, components of RAS have been found to be localized in rather unexpected sites in the body including the kidneys, brain, bone marrow, immune cells, and reproductive system. These discoveries have led to steady, growing evidence of the existence of independent tissue RAS specific to several parts of the body. It is important to understand how RAS regulates these systems for a variety of reasons: It gives a better overall picture of human physiology, helps to understand and mitigate the unintended consequences of RAS-inhibiting or activating drugs, and sets the stage for potential new therapies for a variety of ailments. This review fulfills the need for an updated overview of knowledge about local tissue RAS in several bodily systems, including their components, functions, and medical implications.
Collapse
Affiliation(s)
- Adithi Rao
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California, USA
| | - Shabir A Bhat
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Tomohiro Shibata
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jorge F Giani
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Florian Rader
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Kenneth E Bernstein
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Zakir Khan
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
23
|
Shkreli L, Thoroddsen T, Kobelt M, Martens MA, Browning M, Harmer CJ, Cowen P, Reinecke A. Acute Angiotensin II Receptor Blockade Facilitates Parahippocampal Processing During Memory Encoding in High-Trait-Anxious Individuals. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:100286. [PMID: 38323154 PMCID: PMC10844816 DOI: 10.1016/j.bpsgos.2023.100286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/03/2023] [Accepted: 12/14/2023] [Indexed: 02/08/2024] Open
Abstract
Background Angiotensin II receptor blockers (ARBs) have been associated with preventing posttraumatic stress disorder symptom development and improving memory. However, the underlying neural mechanisms are poorly understood. This study investigated ARB effects on memory encoding and hippocampal functioning that have previously been implicated in posttraumatic stress disorder development. Methods In a double-blind randomized design, 40 high-trait-anxious participants (33 women) received the ARB losartan (50 mg) or placebo. At drug peak level, participants encoded images of animals and landscapes before undergoing functional magnetic resonance imaging, where they viewed the encoded familiar images and unseen novel images to be memorized and classified as animals/landscapes. Memory recognition was assessed 1 hour after functional magnetic resonance imaging. To analyze neural effects, whole-brain analysis, hippocampus region-of-interest analysis, and exploratory multivariate pattern similarity analysis were employed. Results ARBs facilitated parahippocampal processing. In the whole-brain analysis, losartan enhanced brain activity for familiar images in the parahippocampal gyrus (PHC), anterior cingulate cortex, and caudate. For novel images, losartan enhanced brain activity in the PHC only. Pattern similarity analysis showed that losartan increased neural stability in the PHC when processing novel and familiar images. However, there were no drug effects on memory recognition or hippocampal activation. Conclusions Given that the hippocampus receives major input from the PHC, our findings suggest that ARBs may modulate higher-order visual processing through parahippocampal involvement, potentially preserving intact memory input. Future research needs to directly investigate whether this effect may underlie the preventive effects of ARBs in the development of posttraumatic stress disorder.
Collapse
Affiliation(s)
- Lorika Shkreli
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | | | - Malte Kobelt
- Institute of Cognitive Neuroscience, Ruhr-Universität Bochum, Bochum, Germany
| | | | - Michael Browning
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- Oxford Health NHS Foundation Trust, Oxford, United Kingdom
| | - Catherine J. Harmer
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- Oxford Health NHS Foundation Trust, Oxford, United Kingdom
| | - Phil Cowen
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- Oxford Health NHS Foundation Trust, Oxford, United Kingdom
| | - Andrea Reinecke
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- Oxford Health NHS Foundation Trust, Oxford, United Kingdom
| |
Collapse
|
24
|
Barak R, Goshtasbi G, Fatehi R, Firouzabadi N. Signaling pathways and genetics of brain Renin angiotensin system in psychiatric disorders: State of the art. Pharmacol Biochem Behav 2024; 236:173706. [PMID: 38176544 DOI: 10.1016/j.pbb.2023.173706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 12/19/2023] [Accepted: 12/28/2023] [Indexed: 01/06/2024]
Abstract
Along the conventional pathways, Renin-angiotensin system (RAS) plays a key role in the physiology of the CNS and pathogenesis of psychiatric diseases. RAS is a complex regulatory pathway which is composed of several peptides and receptors and comprises two counter-regulatory axes. The classical (ACE1/AngII/AT1 receptor) axis and the contemporary (ACE2/Ang (1-7)/Mas receptor) axis. The genes coding for elements of both axes have been broadly studied. Numerous functional polymorphisms on components of RAS have been identified to serve as informative disease and treatment markers. This review summarizes the role of each peptide and receptor in the pathophysiology of psychiatric disorders (depression, bipolar disorders and schizophrenia), followed by a concise look at the role of genetic polymorphism of the RAS in the pathophysiology of these disorders.
Collapse
Affiliation(s)
- Roya Barak
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ghazal Goshtasbi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reihaneh Fatehi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Firouzabadi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
25
|
Xu T, Chen Z, Zhou X, Wang L, Zhou F, Yao D, Zhou B, Becker B. The central renin-angiotensin system: A genetic pathway, functional decoding, and selective target engagement characterization in humans. Proc Natl Acad Sci U S A 2024; 121:e2306936121. [PMID: 38349873 PMCID: PMC10895353 DOI: 10.1073/pnas.2306936121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 01/02/2024] [Indexed: 02/15/2024] Open
Abstract
Accumulating evidence suggests that the brain renin angiotensin system (RAS) plays a pivotal role in the regulation of cognition and behavior as well as in the neuropathology of neurological and mental disorders. The angiotensin II type 1 receptor (AT1R) mediates most functional and neuropathology-relevant actions associated with the central RAS. However, an overarching comprehension to guide translation and utilize the therapeutic potential of the central RAS in humans is currently lacking. We conducted a comprehensive characterization of the RAS using an innovative combination of transcriptomic gene expression mapping, image-based behavioral decoding, and pre-registered randomized controlled discovery-replication pharmacological resting-state functional magnetic resonance imaging (fMRI) trials (N = 132) with a selective AT1R antagonist. The AT1R exhibited a particular dense expression in a subcortical network encompassing the thalamus, striatum, and amygdalo-hippocampal formation. Behavioral decoding of the AT1R gene expression brain map showed an association with memory, stress, reward, and motivational processes. Transient pharmacological blockade of the AT1R further decreased neural activity in subcortical systems characterized by a high AT1R expression, while increasing functional connectivity in the cortico-basal ganglia-thalamo-cortical circuitry. Effects of AT1R blockade on the network level were specifically associated with the transcriptomic signatures of the dopaminergic, opioid, acetylcholine, and corticotropin-releasing hormone signaling systems. The robustness of the results was supported in an independent pharmacological fMRI trial. These findings present a biologically informed comprehensive characterization of the central AT1R pathways and their functional relevance on the neural and behavioral level in humans.
Collapse
Affiliation(s)
- Ting Xu
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu610054, People’s Republic of China
- Ministry of Education Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology, Chengdu610054, People’s Republic of China
| | - Zhiyi Chen
- Experimental Research Center for Medical and Psychological Science, School of Psychology, Third Military Medical University, Chongqing400037, People’s Republic of China
- Faculty of Psychology, Southwest University, Chongqing400715, People’s Republic of China
- Key Laboratory of Cognition and Personality, Ministry of Education, Faculty of Psychology, Southwest University, Chongqing400715, People’s Republic of China
| | - Xinqi Zhou
- Institute of Brain and Psychological Sciences, Sichuan Normal University, Chengdu 610066, People’s Republic of China
| | - Lan Wang
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu610054, People’s Republic of China
- Ministry of Education Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology, Chengdu610054, People’s Republic of China
| | - Feng Zhou
- Faculty of Psychology, Southwest University, Chongqing400715, People’s Republic of China
- Key Laboratory of Cognition and Personality, Ministry of Education, Faculty of Psychology, Southwest University, Chongqing400715, People’s Republic of China
| | - Dezhong Yao
- Ministry of Education Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology, Chengdu610054, People’s Republic of China
| | - Bo Zhou
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu610054, People’s Republic of China
| | - Benjamin Becker
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu610054, People’s Republic of China
- Ministry of Education Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology, Chengdu610054, People’s Republic of China
- The State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong999077, People’s Republic of China
- Department of Psychology, The University of Hong Kong, Hong Kong999077, People’s Republic of China
| |
Collapse
|
26
|
Alshammari A, Pillai B, Kamat P, Jones TW, Bosomtwi A, Khan MB, Hess DC, Li W, Somanath PR, Sayed MA, Ergul A, Fagan SC. Angiotensin II Type 2 Receptor Agonism Alleviates Progressive Post-stroke Cognitive Impairment in Aged Spontaneously Hypertensive Rats. Transl Stroke Res 2024:10.1007/s12975-024-01232-1. [PMID: 38302738 DOI: 10.1007/s12975-024-01232-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 12/27/2023] [Accepted: 01/17/2024] [Indexed: 02/03/2024]
Abstract
Hypertension and aging are leading risk factors for stroke and vascular contributions to cognitive impairment and dementia (VCID). Most animal models fail to capture the complex interplay between these pathophysiological processes. In the current study, we examined the development of cognitive impairment in 18-month-old spontaneously hypertensive rats (SHR) before and following ischemic stroke. Sixty SHRs were housed for 18 months with cognitive assessments every 6 months and post-surgery. MRI scans were performed at baseline and throughout the study. On day 3 post-stroke, rats were randomized to receive either angiotensin II type 2 receptor (AT2R) agonist Compound 21 (C21) or plain water for 8 weeks. SHRs demonstrated a progressive cognitive decline and significant MRI abnormalities before stroke. Perioperative mortality within 72 h of stroke was low. Stroke resulted in significant acute brain swelling, chronic brain atrophy, and sustained sensorimotor and behavioral deficits. There was no evidence of anhedonia at week 8. C21 enhanced sensorimotor recovery and ischemic lesion resolution at week 8. SHRs represent a clinically relevant animal model to study aging and stroke-associated VCID. This study underscores the importance of translational disease modeling and provides evidence that modulation of the AT2R signaling via C21 may be a useful therapeutic option to improve sensorimotor and cognitive outcomes even in aged animals.
