1
|
Shen Y, Wu Y, Zhuang H, Chen Z, Zhang Q, Li P, Wang J, Huang Z, Zeng Q. Compound 21 Attenuates Isoflurane-Induced Injury in Neonatal Rat Hippocampal Neurons and Primary Rat Neuronal Cells by Upregulating METTL3. J Inflamm Res 2024; 17:10079-10091. [PMID: 39639929 PMCID: PMC11618859 DOI: 10.2147/jir.s483211] [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: 09/03/2024] [Accepted: 11/06/2024] [Indexed: 12/07/2024] Open
Abstract
Background Isoflurane, as an anesthetic drug, has a neurotoxic effect on the developing brain tissue. Compound 21 (C21) has been reported to be neuroprotective and ameliorate stroke effects. However, the mechanism by which C21 protects against nerve damage remains unclear. Methods Animal and cellular models of brain injury were constructed using isoflurane (ISO) in neonatal SD rats and primary rat neuronal cells (PRNCs). After treatment with C21, the ultrastructure and morphology of the hippocampus in the model rats were assessed using the transmission electron microscope and H&E staining. Methylation or apoptosis-related genes or proteins were examined using immunohistochemistry, RT-qPCR, and Western blot. The levels of inflammatory factors were monitored using the ELISA kits. m6A modification was analyzed by Dot blot and MeRIP-qPCR. Cell proliferation and apoptosis were also tested using Edu and TUNEL staining. Results C21 suppresses apoptosis and inflammation and improves hippocampal morphology in ISO-induced neonatal rats. Mechanistically, C21 upregulates m6A modification, PPAR-a, BCL-2, and METTL3 in ISO-induced neonatal rats and ISO-treated PRNCs. C21 promotes cell proliferation, enhances BCL-2 m6A modification, and reduces inflammation by upregulating METTL3 by upregulating METTL3 in ISO-treated PRNCs. Conclusion These findings suggest that C21 enhances neuronal cell survival and morphology and up-regulates methylation and Bc1-2 levels, potentially offering a therapeutic strategy for neuroprotection in clinical settings, particularly in cases of neurotoxic exposure. The mechanism may be related to the upregulation of METTL3.
Collapse
Affiliation(s)
- Yaping Shen
- Department of Interventional Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550002, People’s Republic of China
- Department of Interventional Radiology, The Baiyun Hospital Affiliated to Guizhou Medical University, Guiyang, Guizhou, 550014, People’s Republic of China
| | - Yijiu Wu
- College of Anesthesiology, Guizhou Medical University, Guiyang, 550002, People’s Republic of China
| | - Hai Zhuang
- Department of Infectious, The Baiyun Hospital Affiliated to Guizhou Medical University, Guiyang, Guizhou, 550014, People’s Republic of China
| | - Zhumei Chen
- Department of Anesthesiology, The Baiyun Hospital Affiliated to Guizhou Medical University, Guiyang, Guizhou, 550014, People’s Republic of China
| | - Qiong Zhang
- Department of Anesthesiology, The Baiyun Hospital Affiliated to Guizhou Medical University, Guiyang, Guizhou, 550014, People’s Republic of China
| | - Peixin Li
- Department of Anesthesiology, The Baiyun Hospital Affiliated to Guizhou Medical University, Guiyang, Guizhou, 550014, People’s Republic of China
| | - Jing Wang
- College of Anesthesiology, Guizhou Medical University, Guiyang, 550002, People’s Republic of China
| | - Zhi Huang
- Department of Interventional Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550002, People’s Republic of China
| | - Qingfan Zeng
- College of Anesthesiology, Guizhou Medical University, Guiyang, 550002, People’s Republic of China
- Department of Anesthesiology, The Baiyun Hospital Affiliated to Guizhou Medical University, Guiyang, Guizhou, 550014, People’s Republic of China
| |
Collapse
|
2
|
Buonfiglio F, Pfeiffer N, Gericke A. Glaucoma and the ocular renin-angiotensin-aldosterone system: Update on molecular signalling and treatment perspectives. Cell Signal 2024; 122:111343. [PMID: 39127136 DOI: 10.1016/j.cellsig.2024.111343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/29/2024] [Accepted: 08/08/2024] [Indexed: 08/12/2024]
Abstract
Glaucoma, a leading cause of blindness worldwide, encompasses a group of pathological conditions affecting the optic nerve and is characterized by progressive retinal ganglion cell loss, cupping of the optic nerve head, and distinct visual field defects. While elevated intraocular pressure (IOP) is the main risk factor for glaucoma, many patients do not have elevated IOP. Consequently, other risk factors, such as ocular blood flow abnormalities and immunological factors, have been implicated in its pathophysiology. Traditional therapeutic strategies primarily aim to reduce IOP, but there is growing interest in developing novel treatment approaches to improve disease management and reduce the high rates of severe visual impairment. In this context, targeting the ocular renin-angiotensin-aldosterone system (RAAS) has been found as a potential curative strategy. The RAAS contributes to glaucoma development through key effectors such as prorenin, angiotensin II, and aldosterone. Recent evidence has highlighted the potential of using RAAS modulators to combat glaucoma, yielding encouraging results. Our study aims to explore the molecular pathways linking the ocular RAAS and glaucoma, summarizing recent advances that elucidate the role of the RAAS in triggering oxidative stress, inflammation, and remodelling in the pathogenesis of glaucoma. Additionally, we will present emerging therapeutic approaches that utilize RAAS modulators and antioxidants to slow the progression of glaucoma.
Collapse
Affiliation(s)
- Francesco Buonfiglio
- Departments of Ophthalmology, University Medical Center of the Johannes Gutenberg- University, Langenbeckstr.1, 55131 Mainz, Germany.
| | - Norbert Pfeiffer
- Departments of Ophthalmology, University Medical Center of the Johannes Gutenberg- University, Langenbeckstr.1, 55131 Mainz, Germany.
| | - Adrian Gericke
- Departments of Ophthalmology, University Medical Center of the Johannes Gutenberg- University, Langenbeckstr.1, 55131 Mainz, Germany.
| |
Collapse
|
3
|
Villa-González M, Rubio M, Martín-López G, Mallavibarrena PR, Vallés-Saiz L, Vivien D, Wandosell F, Pérez-Álvarez MJ. Pharmacological inhibition of mTORC1 reduces neural death and damage volume after MCAO by modulating microglial reactivity. Biol Direct 2024; 19:26. [PMID: 38582839 PMCID: PMC10999095 DOI: 10.1186/s13062-024-00470-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/25/2024] [Indexed: 04/08/2024] Open
Abstract
Ischemic stroke is a sudden and acute disease characterized by neuronal death, increment of reactive gliosis (reactive microglia and astrocytes), and a severe inflammatory process. Neuroinflammation is an early event after cerebral ischemia, with microglia playing a leading role. Reactive microglia involve functional and morphological changes that drive a wide variety of phenotypes. In this context, deciphering the molecular mechanisms underlying such reactive microglial is essential to devise strategies to protect neurons and maintain certain brain functions affected by early neuroinflammation after ischemia. Here, we studied the role of mammalian target of rapamycin (mTOR) activity in the microglial response using a murine model of cerebral ischemia in the acute phase. We also determined the therapeutic relevance of the pharmacological administration of rapamycin, a mTOR inhibitor, before and after ischemic injury. Our data show that rapamycin, administered before or after brain ischemia induction, reduced the volume of brain damage and neuronal loss by attenuating the microglial response. Therefore, our findings indicate that the pharmacological inhibition of mTORC1 in the acute phase of ischemia may provide an alternative strategy to reduce neuronal damage through attenuation of the associated neuroinflammation.
Collapse
Affiliation(s)
- Mario Villa-González
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
| | - Marina Rubio
- Physiopathology and Imaging of Neurological Disorders, Normandie University, UNICAEN, UMR-S U1237, INSERM, Institut Blood and Brain @ CaenNormandie, GIP Cyceron, Caen, France
| | - Gerardo Martín-López
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
| | - Paula R Mallavibarrena
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
| | | | - Denis Vivien
- Physiopathology and Imaging of Neurological Disorders, Normandie University, UNICAEN, UMR-S U1237, INSERM, Institut Blood and Brain @ CaenNormandie, GIP Cyceron, Caen, France
- Department of Clinical Research, Caen-Normandie Hospital (CHU), Caen, France
| | - Francisco Wandosell
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain.
- Centro de Investigaciones Biológicas en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| | - Maria José Pérez-Álvarez
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain.
- Instituto Universitario de Biología Molecular (IUBM-UAM), Madrid, Spain.
| |
Collapse
|
4
|
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
|
5
|
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: 3.0] [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
|
6
|
Steckelings UM, Widdop RE, Sturrock ED, Lubbe L, Hussain T, Kaschina E, Unger T, Hallberg A, Carey RM, Sumners C. The Angiotensin AT 2 Receptor: From a Binding Site to a Novel Therapeutic Target. Pharmacol Rev 2022; 74:1051-1135. [PMID: 36180112 PMCID: PMC9553111 DOI: 10.1124/pharmrev.120.000281] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/19/2022] [Accepted: 06/27/2022] [Indexed: 11/22/2022] Open
Abstract
Discovered more than 30 years ago, the angiotensin AT2 receptor (AT2R) has evolved from a binding site with unknown function to a firmly established major effector within the protective arm of the renin-angiotensin system (RAS) and a target for new drugs in development. The AT2R represents an endogenous protective mechanism that can be manipulated in the majority of preclinical models to alleviate lung, renal, cardiovascular, metabolic, cutaneous, and neural diseases as well as cancer. This article is a comprehensive review summarizing our current knowledge of the AT2R, from its discovery to its position within the RAS and its overall functions. This is followed by an in-depth look at the characteristics of the AT2R, including its structure, intracellular signaling, homo- and heterodimerization, and expression. AT2R-selective ligands, from endogenous peptides to synthetic peptides and nonpeptide molecules that are used as research tools, are discussed. Finally, we summarize the known physiological roles of the AT2R and its abundant protective effects in multiple experimental disease models and expound on AT2R ligands that are undergoing development for clinical use. The present review highlights the controversial aspects and gaps in our knowledge of this receptor and illuminates future perspectives for AT2R research. SIGNIFICANCE STATEMENT: The angiotensin AT2 receptor (AT2R) is now regarded as a fully functional and important component of the renin-angiotensin system, with the potential of exerting protective actions in a variety of diseases. This review provides an in-depth view of the AT2R, which has progressed from being an enigma to becoming a therapeutic target.
