1
|
Wong Zhang DE, Gibson Hughes TA, Figueiredo Galvao HB, Lo C, Dinh QN, Zhang SR, Kim HA, Selvaraji S, Clarkson AN, Arumugam TV, Drummond G, Sobey CG, De Silva TM. Post-stroke cognitive impairment and brain hemorrhage are augmented in hypertensive mice. J Cereb Blood Flow Metab 2024; 44:1517-1534. [PMID: 38886874 PMCID: PMC11572097 DOI: 10.1177/0271678x241262127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/19/2024] [Accepted: 05/30/2024] [Indexed: 06/20/2024]
Abstract
Hypertension is a major risk factor for both stroke and cognitive impairment, but it is unclear whether it may specifically affect post-stroke cognitive impairment. We assessed the effect of hypertension and/or stroke on brain injury, cognitive outcome, and the brain transcriptomic profile. C57BL/6J mice (n = 117; 3-5 mo.) received s.c. infusion of either saline or angiotensin II followed by sham surgery or photothrombotic stroke targeting the prefrontal cortex seven days later. Cognitive function was assessed with the Barnes maze and RNA sequencing was used to quantify transcriptomic changes in the brain. Angiotensin II treatment produced spontaneous hemorrhaging after stroke. In the Barnes maze, hypertensive mice that received stroke surgery had an increased escape latency compared to other groups (day 3: hypertensive + stroke = 166.6 ± 6.0 s vs. hypertensive + sham = 122.8 ± 13.8 s vs. normotensive + stroke = 139.9 ± 10.1 s vs. normotensive + sham = 101.9 ± 16.7 s), consistent with impaired cognition. RNA sequencing revealed >1500 differentially expressed genes related to neuroinflammation in hypertensive + stroke vs. normotensive + stroke, which included genes associated with apoptosis, microRNAs, autophagy, anti-cognitive biomarkers and Wnt signaling. Overall, we show that the combination of hypertension and stroke resulted in greater learning impairment and brain injury.
Collapse
Affiliation(s)
- David E Wong Zhang
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Tayla A Gibson Hughes
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Hericka B Figueiredo Galvao
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Cecilia Lo
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Quynh Nhu Dinh
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Shenpeng R Zhang
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Hyun Ah Kim
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Sharmalee Selvaraji
- Department of Physiology, Yong Loo Lin School Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore
| | - Andrew N Clarkson
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Thiruma V Arumugam
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Grant Drummond
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Christopher G Sobey
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - T Michael De Silva
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| |
Collapse
|
2
|
Olasunkanmi OI, Aremu J, Wong ML, Licinio J, Zheng P. Maternal gut-microbiota impacts the influence of intrauterine environmental stressors on the modulation of human cognitive development and behavior. J Psychiatr Res 2024; 180:307-326. [PMID: 39488009 DOI: 10.1016/j.jpsychires.2024.10.028] [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: 07/18/2023] [Revised: 11/01/2023] [Accepted: 10/23/2024] [Indexed: 11/04/2024]
Abstract
This review examines the longstanding debate of nature and intrauterine environmental challenges that shapes human development and behavior, with a special focus on the influence of maternal prenatal gut microbes. Recent research has revealed the critical role of the gut microbiome in human neurodevelopment, and evidence suggest that maternal microbiota can impact fetal gene and microenvironment composition, as well as immunophysiology and neurochemical responses. Furthermore, intrauterine neuroepigenetic regulation may be influenced by maternal microbiota, capable of having long-lasting effects on offspring behavior and cognition. By examining the complex relationship between maternal prenatal gut microbes and human development, this review highlights the importance of early-life environmental factors in shaping neurodevelopment and cognition.
Collapse
Affiliation(s)
- Oluwatayo Israel Olasunkanmi
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education) Chongqing Medical University, Chongqing, China.
| | - John Aremu
- Department of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Ma-Li Wong
- Department of Psychiatry, College of Medicine, Upstate Medical University, Syracuse, NY, USA
| | - Julio Licinio
- Department of Psychiatry, College of Medicine, Upstate Medical University, Syracuse, NY, USA.
| | - Peng Zheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education) Chongqing Medical University, Chongqing, China.
| |
Collapse
|
3
|
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
|
4
|
Gong G, Ganesan K, Wan Y, Liu Y, Huang Y, Luo Y, Wang X, Zhang Z, Zheng Y. Unveiling the neuroprotective properties of isoflavones: current evidence, molecular mechanisms and future perspectives. Crit Rev Food Sci Nutr 2024:1-37. [PMID: 38794836 DOI: 10.1080/10408398.2024.2357701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Neurodegenerative diseases encompass a wide range of debilitating and incurable brain disorders characterized by the progressive deterioration of the nervous system's structure and function. Isoflavones, which are naturally occurring polyphenolic phytochemicals, have been found to regulate various cellular signaling pathways associated with the nervous system. The main objective of this comprehensive review is to explore the neuroprotective effects of isoflavones, elucidate the underlying mechanisms, and assess their potential for treating neurodegenerative disorders. Relevant data regarding isoflavones and their impact on neurodegenerative diseases were gathered from multiple library databases and electronic sources, including PubMed, Google Scholar, Web of Science, and Science Direct. Numerous isoflavones, including genistein, daidzein, biochanin A, and formononetin, have exhibited potent neuroprotective properties against various neurodegenerative diseases. These compounds have been found to modulate neurotransmitters, which in turn contributes to their ability to protect against neurodegeneration. Both in vitro and in vivo experimental studies have provided evidence of their neuroprotection mechanisms, which involve interactions with estrogenic receptors, antioxidant effects, anti-inflammatory properties, anti-apoptotic activity, and modulation of neural plasticity. This review aims to provide current insights into the neuroprotective characteristics of isoflavones and shed light on their potential therapeutic applications in future clinical scenarios.