Collapse
Affiliation(s)
- Abdulkarim Alshammari
- Program in Clinical and Experimental Therapeutics, Charlie Norwood Veterans Affairs Health Care System and College of Pharmacy, University of Georgia, Augusta, GA, USA
- Department of Clinical Pharmacy, Faculty of Pharmacy, Northern Border University, Rafha, Saudi Arabia
| | - Bindu Pillai
- Program in Clinical and Experimental Therapeutics, Charlie Norwood Veterans Affairs Health Care System and College of Pharmacy, University of Georgia, Augusta, GA, USA
| | - Pradip Kamat
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Timothy W Jones
- Program in Clinical and Experimental Therapeutics, Charlie Norwood Veterans Affairs Health Care System and College of Pharmacy, University of Georgia, Augusta, GA, USA
| | - Asamoah Bosomtwi
- Georgia Cancer Center and Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | | | - David C Hess
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Weiguo Li
- Ralph H. Johnson Veterans Affairs Health Care System and Department of Pathology & Lab. Medicine, Medical University of South Carolina, 171 Ashley Ave. MSC 908, Charleston, SC, 29492, USA
| | - Payaningal R Somanath
- Program in Clinical and Experimental Therapeutics, Charlie Norwood Veterans Affairs Health Care System and College of Pharmacy, University of Georgia, Augusta, GA, USA
| | | | - Adviye Ergul
- Ralph H. Johnson Veterans Affairs Health Care System and Department of Pathology & Lab. Medicine, Medical University of South Carolina, 171 Ashley Ave. MSC 908, Charleston, SC, 29492, USA.
| | - Susan C Fagan
- Program in Clinical and Experimental Therapeutics, Charlie Norwood Veterans Affairs Health Care System and College of Pharmacy, University of Georgia, Augusta, GA, USA
| |
Collapse
|
27
|
Al-Kuraishy HM, Al-Hamash SM, Jabir MS, Al-Gareeb AI, Albuhadily AK, Albukhaty S, Sulaiman GM. The classical and non-classical axes of renin-angiotensin system in Parkinson disease: The bright and dark side of the moon. Ageing Res Rev 2024; 94:102200. [PMID: 38237699 DOI: 10.1016/j.arr.2024.102200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/30/2023] [Accepted: 01/12/2024] [Indexed: 01/28/2024]
Abstract
Parkinson disease (PD) is a common brain neurodegenerative disease due to progressive degeneration of the dopaminergic neurons in the substantia nigra pars compacta (SNpc). Of note, the cardio-metabolic disorders such as hypertension are adversely affect PD neuropathology through exaggeration of renin-angiotensin system (RAS). The RAS affects the stability of dopaminergic neurons in the SNpc, and exaggeration of angiotensin II (AngII) is implicated in the development and progression of PD. RAS has two axes classical including angiotensin converting enzyme (ACE)/AngII/AT1R, and the non-classical axis which include ACE2/Ang1-7/Mas receptor, AngIII, AngIV, AT2R, and AT4R. It has been shown that brain RAS is differs from that of systemic RAS that produce specific neuronal effects. As well, there is an association between brain RAS and PD. Therefore, this review aims to revise from published articles the role of brain RAS in the pathogenesis of PD focusing on the non-classical pathway, and how targeting of this axis can modulate PD neuropathology.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Sadiq M Al-Hamash
- Department of Pediatric Cardiology, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Majid S Jabir
- Department of Applied science, University of technology, Iraq.
| | - Ali I Al-Gareeb
- Department of Clinical pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali K Albuhadily
- Department of Clinical pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Salim Albukhaty
- Department of Chemistry, College of Science, University of Misan, Maysan 62001, Iraq
| | | |
Collapse
|
28
|
Sotozawa M, Kinguchi S, Wakui H, Azushima K, Funakoshi K, Nakajima W, Miyazaki T, Takahashi T, Tamura K. Enhancement of angiotensin II type 1 receptor-associated protein in the paraventricular nucleus suppresses angiotensin II-dependent hypertension. Hypertens Res 2024; 47:67-77. [PMID: 37884662 DOI: 10.1038/s41440-023-01480-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/29/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023]
Abstract
The renin-angiotensin system in the brain plays a pivotal role in modulating sympathetic nerve activity and contributes to the pathogenesis of hypertension. Angiotensin II (Ang II) type 1 receptor (AT1R)-associated protein (ATRAP) promotes internalization of AT1R while suppressing pathological overactivation of AT1R signaling. However, the pathophysiological function of ATRAP in the brain remains unknown. Therefore, this study aims to investigate whether ATRAP in the paraventricular nucleus (PVN) is involved in neurogenic hypertension pathogenesis in Ang II-infused rats. The ATRAP/AT1R ratio, which serves as an indicator of tissue AT1R hyperactivity, tended to decrease within the PVN in the Ang II group than in the vehicle group. This suggests an Ang II-induced hyperactivation of the AT1R signaling pathway in the PVN. Lentiviral vectors were generated to stimulate ATRAP expression. At 6 weeks of age, rats were microinjected with LV-Venus (Venus-expressing lentivirus) or LV-ATRAP (Venus-ATRAP-expressing lentivirus). The rats were then randomly divided into four groups: (1) Vehicle/LV-Venus, (2) Vehicle/LV-ATRAP, (3) Ang II/LV-Venus, and (4) Ang II/LV-ATRAP. Two weeks after microinjection, vehicle or Ang II was administered systemically for 2 weeks. In the Ang II/LV-ATRAP group, systolic blood pressure at 1 and 2 weeks following administration was significantly lower than that in the Ang II/LV-Venus group. Furthermore, urinary adrenaline levels tended to decrease in the Ang II/LV-ATRAP group than in the Ang II/LV-Venus group. These findings suggest that enhanced ATRAP expression in the PVN suppresses Ang II-induced hypertension, potentially by suppressing hyperactivation of the tissue AT1R signaling pathway and, subsequently, sympathetic nerve activity.
Collapse
Affiliation(s)
- Mari Sotozawa
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Sho Kinguchi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiromichi Wakui
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Kengo Azushima
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Kengo Funakoshi
- Department of Neuroanatomy, Yokohama City University School of Medicine, Yokohama, Japan
| | - Waki Nakajima
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tomoyuki Miyazaki
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takuya Takahashi
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
29
|
Pedrosa MA, Labandeira CM, Lago-Baameiro N, Valenzuela R, Pardo M, Labandeira-Garcia JL, Rodriguez-Perez AI. Extracellular Vesicles and Their Renin-Angiotensin Cargo as a Link between Metabolic Syndrome and Parkinson's Disease. Antioxidants (Basel) 2023; 12:2045. [PMID: 38136165 PMCID: PMC10741149 DOI: 10.3390/antiox12122045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Several studies showed an association between metabolic syndrome (MetS) and Parkinson's disease (PD). The linking mechanisms remain unclear. MetS promotes low-grade peripheral oxidative stress and inflammation and dysregulation of the adipose renin-angiotensin system (RAS). Interestingly, brain RAS dysregulation is involved in the progression of dopaminergic degeneration and PD. Circulating extracellular vesicles (EVs) from MetS fat tissue can cross the brain-blood barrier and may act as linking signals. We isolated and characterized EVs from MetS and control rats and analyzed their mRNA and protein cargo using RT-PCR and the ExoView R200 platform, respectively. Furthermore, cultures of the N27 dopaminergic cell line and the C6 astrocytic cell line were treated with EVs from MetS rats. EVs were highly increased in MetS rat serum, which was inhibited by treatment of the rats with the angiotensin type-1-receptor blocker candesartan. Furthermore, EVs from MetS rats showed increased pro-oxidative/pro-inflammatory and decreased anti-oxidative/anti-inflammatory RAS components, which were inhibited in candesartan-treated MetS rats. In cultures, EVs from MetS rats increased N27 cell death and modulated C6 cell function, upregulating markers of neuroinflammation and oxidative stress, which were inhibited by the pre-treatment of cultures with candesartan. The results from rat models suggest EVs and their RAS cargo as a mechanism linking Mets and PD.
Collapse
Affiliation(s)
- Maria A. Pedrosa
- Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.A.P.); (R.V.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | | | - Nerea Lago-Baameiro
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, 15706 Santiago de Compostela, Spain; (N.L.-B.); (M.P.)
| | - Rita Valenzuela
- Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.A.P.); (R.V.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | - Maria Pardo
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, 15706 Santiago de Compostela, Spain; (N.L.-B.); (M.P.)
- CIBER Fisiopatología Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jose Luis Labandeira-Garcia
- Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.A.P.); (R.V.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | - Ana I. Rodriguez-Perez
- Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.A.P.); (R.V.)