Collapse
Affiliation(s)
- U Muscha Steckelings
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Robert E Widdop
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Edward D Sturrock
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Lizelle Lubbe
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Tahir Hussain
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Elena Kaschina
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Thomas Unger
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Anders Hallberg
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Robert M Carey
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Colin Sumners
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| |
Collapse
|
7
|
Ahmed HA, Ismael S, Salman M, Devlin P, McDonald MP, Liao FF, Ishrat T. Direct AT2R Stimulation Slows Post-stroke Cognitive Decline in the 5XFAD Alzheimer's Disease Mice. Mol Neurobiol 2022; 59:4124-4140. [PMID: 35486224 PMCID: PMC10947502 DOI: 10.1007/s12035-022-02839-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/01/2022] [Indexed: 10/18/2022]
Abstract
Alzheimer's disease (AD), currently the single leading cause of death still on the rise, almost always coexists alongside vascular cognitive impairment (VCI). In fact, the ischemic disease affects up to 90% of AD patients, with strokes and major infarctions representing over a third of vascular lesions. Studies also confirmed that amyloid plaques, typical of AD, are much more likely to cause dementia if strokes or cerebrovascular damage also exist, leading to the term "mixed pathology" cognitive impairment. Although its incidence is expected to grow, there are no satisfactory treatments. There is hence an urgent need for safe and effective therapies that preserve cognition, maintain function, and prevent the clinical deterioration that results from the progression of this irreversible, neurodegenerative disease. To our knowledge, this is the first study to investigate the effects of long-term treatment with C21, a novel angiotensin II type 2 receptor (AT2R) agonist, on the development of "mixed pathology" cognitive impairment. This was accomplished using a unique model that employs the fundamental elements of both AD and VCI. Treatment with C21/vehicle was started 1 h post-stroke and continued for 5 weeks in mice with concurrent AD pathology. Efficacy was established through a series of functional tests assessing various aspects of cognition, including spatial learning, short-term/working memory, long-term/reference memory, and cognitive flexibility, in addition to the molecular markers characteristic of AD. Our findings demonstrate that C21 treatment preserves cognitive function, maintains cerebral blood flow, and reduces Aβ accumulation and toxic tau phosphorylation in AD animals post-stroke.
Collapse
Affiliation(s)
- Heba A Ahmed
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, 875 Monroe Avenue, Wittenborg Bldg, Room-231, Memphis, TN, 38163, USA
| | - Saifudeen Ismael
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, 875 Monroe Avenue, Wittenborg Bldg, Room-231, Memphis, TN, 38163, USA
| | - Mohd Salman
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, 875 Monroe Avenue, Wittenborg Bldg, Room-231, Memphis, TN, 38163, USA
| | - Patrick Devlin
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, 875 Monroe Avenue, Wittenborg Bldg, Room-231, Memphis, TN, 38163, USA
| | - Michael P McDonald
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, 875 Monroe Avenue, Wittenborg Bldg, Room-231, Memphis, TN, 38163, USA
- Department of Neurology, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Francesca-Fang Liao
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
- Department of Pharmacology, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Tauheed Ishrat
- Department of Anatomy and Neurobiology, College of Medicine, The University of Tennessee Health Science Center, 875 Monroe Avenue, Wittenborg Bldg, Room-231, Memphis, TN, 38163, USA.
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
- Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|
8
|
Gouveia F, Camins A, Ettcheto M, Bicker J, Falcão A, Cruz MT, Fortuna A. Targeting brain Renin-Angiotensin System for the prevention and treatment of Alzheimer's disease: Past, present and future. Ageing Res Rev 2022; 77:101612. [PMID: 35346852 DOI: 10.1016/j.arr.2022.101612] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 02/09/2022] [Accepted: 03/16/2022] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a well-known neurodegenerative disease characterized by the presence of two main hallmarks - Tau hyperphosphorylation and Aβ deposits. Notwithstanding, in the last few years the scientific evidence about the drivers of AD have been changing and nowadays age-related vascular alterations and several cardiovascular risk factors have been shown to trigger the development of AD. In this context, drugs targeting the Renin Angiotensin System (RAS), commonly used for the treatment of hypertension, are evidencing a high potential to delay AD development due to their action on brain RAS. Indeed, the ACE 1/Ang II/AT1R axis is believed to be upregulated in AD and to be responsible for deleterious effects such as increased oxidative stress, neuroinflammation, blood-brain barrier (BBB) hyperpermeability, astrocytes dysfunction and a decrease in cerebral blood flow. In contrast, the alternative axis - ACE 1/Ang II/AT2R; ACE 2/Ang (1-7)/MasR; Ang IV/ AT4R(IRAP) - seems to counterbalance the deleterious effects of the principal axis and to exert beneficial effects on memory and cognition. Accordingly, retrospective studies demonstrate a reduced risk of developing AD among people taking RAS medication as well as several in vitro and in vivo pre-clinical studies as it is herein critically reviewed. In this review, we first revise, at a glance, the pathophysiology of AD focused on its classic hallmarks. Secondly, an overview about the impact of the RAS on the pathophysiology of AD is also provided, focused on their four essential axes ACE 1/Ang II/AT2R; ACE 2/Ang (1-7)/MasR; Ang IV/ AT4R(IRAP) and ACE 1/Ang II/AT1R. Finally, the therapeutic potential of available drugs targeting RAS on AD, namely angiotensin II receptor blockers (ARBs) and angiotensin converting enzyme inhibitors (ACEIs), is highlighted and data supporting this hope will be presented, from in vitro and in vivo pre-clinical to clinical studies.
Collapse
|
9
|
Villa-González M, Martín-López G, Pérez-Álvarez MJ. Dysregulation of mTOR Signaling after Brain Ischemia. Int J Mol Sci 2022; 23:ijms23052814. [PMID: 35269956 PMCID: PMC8911477 DOI: 10.3390/ijms23052814] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/28/2022] [Accepted: 03/01/2022] [Indexed: 02/04/2023] Open
Abstract
In this review, we provide recent data on the role of mTOR kinase in the brain under physiological conditions and after damage, with a particular focus on cerebral ischemia. We cover the upstream and downstream pathways that regulate the activation state of mTOR complexes. Furthermore, we summarize recent advances in our understanding of mTORC1 and mTORC2 status in ischemia–hypoxia at tissue and cellular levels and analyze the existing evidence related to two types of neural cells, namely glia and neurons. Finally, we discuss the potential use of mTORC1 and mTORC2 as therapeutic targets after stroke.
Collapse
Affiliation(s)
- Mario Villa-González
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.V.-G.); (G.M.-L.)
- Centro de Biología Molecular “Severo Ochoa” (CBMSO), Universidad Autónoma de Madrid/CSIC, 28049 Madrid, Spain
| | - Gerardo Martín-López
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.V.-G.); (G.M.-L.)
| | - María José Pérez-Álvarez
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (M.V.-G.); (G.M.-L.)
- Centro de Biología Molecular “Severo Ochoa” (CBMSO), Universidad Autónoma de Madrid/CSIC, 28049 Madrid, Spain
- Correspondence: ; Tel.: +34-91-497-2819
| |
Collapse
|
10
|
Ismael S, Mirzahosseini G, Ahmed HA, Yoo A, Kassan M, Malik KU, Ishrat T. Renin-Angiotensin System Alterations in the Human Alzheimer's Disease Brain. J Alzheimers Dis 2021; 84:1473-1484. [PMID: 34690145 DOI: 10.3233/jad-215051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Understanding Alzheimer's disease (AD) in terms of its various pathophysiological pathways is essential to unravel the complex nature of the disease process and identify potential therapeutic targets. The renin-angiotensin system (RAS) has been implicated in several brain diseases, including traumatic brain injury, ischemic stroke, and AD. OBJECTIVE This study was designed to evaluate the protein expression levels of RAS components in postmortem cortical and hippocampal brain samples obtained from AD versus non-AD individuals. METHODS We analyzed RAS components in the cortex and hippocampus of postmortem human brain samples by western blotting and immunohistochemical techniques in comparison with age-matched non-demented controls. RESULTS The expression of AT1R increased in the hippocampus, whereas AT2R expression remained almost unchanged in the cortical and hippocampal regions of AD compared to non-AD brains. The Mas receptor was downregulated in the hippocampus. We also detected slight reductions in ACE-1 protein levels in both the cortex and hippocampus of AD brains, with minor elevations in ACE-2 in the cortex. We did not find remarkable differences in the protein levels of angiotensinogen and Ang II in either the cortex or hippocampus of AD brains, whereas we observed a considerable increase in the expression of brain-derived neurotrophic factor in the hippocampus. CONCLUSION The current findings support the significant contribution of RAS components in AD pathogenesis, further suggesting that strategies focusing on the AT1R and AT2R pathways may lead to novel therapies for the management of AD.