Collapse
Affiliation(s)
- Guowei Gong
- Department of Bioengineering, Zunyi Medical University, Zhuhai Campus, China
- Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, China
| | - Kumar Ganesan
- School of Chinese Medicine, The Hong Kong University, Hong Kong SAR, China
| | - Yukai Wan
- Second Clinical Medical College of Guangzhou, University of Traditional Chinese Medicine, Guangzhou, China
| | - Yaqun Liu
- Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, China
| | - Yongping Huang
- Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, China
| | - Yuting Luo
- Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, China
| | - Xuexu Wang
- Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, China
| | - Zhenxia Zhang
- Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, China
| | - Yuzhong Zheng
- Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food Engineering, Hanshan Normal University, Chaozhou, China
- Guangdong East Drug and Food and Health Branch, Chaozhou, China
| |
Collapse
|
5
|
Shahror RA, Morris CA, Mohammed AA, Wild M, Zaman B, Mitchell CD, Phillips PH, Rusch NJ, Shosha E, Fouda AY. Role of myeloid cells in ischemic retinopathies: recent advances and unanswered questions. J Neuroinflammation 2024; 21:65. [PMID: 38454477 PMCID: PMC10918977 DOI: 10.1186/s12974-024-03058-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/28/2024] [Indexed: 03/09/2024] Open
Abstract
Myeloid cells including microglia and macrophages play crucial roles in retinal homeostasis by clearing cellular debris and regulating inflammation. These cells are activated in several blinding ischemic retinal diseases including diabetic retinopathy, where they may exert both beneficial and detrimental effects on neurovascular function and angiogenesis. Myeloid cells impact the progression of retinal pathologies and recent studies suggest that targeting myeloid cells is a promising therapeutic strategy to mitigate diabetic retinopathy and other ischemic retinal diseases. This review summarizes the recent advances in our understanding of the role of microglia and macrophages in retinal diseases and focuses on the effects of myeloid cells on neurovascular injury and angiogenesis in ischemic retinopathies. We highlight gaps in knowledge and advocate for a more detailed understanding of the role of myeloid cells in retinal ischemic injury to fully unlock the potential of targeting myeloid cells as a therapeutic strategy for retinal ischemia.
Collapse
Affiliation(s)
- Rami A Shahror
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
| | - Carol A Morris
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
| | - Aya A Mohammed
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
| | - Melissa Wild
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
| | - Bushra Zaman
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
| | - Christian D Mitchell
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
| | - Paul H Phillips
- Department of Ophthalmology, Harvey & Bernice Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Nancy J Rusch
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
| | - Esraa Shosha
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA
- Clinical Pharmacy Department, Cairo University, Cairo, Egypt
| | - Abdelrahman Y Fouda
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences (UAMS), 4301 West Markham Street, Slot 611, BIOMED-1, B306, Little Rock, AR, 72205, USA.
- Clinical Pharmacy Department, Cairo University, Cairo, Egypt.
| |
Collapse
|
6
|
Fatima N, Ali R, Faisal T, Kulkarni K, Patel S, Hussain T. Macrophage angiotensin AT 2 receptor activation is protective against early phases of LPS-induced acute kidney injury. Am J Physiol Renal Physiol 2023; 325:F552-F563. [PMID: 37615049 PMCID: PMC10878726 DOI: 10.1152/ajprenal.00177.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/27/2023] [Accepted: 08/17/2023] [Indexed: 08/25/2023] Open
Abstract
Lipopolysaccharide (LPS)-induced acute kidney injury (AKI) is characterized by inflammation and infiltration of immune cells, mainly neutrophils and macrophages, and results in sudden renal dysfunction. Previously, we have reported the anti-inflammatory and renoprotective role of the angiotensin II type 2 receptor (AT2R), expressed on kidney tubular cells and immune cells, in LPS-induced AKI. Moreover, in vitro studies revealed macrophage AT2R activation shifts the cells to the anti-inflammatory M2 subtype. However, the protective role of the macrophage AT2R in a model of AKI is unknown. The present study addressed this question by adoptive transfer of bone marrow-derived macrophages (BMDMs) in systemic macrophage-depleted mice. We acquired significant systemic macrophage depletion by two doses of liposomal clodronate (CLD), and the mice were repopulated with BMDMs (CD11b+F4/80+, double positive) primed with AT2R agonist C21 (CLD + MacC21 + LPS) or vehicle (CLD + Mac + LPS) in vitro for 60 min, followed by LPS (5 mg/kg body wt ip) challenge. We observed a gradual increase in the CD11b+ cells at 2 and 24 h after the LPS challenge. However, kidney CD11b+ cells in the CLD + Mac + LPS group were elevated compared with the CLD + MacC21 + LPS group at 2 h after the LPS challenge. The level of inflammatory cytokine (tumor necrosis factor-α) was elevated at 2 h, which was reduced significantly in CLD + MacC21 + LPS-treated animals. Also, CLD + MacC21 + LPS-treated animals had elevated plasma and renal IL-10, indicating an anti-inflammatory role of C21-treated BMDMs. Renal functional injury in CLD + MacC21 + LPS-treated animals was partially improved. Collectively, the data demonstrate that BMDM AT2R stimulation results in anti-inflammation and partial renoprotection against early stages of LPS-induced AKI.NEW & NOTEWORTHY Endotoxin such as lipopolysaccharide (LPS) induces acute kidney injury (AKI), which is a risk factor for and often leads to chronic kidney diseases. The present study revealed that bone marrow-derived macrophage activation of the angiotensin II type 2 receptor (AT2R) contributes to the anti-inflammation and partial renoprotection against early stages of LPS-induced AKI. Since AT2R is an emerging anti-inflammatory and organ-protective target, this study advances our understanding of AT2R's anti-inflammatory mechanisms associated with renoprotection.