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| |
Collapse
|
30
|
Smith HC, Yu Z, Iyer L, Marvar PJ. Sex-dependent effects of angiotensin type 2 receptor expressing medial prefrontal cortex (mPFC) interneurons in fear extinction learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.21.568156. [PMID: 38045293 PMCID: PMC10690250 DOI: 10.1101/2023.11.21.568156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Background The renin-angiotensin system (RAS) has been identified as a potential therapeutic target for PTSD, though its mechanisms are not well understood. Brain angiotensin type 2 receptors (AT2Rs) are a subtype of angiotensin II receptors located in stress and anxiety-related regions, including the medial prefrontal cortex (mPFC), but their function and mechanism in the mPFC remain unexplored. We therefore used a combination of imaging, cre/lox, and behavioral methods to investigate mPFC-AT2R-expressing neuron involvement in fear learning. Methods To characterize mPFC-AT2R-expressing neurons in the mPFC, AT2R-Cre/td-Tomato male and female mice were used for immunohistochemistry (IHC). mPFC brain sections were stained with glutamatergic or interneuron markers, and density of AT2R+ cells and colocalization with each marker was quantified. To assess fear-related behaviors in AT2R-flox mice, we selectively deleted AT2R from mPFC neurons using an AAV-Cre virus. Mice then underwent Pavlovian auditory fear conditioning, approach/avoidance, and locomotion testing. Results IHC results revealed that AT2R is densely expressed in the mPFC. Furthermore, AT2R is primarily expressed in somatostatin interneurons in females but not males. Following fear conditioning, mPFC-AT2R deletion impaired extinction in female but not male mice. Locomotion was unaltered by mPFC-AT2R deletion in males or females, while AT2R-deleted females had increased exploratory behavior. Conclusion These results lend support for mPFC-AT2R+ neurons as a novel subgroup of somatostatin interneurons that influence fear extinction in a sex-dependent manner. This furthers underscores the role of mPFC in top-down regulation and a unique role for peptidergic (ie., angiotensin) mPFC regulation of fear and sex differences.
Collapse
Affiliation(s)
- Hannah C. Smith
- Department of Neuroscience, George Washington University, Washington, DC
| | - Zhe Yu
- Department of Pharmacology & Physiology, George Washington University, Washington, DC
| | - Laxmi Iyer
- Department of Pharmacology & Physiology, George Washington University, Washington, DC
| | - Paul J. Marvar
- Department of Neuroscience, George Washington University, Washington, DC
- Department of Pharmacology & Physiology, George Washington University, Washington, DC
- Department of Psychiatry and Behavioral Sciences, George Washington University, Washington DC
| |
Collapse
|
31
|
Cappelletti P, Gallo G, Marino R, Palaniappan S, Corbo M, Savoia C, Feligioni M. From cardiovascular system to brain, the potential protective role of Mas Receptors in COVID-19 infection. Eur J Pharmacol 2023; 959:176061. [PMID: 37775018 DOI: 10.1016/j.ejphar.2023.176061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 10/01/2023]
Abstract
Coronavirus disease 2019 (COVID-19) has been declared a new pandemic in March 2020. Although most patients are asymptomatic, those with underlying cardiovascular comorbidities may develop a more severe systemic infection which is often associated with fatal pneumonia. Nonetheless, neurological and cardiovascular manifestations could be present even without respiratory symptoms. To date, no COVID-19-specific drugs are able for preventing or treating the infection and generally, the symptoms are relieved with general anti-inflammatory drugs. Angiotensin-converting-enzyme 2 (ACE2) may function as the receptor for virus entry within the cells favoring the progression of infection in the organism. On the other hand, ACE2 is a relevant enzyme in renin angiotensin system (RAS) cascade fostering Ang1-7/Mas receptor activation which promotes protective effects in neurological and cardiovascular systems. It is known that RAS is composed by two functional countervailing axes the ACE/AngII/AT1 receptor and the ACE/AngII/AT2 receptor which counteracts the actions mediated by AngII/AT1 receptor by inducing anti-inflammatory, antioxidant and anti-growth functions. Subsequently an "alternative" ACE2/Ang1-7/Mas receptor axis has been described with functions similar to the latter protective arm. Here, we discuss the neurological and cardiovascular effects of COVID-19 highlighting the role of the stimulation of the RAS "alternative" protective arm in attenuating pulmonary, cerebral and cardiovascular damages. In conclusion, only two clinical trials are running for Mas receptor agonists but few other molecules are in preclinical phase and if successful these drugs might represent a successful strategy for the treatment of the acute phase of COVID-19 infection.
Collapse
Affiliation(s)
- Pamela Cappelletti
- Department of Neuro-Rehabilitation Sciences, Casa di Cura Igea, Milan, Italy.
| | - Giovanna Gallo
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Rachele Marino
- European Brain Research Institute (EBRI) Rita Levi Montalcini Foundation, Rome, Italy
| | | | - Massimo Corbo
- Department of Neuro-Rehabilitation Sciences, Casa di Cura Igea, Milan, Italy
| | - Carmine Savoia
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Marco Feligioni
- Department of Neuro-Rehabilitation Sciences, Casa di Cura Igea, Milan, Italy; European Brain Research Institute (EBRI) Rita Levi Montalcini Foundation, Rome, Italy.
| |
Collapse
|
32
|
Fleisher-Berkovich S, Battaglia V, Baratta F, Brusa P, Ventura Y, Sharon N, Dahan A, Collino M, Ben-Shabat S. An Emerging Strategy for Neuroinflammation Treatment: Combined Cannabidiol and Angiotensin Receptor Blockers Treatments Effectively Inhibit Glial Nitric Oxide Release. Int J Mol Sci 2023; 24:16254. [PMID: 38003444 PMCID: PMC10671332 DOI: 10.3390/ijms242216254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/06/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Cannabidiol (CBD), the major non-psychoactive phytocannabinoid found in cannabis, has anti-neuroinflammatory properties. Despite the increasing use of CBD, little is known about its effect in combination with other substances. Combination therapy has been gaining attention recently, aiming to produce more efficient effects. Angiotensin II activates the angiotensin 1 receptor and regulates neuroinflammation and cognition. Angiotensin receptor 1 blockers (ARBs) were shown to be neuroprotective and prevent cognitive decline. The present study aimed to elucidate the combined role of CBD and ARBs in the modulation of lipopolysaccharide (LPS)-induced glial inflammation. While LPS significantly enhanced nitric oxide synthesis vs. the control, telmisartan and CBD, when administered alone, attenuated this effect by 60% and 36%, respectively. Exposure of LPS-stimulated cells to both compounds resulted in the 95% inhibition of glial nitric oxide release (additive effect). A synergistic inhibitory effect on nitric oxide release was observed when cells were co-treated with losartan (5 μM) and CBD (5 μM) (by 80%) compared to exposure to each compound alone (by 22% and 26%, respectively). Telmisartan and CBD given alone increased TNFα levels by 60% and 40%, respectively. CBD and telmisartan, when given together, attenuated the LPS-induced increase in TNFα levels without statistical significance. LPS-induced IL-17 release was attenuated by CBD with or without telmisartan (by 75%) or telmisartan alone (by 60%). LPS-induced Interferon-γ release was attenuated by 80% when telmisartan was administered in the absence or presence of CBD. Anti-inflammatory effects were recorded when CBD was combined with the known anti-inflammatory agent dimethyl fumarate (DMF)/monomethyl fumarate (MMF). A synergistic inhibitory effect of CBD and MMF on glial release of nitric oxide (by 77%) was observed compared to cells exposed to MMF (by 35%) or CBD (by 12%) alone. Overall, this study highlights the potential of new combinations of CBD (5 μM) with losartan (5 μM) or MMF (1 μM) to synergistically attenuate glial NO synthesis. Additive effects on NO production were observed when telmisartan (5 μM) and CBD (5 μM) were administered together to glial cells.
Collapse
Affiliation(s)
- Sigal Fleisher-Berkovich
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel; (Y.V.); (N.S.); (A.D.)
| | - Veronica Battaglia
- Department of Drug Science and Technology, University of Torino, 10124 Torino, Italy; (V.B.); (F.B.); (P.B.)
| | - Francesca Baratta
- Department of Drug Science and Technology, University of Torino, 10124 Torino, Italy; (V.B.); (F.B.); (P.B.)
| | - Paola Brusa
- Department of Drug Science and Technology, University of Torino, 10124 Torino, Italy; (V.B.); (F.B.); (P.B.)
| | - Yvonne Ventura
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel; (Y.V.); (N.S.); (A.D.)
| | - Nitzan Sharon
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel; (Y.V.); (N.S.); (A.D.)
| | - Arik Dahan
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel; (Y.V.); (N.S.); (A.D.)
| | - Massimo Collino
- Department of Neuroscience “Rita Levi Montalcini”, University of Torino, 10124 Torino, Italy;
| | - Shimon Ben-Shabat
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel; (Y.V.); (N.S.); (A.D.)