Collapse
Affiliation(s)
- Saifudeen Ismael
- Departments of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Golnoush Mirzahosseini
- Departments of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA.,Departments of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Heba A Ahmed
- Departments of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Arum Yoo
- Departments of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Modar Kassan
- Departments of Physiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kafait U Malik
- Departments of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA.,Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Tauheed Ishrat
- Departments of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA.,Departments of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA.,Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
11
|
Rabab’h O, Al-Ramadan A, Gharaibeh A. Comment on "Vascular Endothelial Growth Factor (VEGF) as a Vital Target for Brain Inflammation during the COVID-19 Outbreak". ACS Chem Neurosci 2021; 12:2712-2713. [PMID: 34197067 DOI: 10.1021/acschemneuro.1c00359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Omar Rabab’h
- Insight Research Institute, Flint, Michigan 48507, United States
- Center for Cognition & Neuroethics, University of Michigan—Flint, Flint, Michigan 48502, United States
| | - Ali Al-Ramadan
- Insight Research Institute, Flint, Michigan 48507, United States
- Center for Cognition & Neuroethics, University of Michigan—Flint, Flint, Michigan 48502, United States
| | - Abeer Gharaibeh
- Insight Research Institute, Flint, Michigan 48507, United States
- Center for Cognition & Neuroethics, University of Michigan—Flint, Flint, Michigan 48502, United States
- Insight Institute of Neurosurgery & Neuroscience, Flint, Michigan 48507, United States
| |
Collapse
|
12
|
Tuo QZ, Zhang ST, Lei P. Mechanisms of neuronal cell death in ischemic stroke and their therapeutic implications. Med Res Rev 2021; 42:259-305. [PMID: 33957000 DOI: 10.1002/med.21817] [Citation(s) in RCA: 355] [Impact Index Per Article: 88.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 03/31/2021] [Accepted: 04/23/2021] [Indexed: 02/05/2023]
Abstract
Ischemic stroke caused by arterial occlusion is the most common type of stroke, which is among the most frequent causes of disability and death worldwide. Current treatment approaches involve achieving rapid reperfusion either pharmacologically or surgically, both of which are time-sensitive; moreover, blood flow recanalization often causes ischemia/reperfusion injury. However, even though neuroprotective intervention is urgently needed in the event of stroke, the exact mechanisms of neuronal death during ischemic stroke are still unclear, and consequently, the capacity for drug development has remained limited. Multiple cell death pathways are implicated in the pathogenesis of ischemic stroke. Here, we have reviewed these potential neuronal death pathways, including intrinsic and extrinsic apoptosis, necroptosis, autophagy, ferroptosis, parthanatos, phagoptosis, and pyroptosis. We have also reviewed the latest results of pharmacological studies on ischemic stroke and summarized emerging drug targets with a focus on clinical trials. These observations may help to further understand the pathological events in ischemic stroke and bridge the gap between basic and translational research to reveal novel neuroprotective interventions.
Collapse
Affiliation(s)
- Qing-Zhang Tuo
- Department of Geriatrics and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Shu-Ting Zhang
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
13
|
The Angiotensin II Type 2 Receptor, a Target for Protection and Regeneration of the Peripheral Nervous System? Pharmaceuticals (Basel) 2021; 14:ph14030175. [PMID: 33668331 PMCID: PMC7996246 DOI: 10.3390/ph14030175] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 02/07/2023] Open
Abstract
Preclinical evidence, accumulated over the past decade, indicates that the angiotensin II type 2 receptor (AT2R) stimulation exerts significant neuroprotective effects in various animal models of neuronal injury, notably in the central nervous system. While the atypical G protein-coupled receptor superfamily nature of AT2R and its related signaling are still under investigation, pharmacological studies have shown that stimulation of AT2R leads to neuritogenesis in vitro and in vivo. In this review, we focus on the potential neuroprotective and neuroregenerative roles of AT2R specifically in the peripheral nervous system (PNS). The first section describes the evidence for AT2R expression in the PNS and highlights current controversies concerning the cellular distribution of the receptor. The second section focuses on AT2R signaling implicated in neuronal survival and in neurite outgrowth. The following sections review the relatively few preclinical studies highlighting the putative neuroprotective and neuroregenerative effects of AT2R stimulation in the context of peripheral neuropathy.
Collapse
|
14
|
Scotti L, Bassi L, Soranna D, Verde F, Silani V, Torsello A, Parati G, Zambon A. Association between renin-angiotensin-aldosterone system inhibitors and risk of dementia: A meta-analysis. Pharmacol Res 2021; 166:105515. [PMID: 33636351 DOI: 10.1016/j.phrs.2021.105515] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/04/2021] [Accepted: 02/21/2021] [Indexed: 10/22/2022]
Abstract
OBJECTIVE To evaluate the association of all RAAS inhibitors, ACE inhibitors (ACEIs) and angiotensin II receptor blockers (ARBs) on dementia onset (any dementia, Alzheimer's disease and vascular dementia) using a meta-analytic approach. METHODS A systematic MEDLINE search was carried out to identify all observational studies published up to the 30th September 2020 evaluating the association between RAAS inhibitors and risk of dementia. Studies were included if original investigations considering incident dementia cases, with ACEIs and/or ARBs as exposure and other antihypertensives (AHs) use as reference, and if reporting association estimates and relative variability measures. Random effect pooled relative risks (pRR) and the corresponding 95% confidence intervals (95%CI) were calculated according to DerSimonian and Laird's (DL) or to Hartung Knapp Sidik Jonkman (HKSJ) method depending on the number of studies and between-studies heterogeneity. A linear mixed meta-regression model (MM) was applied to take into account correlation among association estimates from the same study. RESULTS 15 studies were included in the meta-analysis. ARBs but not ACEIs' use led to a significant reduction of the risk of any dementia (pRR 0.78, 95%CIMM 0.70-0.87) and Alzheimer's disease (pRR 0.73, 95%CIMM 0.60-0.90). Moreover, when compared to ACEIs, ARBs reduced of 14% the risk of any dementia (pRR 0.86, 95%CIDL 0.79-0.94). CONCLUSIONS ARBs but not ACEIs led to a reduction in the risk of any dementia. The difference between ARBs and ACEIs in terms of preventive effectiveness could be due to distinct profiles of antagonism towards independent receptor pathways or to differential influences on amyloid metabolism.
Collapse
Affiliation(s)
- Lorenza Scotti
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy.
| | | | - Davide Soranna
- Biostatistic Unit, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Federico Verde
- Department of Neurology - Stroke Unit and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy; Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, University of Milano, Milan, Italy
| | - Vincenzo Silani
- Department of Neurology - Stroke Unit and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy; Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, University of Milano, Milan, Italy
| | - Antonio Torsello
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Gianfranco Parati
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy; Department of Cardiovascular Neural and Metabolic Sciences, IRCCS Istituto Auxologico Italiano, S. Luca Hospital, Milan, Italy
| | - Antonella Zambon
- Biostatistic Unit, IRCCS Istituto Auxologico Italiano, Milan, Italy; Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
15
|
Hua H, Zhang W, Li J, Li J, Liu C, Guo Y, Cheng Y, Pi F, Xie Y, Yao W, Gao Y, Qian H. Neuroprotection against cerebral ischemia/reperfusion by dietary phytochemical extracts from Tibetan turnip (Brassica rapa L.). JOURNAL OF ETHNOPHARMACOLOGY 2021; 265:113410. [PMID: 32980487 DOI: 10.1016/j.jep.2020.113410] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 08/21/2020] [Accepted: 09/17/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Tibetan turnip (Brassica rapa L.) has a wide array of medicine properties including heat-clearing, detoxifying and anti-hypoxia as listed in the famous centuries-old Tibetan medicine classic "The Four Medical Tantras". Evidence-based medicine also indicated the anti-hypoxic effect of turnips, suggesting a potential link to neuroprotective effect on ischemic stroke. This thereby enables turnips to serve as a novel nontoxic agent in related treatment. AIM OF THE STUDY This study aimed to investigate the neuroprotective effect and elucidate the mechanism of aqueous extract of turnip (AET) on cerebral ischemia/reperfusion. MATERIALS AND METHODS The experimental models of cerebral ischemia included transient middle cerebral artery occlusion/reperfusion (MCAO) in C57BL/6J mice and oxygen-glucose deprivation/reoxygenation (OGD/R) in HT-22 cells. Long-term effect of AET on infarct volume was evaluated by microtubule-associated protein 2 (MAP2) immunofluorescence 28 days after MCAO, and on neurofunctional outcomes determined by rotarod, grid walking, and cylinder tests in the meantime. Efficacy of AET was determined by the cell viability, the release of lactate dehydrogenase (LDH) and reactive oxygen species (ROS) in neurons. The underlying mechanism of AET rescued OGD/R cells were characterized by PI3K, Akt and mTOR expressions, which were further used to validate AET's role in the pathway. RESULTS AET can reduce cerebral infarct volume and ameliorate behavioral deficits of MCAO/R mice dose-dependently. In vitro experiment further demonstrated that suitable concentrations of AET inhibited ROS, LDH production and restored mitochondrial expression induced by OGD/R. AET pretreatment can reverse the OGD/R-induced decreased level of phosphorylation of PI3K, Akt, mTOR, whereas this effect was blocked in the LY294002 (PI3K inhibitor) treatment group. CONCLUSIONS AET improved the survival of OGD/R-injured HT-22 cells by activating the PI3K/Akt/mTOR pathway. Based on the results above, aqueous extract of turnip has a protective effect on focal cerebral ischemic injury.