Collapse
Affiliation(s)
- Naureen Fatima
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, United States
| | - Riyasat Ali
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, United States
| | - Tahmid Faisal
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, United States
| | - Kalyani Kulkarni
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, United States
| | - Sanket Patel
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, United States
| | - Tahir Hussain
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, United States
| |
Collapse
|
7
|
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
|
8
|
Ma K, Zheng ZR, Meng Y. Pathogenesis of Chronic Kidney Disease Is Closely Bound up with Alzheimer's Disease, Especially via the Renin-Angiotensin System. J Clin Med 2023; 12:jcm12041459. [PMID: 36835994 PMCID: PMC9966558 DOI: 10.3390/jcm12041459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/01/2023] [Accepted: 02/07/2023] [Indexed: 02/15/2023] Open
Abstract
Chronic kidney disease (CKD) is a clinical syndrome secondary to the definitive change in function and structure of the kidney, which is characterized by its irreversibility and slow and progressive evolution. Alzheimer's disease (AD) is characterized by the extracellular accumulation of misfolded β-amyloid (Aβ) proteins into senile plaques and the formation of neurofibrillary tangles (NFTs) containing hyperphosphorylated tau. In the aging population, CKD and AD are growing problems. CKD patients are prone to cognitive decline and AD. However, the connection between CKD and AD is still unclear. In this review, we take the lead in showing that the development of the pathophysiology of CKD may also cause or exacerbate AD, especially the renin-angiotensin system (RAS). In vivo studies had already shown that the increased expression of angiotensin-converting enzyme (ACE) produces a positive effect in aggravating AD, but ACE inhibitors (ACEIs) have protective effects against AD. Among the possible association of risk factors in CKD and AD, we mainly discuss the RAS in the systemic circulation and the brain.
Collapse
Affiliation(s)
- Ke Ma
- The First Affiliated Hospital of Jinan University, Guangzhou 510000, China
| | - Zi-Run Zheng
- The First Affiliated Hospital of Jinan University, Guangzhou 510000, China
| | - Yu Meng
- The First Affiliated Hospital of Jinan University, Guangzhou 510000, China
- Central Laboratory, The Fifth Affiliated Hospital of Jinan University, Heyuan 517000, China
- Institute of Nephrology, Jinan University, Guangzhou 510000, China
- Correspondence:
| |
Collapse
|
9
|
Jabber H, Mohammed B, Hadi NR. Investigating the renoprotective effect of C21 in male mice with sepsis via modulation of p-AKT/PI3K expression. J Med Life 2023; 16:203-209. [PMID: 36937479 PMCID: PMC10015579 DOI: 10.25122/jml-2022-0299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/20/2022] [Indexed: 03/21/2023] Open
Abstract
This study aimed to investigate if C21 could prevent acute renal injury induced by sepsis by regulating the expression of p-AKT/PI3K. Five equal groups of 25 adult male Swiss-albino mice were randomly divided (n=5): sham (laparotomy without CLP), CLP, vehicle (equivalent amount of DMSO one hour before CLP), and C21 (0.03 mg/kg, one hour before CLP). ELISA was used to measure serum inflammatory mediators, and the expression of PI3K and P-AKT was determined using PCR and immunohistochemistry (IHC), respectively. TNF, TNF receptor, F8-isoprostane, urea, creatinine, and IL-6 blood levels were considerably lower in the CLP group (p<0.05) compared to the sham group, whereas the C21 treated group had significantly (p<0.05) greater levels of these inflammatory mediators. The IHC analysis revealed that P-AKT expression was significantly lower (p<0.05) in the CLP group compared to the sham group, while the C21 pretreatment group had significantly higher levels of P-AKT expression compared to the CLP group (p<0.05). The PI3K expression in the CLP group was significantly lower than in the sham group (p<0.05), according to PCR results, whereas the PI3K expression in the C21 pretreatment group was significantly greater than in the CLP group (p<0.05). This study showed that C21 might reduce levels of pro-inflammatory cytokines, including TNF-, IL-6, and TNF receptor, by modulating the PI3K/AKT signaling pathways, which can, in turn, reduce renal dysfunction during CLP-induced sepsis in male mice.
Collapse
Affiliation(s)
- Huda Jabber
- Department of Pharmacology and Therapeutics, College of Medicine, University of Al-Qadisiyah, Iraq
| | - Bassim Mohammed
- Department of Pharmacology and Therapeutics, College of Medicine, University of Al-Qadisiyah, Iraq
| | - Najah Rayish Hadi
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Kufa, Kufa, Iraq
- Corresponding Author: Najah Rayish Hadi, Department of Pharmacology & Therapeutics, Faculty of Medicine, University of Kufa, Kufa, Iraq. E-mail:
| |
Collapse
|
10
|
Garrido-Gil P, Pedrosa MA, Garcia-Garrote M, Pequeño-Valtierra A, Rodríguez-Castro J, García-Souto D, Rodríguez-Pérez AI, Labandeira-Garcia JL. Microglial angiotensin type 2 receptors mediate sex-specific expression of inflammatory cytokines independently of circulating estrogen. Glia 2022; 70:2348-2360. [PMID: 35943203 DOI: 10.1002/glia.24255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 07/17/2022] [Accepted: 07/25/2022] [Indexed: 01/07/2023]
Abstract
There are sex differences in microglia, which can maintain sex-related gene expression and functional differences in the absence of circulating sex steroids. The angiotensin type 2 (AT2) receptors mediate anti-inflammatory actions in different tissues, including brain. In mice, we performed RT-PCR analysis of microglia isolated from adult brains and RNA scope in situ hybridization from males, females, ovariectomized females, orchiectomized males and brain masculinized females. We also compared wild type and AT2 knockout mice. The expression of AT2 receptors in microglial cells showed sex differences with much higher AT2 mRNA expression in females than in males, and this was not dependent on circulating gonadal hormones, as observed using ovariectomized females, brain masculinized females and orchiectomized males. These results suggest genomic reasons, possibly related to sex chromosome complement, for sex differences in AT2 expression in microglia, as the AT2 receptor gene is located in the X chromosome. Furthermore, sex differences in expression of AT2 receptors were associated to sex differences in microglial expression of key anti-inflammatory cytokines such as interleukin-10 and pro-inflammatory cytokines such as interleukin-1β and interleukin-6. In conclusion, sex differences in microglial AT2 receptor expression appear as a major factor contributing to sex differences in the neuroinflammatory responses beyond the effects of circulating steroids.