| |
Collapse
|
33
|
Prieto I, Segarra AB, Banegas I, Martínez-Cañamero M, Durán R, Vives F, Domínguez-Vías G, Ramírez-Sánchez M. Correlational Study of Aminopeptidase Activities between Left or Right Frontal Cortex versus the Hypothalamus, Pituitary, Adrenal Axis of Spontaneously Hypertensive Rats Treated with Hypotensive or Hypertensive Agents. Int J Mol Sci 2023; 24:16007. [PMID: 37958990 PMCID: PMC10647763 DOI: 10.3390/ijms242116007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/27/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023] Open
Abstract
It has been suggested that the neuro-visceral integration works asymmetrically and that this asymmetry is dynamic and modifiable by physio-pathological influences. Aminopeptidases of the renin-angiotensin system (angiotensinases) have been shown to be modifiable under such conditions. This article analyzes the interactions of these angiotensinases between the left or right frontal cortex (FC) and the same enzymes in the hypothalamus (HT), pituitary (PT), adrenal (AD) axis (HPA) in control spontaneously hypertensive rats (SHR), in SHR treated with a hypotensive agent in the form of captopril (an angiotensin-converting enzyme inhibitor), and in SHR treated with a hypertensive agent in the form of the L-Arginine hypertensive analogue L-NG-Nitroarginine Methyl Ester (L-NAME). In the control SHR, there were significant negative correlations between the right FC with HPA and positive correlations between the left FC and HPA. In the captopril group, the predominance of negative correlations between the right FC and HPA and positive correlations between the HPA and left FC was maintained. In the L-NAME group, a radical change in all types of interactions was observed; particularly, there was an inversion in the predominance of negative correlations between the HPA and left FC. These results indicated a better balance of neuro-visceral interactions after captopril treatment and an increase in these interactions in the hypertensive animals, especially in those treated with L-NAME.
Collapse
Affiliation(s)
- Isabel Prieto
- Department of Health Sciences, University of Jaén, 23071 Jaén, Spain; (I.P.); (A.B.S.); (I.B.); (M.M.-C.)
| | - Ana Belén Segarra
- Department of Health Sciences, University of Jaén, 23071 Jaén, Spain; (I.P.); (A.B.S.); (I.B.); (M.M.-C.)
| | - Inmaculada Banegas
- Department of Health Sciences, University of Jaén, 23071 Jaén, Spain; (I.P.); (A.B.S.); (I.B.); (M.M.-C.)
| | | | - Raquel Durán
- Department of Physiology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; (R.D.); (F.V.)
| | - Francisco Vives
- Department of Physiology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; (R.D.); (F.V.)
| | - Germán Domínguez-Vías
- Department of Physiology, Faculty of Health Sciences, Ceuta Campus, University of Granada, 18071 Granada, Spain;
| | - Manuel Ramírez-Sánchez
- Department of Health Sciences, University of Jaén, 23071 Jaén, Spain; (I.P.); (A.B.S.); (I.B.); (M.M.-C.)
| |
Collapse
|
34
|
Shehata NI, Abd EL-Salam DM, Hussein RM, Rizk SM. Effect of safranal or candesartan on 3-nitropropionicacid-induced biochemical, behavioral and histological alterations in a rat model of Huntington's disease. PLoS One 2023; 18:e0293660. [PMID: 37910529 PMCID: PMC10619823 DOI: 10.1371/journal.pone.0293660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 10/17/2023] [Indexed: 11/03/2023] Open
Abstract
3-nitropropionic acid (3-NP) is a potent mitochondrial inhibitor mycotoxin. Systemic administration of 3-NP can induce Huntington's disease (HD)-like symptoms in experimental animals. Safranal (Safr) that is found in saffron essential oil has antioxidant, anti-inflammatory and anti-apoptotic actions. Candesartan (Cands) is an angiotensin receptor blocker that has the potential to prevent cognitive deficits. The present study aims to investigate the potential neuroprotective efficacy of Safr or Cands in 3-NP-induced rat model of HD. The experiments continued for nine consecutive days. Rats were randomly assigned into seven groups. The first group (Safr-control) was daily intraperitoneally injected with paraffin oil. The second group (Cands- and 3-NP-control) daily received an oral dose of 0.5% carboxymethylcellulose followed by an intraperitoneal injection of 0.9% saline. The third and fourth groups received a single daily dose of 50 mg/kg Safr (intraperitoneal) and 1 mg/kg Cands (oral), respectively. The sixth group was daily treated with 50 mg Safr kg/day (intraperitoneal) and was intraperitoneally injected with 20 mg 3-NP/ kg, from the 3rd till the 9th day. The seventh group was daily treated with 1 mg Cands /kg/day (oral) and was intraperitoneally injected with 20 mg 3-NP/ kg, from the 3rd till the 9th day. The present results revealed that 3-NP injection induced a considerable body weight loss, impaired memory and locomotor activity, reduced striatal monoamine levels. Furthermore, 3-NP administration remarkably increased striatal malondialdehyde and nitric oxide levels, whereas markedly decreased the total antioxidant capacity. Moreover, 3-NP significantly upregulated the activities of inducible nitric oxide synthase and caspase-3 as well as the Fas ligand, in striatum. On the contrary, Safr and Cands remarkably alleviated the above-mentioned 3-NP-induced alterations. In conclusion, Safr and Cands may prevent or delay the progression of HD and its associated impairments through their antioxidant, anti-inflammatory, anti-apoptotic and neuromodulator effects.
Collapse
Affiliation(s)
| | | | | | - Sherine Maher Rizk
- Faculty of Pharmacy, Biochemistry Department, Cairo University, Cairo, Egypt
| |
Collapse
|
35
|
Zambrano AK, Cadena-Ullauri S, Guevara-Ramírez P, Ruiz-Pozo VA, Tamayo-Trujillo R, Paz-Cruz E, Ibarra-Rodríguez AA, Doménech N. Genetic diet interactions of ACE: the increased hypertension predisposition in the Latin American population. Front Nutr 2023; 10:1241017. [PMID: 37964928 PMCID: PMC10640988 DOI: 10.3389/fnut.2023.1241017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023] Open
Abstract
Hypertension is one of the primary risk factors associated with cardiovascular diseases (CVDs). It is a condition that affects people worldwide, and its prevalence is increasing due to several factors, such as lack of physical activity, population aging, and unhealthy diets. Notably, this increase has primarily occurred in low and middle-income countries (LMICs). In Latin America, approximately 40% of adults have been diagnosed with hypertension. Moreover, reports have shown that the Latin American genetic composition is highly diverse, and this genetic background can influence various biological processes, including disease predisposition and treatment effectiveness. Research has shown that Western dietary patterns, which include increased consumption of red meat, refined grains, sugar, and ultra-processed food, have spread across the globe, including Latin America, due to globalization processes. Furthermore, a higher than recommended sodium consumption, which has been associated with hypertension, has been identified across different regions, including Asia, Europe, America, Oceania, and Africa. In conclusion, hypertension is a multifactorial disease involving environmental and genetic factors. In Latin America, hypertension prevalence is increasing due to various factors, including age, the adoption of a "Westernized" diet, and potential genetic predisposition factors involving the ACE gene. Furthermore, identifying the genetic and molecular mechanisms of the disease, its association with diet, and how they interact is essential for the development of personalized treatments to increase its efficacy and reduce side effects.
Collapse
Affiliation(s)
- Ana Karina Zambrano
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Santiago Cadena-Ullauri
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Patricia Guevara-Ramírez
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Viviana A. Ruiz-Pozo
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Rafael Tamayo-Trujillo
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Elius Paz-Cruz
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | | | - Nieves Doménech
- Instituto de Investigación Biomédica de A Coruña (INIBIC)-CIBERCV, Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidad da Coruña (UDC), La Coruña, Spain
| |
Collapse
|
36
|
Franco R, Serrano-Marín J, Navarro G, Rivas-Santisteban R. The NADPH Link between the Renin Angiotensin System and the Antioxidant Mechanisms in Dopaminergic Neurons. Antioxidants (Basel) 2023; 12:1869. [PMID: 37891948 PMCID: PMC10604245 DOI: 10.3390/antiox12101869] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
The renin angiotensin system (RAS) has several components including signaling peptides, enzymes, and membrane receptors. The effort in characterizing this system in the periphery has led to the approval of a class of antihypertensives. Much less is known about RAS in the central nervous system. The production of RAS peptides and the expression of several RAS enzymes and receptors in dopaminergic neurons of the substantia nigra has raised expectations in the therapy of Parkinson's disease, a neurodegenerative condition characterized by lack of dopamine in the striatum, the motor control region of the mammalian brain. On the one hand, dopamine production requires reducing power. On the other hand, reducing power is required by mechanisms involved in REDOX homeostasis. This review focuses on the potential role of RAS in the regulation of neuronal/glial expression of glucose-6-phosphate dehydrogenase, which produces the NADPH required for dopamine synthesis and for reactive oxygen species (ROS) detoxification. It is known that transgenic expression of the gene coding for glucose-6-phosphate dehydrogenase prevents the death of dopaminergic nigral neurons. Signaling via angiotensin II G protein-coupled receptors, AT1 or AT2, leads to the activation of protein kinase A and/or protein kinase C that in turn can regulate glucose-6- phosphate dehydrogenase activity, by Ser/Thr phosphorylation/dephosphorylation events. Long-term effects of AT1 or AT2 receptor activation may also impact on the concentration of the enzyme via activation of transcription factors that participate in the regulation of gene expression in neurons (or glia). Future research is needed to determine how the system can be pharmacologically manipulated to increase the availability of NADPH to neurons degenerating in Parkinson's disease and to neuroprotective glia.