Collapse
Affiliation(s)
- Hanyi Hua
- Department of School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Wenyi Zhang
- Department of School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Jiaying Li
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Jiayi Li
- Department of School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Chang Liu
- Department of School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Yahui Guo
- Department of School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Yuliang Cheng
- Department of School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Fuwei Pi
- Department of School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Yunfei Xie
- Department of School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Weirong Yao
- Department of School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| | - He Qian
- Department of School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
16
|
McFall A, Nicklin SA, Work LM. The counter regulatory axis of the renin angiotensin system in the brain and ischaemic stroke: Insight from preclinical stroke studies and therapeutic potential. Cell Signal 2020; 76:109809. [PMID: 33059037 PMCID: PMC7550360 DOI: 10.1016/j.cellsig.2020.109809] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 01/01/2023]
Abstract
Stroke is the 2nd leading cause of death worldwide and the leading cause of physical disability and cognitive issues. Although we have made progress in certain aspects of stroke treatment, the consequences remain substantial and new treatments are needed. Hypertension has long been recognised as a major risk factor for stroke, both haemorrhagic and ischaemic. The renin angiotensin system (RAS) plays a key role in blood pressure regulation and this, plus local expression and signalling of RAS in the brain, both support the potential for targeting this axis therapeutically in the setting of stroke. While historically, focus has been on suppressing classical RAS signalling through the angiotensin type 1 receptor (AT1R), the identification of a counter-regulatory axis of the RAS signalling via the angiotensin type 2 receptor (AT2R) and Mas receptor has renewed interest in targeting the RAS. This review describes RAS signalling in the brain and the potential of targeting the Mas receptor and AT2R in preclinical models of ischaemic stroke. The animal and experimental models, and the route and timing of intervention, are considered from a translational perspective.
Collapse
Affiliation(s)
- Aisling McFall
- Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Stuart A Nicklin
- Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK
| | - Lorraine M Work
- Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
17
|
Panariello F, Cellini L, Speciani M, De Ronchi D, Atti AR. How Does SARS-CoV-2 Affect the Central Nervous System? A Working Hypothesis. Front Psychiatry 2020; 11:582345. [PMID: 33304284 PMCID: PMC7701095 DOI: 10.3389/fpsyt.2020.582345] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 10/07/2020] [Indexed: 12/15/2022] Open
Abstract
Interstitial pneumonia was the first manifestation to be recognized as caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2); however, in just a few weeks, it became clear that the coronavirus disease-2019 (COVID-19) overrun tissues and more body organs than just the lungs, so much so that it could be considered a systemic pathology. Several studies reported the involvement of the conjunctiva, the gut, the heart and its pace, and vascular injuries such as thromboembolic complications and Kawasaki disease in children and toddlers were also described. More recently, it was reported that in a sample of 214 SARS-CoV-2 positive patients, 36.4% complained of neurological symptoms ranging from non-specific manifestations (dizziness, headache, and seizures), to more specific symptoms such hyposmia or hypogeusia, and stroke. Older individuals, especially males with comorbidities, appear to be at the highest risk of developing such severe complications related to the Central Nervous System (CNS) involvement. Neuropsychiatric manifestations in COVID-19 appear to develop in patients with and without pre-existing neurological disorders. Growing evidence suggests that SARS-CoV-2 binds to the human Angiotensin-Converting Enzyme 2 (ACE2) for the attachment and entrance inside host cells. By describing ACE2 and the whole Renin Angiotensin Aldosterone System (RAAS) we may better understand whether specific cell types may be affected by SARS-CoV-2 and whether their functioning can be disrupted in case of an infection. Since clear evidences of neurological interest have already been shown, by clarifying the topographical distribution and density of ACE2, we will be able to speculate how SARS-CoV-2 may affect the CNS and what is the pathogenetic mechanism by which it contributes to the specific clinical manifestations of the disease. Based on such evidences, we finally hypothesize the process of SARS-CoV-2 invasion of the CNS and provide a possible explanation for the onset or the exacerbation of some common neuropsychiatric disorders in the elderly including cognitive impairment and Alzheimer disease.
Collapse
Affiliation(s)
- Fabio Panariello
- Department of Mental Health, Local Health Authorities, Bologna, Italy
| | - Lorenzo Cellini
- Department of Biomedical and Neuromotor Sciences, Psychiatry, Bologna University, Bologna, Italy
| | - Maurizio Speciani
- Department of Biomedical and Neuromotor Sciences, Psychiatry, Bologna University, Bologna, Italy
| | - Diana De Ronchi
- Department of Biomedical and Neuromotor Sciences, Psychiatry, Bologna University, Bologna, Italy
| | - Anna Rita Atti
- Department of Biomedical and Neuromotor Sciences, Psychiatry, Bologna University, Bologna, Italy
| |
Collapse
|
18
|
Patel SN, Fatima N, Ali R, Hussain T. Emerging Role of Angiotensin AT2 Receptor in Anti-Inflammation: An Update. Curr Pharm Des 2020; 26:492-500. [PMID: 31939729 DOI: 10.2174/1381612826666200115092015] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 11/21/2019] [Indexed: 12/16/2022]
Abstract
The hyperactive RAS and inflammation are closely associated. The angiotensin-II/AT1R axis of the RAS has been explored extensively for its role in inflammation and a plethora of pathological conditions. Understanding the role of AT2R in inflammation is an emerging area of research. The AT2R is expressed on a variety of immune and non-immune cells, which upon activation triggers the release of a host of cytokines and has multiple effects that coalesce to anti-inflammation and prevents maladaptive repair. The anti-inflammatory outcomes of AT2R activation are linked to its well-established signaling pathways involving formation of nitric oxide and activation of phosphatases. Collectively, these effects promote cell survival and tissue function. The consideration of AT2R as a therapeutic target requires further investigations.
Collapse
Affiliation(s)
- Sanket N Patel
- Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, United States
| | - Naureen Fatima
- Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, United States
| | - Riyasat Ali
- Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, United States
| | - Tahir Hussain
- Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, United States
| |
Collapse
|
19
|
Cosarderelioglu C, Nidadavolu LS, George CJ, Oh ES, Bennett DA, Walston JD, Abadir PM. Brain Renin-Angiotensin System at the Intersect of Physical and Cognitive Frailty. Front Neurosci 2020; 14:586314. [PMID: 33117127 PMCID: PMC7561440 DOI: 10.3389/fnins.2020.586314] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 08/25/2020] [Indexed: 12/15/2022] Open
Abstract
The renin–angiotensin system (RAS) was initially considered to be part of the endocrine system regulating water and electrolyte balance, systemic vascular resistance, blood pressure, and cardiovascular homeostasis. It was later discovered that intracrine and local forms of RAS exist in the brain apart from the endocrine RAS. This brain-specific RAS plays essential roles in brain homeostasis by acting mainly through four angiotensin receptor subtypes; AT1R, AT2R, MasR, and AT4R. These receptors have opposing effects; AT1R promotes vasoconstriction, proliferation, inflammation, and oxidative stress while AT2R and MasR counteract the effects of AT1R. AT4R is critical for dopamine and acetylcholine release and mediates learning and memory consolidation. Consequently, aging-associated dysregulation of the angiotensin receptor subtypes may lead to adverse clinical outcomes such as Alzheimer’s disease and frailty via excessive oxidative stress, neuroinflammation, endothelial dysfunction, microglial polarization, and alterations in neurotransmitter secretion. In this article, we review the brain RAS from this standpoint. After discussing the functions of individual brain RAS components and their intracellular and intracranial locations, we focus on the relationships among brain RAS, aging, frailty, and specific neurodegenerative diseases, such as Alzheimer’s disease, Parkinson’s disease, and vascular cognitive impairment, through oxidative stress, neuroinflammation, and vascular dysfunction. Finally, we discuss the effects of RAS-modulating drugs on the brain RAS and their use in novel treatment approaches.
Collapse
Affiliation(s)
- Caglar Cosarderelioglu
- Division of Geriatrics, Department of Internal Medicine, Ankara University School of Medicine, Ankara, Turkey.,Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lolita S Nidadavolu
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Claudene J George
- Division of Geriatrics, Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, United States
| | - Esther S Oh
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, United States
| | - Jeremy D Walston
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Peter M Abadir
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
20
|
Ahmed HA, Ishrat T. The Brain AT2R-a Potential Target for Therapy in Alzheimer's Disease and Vascular Cognitive Impairment: a Comprehensive Review of Clinical and Experimental Therapeutics. Mol Neurobiol 2020; 57:3458-3484. [PMID: 32533467 PMCID: PMC8109287 DOI: 10.1007/s12035-020-01964-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 05/28/2020] [Indexed: 10/24/2022]
Abstract
Dementia is a potentially avertable tragedy, currently considered among the top 10 greatest global health challenges of the twenty-first century. Dementia not only robs individuals of their dignity and independence, it also has a ripple effect that starts with the inflicted individual's family and projects to the society as a whole. The constantly growing number of cases, along with the lack of effective treatments and socioeconomic impact, poses a serious threat to the sustainability of our health care system. Hence, there is a worldwide effort to identify new targets for the treatment of Alzheimer's disease (AD), the leading cause of dementia. Due to its multifactorial etiology and the recent clinical failure of several novel amyloid-β (Aβ) targeting therapies, a comprehensive "multitarget" approach may be most appropriate for managing this condition. Interestingly, renin angiotensin system (RAS) modulators were shown to positively impact all the factors involved in the pathophysiology of dementia including vascular dysfunction, Aβ accumulation, and associated cholinergic deficiency, in addition to tau hyperphosphorylation and insulin derangements. Furthermore, for many of these drugs, the preclinical evidence is also supported by epidemiological data and/or preliminary clinical trials. The purpose of this review is to provide a comprehensive update on the major causes of dementia including the risk factors, current diagnostic criteria, pathophysiology, and contemporary treatment strategies. Moreover, we highlight the angiotensin II receptor type 2 (AT2R) as an effective drug target and present ample evidence supporting its potential role and clinical applications in cognitive impairment to encourage further investigation in the clinical setting.