Collapse
Affiliation(s)
- Pablo Garrido-Gil
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CiberNed), Madrid, Spain
| | - Maria A Pedrosa
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CiberNed), Madrid, Spain
| | - Maria Garcia-Garrote
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CiberNed), Madrid, Spain
| | - Ana Pequeño-Valtierra
- Laboratory of Genomes and Disease, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Jorge Rodríguez-Castro
- Laboratory of Genomes and Disease, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Daniel García-Souto
- Laboratory of Genomes and Disease, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ana I Rodríguez-Pérez
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CiberNed), Madrid, Spain
| | - Jose L Labandeira-Garcia
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's disease, Research Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CiberNed), Madrid, Spain
| |
Collapse
|
11
|
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: 36] [Impact Index Per Article: 12.0] [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
|
12
|
Hur HJ, Lee JY, Kim DH, Cho MS, Lee S, Kim HS, Kim DW. Conditioned Medium of Human Pluripotent Stem Cell-Derived Neural Precursor Cells Exerts Neurorestorative Effects against Ischemic Stroke Model. Int J Mol Sci 2022; 23:7787. [PMID: 35887140 PMCID: PMC9319001 DOI: 10.3390/ijms23147787] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/09/2022] [Accepted: 07/12/2022] [Indexed: 02/01/2023] Open
Abstract
Previous studies have shown that early therapeutic events of neural precursor cells (NPCs) transplantation to animals with acute ischemic stroke readily protected neuronal cell damage and improved behavioral recovery through paracrine mechanisms. In this study, we tested the hypothesis that administration of conditioned medium from NPCs (NPC-CMs) could recapitulate the beneficial effects of cell transplantation. Rats with permanent middle cerebral artery occlusion (pMCAO) were randomly assigned to one of the following groups: PBS control, Vehicle (medium) controls, single (NPC-CM(S)) or multiple injections of NPC-CM(NPC-CM(M)) groups. A single intravenous injection of NPC-CM exhibited strong neuroregenerative potential to induce behavioral recovery, and multiple injections enhanced this activity further by suppressing inflammatory damage and inducing endogenous neurogenesis leading to histopathological and functional recovery. Proteome analysis of NPC-CM identified a number of proteins that are known to be associated with nervous system development, neurogenesis, and angiogenesis. In addition, transcriptome analysis revealed the importance of the inflammatory response during stroke recovery and some of the key hub genes in the interaction network were validated. Thus, our findings demonstrated that NPC-CM promoted functional recovery and reduced cerebral infarct and inflammation with enhanced endogenous neurogenesis, and the results highlighted the potency of NPC-CM in stroke therapy.
Collapse
Affiliation(s)
- Hye-Jin Hur
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Korea; (H.-J.H.); (D.-H.K.)
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Ji Yong Lee
- Research Institute of Hyperbaric Medicine and Science, Yonsei University Wonju College of Medicine, Wonju-si 26426, Korea;
| | - Do-Hun Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Korea; (H.-J.H.); (D.-H.K.)
- S. Biomedics Co., Ltd., Seoul 04979, Korea;
| | | | - Sangsik Lee
- Department of Biomedical Engineering, College of Medical Convergence, Catholic Kwandong University, Gangneung-si 25601, Korea;
| | - Han-Soo Kim
- Department of Biomedical Sciences, College of Medical Convergence, Catholic Kwandong University, Gangneung-si 25601, Korea
| | - Dong-Wook Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Korea; (H.-J.H.); (D.-H.K.)
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
13
|
Lansdell TA, Chambers LC, Dorrance AM. Endothelial Cells and the Cerebral Circulation. Compr Physiol 2022; 12:3449-3508. [PMID: 35766836 DOI: 10.1002/cphy.c210015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Endothelial cells form the innermost layer of all blood vessels and are the only vascular component that remains throughout all vascular segments. The cerebral vasculature has several unique properties not found in the peripheral circulation; this requires that the cerebral endothelium be considered as a unique entity. Cerebral endothelial cells perform several functions vital for brain health. The cerebral vasculature is responsible for protecting the brain from external threats carried in the blood. The endothelial cells are central to this requirement as they form the basis of the blood-brain barrier. The endothelium also regulates fibrinolysis, thrombosis, platelet activation, vascular permeability, metabolism, catabolism, inflammation, and white cell trafficking. Endothelial cells regulate the changes in vascular structure caused by angiogenesis and artery remodeling. Further, the endothelium contributes to vascular tone, allowing proper perfusion of the brain which has high energy demands and no energy stores. In this article, we discuss the basic anatomy and physiology of the cerebral endothelium. Where appropriate, we discuss the detrimental effects of high blood pressure on the cerebral endothelium and the contribution of cerebrovascular disease endothelial dysfunction and dementia. © 2022 American Physiological Society. Compr Physiol 12:3449-3508, 2022.