Collapse
Affiliation(s)
- Rafael Franco
- Department of Biochemistry and Molecular Biomedicine, School of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
- CiberNed, Network Center for Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28029 Madrid, Spain;
- School of Chemistry, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Joan Serrano-Marín
- Department of Biochemistry and Molecular Biomedicine, School of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Gemma Navarro
- CiberNed, Network Center for Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28029 Madrid, Spain;
- Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, 08028 Barcelona, Spain
- Institute of Neurosciences, Universitat de Barcelona, 08007 Barcelona, Spain
| | - Rafael Rivas-Santisteban
- CiberNed, Network Center for Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28029 Madrid, Spain;
- Campus Bellaterra, Autonomous University of Barcelona, Cerdanyola del Vallés, 08193 Barcelona, Spain
| |
Collapse
|
37
|
Park MS, Kim EJ. A Correlative Relationship Between Heart Failure and Cognitive Impairment: A Narrative Review. J Korean Med Sci 2023; 38:e334. [PMID: 37821090 PMCID: PMC10562184 DOI: 10.3346/jkms.2023.38.e334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/11/2023] [Indexed: 10/13/2023] Open
Abstract
Heart failure (HF) is a chronic condition affecting millions of people worldwide. While the cardinal manifestations of HF are related to the cardiovascular system, it has become progressively evident that mild cognitive impairment (MCI) is also a significant complication of the disease. In fact, a significant number of patients with HF may experience MCI, which can manifest as deficits in attention, memory, executive function, and processing speed. The mechanisms responsible for cognitive dysfunction in HF are intricate and multifactorial. Possible factors contributing to this condition include decreased cerebral blood flow, thrombogenicity associated with HF, systemic inflammatory conditions, and proteotoxicity. MCI in HF has significant clinical implications, as it is linked to poorer quality of life, increased morbidity and mortality, and higher healthcare costs. Additionally, MCI can disrupt self-care behaviors, adherence to medication, and decision-making abilities, all of which are crucial for effectively managing HF. However, there is currently no gold standard diagnostic tool and follow-up strategy for MCI in HF patients. There is limited knowledge on the prevention and treatment of MCI. In conclusion, MCI is a common and clinically important complication of HF. Considering the substantial influence of MCI on patient outcomes, it is imperative for healthcare providers to be cognizant of this issue and integrate cognitive screening and management strategies into the care of HF patients.
Collapse
Affiliation(s)
- Myung Soo Park
- Department of Medicine, Korea University Graduate School, Seoul, Korea
- Division of Cardiology, Department of Internal Medicine, Dongtan Sacred Heart Hospital, Hwaseong, Korea
| | - Eung Ju Kim
- Division of Cardiology, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea.
| |
Collapse
|
38
|
Reveret L, Leclerc M, Emond V, Tremblay C, Loiselle A, Bourassa P, Bennett DA, Hébert SS, Calon F. Higher angiotensin-converting enzyme 2 (ACE2) levels in the brain of individuals with Alzheimer's disease. Acta Neuropathol Commun 2023; 11:159. [PMID: 37784209 PMCID: PMC10544218 DOI: 10.1186/s40478-023-01647-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 10/04/2023] Open
Abstract
Cognitive decline due to Alzheimer's disease (AD) is frequent in the geriatric population, which has been disproportionately affected by the COVID-19 pandemic. In this study, we investigated the levels of angiotensin-converting enzyme 2 (ACE2), a regulator of the renin-angiotensin system and the main entry receptor of SARS-CoV-2 in host cells, in postmortem parietal cortex samples from two independent AD cohorts, totalling 142 persons. Higher concentrations of ACE2 protein (p < 0.01) and mRNA (p < 0.01) were found in individuals with a neuropathological diagnosis of AD compared to age-matched healthy control subjects. Brain levels of soluble ACE2 were inversely associated with cognitive scores (p = 0.02) and markers of pericytes (PDGFRβ, p = 0.02 and ANPEP, p = 0.007), but positively correlated with concentrations of soluble amyloid-β peptides (Aβ) (p = 0.01) and insoluble phospho-tau (S396/404, p = 0.002). However, no significant differences in ACE2 were observed in the 3xTg-AD mouse model of tau and Aβ neuropathology. Results from immunofluorescence and Western blots showed that ACE2 protein is predominantly localized in microvessels in the mouse brain whereas it is more frequently found in neurons in the human brain. The present data suggest that higher levels of soluble ACE2 in the human brain may contribute to AD, but their role in CNS infection by SARS-CoV-2 remains unclear.
Collapse
Affiliation(s)
- Louise Reveret
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada
| | - Manon Leclerc
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada
| | - Vincent Emond
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada
| | - Cyntia Tremblay
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada
| | - Andréanne Loiselle
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada
| | - Philippe Bourassa
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Sébastien S Hébert
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada
- Faculty of Medicine, Laval University, Quebec, QC, Canada
| | - Frédéric Calon
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada.
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada.
| |
Collapse
|
39
|
Ortiz-Nazario E, Denton-Ortiz CM, Soto-Escobar LDM, Mateo-Mayol Z, Colon-Romero M, Hernandez-Lopez A, Porter JT. Sex-dependent effects of angiotensin II type 1 receptor blocker on molecular and behavioral changes induced by single prolonged stress. Behav Brain Res 2023; 454:114639. [PMID: 37652238 PMCID: PMC10530531 DOI: 10.1016/j.bbr.2023.114639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/02/2023]
Abstract
Post-traumatic stress disorder (PTSD) is a neuropsychiatric disorder that not only entails alterations in fear behavior and anxiety but also includes neuroendocrine dysfunctions involving the hypothalamic pituitary adrenal (HPA) axis and the renin-angiotensin system. Recent preclinical studies demonstrate that activation of the angiotensin type 1 receptor (AT1R) in the paraventricular region of the hypothalamus (PVR) promotes anxiety-like behaviors and enables microglia proliferation. An increase in microglia and anxiety-like behavior also occurs in the PTSD animal model single-prolonged stress (SPS). In the present study, we tested whether AT1Rs contribute to the effects of SPS on behavior and microglia in brain structures important for HPA axis regulation and fear behavior. To test this, male and female animals were exposed to SPS and then given the oral AT1R antagonist candesartan beginning one week later. Candesartan did not alter auditory fear conditioning or extinction in SPS-exposed male or female animals. However, we found that the male animals exposed to SPS showed increased anxiety-like behavior, which was reversed by candesartan. In contrast, neither SPS nor candesartan altered anxiety-like behavior in the female animals. At the molecular level, SPS increased the cellular expression of AT1Rs in the PVR of male animals and candesartan reversed this effect, whereas AT1Rs in the PVR of females were unaltered by either SPS or candesartan. Iba1-expressing microglia increased in the PVR after SPS exposure and was reversed by candesartan in both sexes suggesting that SPS stimulates AT1Rs to increase microglia in the PVR. Collectively, these results suggest that the contribution of AT1Rs to the molecular and behavioral effects of SPS is sex-dependent.
Collapse
Affiliation(s)
- Emily Ortiz-Nazario
- Dept of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Puerto Rico, Pontifical Catholic University of Puerto Rico, Ponce 00732, Puerto Rico
| | - Carla M Denton-Ortiz
- Dept of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Puerto Rico, Pontifical Catholic University of Puerto Rico, Ponce 00732, Puerto Rico
| | - Lawry D M Soto-Escobar
- Dept of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Puerto Rico, Pontifical Catholic University of Puerto Rico, Ponce 00732, Puerto Rico
| | - Zaira Mateo-Mayol
- Dept of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Puerto Rico, Pontifical Catholic University of Puerto Rico, Ponce 00732, Puerto Rico
| | - Maria Colon-Romero
- Dept of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Puerto Rico, Pontifical Catholic University of Puerto Rico, Ponce 00732, Puerto Rico
| | - Anixa Hernandez-Lopez
- Dept of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Puerto Rico, Pontifical Catholic University of Puerto Rico, Ponce 00732, Puerto Rico
| | - James T Porter
- Dept of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Puerto Rico, Pontifical Catholic University of Puerto Rico, Ponce 00732, Puerto Rico.
| |
Collapse
|
40
|
Villapol S, Janatpour ZC, Affram KO, Symes AJ. The Renin Angiotensin System as a Therapeutic Target in Traumatic Brain Injury. Neurotherapeutics 2023; 20:1565-1591. [PMID: 37759139 PMCID: PMC10684482 DOI: 10.1007/s13311-023-01435-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Traumatic brain injury (TBI) is a major public health problem, with limited pharmacological options available beyond symptomatic relief. The renin angiotensin system (RAS) is primarily known as a systemic endocrine regulatory system, with major roles controlling blood pressure and fluid homeostasis. Drugs that target the RAS are used to treat hypertension, heart failure and kidney disorders. They have now been used chronically by millions of people and have a favorable safety profile. In addition to the systemic RAS, it is now appreciated that many different organ systems, including the brain, have their own local RAS. The major ligand of the classic RAS, Angiotensin II (Ang II) acts predominantly through the Ang II Type 1 receptor (AT1R), leading to vasoconstriction, inflammation, and heightened oxidative stress. These processes can exacerbate brain injuries. Ang II receptor blockers (ARBs) are AT1R antagonists. They have been shown in several preclinical studies to enhance recovery from TBI in rodents through improvements in molecular, cellular and behavioral correlates of injury. ARBs are now under consideration for clinical trials in TBI. Several different RAS peptides that signal through receptors distinct from the AT1R, are also potential therapeutic targets for TBI. The counter regulatory RAS pathway has actions that oppose those stimulated by AT1R signaling. This alternative pathway has many beneficial effects on cells in the central nervous system, bringing about vasodilation, and having anti-inflammatory and anti-oxidative stress actions. Stimulation of this pathway also has potential therapeutic value for the treatment of TBI. This comprehensive review will provide an overview of the various components of the RAS, with a focus on their direct relevance to TBI pathology. It will explore different therapeutic agents that modulate this system and assess their potential efficacy in treating TBI patients.