Collapse
Affiliation(s)
- Heba A Ahmed
- Department of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, 855 Monroe Avenue, Wittenborg Bldg, Room-231, Memphis, TN, 38163, USA
| | - Tauheed Ishrat
- Department of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, 855 Monroe Avenue, Wittenborg Bldg, Room-231, Memphis, TN, 38163, USA.
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA.
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
21
|
Cespedes A, Villa M, Benito-Cuesta I, Perez-Alvarez MJ, Ordoñez L, Wandosell F. Energy-Sensing Pathways in Ischemia: The Counterbalance Between AMPK and mTORC. Curr Pharm Des 2020; 25:4763-4770. [PMID: 31820693 DOI: 10.2174/1381612825666191210152156] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 12/06/2019] [Indexed: 01/02/2023]
Abstract
Stroke is an important cause of death and disability, and it is the second leading cause of death worldwide. In humans, middle cerebral artery occlusion (MCAO) is the most common cause of ischemic stroke. The damage occurs due to the lack of nutrients and oxygen contributed by the blood flow. The present review aims to analyze to what extent the lack of each of the elements of the system leads to damage and which mechanisms are unaffected by this deficiency. We believe that the specific analysis of the effect of lack of each component could lead to the emergence of new therapeutic targets for this important brain pathology.
Collapse
Affiliation(s)
- Angel Cespedes
- Centro de Biología Molecular "Severo Ochoa". CSIC-UAM. Nicolás Cabrera 1, 28049 Madrid, Spain.,Research Group of Neurodegenerative Diseases, Department of Animal Health, Faculty of Veterinary Medicine and Zootechnics - Tolima University, Santa Helena - 730006299, Ibagué, Colombia
| | - Mario Villa
- Centro de Biología Molecular "Severo Ochoa". CSIC-UAM. Nicolás Cabrera 1, 28049 Madrid, Spain.,Departamento de Biología (Fisiología Animal). Facultad de Ciencias. Universidad Autónoma de Madrid. C/Darwin 2. 28049 Madrid, Spain
| | - Irene Benito-Cuesta
- Centro de Biología Molecular "Severo Ochoa". CSIC-UAM. Nicolás Cabrera 1, 28049 Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Maria J Perez-Alvarez
- Centro de Biología Molecular "Severo Ochoa". CSIC-UAM. Nicolás Cabrera 1, 28049 Madrid, Spain.,Departamento de Biología (Fisiología Animal). Facultad de Ciencias. Universidad Autónoma de Madrid. C/Darwin 2. 28049 Madrid, Spain
| | - Lara Ordoñez
- Centro de Biología Molecular "Severo Ochoa". CSIC-UAM. Nicolás Cabrera 1, 28049 Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Francisco Wandosell
- Centro de Biología Molecular "Severo Ochoa". CSIC-UAM. Nicolás Cabrera 1, 28049 Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
22
|
Noureddine FY, Altara R, Fan F, Yabluchanskiy A, Booz GW, Zouein FA. Impact of the Renin-Angiotensin System on the Endothelium in Vascular Dementia: Unresolved Issues and Future Perspectives. Int J Mol Sci 2020; 21:E4268. [PMID: 32560034 PMCID: PMC7349348 DOI: 10.3390/ijms21124268] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 06/12/2020] [Accepted: 06/14/2020] [Indexed: 12/11/2022] Open
Abstract
The effects of the renin-angiotensin system (RAS) surpass the renal and cardiovascular systems to encompass other body tissues and organs, including the brain. Angiotensin II (Ang II), the most potent mediator of RAS in the brain, contributes to vascular dementia via different mechanisms, including neuronal homeostasis disruption, vascular remodeling, and endothelial dysfunction caused by increased inflammation and oxidative stress. Other RAS components of emerging significance at the level of the blood-brain barrier include angiotensin-converting enzyme 2 (ACE2), Ang(1-7), and the AT2, Mas, and AT4 receptors. The various angiotensin hormones perform complex actions on brain endothelial cells and pericytes through specific receptors that have either detrimental or beneficial actions. Increasing evidence indicates that the ACE2/Ang(1-7)/Mas axis constitutes a protective arm of RAS on the blood-brain barrier. This review provides an update of studies assessing the different effects of angiotensins on cerebral endothelial cells. The involved signaling pathways are presented and help highlight the potential pharmacological targets for the management of cognitive and behavioral dysfunctions associated with vascular dementia.
Collapse
Affiliation(s)
- Fatima Y. Noureddine
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon;
| | - Raffaele Altara
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, and KG Jebsen Center for Cardiac Research, 0424 Oslo, Norway;
| | - Fan Fan
- Department of Pharmacology and Toxicology, School of Medicine, The University of Mississippi Medical Center, Jackson, MS 39216, USA; (F.F.); (G.W.B.)
| | - Andriy Yabluchanskiy
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - George W. Booz
- Department of Pharmacology and Toxicology, School of Medicine, The University of Mississippi Medical Center, Jackson, MS 39216, USA; (F.F.); (G.W.B.)
| | - Fouad A. Zouein
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon;
| |
Collapse
|
23
|
Villa González M, Vallés-Saiz L, Hernández IH, Avila J, Hernández F, Pérez-Alvarez MJ. Focal cerebral ischemia induces changes in oligodendrocytic tau isoforms in the damaged area. Glia 2020; 68:2471-2485. [PMID: 32515854 DOI: 10.1002/glia.23865] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 05/15/2020] [Accepted: 05/18/2020] [Indexed: 12/31/2022]
Abstract
Ischemic stroke is a major cause of death and the first leading cause of long-term disability worldwide. The only therapeutic strategy available to date is reperfusion and not all the patients are suitable for this treatment. Blood flow blockage or reduction leads to considerable brain damage, affecting both gray and white matter. The detrimental effects of ischemia have been studied extensively in the former but not in the latter. Previous reports indicate that preservation of white matter integrity reduces deleterious effect of ischemia on the brain. Oligodendrocytes are sensitive to ischemic damage, however, some reports demonstrate that oligodendrogenesis occurs after ischemia. These glial cells have a complex cytoskeletal network, including tau, that plays a key role to proper myelination. 4R-Tau/3R-Tau, which differ in the presence/absence of Exon 10, are found in oligodendrocytes; but the precise role of each isoform is not understood. Using permanent middle cerebral artery occlusion model and immunofluorescence, we demonstrate that cerebral ischemia induces an increase in 3R-Tau versus 4R-Tau in oligodendrocytes in the damaged area. In addition, cellular distribution of Tau undergoes a change after ischemia, with some oligodendrocytic processes showing positive staining for 3R-Tau. This occurs simultaneously with the amelioration of neurological damage in ischemic rats. We propose that ischemia triggers an endogenous mechanism involving 3R-Tau, that induces colonization of the ischemic damaged area by oligodendrocytes in an attempt to myelinate-injured axons. Understanding the molecular mechanism of this phenomenon could pave the way for the design of therapeutic strategies that exploit glial cells for the treatment of ischemia.
Collapse
Affiliation(s)
- Mario Villa González
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Neuropatología Molecular CSIC-UAM, Centro de Biología Molecular "Severo Ochoa", Madrid, Spain
| | - Laura Vallés-Saiz
- Departamento de Neuropatología Molecular CSIC-UAM, Centro de Biología Molecular "Severo Ochoa", Madrid, Spain
| | - Ivó H Hernández
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Neuropatología Molecular CSIC-UAM, Centro de Biología Molecular "Severo Ochoa", Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jesús Avila
- Departamento de Neuropatología Molecular CSIC-UAM, Centro de Biología Molecular "Severo Ochoa", Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Félix Hernández
- Departamento de Neuropatología Molecular CSIC-UAM, Centro de Biología Molecular "Severo Ochoa", Madrid, Spain.,Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - María José Pérez-Alvarez
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain.,Departamento de Neuropatología Molecular CSIC-UAM, Centro de Biología Molecular "Severo Ochoa", Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
24
|
Nozohouri S, Sifat AE, Vaidya B, Abbruscato TJ. Novel approaches for the delivery of therapeutics in ischemic stroke. Drug Discov Today 2020; 25:535-551. [PMID: 31978522 DOI: 10.1016/j.drudis.2020.01.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/20/2019] [Accepted: 01/15/2020] [Indexed: 02/06/2023]
Abstract
Here, we review novel approaches to deliver neuroprotective drugs to salvageable penumbral brain areas of stroke injury with the goals of offsetting ischemic brain injury and enhancing recovery.
Collapse
Affiliation(s)
- Saeideh Nozohouri
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Ali Ehsan Sifat
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Bhuvaneshwar Vaidya
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| | - Thomas J Abbruscato
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| |
Collapse
|
25
|
Guo S, Som AT, Arai K, Lo EH. Effects of angiotensin-II on brain endothelial cell permeability via PPARalpha regulation of para- and trans-cellular pathways. Brain Res 2019; 1722:146353. [PMID: 31356784 PMCID: PMC6755037 DOI: 10.1016/j.brainres.2019.146353] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/24/2019] [Accepted: 07/25/2019] [Indexed: 12/24/2022]
Abstract
Angiotensin-II (Ang-II) is a key factor in hypertension, diabetes and aging, which are all primary risk factors for CNS disease. Furthermore, Ang-II may play under-appreciated roles in neurogenesis, angiogenesis and CNS remodeling. Therefore, any contemplated attempts for neurorestorative therapies in the CNS should consider the context of Ang-II signaling. Here, we investigate how Ang-II may regulate cerebral endothelial permeability, a key functional feature of the neurovascular unit. Exposure of human brain endothelial cell cultures to Ang-II increased its permeability to BSA-Alexa488 tracer. Immunocytochemistry and pulse-chase experiments suggested that both para-cellular as well as trans-cellular pathways were involved. Candesartan but not PD123319 blocked Ang-II permeability effects, suggesting that Ang-II effects may be mediated via type 1 receptor. Immunocytochemistry and western blots showed that Ang-II disrupted the membrane distributions of ZO-1 and VE-Cad, decreased total levels of JAM-A and Mfsd2a, and increased Cav1. These effects of Ang-II were accompanied by dephosphorylation of PPARalpha. Finally, Ang-II-induced increases in endothelial permeability were ameliorated by PPARalpha agonists. Taken together, these studies suggest that Ang-II may disrupt both para- and trans-cellular permeability in cerebral endothelium, and PPARalpha-related pathways may offer potential therapeutic targets for ameliorating these effects in cell-based regenerative medicine.