Collapse
Affiliation(s)
- Theresa A Lansdell
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Laura C Chambers
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| |
Collapse
|
14
|
He F, Ye B, Chen J, Li C. Effect of EGCG on inflammatory reaction in rats suffered cerebral ischemia/reperfusion injury. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2021; 46:1325-1331. [PMID: 35232900 PMCID: PMC10930569 DOI: 10.11817/j.issn.1672-7347.2021.210245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Indexed: 06/14/2023]
Abstract
OBJECTIVES Epigallocatechin gallate (EGCG) is the main bioactive component of polyphenols in tea, which has a variety of biological effects. The neurologic injury caused by cerebral ischemia/reperfusion (I/R) is closely related to the inflammatory reaction. The pro-inflammatory factor interleukin-1β (IL-1β) and the anti-inflammatory factor interleukin-10 (IL-10) play important roles in the regulation of inflammatory reaction. This study aims to explore the effect of EGCG on water content, myeloperoxidase (MPO) activity, IL-1β and IL-10 in brain tissues of rats suffered cerebral I/R injury. METHODS Middle cerebral artery occlusion model of SD rats was established by suture embolic method. After ischemia for 1.5 h, the nylon thread was pulled out and reperfusion was performed for 24.0 h. SD rats were randomly divided into 5 groups: a sham group, an I/R group, a EGCG1 group (I/R+12.5 mg/kg EGCG), a EGCG2 (I/R+25.0 mg/kg EGCG), and a EGCG3 group (I/R+50.0 mg/kg EGCG). The sham group was the same as the I/R group except that the middle cerebral artery was not blocked. The water content in brain tissues of rats was measured by dry and wet weight method, and MPO activity was detected by colorimetry. The levels of IL-1β and IL-10 mRNA were determined by RT-PCR, and the contents of IL-1β and IL-10 were determined by ELISA. In addition, primary cultured cortical neurons of SD rats were randomly divided into 3 groups: a control group, an oxygen-glucose deprivation/reperfusion (OGD/R) group, and an OGD/R+EGCG group (OGD/R+50.0 μmol/L EGCG). The effects of EGCG on the levels of IL-1β and IL-10 protein in neurons were assessed by Western blotting. RESULTS Compared with the sham group, water content, MPO activity, the contents of IL-1β and IL-10 were significantly increased (P<0.05 or P<0.01), the mRNA expression of IL-1β and IL-10 were obviously up-regulated (both P<0.01) in cerebral tissues of rats in the I/R group. Compared with the I/R group, water content and MPO activity in cerebral tissues of rats were significantly decreased (both P<0.01), the content of IL-1β (P<0.01) were significantly decreased and IL-10 (P<0.01) were significantly increased in the EGCG3 group. Compared with the I/R group, the mRNA expression of IL-1β was obviously down-regulated and the mRNA expression of IL-10 was obviously up-regulated in the EGCG2 and the EGCG3 group (P<0.05 or P<0.01). Compared with the control group, the protein levels of IL-1β and IL-10 in neurons were significantly increased in the OGD/R group (both P<0.01). Compared with the OGD/R group, the protein expression of IL-1β was obviously down-regulated and the protein expression of IL-10 was obviously up-regulated in neurons in the OGD/R+EGCG group (both P<0.01). CONCLUSIONS EGCG can inhibit the inflammatory reaction induced by cerebral I/R, which may be related to down-regulating the expression of pro-inflammatory factor IL-1β and up-regulating anti-inflammatory factor IL-10.
Collapse
Affiliation(s)
- Fang He
- Department of Basic Medicine, Suzhou Vocational Health College, Suzhou Jiangsu 215009.
| | - Bei Ye
- Department of Basic Medicine, Suzhou Vocational Health College, Suzhou Jiangsu 215009
| | - Jianzhen Chen
- Department of Basic Medicine, Suzhou Vocational Health College, Suzhou Jiangsu 215009
| | - Chang Li
- Department of Thoracic Surgery, First Affiliated Hospital of Soochow University, Suzhou Jiangsu 215006, China
| |
Collapse
|
15
|
Trichosanthis Semen Suppresses Lipopolysaccharide-Induced Neuroinflammation by Regulating the NF-κB Signaling Pathway and HO-1 Expression in Microglia. Toxins (Basel) 2021; 13:toxins13120898. [PMID: 34941735 PMCID: PMC8704237 DOI: 10.3390/toxins13120898] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 12/10/2021] [Indexed: 01/08/2023] Open
Abstract
Neuroinflammation, which is mediated by microglia that release various inflammatory cytokines, is a typical feature of neurodegenerative diseases (NDDs), such as Alzheimer’s disease and Parkinson’s disease. Hence, alleviating neuroinflammation by downregulating pro-inflammatory action, and upregulating anti-inflammatory action of microglia is an efficient therapeutic target for NDDs. In this study, we evaluated whether trichosanthis semen (TS), a dried ripe seed of Trichosanthes kirilowii Maximowicz, reduces lipopolysaccharide (LPS)-induced neuroinflammation by regulating microglial responses in vitro and in vivo. Our results presented that TS reduced the release of pro-inflammatory mediators, such as nitric oxide (NO), inducible NO synthase, tumor necrosis factor-α, interleukin-1β, and interleukin-6 via inhibition of the nuclear factor kappa B (NF-κB) signaling pathway in LPS-treated BV2 microglial cells. Moreover, TS induced anti-inflammatory mediators, such as interleukin-10, found in inflammatory zone 1, and chitinase 3-like 3 by the upregulation of heme oxygenase 1 (HO-1). We further confirmed that TS administration suppressed microglial activation, but enhanced HO-1 expression in LPS-injected mice. These results suggest that TS has anti-neuroinflammatory effects via inhibition of NF-κB signaling through the activation of HO-1, and that TS may be a therapeutical candidate for NDDs treatment.