Collapse
Affiliation(s)
- Sonia Villapol
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, USA
| | - Zachary C Janatpour
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Kwame O Affram
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Aviva J Symes
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
41
|
Quan W, Xu CS, Li XC, Yang C, Lan T, Wang MY, Yu DH, Tang F, Wang ZF, Li ZQ. Telmisartan inhibits microglia-induced neurotoxic A1 astrocyte conversion via PPARγ-mediated NF-κB/p65 degradation. Int Immunopharmacol 2023; 123:110761. [PMID: 37544025 DOI: 10.1016/j.intimp.2023.110761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/25/2023] [Accepted: 08/01/2023] [Indexed: 08/08/2023]
Abstract
Astrocytes are crucially involved in neuroinflammation. Activated astrocytes exhibit at least two phenotypes, A1 (neurotoxic) and A2 (neuroprotective). The A1 phenotype is the major reactive astrocyte phenotype involved in aging and neurodegenerative diseases. Telmisartan, which is an antihypertensive agent, is a promising neuroprotective agent. This study aimed to investigate the effects of telmisartan on the phenotype of reactive astrocytes. Astrocytes were activated by culturing with the conditioned medium derived from lipopolysaccharide-stimulated microglia. This conditioned medium induced early, transient A2 astrocyte conversion (within 24 h) and late, sustained A1 conversion (beginning at 24 h and lasting up to 7 days), with a concomitant increase in the production of pro-inflammatory cytokines (interleukin [IL]-1β, tumor necrosis factor [TNF]α, and IL-6) and phosphorylation of nuclear factor-κB (NF-κB)/p65. Telmisartan treatment promoted and inhibited A2 and A1 conversion, respectively. Telmisartan reduced total and phosphorylated p65 protein levels. Losartan, a specific angiotensin II type-1 receptor (AT1R) blocker, did not influence the reactive state of astrocytes. Additionally, AT1R activation by angiotensin II did not induce the expression of pro-inflammatory cytokines and A1/A2 markers, indicating that the AT1R signaling pathway is not involved in the astrocyte-mediated inflammatory response. A peroxisome proliferator-activated receptor γ (PPARγ) antagonist reversed the effects of telmisartan. Moreover, telmisartan-induced p65 downregulation was reversed by the proteasome inhibitor MG132. These results indicate that telmisartan suppresses activated microglia-induced neurotoxic A1 astrocyte conversion through p65 degradation. Our findings contribute towards the elucidation of the anti-inflammatory activity of telmisartan in brain disorders.
Collapse
Affiliation(s)
- Wei Quan
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Cheng-Shi Xu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiao-Chong Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Chao Yang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Tian Lan
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Meng-Yue Wang
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei, China
| | - Dong-Hu Yu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Feng Tang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ze-Fen Wang
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan, Hubei, China.
| | - Zhi-Qiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China; Brain Glioma Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
42
|
Cosarderelioglu C, Kreimer S, Plaza‐Rodriguez AI, Iglesias PA, Talbot CC, Siragy HM, Carey RM, Ubaida‐Mohien C, O'Rourke B, Ferrucci L, Bennett DA, Walston J, Abadir P. Decoding Angiotensin Receptors: TOMAHAQ-Based Detection and Quantification of Angiotensin Type-1 and Type-2 Receptors. J Am Heart Assoc 2023; 12:e030791. [PMID: 37681524 PMCID: PMC10547273 DOI: 10.1161/jaha.123.030791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/20/2023] [Indexed: 09/09/2023]
Abstract
Background The renin-angiotensin system plays a crucial role in human physiology, and its main hormone, angiotensin, activates 2 G-protein-coupled receptors, the angiotensin type-1 and type-2 receptors, in almost every organ. However, controversy exists about the location, distribution, and expression levels of these receptors. Concerns have been raised over the low sensitivity, low specificity, and large variability between lots of commercially available antibodies for angiotensin type-1 and type-2 receptors, which makes it difficult to reconciliate results of different studies. Here, we describe the first non-antibody-based sensitive and specific targeted quantitative mass spectrometry assay for angiotensin receptors. Methods and Results Using a technique that allows targeted analysis of multiple peptides across multiple samples in a single mass spectrometry analysis, known as TOMAHAQ (triggered by offset, multiplexed, accurate mass, high resolution, and absolute quantification), we have identified and validated specific human tryptic peptides that permit identification and quantification of angiotensin type-1 and type-2 receptors in biological samples. Several peptide sequences are conserved in rodents, making these mass spectrometry assays amenable to both preclinical and clinical studies. We have used this method to quantify angiotensin type-1 and type-2 receptors in postmortem frontal cortex samples of older adults (n=28) with Alzheimer dementia. We correlated levels of angiotensin receptors to biomarkers classically linked to renin-angiotensin system activation, including oxidative stress, inflammation, amyloid-β load, and paired helical filament-tau tangle burden. Conclusions These robust high-throughput assays will not only catalyze novel mechanistic studies in the angiotensin research field but may also help to identify patients with an unbalanced angiotensin receptor distribution who would benefit from angiotensin receptor blocker treatment.
Collapse
Affiliation(s)
- Caglar Cosarderelioglu
- Division of Geriatric Medicine and GerontologyJohns Hopkins University School of MedicineBaltimoreMD
- Division of Geriatrics, Department of Internal MedicineAnkara University School of MedicineAnkaraTurkey
| | - Simion Kreimer
- The Mass Spectrometry and Proteomics FacilityJohns Hopkins University School of MedicineBaltimoreMD
| | | | - Pablo A. Iglesias
- Department of Electrical and Computer Engineering, Whiting School of EngineeringJohns Hopkins UniversityBaltimoreMD
| | - C. Conover Talbot
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of MedicineBaltimoreMD
| | - Helmy M. Siragy
- Division of Endocrinology and Metabolism, Department of MedicineUniversity of VirginiaCharlottesvilleVA
| | - Robert M. Carey
- Division of Endocrinology and Metabolism, Department of MedicineUniversity of VirginiaCharlottesvilleVA
| | | | - Brian O'Rourke
- Division of Cardiology, Department of MedicineJohns Hopkins UniversityBaltimoreMD
| | - Luigi Ferrucci
- National Institute on Aging, National Institutes of HealthBaltimoreMD
| | - David A. Bennett
- Rush Alzheimer’s Disease CenterRush University Medical CenterChicagoIL
| | - Jeremy Walston
- Division of Geriatric Medicine and GerontologyJohns Hopkins University School of MedicineBaltimoreMD
| | - Peter Abadir
- Division of Geriatric Medicine and GerontologyJohns Hopkins University School of MedicineBaltimoreMD
| |
Collapse
|
43
|
Ababei DC, Bild V, Macadan I, Vasincu A, Rusu RN, Blaj M, Stanciu GD, Lefter RM, Bild W. Therapeutic Implications of Renin-Angiotensin System Modulators in Alzheimer's Dementia. Pharmaceutics 2023; 15:2290. [PMID: 37765259 PMCID: PMC10538010 DOI: 10.3390/pharmaceutics15092290] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
The Renin-Angiotensin System (RAS) has attracted considerable interest beyond its traditional cardiovascular role due to emerging data indicating its potential involvement in neurodegenerative diseases, including Alzheimer's dementia (AD). This review investigates the therapeutic implications of RAS modulators, specifically focusing on angiotensin-converting enzyme inhibitors (ACEIs), angiotensin receptor blockers (ARBs), and renin inhibitors in AD. ACEIs, commonly used for hypertension, show promise in AD by reducing angiotensin (Ang) II levels. This reduction is significant as Ang II contributes to neuroinflammation, oxidative stress, and β-amyloid (Aβ) accumulation, all implicated in AD pathogenesis. ARBs, known for vasodilation, exhibit neuroprotection by blocking Ang II receptors, improving cerebral blood flow and cognitive decline in AD models. Renin inhibitors offer a novel approach by targeting the initial RAS step, displaying anti-inflammatory and antioxidant effects that mitigate AD degeneration. Preclinical studies demonstrate RAS regulation's favorable impact on neuroinflammation, neuronal damage, cognitive function, and Aβ metabolism. Clinical trials on RAS modulators in AD are limited, but with promising results, ARBs being more effective that ACEIs in reducing cognitive decline. The varied roles of ACEIs, ARBs, and renin inhibitors in RAS modulation present a promising avenue for AD therapeutic intervention, requiring further research to potentially transform AD treatment strategies.
Collapse
Affiliation(s)
- Daniela-Carmen Ababei
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (D.-C.A.); (A.V.); (R.-N.R.)
| | - Veronica Bild
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (D.-C.A.); (A.V.); (R.-N.R.)