Collapse
Affiliation(s)
- Shuzhen Guo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, United States.
| | | | | | | |
Collapse
|
26
|
Herr D, Sauer C, Holzheu I, Sauter R, Janni W, Wöckel A, Wulff C. Role of Renin-Angiotensin-System in Human Breast Cancer Cells: Is There a Difference in Regulation of Angiogenesis between Hormone-Receptor Positive and Negative Breast Cancer Cells? Geburtshilfe Frauenheilkd 2019; 79:626-634. [PMID: 31217631 PMCID: PMC6570612 DOI: 10.1055/a-0887-7313] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/02/2019] [Accepted: 04/02/2019] [Indexed: 12/14/2022] Open
Abstract
Objective This study examined the role of the RAS in human breast cancer cells to question if there are differences between HR-positive and HR-negative cells with regard to regulation of VEGF. Methods Expression of different RAS components in hormone receptor (HR)-positive and HR-negative breast cancer cells was investigated using RT-PCR. Different stimulation protocols with different RAS inhibitors were used to investigate the effect on VEGF expression. Angiotensin II-dependent expression of VEGF was quantified by real time PCR. In addition, the effect of intrinsic RAS was studied performing siRNA knockdown of angiotensinogen (AGT). Statistical analysis were calculated using IBM SPSS Statistics Version 21. Results Expression of AT 1 R, AT 2 R, AGT and ACE was shown in HR-positive and HR-negative breast cancer cell lines. Extrinsic stimulation with angiotensin II increased VEGF significantly. After treatment with captopril or AT 1 R-inhibitor candesartan, VEGF-expression decreased significantly in HR-positive and HR-negative cell lines. However, inhibition of AT 2 R using PD 123,319 did not show any significant changes of VEGF. After prevention of intrinsic angiotensin II, extrinsic angiotensin II as well as the combination with inhibitors of the receptors caused a significant reduction of VEGF. Surprisingly, the overall effect of the RAS after knockdown of AGT revealed a significant increase of VEGF in HR-positive cells at any time while a significant decrease was observed in HR-negative cells after 144 hours incubation. Conclusion The RAS-dependent regulation of VEGF between HR-positive and HR-negative breast cancer cells seems do be different. These findings provide evidence for a possible future therapeutic strategy.
Collapse
Affiliation(s)
- Daniel Herr
- Department of Obstetrics and Gynaecology, Würzburg University Medical Centre, Würzburg, Germany
| | - Christof Sauer
- Department of Obstetrics and Gynaecology, Ulm University Medical Centre, Ulm, Germany
| | - Iris Holzheu
- Department of Obstetrics and Gynaecology, Ulm University Medical Centre, Ulm, Germany
| | - Regina Sauter
- Department of Obstetrics and Gynaecology, Ulm University Medical Centre, Ulm, Germany
| | - Wolfgang Janni
- Department of Obstetrics and Gynaecology, Ulm University Medical Centre, Ulm, Germany
| | - Achim Wöckel
- Department of Obstetrics and Gynaecology, Würzburg University Medical Centre, Würzburg, Germany
| | - Christine Wulff
- Department of Obstetrics and Gynaecology, Würzburg University Medical Centre, Würzburg, Germany
| |
Collapse
|
27
|
Jian R, Yang M, Xu F. Lentiviral‐mediated silencing of mast cell‐expressed membrane protein 1 promotes angiogenesis of rats with cerebral ischemic stroke. J Cell Biochem 2019; 120:16786-16797. [PMID: 31104315 DOI: 10.1002/jcb.28937] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/10/2019] [Accepted: 04/18/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Rui Jian
- Rehabilitation Medicine Department Affiliated Hospital of Southwest Medical University Luzhou P.R. China
| | - Min Yang
- Rehabilitation Medicine Department Affiliated Hospital of Southwest Medical University Luzhou P.R. China
| | - Fangyuan Xu
- Rehabilitation Medicine Department Affiliated Hospital of Southwest Medical University Luzhou P.R. China
| |
Collapse
|
28
|
Forrester SJ, Booz GW, Sigmund CD, Coffman TM, Kawai T, Rizzo V, Scalia R, Eguchi S. Angiotensin II Signal Transduction: An Update on Mechanisms of Physiology and Pathophysiology. Physiol Rev 2018; 98:1627-1738. [PMID: 29873596 DOI: 10.1152/physrev.00038.2017] [Citation(s) in RCA: 718] [Impact Index Per Article: 102.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The renin-angiotensin-aldosterone system plays crucial roles in cardiovascular physiology and pathophysiology. However, many of the signaling mechanisms have been unclear. The angiotensin II (ANG II) type 1 receptor (AT1R) is believed to mediate most functions of ANG II in the system. AT1R utilizes various signal transduction cascades causing hypertension, cardiovascular remodeling, and end organ damage. Moreover, functional cross-talk between AT1R signaling pathways and other signaling pathways have been recognized. Accumulating evidence reveals the complexity of ANG II signal transduction in pathophysiology of the vasculature, heart, kidney, and brain, as well as several pathophysiological features, including inflammation, metabolic dysfunction, and aging. In this review, we provide a comprehensive update of the ANG II receptor signaling events and their functional significances for potential translation into therapeutic strategies. AT1R remains central to the system in mediating physiological and pathophysiological functions of ANG II, and participation of specific signaling pathways becomes much clearer. There are still certain limitations and many controversies, and several noteworthy new concepts require further support. However, it is expected that rigorous translational research of the ANG II signaling pathways including those in large animals and humans will contribute to establishing effective new therapies against various diseases.
Collapse
Affiliation(s)
- Steven J Forrester
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - George W Booz
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Curt D Sigmund
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Thomas M Coffman
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| |
Collapse
|
29
|
He Q, Li S, Li L, Hu F, Weng N, Fan X, Kuang S. Total Flavonoids in Caragana (TFC) Promotes Angiogenesis and Enhances Cerebral Perfusion in a Rat Model of Ischemic Stroke. Front Neurosci 2018; 12:635. [PMID: 30258350 PMCID: PMC6143657 DOI: 10.3389/fnins.2018.00635] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 08/23/2018] [Indexed: 02/02/2023] Open
Abstract
Previous studies have demonstrated that total flavonoid extracts from Caragana sinica (TFC) exert multiple therapeutic effects, promote blood flow, and exhibit anti-inflammatory and antioxidant properties. The present study aimed to investigate whether TFC promotes angiogenesis and exerts neuroprotective effects in a rat model of transient middle cerebral artery occlusion (tMCAO). Male Wistar rats were subjected to tMCAO for 1.5 h, followed by 24 h of reperfusion. TFC (15, 30, 60 mg/kg) was administered for 14 days. Evaluations of neurological function were performed following reperfusion, and infarct volumes were assessed in brain slices stained with 2,3,5-triphenyltetrazolium chloride (TTC). Our results indicated that TFC significantly attenuated cerebral infarct volume and neurological deficits following tMCAO. Laser Doppler, micro-PET/CT, and MRI analyses further demonstrated that TFC reduced infarct volume and enhanced cerebral blood flow in a dose-dependent manner, with the most significant effects occurring at a concentration of 60 mg/kg. Significant up-regulation of CD31, VEGF, Ang-1, HIF-1α, delta-like 4 (Dll4), and Notch1 expression was also observed in the experimental groups, relative to that in the vehicle group. In summary, the results of the present study indicate that TFC (15, 30, 60 mg/kg) attenuates neurological deficits, reduces infarct volume, and promotes angiogenesis following MCAO in a concentration-dependent manner, likely via increases in the expression of CD31, VEGF, Ang-1, HIF-1α, Dll4, and Notch1. Further studies are required to determine the clinical usefulness and potential mechanisms of TFC in patients with cerebral focal ischemic stroke.
Collapse
Affiliation(s)
- Qiansong He
- Guiyang College of Traditional Chinese Medicine, Guiyang, China
| | - Shirong Li
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Lailai Li
- Guiyang College of Traditional Chinese Medicine, Guiyang, China
| | - Feiran Hu
- Guiyang College of Traditional Chinese Medicine, Guiyang, China
| | - Ning Weng
- Guiyang College of Traditional Chinese Medicine, Guiyang, China
| | - Xiaodi Fan
- Department of Experimental Research Center, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Shixiang Kuang
- Guiyang College of Traditional Chinese Medicine, Guiyang, China
| |
Collapse
|
30
|
Neuroprotection via AT2 receptor agonists in ischemic stroke. Clin Sci (Lond) 2018; 132:1055-1067. [DOI: 10.1042/cs20171549] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 04/30/2018] [Accepted: 05/01/2018] [Indexed: 12/12/2022]
Abstract
Stroke is a devastating disease that afflicts millions of people each year worldwide. Ischemic stroke, which accounts for ~88% of cases, occurs when blood supply to the brain is decreased, often because of thromboembolism or atherosclerotic occlusion. This deprives the brain of oxygen and nutrients, causing immediate, irreversible necrosis within the core of the ischemic area, but more delayed and potentially reversible neuronal damage in the surrounding brain tissue, the penumbra. The only currently approved therapies for ischemic stroke, the thrombolytic agent recombinant tissue plasminogen activator (rtPA) and the endovascular clot retrieval/destruction processes, are aimed at restoring blood flow to the infarcted area, but are only available for a minority of patients and are not able in most cases to completely restore neurological deficits. Consequently, there remains a need for agents that will protect neurones against death following ischemic stroke. Here, we evaluate angiotensin II (Ang II) type 2 (AT2) receptor agonists as a possible therapeutic target for this disease. We first provide an overview of stroke epidemiology, pathophysiology, and currently approved therapies. We next review the large amount of preclinical evidence, accumulated over the past decade and a half, which indicates that AT2 receptor agonists exert significant neuroprotective effects in various animal models, and discuss the potential mechanisms involved. Finally, after discussing the challenges of delivering blood–brain barrier (BBB) impermeable AT2 receptor agonists to the infarcted areas of the brain, we summarize the evidence for and against the development of these agents as a promising therapeutic strategy for ischemic stroke.