Collapse
|
16
|
Marzano LAS, de Castro FLM, Machado CA, de Barros JLVM, Macedo E Cordeiro T, Simões E Silva AC, Teixeira AL, Silva de Miranda A. Potential Role of Adult Hippocampal Neurogenesis in Traumatic Brain Injury. Curr Med Chem 2021; 29:3392-3419. [PMID: 34561977 DOI: 10.2174/0929867328666210923143713] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/28/2021] [Accepted: 08/18/2021] [Indexed: 11/22/2022]
Abstract
Traumatic brain injury (TBI) is a serious cause of disability and death among young and adult individuals, displaying complex pathophysiology including cellular and molecular mechanisms that are not fully elucidated. Many experimental and clinical studies investigated the potential relationship between TBI and the process by which neurons are formed in the brain, known as neurogenesis. Currently, there are no available treatments for TBI's long-term consequences being the search for novel therapeutic targets, a goal of highest scientific and clinical priority. Some studies evaluated the benefits of treatments aimed at improving neurogenesis in TBI. In this scenario, herein, we reviewed current pre-clinical studies that evaluated different approaches to improving neurogenesis after TBI while achieving better cognitive outcomes, which may consist in interesting approaches for future treatments.
Collapse
Affiliation(s)
- Lucas Alexandre Santos Marzano
- Laboratório Interdisciplinar de Investigação Médica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Brazil
| | | | - Caroline Amaral Machado
- Laboratório de Neurobiologia, Departamento de Morfologia, Instituto de Ciências Biológicas, UFMG, Brazil
| | | | - Thiago Macedo E Cordeiro
- Laboratório Interdisciplinar de Investigação Médica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Ana Cristina Simões E Silva
- Laboratório Interdisciplinar de Investigação Médica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Antônio Lúcio Teixeira
- Neuropsychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, United States
| | - Aline Silva de Miranda
- Laboratório Interdisciplinar de Investigação Médica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Brazil
| |
Collapse
|
17
|
Ismael S, Ishrat T. Compound 21, a Direct AT2R Agonist, Induces IL-10 and Inhibits Inflammation in Mice Following Traumatic Brain Injury. Neuromolecular Med 2021; 24:274-278. [PMID: 34542832 DOI: 10.1007/s12017-021-08687-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/13/2021] [Indexed: 10/20/2022]
Abstract
Recent studies demonstrated that the angiotensin type 2 receptor (AT2R) agonist, compound 21 (C21), provides neuroprotection and enhances recovery in experimental stroke. However, C21 has never been tested in traumatic brain injury (TBI). Here, we aim to examine whether C21 confers protection after TBI. Unilateral cortical impact injury was induced in young adult C57BL/6 mice. C21 (0.03 mg/kg, i.p.) was administered at 1 h and 3 h post-TBI. After neurological severity score (NSS) assessments, all animals were sacrificed for immunoblotting analysis at 24 h post-TBI. C21 treatment significantly ameliorated NSS and reduced TBI's biomarkers [high mobility group box 1 (HMGB1), aquaporin-4 (AQ4)] and inflammatory markers [interlukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α)] in the pericontusional areas compared to saline TBI. Further, C21 treatment induced interleukin-10 (IL-10) and phosphorylation of endothelial nitric oxide synthase (eNOS) after TBI. C21 also attenuated pro-apoptotic activation of poly (ADP-ribose) polymerase (PARP) and caspase-3. These findings support the therapeutic potential of C21 against TBI.
Collapse
Affiliation(s)
- Saifudeen Ismael
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Tauheed Ishrat
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA. .,Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA. .,Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, 38163, USA. .,Department of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, 875 Monroe Avenue, Wittenborg Bldg, Room-231, Memphis, TN, 38163, USA.
| |
Collapse
|
18
|
Wang Y, Emre C, Gyllenhammar-Schill H, Fjellman K, Eyjolfsdottir H, Eriksdotter M, Schultzberg M, Hjorth E. Cerebrospinal Fluid Inflammatory Markers in Alzheimer's Disease: Influence of Comorbidities. Curr Alzheimer Res 2021; 18:157-170. [PMID: 33784960 DOI: 10.2174/1567205018666210330162207] [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] [Received: 02/10/2021] [Revised: 03/20/2021] [Accepted: 03/26/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) develops into dementia after several years, and subjective cognitive impairment (SCI) and mild cognitive impairment (MCI) are used as intermediary diagnoses of increasing severity. Inflammation is an important part of AD pathology and provides potential novel biomarkers and treatment targets. OBJECTIVE To identify novel potential biomarkers of AD in cerebrospinal fluid (CSF) and create a molecular pattern of inflammatory factors providing differentiation between AD and SCI. METHODS We analyzed 43 inflammatory-related mediators in CSF samples from a cohort of SCI and AD cases vetted for confounding factors (Training cohort). Using multivariate analysis (MVA), a model for discrimination between SCI and AD was produced, which we then applied to a larger nonvetted cohort (named Test cohort). The data were analyzed for factors showing differences between diagnostic groups and factors that differed between the vetted and non-vetted cohorts. The relationship of the factors to the agreement between model and clinical diagnosis was investigated. RESULTS A good MVA model able to discriminate AD from SCI without including tangle and plaque biomarkers was produced from the Training cohort. The model showed 50% agreement with clinical diagnosis in the Test cohort. Comparison of the cohorts indicated different patterns of factors distinguishing SCI from AD. As an example, soluble interleukin (IL)-6Rα showed lower levels in AD cases in the Training cohort, whereas placental growth factor (PlGF) and serum amyloid A (SAA) levels were higher in AD cases of the Test cohort. The levels of p-tau were also higher in the Training cohort. CONCLUSION This study provides new knowledge regarding the involvement of inflammation in AD by indicating different patterns of factors in CSF depending on whether potential confounding comorbidities are present or not, and presents sIL-6Rα as a potential new biomarker for improved diagnosis of AD.