- Center of Biomedical Research, Romanian Academy, Iasi Branch, 8 Carol I Avenue, 700506 Iasi, Romania; (R.-M.L.); (W.B.)
| | - Ioana Macadan
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (D.-C.A.); (A.V.); (R.-N.R.)
| | - Alexandru Vasincu
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (D.-C.A.); (A.V.); (R.-N.R.)
| | - Răzvan-Nicolae Rusu
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (D.-C.A.); (A.V.); (R.-N.R.)
| | - Mihaela Blaj
- Department of Anaesthesiology and Intensive Therapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania;
| | - Gabriela Dumitrița Stanciu
- Center for Advanced Research and Development in Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania;
| | - Radu-Marian Lefter
- Center of Biomedical Research, Romanian Academy, Iasi Branch, 8 Carol I Avenue, 700506 Iasi, Romania; (R.-M.L.); (W.B.)
| | - Walther Bild
- Center of Biomedical Research, Romanian Academy, Iasi Branch, 8 Carol I Avenue, 700506 Iasi, Romania; (R.-M.L.); (W.B.)
- Department of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
| |
Collapse
|
44
|
Celiker FB, Kanat A, Turan A, Beyazal M, Burakgazi G, Hursoy N, Gundogdu H, Polat HB. Could the Cerebral Involvement of COVID-19 Disease be Related to Microstructural Changes that are not Reflected in Conventional MRI Images? Neurol India 2023; 71:699-704. [PMID: 37635501 DOI: 10.4103/0028-3886.383878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
Abstract
In patients with COVID-19, neurodegeneration may develop before clinical symptoms appear. Diffusion-weighted (DW) MRI is an important technique for analyzing microstructural changes such as gliosis. In this study, a quantitative evaluation of microstructural changes in the brain with apparent diffusion coefficient (ADC) values in patients presenting with a headache after the COVID-19 disease was analyzed and compared. DW MR images of patients of 20 COVID-19 patients (13 females, 7 males) who required imaging due to headache; 20 controls (16 females, 4 males) were retrospectively reevaluated. ADC measurements were taken from 16 regions of the brain, including right and left symmetrical in patients with COVID-19 infections and control groups. All regions of interest (ROIs) were taken from the hypothalamus, parahippocampus, thalamus, corpus striatum, cingulate gyrus, occipital gyrus, dentate nucleus, and medulla oblongata posterior. ADC values in the dentate nucleus right (784.6 ± 75.7 vs. 717.25 ± 50.75), dentate nucleus left (768.05 ± 69.76 vs. 711.40 ± 52.99), right thalamus (731.15 ± 38.14 vs. 701.60 ± 43.65), left thalamus (744.05 ± 39.00 vs. 702.85 ± 28.88), right parahippocampus (789.10 ± 56.35 vs. 754.75 ± 33.78), right corpus striatum (710.00 ± 39.81 vs. 681.55 ± 39.84) were significantly higher than those in the control group. No significant changes were observed in other areas. A significant increase in ADC values at many levels in the brain in patients with COVID-19 disease and headache was observed. Thus, this study indicates that cerebral involvement in COVID-19 disease may be related to microstructural changes that are not reflected in conventional MRI images.
Collapse
Affiliation(s)
- Fatma B Celiker
- Department of Radiology, Medical Faculty, Recep Tayyip Erdogan University, Rize, Turkey
| | - Ayhan Kanat
- Department of Neurosurgery, Medical Faculty, Recep Tayyip Erdogan University, Rize, Turkey
| | - Arzu Turan
- Acibadem Hospital, The Department of Radiology, Bodrum, Turkey
| | - Mehmet Beyazal
- Department of Radiology, Medical Faculty, Recep Tayyip Erdogan University, Rize, Turkey
| | - Gulen Burakgazi
- Department of Radiology, Medical Faculty, Recep Tayyip Erdogan University, Rize, Turkey
| | - Nur Hursoy
- Department of Radiology, Medical Faculty, Recep Tayyip Erdogan University, Rize, Turkey
| | - Hasan Gundogdu
- Department of Radiology, Medical Faculty, Recep Tayyip Erdogan University, Rize, Turkey
| | - Hatice B Polat
- Department of Internal Medicine, Medical Faculty, Recep Tayyip Erdogan University, Rize, Turkey
| |
Collapse
|
45
|
Liu W, Tan Z, Geng M, Jiang X, Xin Y. Impact of the gut microbiota on angiotensin Ⅱ-related disorders and its mechanisms. Biochem Pharmacol 2023:115659. [PMID: 37330020 DOI: 10.1016/j.bcp.2023.115659] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 06/19/2023]
Abstract
The renin-angiotensin system (RAS) consists of multiple angiotensin peptides and performs various biological functions mediated by distinct receptors. Angiotensin II (Ang II) is the major effector of the RAS and affects the occurrence and development of inflammation, diabetes mellitus and its complications, hypertension, and end-organ damage via the Ang II type 1 receptor. Recently, considerable interest has been given to the association and interaction between the gut microbiota and host. Increasing evidence suggests that the gut microbiota may contribute to cardiovascular diseases, obesity, type 2 diabetes mellitus, chronic inflammatory diseases, and chronic kidney disease. Recent data have confirmed that Ang II can induce an imbalance in the intestinal flora and further aggravate disease progression. Furthermore, angiotensin converting enzyme 2 is another player in RAS, alleviates the deleterious effects of Ang II, modulates gut microbial dysbiosis, local and systemic immune responses associated with coronavirus disease 19. Due to the complicated etiology of pathologies, the precise mechanisms that link disease processes with specific characteristics of the gut microbiota remain obscure. This review aims to highlight the complex interactions between the gut microbiota and its metabolites in Ang II-related disease progression, and summarize the possible mechanisms. Deciphering these mechanisms will provide a theoretical basis for novel therapeutic strategies for disease prevention and treatment. Finally, we discuss therapies targeting the gut microbiota to treat Ang II-related disorders.
Collapse
Affiliation(s)
- Wei Liu
- Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Zining Tan
- Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Mengrou Geng
- Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Xin Jiang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy and Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China.
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
46
|
Shen L, Chen DY, Lou QQ, Cao P, Hu R, Jin Y, Wang D, Hu SS. Angiotensin Type 2 Receptor Pharmacological Agonist Relieves Neurocognitive Deficits via Reducing Neuroinflammation and Microglial Engulfment of Dendritic Spines. J Neuroimmune Pharmacol 2023; 18:41-57. [PMID: 36464726 PMCID: PMC9734469 DOI: 10.1007/s11481-022-10054-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 11/08/2022] [Indexed: 12/07/2022]
Abstract
Mechanically ventilated patients suffering critical illness are at high risk of developing neurocognitive impairments. Angiotensin type 2 receptor (AGTR2) has been demonstrated to be anti-inflammatory and neuroprotective. The present study thus aimed to investigate whether AGTR2 can alleviate cerebral dysfunction in mice subjected to cochallenge with lipopolysaccharide (LPS) and mechanical ventilation (MV), and to reveal the underlying mechanism. We utilized a mice model that received a single injection of LPS (1 mg/kg, intraperitoneally) followed 2 h later by MV (10 ml/kg, lasting for 2 h). Pretreatment with the AGTR2 pharmacological agonist C21 (0.03, 0.3, and 3 mg/kg, intraperitoneally, once daily, lasting for 10 days). Locomotor activity and behavioral deficits were evaluated 24 h post-MV by open-field and fear-condition tests. Brain hippocampus and prefrontal cortex tissues were collected for immunofluorescence staining and western blotting to evaluate the resulting impacts on microglia, including morphological traits, functional markers, synaptic engulfment, superoxide production, and signaling molecules. Compared with vehicle-control, pre-administrated C21 reduced the branch endpoints and length of microglia processes in a dose-dependent manner in mice subjected to LPS/MV. The neuroprotective effect of AGTR2 was behaviorally confirmed by the improvement of memory decline in LPS/MV-treated mice following C21 pretreatment. In addition to morphological alterations, C21 reduced microglial functional markers and reduced microglial-dendrite contact and microglial engulfment of synaptic protein markers. In terms of the underlying molecular mechanism, AGTR2 stimulation by C21 leads to activation of protein phosphatase 2A, which subsequently mitigates microglial PKCδ and NF-κB activation, and inhibites NOX2-derived ROS production. The AGTR2 agonist C21 alleviates behavioral deficits in those mice subjected to LPS/MV, via mechanisms that involve reactive microglia and abnormal synaptic plasticity in NOX2-derived ROS and the PKCδ-NFκB pathway.