Collapse
|
31
|
Jackson L, Eldahshan W, Fagan SC, Ergul A. Within the Brain: The Renin Angiotensin System. Int J Mol Sci 2018; 19:E876. [PMID: 29543776 PMCID: PMC5877737 DOI: 10.3390/ijms19030876] [Citation(s) in RCA: 224] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 03/10/2018] [Accepted: 03/11/2018] [Indexed: 02/07/2023] Open
Abstract
For many years, modulators of the renin angiotensin system (RAS) have been trusted by clinicians for the control of essential hypertension. It was recently demonstrated that these modulators have other pleiotropic properties independent of their hypotensive effects, such as enhancement of cognition. Within the brain, different components of the RAS have been extensively studied in the context of neuroprotection and cognition. Interestingly, a crosstalk between the RAS and other systems such as cholinergic, dopaminergic and adrenergic systems have been demonstrated. In this review, the preclinical and clinical evidence for the impact of RAS modulators on cognitive impairment of multiple etiologies will be discussed. In addition, the expression and function of different receptor subtypes within the RAS such as: Angiotensin II type I receptor (AT1R), Angiotensin II type II receptor (AT2R), Angiotensin IV receptor (AT4R), Mas receptor (MasR), and Mas-related-G protein-coupled receptor (MrgD), on different cell types within the brain will be presented. We aim to direct the attention of the scientific community to the plethora of evidence on the importance of the RAS on cognition and to the different disease conditions in which these agents can be beneficial.
Collapse
Affiliation(s)
- LaDonya Jackson
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA.
| | - Wael Eldahshan
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA.
| | - Susan C Fagan
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA.
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| | - Adviye Ergul
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA.
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30904, USA.
| |
Collapse
|
32
|
Protective effects of the angiotensin II AT 2 receptor agonist compound 21 in ischemic stroke: a nose-to-brain delivery approach. Clin Sci (Lond) 2018; 132:581-593. [PMID: 29500223 DOI: 10.1042/cs20180100] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 02/28/2018] [Accepted: 03/01/2018] [Indexed: 02/07/2023]
Abstract
Significant neuroprotective effects of angiotensin II type 2 (AT2) receptor (AT2 receptor) agonists in ischemic stroke have been previously demonstrated in multiple studies. However, the routes of agonist application used in these pre-clinical studies, direct intracerebroventricular (ICV) and systemic administration, are unsuitable for translation into humans; in the latter case because AT2 receptor agonists are blood-brain barrier (BBB) impermeable. To circumvent this problem, in the current study we utilized the nose-to-brain (N2B) route of administration to bypass the BBB and deliver the selective AT2 receptor agonist Compound 21 (C21) to naïve rats or rats that had undergone endothelin 1 (ET-1)-induced ischemic stroke. The results obtained from the present study indicated that C21 applied N2B entered the cerebral cortex and striatum within 30 min in amounts that are therapeutically relevant (8.4-9 nM), regardless of whether BBB was intact or disintegrated. C21 was first applied N2B at 1.5 h after stroke indeed provided neuroprotection, as evidenced by a highly significant, 57% reduction in cerebral infarct size and significant improvements in Bederson and Garcia neurological scores. N2B-administered C21 did not affect blood pressure or heart rate. Thus, these data provide proof-of-principle for the idea that N2B application of an AT2 receptor agonist can exert neuroprotective actions when administered following ischemic stroke. Since N2B delivery of other agents has been shown to be effective in certain human central nervous system diseases, the N2B application of AT2 receptor agonists may become a viable mode of delivering these neuroprotective agents for human ischemic stroke patients.
Collapse
|
33
|
Perez-Alvarez MJ, Villa Gonzalez M, Benito-Cuesta I, Wandosell FG. Role of mTORC1 Controlling Proteostasis after Brain Ischemia. Front Neurosci 2018; 12:60. [PMID: 29497356 PMCID: PMC5818460 DOI: 10.3389/fnins.2018.00060] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/24/2018] [Indexed: 01/24/2023] Open
Abstract
Intense efforts are being undertaken to understand the pathophysiological mechanisms triggered after brain ischemia and to develop effective pharmacological treatments. However, the underlying molecular mechanisms are complex and not completely understood. One of the main problems is the fact that the ischemic damage is time-dependent and ranges from negligible to massive, involving different cell types such as neurons, astrocytes, microglia, endothelial cells, and some blood-derived cells (neutrophils, lymphocytes, etc.). Thus, approaching such a complicated cellular response generates a more complex combination of molecular mechanisms, in which cell death, cellular damage, stress and repair are intermixed. For this reason, animal and cellular model systems are needed in order to dissect and clarify which molecular mechanisms have to be promoted and/or blocked. Brain ischemia may be analyzed from two different perspectives: that of oxygen deprivation (hypoxic damage per se) and that of deprivation of glucose/serum factors. For investigations of ischemic stroke, middle cerebral artery occlusion (MCAO) is the preferred in vivo model, and uses two different approaches: transient (tMCAO), where reperfusion is permitted; or permanent (pMCAO). As a complement to this model, many laboratories expose different primary cortical neuron or neuronal cell lines to oxygen-glucose deprivation (OGD). This ex vivo model permits the analysis of the impact of hypoxic damage and the specific response of different cell types implicated in vivo, such as neurons, glia or endothelial cells. Using in vivo and neuronal OGD models, it was recently established that mTORC1 (mammalian Target of Rapamycin Complex-1), a protein complex downstream of PI3K-Akt pathway, is one of the players deregulated after ischemia and OGD. In addition, neuroprotective intervention either by estradiol or by specific AT2R agonists shows an important regulatory role for the mTORC1 activity, for instance regulating vascular endothelial growth factor (VEGF) levels. This evidence highlights the importance of understanding the role of mTORC1 in neuronal death/survival processes, as it could be a potential therapeutic target. This review summarizes the state-of-the-art of the complex kinase mTORC1 focusing in upstream and downstream pathways, their role in central nervous system and their relationship with autophagy, apoptosis and neuroprotection/neurodegeneration after ischemia/hypoxia.
Collapse
Affiliation(s)
- Maria J Perez-Alvarez
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain.,Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain.,Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | - Mario Villa Gonzalez
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain.,Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
| | - Irene Benito-Cuesta
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain.,Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | - Francisco G Wandosell
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain.,Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, Madrid, Spain
| |
Collapse
|
34
|
Chen L, Ren Z, Wei X, Wang S, Wang Y, Cheng Y, Gao H, Liu H. Losartan protects against cerebral ischemia/reperfusion-induced apoptosis through β-arrestin1-mediated phosphorylation of Akt. Eur J Pharmacol 2017; 815:98-108. [DOI: 10.1016/j.ejphar.2017.08.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 08/12/2017] [Accepted: 08/23/2017] [Indexed: 01/31/2023]
|
35
|
Paquin-Veillette J, Lizotte F, Robillard S, Béland R, Breton MA, Guay A, Despatis MA, Geraldes P. Deletion of AT2 Receptor Prevents SHP-1-Induced VEGF Inhibition and Improves Blood Flow Reperfusion in Diabetic Ischemic Hindlimb. Arterioscler Thromb Vasc Biol 2017; 37:2291-2300. [PMID: 29074590 DOI: 10.1161/atvbaha.117.309977] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 10/05/2017] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Ischemia caused by narrowing of femoral artery is a major cause of peripheral arterial disease and morbidity affecting patients with diabetes mellitus. We have previously reported that the inhibition of the angiogenic response to VEGF (vascular endothelial growth factor) in diabetic mice was associated with the increased expression of SHP-1 (SH2 domain-containing phosphatase 1), a protein that can be activated by the AT2 (angiotensin II type 2) receptor. Deletion of AT2 receptor has been shown to promote angiogenesis within the ischemic muscle. However, the relative impact of AT2 receptor in diabetic condition remains unknown. APPROACH AND RESULTS Nondiabetic and diabetic AT2 null (Atgr2-/Y) mice underwent femoral artery ligation after 2 months of diabetes mellitus. Blood perfusion was measured every week ≤4 weeks post-surgery. Expression of the VEGF, SHP-1, and renin-angiotensin pathways was evaluated. Blood flow in the ischemic muscle of diabetic Atgr2-/Y mice recovered faster and ≤80% after 4 weeks compared with 51% recovery in diabetic control littermates. Diabetic Atgr2-/Y had reduced apoptotic endothelial cells and elevated small vessel formation compared with diabetic Atgr2+/Y mice, as well as increased SHP-1 expression and reduced VEGF receptor activity. In endothelial cells, high glucose levels and AT2 agonist treatment did not change SHP-1, VEGF, and VEGF receptor expression. However, the activity of SHP-1 and its association with the VEGF receptors were increased, causing inhibition of the VEGF action in endothelial cell proliferation and migration. CONCLUSIONS Our results suggest that the deletion of AT2 receptor reduced SHP-1 activity and restored VEGF actions, leading to an increased blood flow reperfusion after ischemia in diabetes mellitus.