Collapse
Affiliation(s)
- Ying Wang
- Department of Neurobiology, Care Sciences & Society, Karolinska Institutet, Center for Alzheimer Research, BioClinicum J9:20, Division of Neurogeriatrics, Visionsgatan 4, SE-171 64 Solna, Sweden
| | - Ceren Emre
- Department of Neurobiology, Care Sciences & Society, Karolinska Institutet, Center for Alzheimer Research, BioClinicum J9:20, Division of Neurogeriatrics, Visionsgatan 4, SE-171 64 Solna, Sweden
| | | | - Karin Fjellman
- Karolinska University Hospital, Theme Clinical Pharmacology, SE-141 86 Huddinge, Sweden
| | | | - Maria Eriksdotter
- Karolinska University Hospital, Theme Aging, SE-141 86 Huddinge, Sweden
| | - Marianne Schultzberg
- Department of Neurobiology, Care Sciences & Society, Karolinska Institutet, Center for Alzheimer Research, BioClinicum J9:20, Division of Neurogeriatrics, Visionsgatan 4, SE-171 64 Solna, Sweden
| | - Erik Hjorth
- Department of Neurobiology, Care Sciences & Society, Karolinska Institutet, Center for Alzheimer Research, BioClinicum J9:20, Division of Neurogeriatrics, Visionsgatan 4, SE-171 64 Solna, Sweden
| |
Collapse
|
19
|
Ali R, Patel S, Hussain T. Angiotensin type 2 receptor activation limits kidney injury during the early phase and induces Treg cells during the late phase of renal ischemia. Am J Physiol Renal Physiol 2021; 320:F814-F825. [PMID: 33719572 PMCID: PMC8424555 DOI: 10.1152/ajprenal.00507.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 02/12/2021] [Accepted: 03/08/2021] [Indexed: 01/30/2023] Open
Abstract
Kidney infiltrating immune cells such as monocytes, neutrophils, and T cells play critical roles in renal ischemia-reperfusion (IR) injury and repair. Recently, the angiotensin II type 2 receptor (AT2R) has been implicated in protecting kidneys against injury and monocyte infiltration, particularly in chronic kidney disease. However, the role of AT2R in IR injury and repair phases and T cell modulation is unknown. To address this question, Sprague-Dawley rats were subjected to IR with or without AT2R agonist C21 treatment. IR caused early (2 h postreperfusion) renal functional injury (proteinuria, plasma urea, and creatinine) and enhanced immune cells (T cells and CD4 T cells) infiltration and levels of the proinflammatory cytokines monocyte chemoattractant protein-1, TNF-α, and IL-6. C21 treatment reversed these changes but increased the anti-inflammatory IL-10 level. On day 3, C21 treatment increased CD4+FoxP3+ (regulatory T cells) and CD4+IL-10+ cells and reduced kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin in the kidney compared with the IR control, suggesting the involvement of AT2R in kidney repair. These data indicate that AT2R activation protects the kidney against IR injury and immune cell infiltration in the early phase and modulates CD4 T cells toward the regulatory T cell phenotype, which may have long-term beneficial effects on kidney function.NEW & NOTEWORTHY The angiotensin II type 2 receptor agonist C21 has been known to have a renoprotective role in various kidney pathologies. C21 treatment (before renal ischemia) attenuated postischemic kidney injury, kidney dysfunction, and immune cell infiltration during the injury phase. Also, C21 treatment modulated the kidney microenvironment by enhancing anti-inflammatory responses mainly mediated by IL-10. During the repair phase, C21 treatment enhanced IL-10-secreting CD4 T cells and FoxP3-secreting regulatory T cells in Sprague-Dawley rats.
Collapse
MESH Headings
- Acute Kidney Injury/immunology
- Acute Kidney Injury/metabolism
- Acute Kidney Injury/pathology
- Acute Kidney Injury/prevention & control
- Animals
- Anti-Inflammatory Agents/pharmacology
- Chemotaxis, Leukocyte/drug effects
- Cytokines/metabolism
- Disease Models, Animal
- Kidney/drug effects
- Kidney/immunology
- Kidney/metabolism
- Kidney/pathology
- Phenotype
- Rats, Sprague-Dawley
- Receptor, Angiotensin, Type 2/agonists
- Receptor, Angiotensin, Type 2/metabolism
- Reperfusion Injury/immunology
- Reperfusion Injury/metabolism
- Reperfusion Injury/pathology
- Reperfusion Injury/prevention & control
- Signal Transduction
- Sulfonamides/pharmacology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Thiophenes/pharmacology
- Time Factors
- Rats
Collapse
Affiliation(s)
- Riyasat Ali
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| | - Sanket Patel
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| | - Tahir Hussain
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| |
Collapse
|
20
|
Rahman Z, Dandekar MP. Crosstalk between gut microbiome and immunology in the management of ischemic brain injury. J Neuroimmunol 2021; 353:577498. [PMID: 33607506 DOI: 10.1016/j.jneuroim.2021.577498] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 12/30/2020] [Accepted: 01/21/2021] [Indexed: 02/06/2023]
Abstract
Ischemic brain injury is a serious neurological complication, which accrues an immense activation of neuroinflammatory responses. Several lines of research suggested the interconnection of gut microbiota perturbation with the activation of proinflammatory mediators. Intestinal microbial communities also interchange information with the brain through various afferent and efferent channels and microbial by-products. Herein, we discuss the different microelements of gut microbiota and its connection with the host immune system and how change in immune-microbial signatures correlates with the stroke incidence and post-injury neurological sequelae. The activated inflammatory cells increase the production of proinflammatory cytokines, chemokines, proteases and adhesive proteins that are involved in the systemic inflammation, blood brain barrier disruption, gut dysbiosis and aggravation of ischemic brain injury. We suggest that fine-tuning of commensal gut microbiota (eubiosis) may regulate the activation of CNS resident cells like microglial, astrocytes, mast cells and natural killer cells.