Collapse
Affiliation(s)
- Liang Shen
- Anhui Provincial Hospita, Anhui Provincial Hospital Affiliated to Anhui Medical University, Anhui Medical University, Hefei, 230036, China
| | - Dan-Yang Chen
- Department of Neurobiology, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Qian-Qian Lou
- Department of Neurobiology, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Peng Cao
- Department of Neurobiology, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Rui Hu
- Department of Neurobiology, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Yan Jin
- Department of Neurobiology, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Di Wang
- Anhui Provincial Hospita, Anhui Provincial Hospital Affiliated to Anhui Medical University, Anhui Medical University, Hefei, 230036, China
- Department of Anesthesiology, First Affiliated Hospital of USTC (Anhui Provincial Hospita), Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, 230001, China
| | - Shan-Shan Hu
- Department of Clinical Laboratory, First Affiliated Hospital of USTC (Anhui Provincial Hospita), Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, 230001, China.
| |
Collapse
|
47
|
Kanugula AK, Kaur J, Batra J, Ankireddypalli AR, Velagapudi R. Renin-Angiotensin System: Updated Understanding and Role in Physiological and Pathophysiological States. Cureus 2023; 15:e40725. [PMID: 37350982 PMCID: PMC10283427 DOI: 10.7759/cureus.40725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2023] [Indexed: 06/24/2023] Open
Abstract
The classical view of the renin-angiotensin system (RAS) is that of the circulating hormone pathway involved in salt and water homeostasis and blood pressure regulation. It is also involved in the pathogenesis of cardiac and renal disorders. This led to the creation of drugs blocking the actions of this classical pathway, which improved cardiac and renal outcomes. Our understanding of the RAS has significantly expanded with the discovery of new peptides involved in this complex pathway. Over the last two decades, a counter-regulatory or protective pathway has been discovered that opposes the effects of the classical pathway. Components of RAS are also implicated in the pathogenesis of obesity and its metabolic diseases. The continued discovery of newer molecules also provides novel therapeutic targets to improve disease outcomes. This article aims to provide an overview of an updated understanding of the RAS, its role in physiological and pathological processes, and potential novel therapeutic options from RAS for managing cardiorenal disorders, obesity, and related metabolic disorders.
Collapse
Affiliation(s)
- Ashok Kumar Kanugula
- Department of Internal Medicine, Wellstar Health System - Spalding Regional Hospital, Griffin, USA
| | - Jasleen Kaur
- Department of Endocrinology, Diabetes, and Metabolism, HealthPartners, Minneapolis, USA
| | - Jaskaran Batra
- Department of Internal Medicine, Univerity of Pittsburg Medical Center (UPMC) McKeesport, McKeesport, USA
| | | | - Ravikanth Velagapudi
- Department of Pulmonary and Critical Care Medicine, Spectrum Health/Michigan State University, Grand Rapids, USA
| |
Collapse
|
48
|
Alshammari A, Han Y, Jones TW, Pillai B, Zhang D, Ergul A, Somanath PR, Fagan SC. Stimulation of Angiotensin II Type 2 Receptor Modulates Pro-Inflammatory Response in Microglia and Macrophages: Therapeutic Implications for the Treatment of Stroke. Life (Basel) 2023; 13:1274. [PMID: 37374057 DOI: 10.3390/life13061274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/10/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Sustained microglial activation contributes to the development of post-stroke cognitive impairment (PSCI). Compound 21 (C21), an angiotensin II type 2 receptor agonist, has shown some neurovascular protection after stroke. This study aimed to investigate the direct anti-inflammatory effects of C21 on macrophages, as well as brain innate immune cells. METHODS Murine microglial cell line (C8-B4) and RAW 264.7 macrophages were exposed to lipopolysaccharide (LPS) and co-treated with C21. Pro-inflammatory mediators were assessed via RT-qPCR and ELISA. Cellular reactive oxygen species (ROS) were evaluated via CellROXGreen staining, and nitrate production was assessed using Griess assay. RESULTS C21 suppressed LPS-induced inflammation and ROS generation in both cells. In microglia, C21 blunted LPS-induced mRNA expression of IL-1β, IL-12b, COX-1, iNOS, and IL-6. A similar pattern was observed in macrophages, where C21 suppressed LPS-induced IL-1β, TNF-α, and CXCL1 expression. These anti-inflammatory effects in microglia and macrophages were associated with increased neuroprotective gene expression, including GDNF and BDNF, in a dose-dependent manner. CONCLUSIONS Our findings suggest a protective effect of C21 against the inflammatory response, in both macrophages and microglia, via suppression of the release of pro-inflammatory cytokines/chemokines and the generation of ROS while stimulating the production of neurotrophic factors.
Collapse
Affiliation(s)
- Abdulkarim Alshammari
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30602, USA
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
- Department of Clinical Pharmacy, Faculty of Pharmacy, Northern Border University, Rafha 76313, Saudi Arabia
| | - Yohan Han
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30602, USA
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Timothy W Jones
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30602, USA
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Bindu Pillai
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30602, USA
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Duo Zhang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30602, USA
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| | - Adviye Ergul
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
- Ralph H. Johnson VA Health Care System, Charleston, SC 29401, USA
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30602, USA
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| | - Susan C Fagan
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30602, USA
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| |
Collapse
|
49
|
Scheinman SB, Tseng KY, Alford S, Tai LM. Higher Neuronal Facilitation and Potentiation with APOE4 Suppressed by Angiotensin II. RESEARCH SQUARE 2023:rs.3.rs-2960437. [PMID: 37292788 PMCID: PMC10246245 DOI: 10.21203/rs.3.rs-2960437/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Progressive hippocampal degeneration is a key component of Alzheimer's disease (AD) progression. Therefore, identifying how hippocampal neuronal function is modulated early in AD is an important approach to eventually prevent degeneration. AD-risk factors and signaling molecules likely modulate neuronal function, including APOE genotype and angiotensin II. Compared to APOE3 , APOE4 increases AD risk up to 12-fold, and high levels of angiotensin II are hypothesized to disrupt neuronal function in AD. However, the extent that APOE and angiotensin II modulates the hippocampal neuronal phenotype in AD-relevant models is unknown. To address this issue, we used electrophysiological techniques to assess the impact of APOE genotype and angiotensin II on basal synaptic transmission, presynaptic and post-synaptic activity in mice that express human APOE3 (E3FAD) or APOE4 (E4FAD) and overproduce Aβ. We found that compared to E3FAD mice, E4FAD mice had lower basal synaptic activity, but higher levels of paired pulse facilitation (PPF) and Long-Term Potentiation (LTP) in the Schaffer Collateral Commissural Pathway (SCCP) of the hippocampus. We also found that exogenous angiotensin II has a profound inhibitory effect on hippocampal LTP in both E3FAD and E4FAD mice. Collectively, our data suggests that APOE4 and Aβ are associated with a hippocampal phenotype comprised of lower basal activity and higher responses to high frequency stimulation, the latter of which is suppressed by angiotensin II. These novel data suggest a potential mechanistic link between hippocampal activity, APOE4 genotype and angiotensin II in AD.
Collapse
Affiliation(s)
| | - Kuei Y Tseng
- University of Illinois at Chicago College of Medicine
| | - Simon Alford
- University of Illinois at Chicago College of Medicine
| | - Leon M Tai
- University of Illinois at Chicago College of Medicine
| |
Collapse
|
50
|
Rivas-Santisteban R, Lillo J, Raïch I, Muñoz A, Lillo A, Rodríguez-Pérez AI, Labandeira-García JL, Navarro G, Franco R. The cannabinoid CB 1 receptor interacts with the angiotensin AT 2 receptor. Overexpression of AT 2-CB 1 receptor heteromers in the striatum of 6-hydroxydopamine hemilesioned rats. Exp Neurol 2023; 362:114319. [PMID: 36632949 DOI: 10.1016/j.expneurol.2023.114319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/14/2022] [Accepted: 01/06/2023] [Indexed: 01/11/2023]
Abstract
It is of particular interest the potential of cannabinoid and angiotensin receptors as targets in the therapy of Parkinson's disease (PD). While endocannabinoids are neuromodulators that act through the CB1 and CB2 cannabinoid receptors, the renin angiotensin-system is relevant for regulation of the correct functioning of several brain circuits. Resonance energy transfer assays in a heterologous system showed that the CB1 receptor (CB1R) can directly interact with the angiotensin AT2 receptor (AT2R). Coactivation of the two receptors results in increased Gi-signaling. The AT2-CB1 receptor heteromer imprint consists of a blockade of AT2R-mediated signaling by rimonabant, a CB1R antagonist. Interestingly, the heteromer imprint, discovered in the heterologous system, was also found in primary striatal neurons thus demonstrating the expression of the heteromer in these cells. In situ proximity ligation assays confirmed the occurrence of AT2-CB1 receptor heteromers in striatal neurons. In addition, increased expression of the AT2-CB1 receptor heteromeric complexes was detected in the striatum of a rodent PD model consisting of rats hemilesioned using 6-hydroxydopamine. Expression of the heteromer was upregulated in the striatum of lesioned animals and, also, of lesioned animals that upon levodopa treatment became dyskinetic. In contrast, there was no upregulation in the striatum of lesioned rats that did not become dyskinetic upon chronic levodopa treatment. The results suggest that therapeutic developments focused on the CB1R should consider that this receptor can interact with the AT2R, which in the CNS is involved in mechanisms related to addictive behaviors and to neurodegenerative and neuroinflammatory diseases.
Collapse
Affiliation(s)
- Rafael Rivas-Santisteban
- Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain; CiberNed. Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
| | - Jaume Lillo
- Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain; CiberNed. Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
| | - Iu Raïch
- Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain; CiberNed. Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
| | - Ana Muñoz
- CiberNed. Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain; Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Alejandro Lillo
- Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain; CiberNed. Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
| | - Ana I Rodríguez-Pérez
- CiberNed. Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain; Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - José L Labandeira-García
- CiberNed. Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain; Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Gemma Navarro
- Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain; CiberNed. Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain; Neurosciences Institute, University of Barcelona (NeuroUB), Facultad de Psicología Campus de Mundet Paseo de la Vall d'Hebron, 171 08035 Barcelona, Spain.
| | - Rafael Franco
- CiberNed. Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain; Molecular Neurobiology laboratory, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain; School of Chemistry, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|