Collapse
MESH Headings
- Animals
- Blood Glucose/metabolism
- Cattle
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Diabetes Mellitus/genetics
- Diabetes Mellitus/metabolism
- Diabetes Mellitus/physiopathology
- Diabetic Angiopathies/genetics
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/physiopathology
- Disease Models, Animal
- Endothelial Cells/metabolism
- Gene Deletion
- Genotype
- Hindlimb
- Ischemia/genetics
- Ischemia/metabolism
- Ischemia/physiopathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Skeletal/blood supply
- Neovascularization, Physiologic
- Phenotype
- Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism
- Receptor, Angiotensin, Type 2/deficiency
- Receptor, Angiotensin, Type 2/genetics
- Receptors, Vascular Endothelial Growth Factor/metabolism
- Recovery of Function
- Regional Blood Flow
- Renin-Angiotensin System
- Signal Transduction
- Time Factors
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Judith Paquin-Veillette
- From the Research Center of the Centre Hospitalier Universitaire de Sherbrooke (J.P.-V., F.L., S.R., R.B., M.-A.B., A.G., P.G.), Québec, Canada; and Departments of Surgery (M.-A.D.) and Medicine (P.G.), Division of Endocrinology, Université de Sherbrooke, Québec, Canada
| | - Farah Lizotte
- From the Research Center of the Centre Hospitalier Universitaire de Sherbrooke (J.P.-V., F.L., S.R., R.B., M.-A.B., A.G., P.G.), Québec, Canada; and Departments of Surgery (M.-A.D.) and Medicine (P.G.), Division of Endocrinology, Université de Sherbrooke, Québec, Canada
| | - Stéphanie Robillard
- From the Research Center of the Centre Hospitalier Universitaire de Sherbrooke (J.P.-V., F.L., S.R., R.B., M.-A.B., A.G., P.G.), Québec, Canada; and Departments of Surgery (M.-A.D.) and Medicine (P.G.), Division of Endocrinology, Université de Sherbrooke, Québec, Canada
| | - Raphaël Béland
- From the Research Center of the Centre Hospitalier Universitaire de Sherbrooke (J.P.-V., F.L., S.R., R.B., M.-A.B., A.G., P.G.), Québec, Canada; and Departments of Surgery (M.-A.D.) and Medicine (P.G.), Division of Endocrinology, Université de Sherbrooke, Québec, Canada
| | - Marc-André Breton
- From the Research Center of the Centre Hospitalier Universitaire de Sherbrooke (J.P.-V., F.L., S.R., R.B., M.-A.B., A.G., P.G.), Québec, Canada; and Departments of Surgery (M.-A.D.) and Medicine (P.G.), Division of Endocrinology, Université de Sherbrooke, Québec, Canada
| | - Andréanne Guay
- From the Research Center of the Centre Hospitalier Universitaire de Sherbrooke (J.P.-V., F.L., S.R., R.B., M.-A.B., A.G., P.G.), Québec, Canada; and Departments of Surgery (M.-A.D.) and Medicine (P.G.), Division of Endocrinology, Université de Sherbrooke, Québec, Canada
| | - Marc-Antoine Despatis
- From the Research Center of the Centre Hospitalier Universitaire de Sherbrooke (J.P.-V., F.L., S.R., R.B., M.-A.B., A.G., P.G.), Québec, Canada; and Departments of Surgery (M.-A.D.) and Medicine (P.G.), Division of Endocrinology, Université de Sherbrooke, Québec, Canada
| | - Pedro Geraldes
- From the Research Center of the Centre Hospitalier Universitaire de Sherbrooke (J.P.-V., F.L., S.R., R.B., M.-A.B., A.G., P.G.), Québec, Canada; and Departments of Surgery (M.-A.D.) and Medicine (P.G.), Division of Endocrinology, Université de Sherbrooke, Québec, Canada.
| |
Collapse
|
36
|
Bennion DM, Isenberg JD, Harmel AT, DeMars K, Dang AN, Jones CH, Pignataro ME, Graham JT, Steckelings UM, Alexander JC, Febo M, Krause EG, de Kloet AD, Candelario-Jalil E, Sumners C. Post-stroke angiotensin II type 2 receptor activation provides long-term neuroprotection in aged rats. PLoS One 2017; 12:e0180738. [PMID: 28671997 PMCID: PMC5495490 DOI: 10.1371/journal.pone.0180738] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 06/20/2017] [Indexed: 11/18/2022] Open
Abstract
Activation of the angiotensin II type 2 receptor (AT2R) by administration of Compound 21 (C21), a selective AT2R agonist, induces neuroprotection in models of ischemic stroke in young adult animals. The mechanisms of this neuroprotective action are varied, and may include direct and indirect effects of AT2R activation. Our objectives were to assess the long-term protective effects of post-stroke C21 treatments in a clinically-relevant model of stroke in aged rats and to characterize the cellular localization of AT2Rs in the mouse brain of transgenic reporter mice following stroke. Intraperitoneal injections of C21 (0.03mg/kg) after ischemic stroke induced by transient monofilament middle cerebral artery occlusion resulted in protective effects that were sustained for up to at least 3-weeks post-stroke. These included improved neurological function across multiple assessments and a significant reduction in infarct volume as assessed by magnetic resonance imaging. We also found AT2R expression to be on neurons, not astrocytes or microglia, in normal female and male mouse brains. Stroke did not induce altered cellular localization of AT2R when assessed at 7 and 14 days post-stroke. These findings demonstrate that the neuroprotection previously characterized only during earlier time points using stroke models in young animals is sustained long-term in aged rats, implying even greater clinical relevance for the study of AT2R agonists for the acute treatment of ischemic stroke in human disease. Further, it appears that this sustained neuroprotection is likely due to a mix of both direct and indirect effects stemming from selective activation of AT2Rs on neurons or other cells besides astrocytes and microglia.
Collapse
Affiliation(s)
- Douglas M. Bennion
- Department of Physiology and Functional Genomics and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Jacob D. Isenberg
- Department of Physiology and Functional Genomics and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Allison T. Harmel
- Department of Physiology and Functional Genomics and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Kelly DeMars
- Department of Neuroscience and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Alex N. Dang
- Department of Physiology and Functional Genomics and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Chad H. Jones
- Department of Physiology and Functional Genomics and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Megan E. Pignataro
- Department of Physiology and Functional Genomics and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Justin T. Graham
- Department of Physiology and Functional Genomics and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - U. Muscha Steckelings
- Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | - Jon C. Alexander
- Department of Psychiatry and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Marcelo Febo
- Department of Psychiatry and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Eric G. Krause
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, United States of America
| | - Annette D. de Kloet
- Department of Physiology and Functional Genomics and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Eduardo Candelario-Jalil
- Department of Neuroscience and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Colin Sumners
- Department of Physiology and Functional Genomics and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida, United States of America
- * E-mail:
| |
Collapse
|
37
|
Hallberg M, Sumners C, Steckelings UM, Hallberg A. Small-molecule AT2 receptor agonists. Med Res Rev 2017; 38:602-624. [DOI: 10.1002/med.21449] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 05/03/2017] [Accepted: 05/16/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Mathias Hallberg
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, BMC; Uppsala University; P.O. Box 591 SE751 24 Uppsala Sweden
| | - Colin Sumners
- Department of Physiology and Functional Genomics, University of Florida; College of Medicine and McKnight Brain Institute; Gainesville FL 32611
| | - U. Muscha Steckelings
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research; University of Southern Denmark; P.O. Box 5230 Odense Denmark
| | - Anders Hallberg
- Department of Medicinal Chemistry, BMC; Uppsala University; P.O. Box 574 SE-751 23 Uppsala Sweden
| |
Collapse
|
38
|
Arroja MMC, Reid E, McCabe C. Therapeutic potential of the renin angiotensin system in ischaemic stroke. EXPERIMENTAL & TRANSLATIONAL STROKE MEDICINE 2016; 8:8. [PMID: 27761230 PMCID: PMC5054604 DOI: 10.1186/s13231-016-0022-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 09/29/2016] [Indexed: 12/24/2022]
Abstract
The renin angiotensin system (RAS) consists of the systemic hormone system, critically involved in regulation and homeostasis of normal physiological functions [i.e. blood pressure (BP), blood volume regulation], and an independent brain RAS, which is involved in the regulation of many functions such as memory, central control of BP and metabolic functions. In general terms, the RAS consists of two opposing axes; the ‘classical axis’ mediated primarily by Angiotensin II (Ang II), and the ‘alternative axis’ mediated mainly by Angiotensin-(1–7) (Ang-(1–7)). An imbalance of these two opposing axes is thought to exist between genders and is thought to contribute to the pathology of cardiovascular conditions such as hypertension, a stroke co-morbidity. Ischaemic stroke pathophysiology has been shown to be influenced by components of the RAS with specific RAS receptor antagonists and agonists improving outcome in experimental models of stroke. Manipulation of the two opposing axes following acute ischaemic stroke may provide an opportunity for protection of the neurovascular unit, particularly in the presence of pre-existing co-morbidities where the balance may be shifted. In the present review we will give an overview of the experimental stroke studies that have investigated pharmacological interventions of the RAS.
Collapse
Affiliation(s)
- Mariana Moreira Coutinho Arroja
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Garscube Estate, Glasgow, G61 1QH UK
| | - Emma Reid
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Garscube Estate, Glasgow, G61 1QH UK
| | - Christopher McCabe
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Garscube Estate, Glasgow, G61 1QH UK
| |
Collapse
|