Collapse
Affiliation(s)
- Ziaur Rahman
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Manoj P Dandekar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India.
| |
Collapse
|
21
|
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
|
22
|
Correcting the imbalanced protective RAS in COVID-19 with angiotensin AT2-receptor agonists. Clin Sci (Lond) 2020; 134:2987-3006. [PMID: 33210709 DOI: 10.1042/cs20200922] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/22/2020] [Accepted: 11/02/2020] [Indexed: 12/13/2022]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that is responsible for the global corona virus disease 2019 (COVID-19) pandemic enters host cells via a mechanism that includes binding to angiotensin converting enzyme (ACE) 2 (ACE2). Membrane-bound ACE2 is depleted as a result of this entry mechanism. The consequence is that the protective renin-angiotensin system (RAS), of which ACE2 is an essential component, is compromised through lack of production of the protective peptides angiotensin-(1-7) and angiotensin-(1-9), and therefore decreased stimulation of Mas (receptor Mas) and angiotensin AT2-receptors (AT2Rs), while angiotensin AT1-receptors (AT1Rs) are overstimulated due to less degradation of angiotensin II (Ang II) by ACE2. The protective RAS has numerous beneficial actions, including anti-inflammatory, anti-coagulative, anti-fibrotic effects along with endothelial and neural protection; opposite to the deleterious effects caused by heightened stimulation of angiotensin AT1R. Given that patients with severe COVID-19 exhibit an excessive immune response, endothelial dysfunction, increased clotting, thromboses and stroke, enhancing the activity of the protective RAS is likely beneficial. In this article, we discuss the evidence for a dysfunctional protective RAS in COVID and develop a rationale that the protective RAS imbalance in COVID-19 may be corrected by using AT2R agonists. We further review preclinical studies with AT2R agonists which suggest that AT2R stimulation may be therapeutically effective to treat COVID-19-induced disorders of various organ systems such as lung, vasculature, or the brain. Finally, we provide information on the design of a clinical trial in which patients with COVID-19 were treated with the AT2R agonist Compound 21 (C21). This trial has been completed, but results have not yet been reported.
Collapse
|
23
|
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
|
24
|
Shi L, Tian Z, Fu Q, Li H, Zhang L, Tian L, Mi W. miR-217-regulated MEF2D-HDAC5/ND6 signaling pathway participates in the oxidative stress and inflammatory response after cerebral ischemia. Brain Res 2020; 1739:146835. [PMID: 32311345 DOI: 10.1016/j.brainres.2020.146835] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 03/03/2020] [Accepted: 04/13/2020] [Indexed: 12/22/2022]
Abstract
Multiple factors are known to contribute to the pathogenesis of cerebral ischemic injury, including microRNAs (miRNAs). However, the precise mechanism of miRNAs involvement in cerebral ischemia remains largely unclear. In the current study, we found that miR-217 was significantly upregulated in ischemic stroke models, and the upregulation of miR-217 was associated with the development of post-stroke cognitive impairment. Further investigation revealed that myocyte enhancer factor 2D (MEF2D) was the direct target of miR-217. In vitro experiments showed that miR-217 promoted aggregation of histone deacetylase 5 (HDAC5) in cell nuclei by targeting MEF2D, which led to decreased expression of interleukin (IL)-10. In addition, miR-217 inhibited the expression of NADH dehydrogenase subunit 6 (ND6) in a MEF2D-dependent manner. Overexpression of MEF2D can reverse oxygen-glucose deprivation (OGD)-induced downregulation of ND6 and OGD-mediated neuronal apoptosis, and also reduce the elevated generation of reactive oxygen species (ROS) induced by OGD. Additionally, we found that in vivo administration of MEF2D overexpression plasmids increased IL-10 production and ameliorated cognitive impairment after cerebral ischemia. Taken together, these findings reveal a novel pathogenetic mechganism of cerebral ischemia-related brain injury involving the miR-217/MEF2D/HDAC5 axis and the miR-217/MEF2D/ND6 axis.
Collapse
Affiliation(s)
- Likai Shi
- Department of Anesthesiology, The First Medical Center of the Chinese People's Liberation Army (PLA) General Hospital, No. 28 Fuxing Road, Beijing 100853, China
| | - Zhenpu Tian
- Department of Anesthesiology, Hainan Hospital of the Chinese People's Liberation Army (PLA) General Hospital, Jianglin Road, Haitang District, Sanya, Hainan 572013, China
| | - Qiang Fu
- Department of Anesthesiology, The First Medical Center of the Chinese People's Liberation Army (PLA) General Hospital, No. 28 Fuxing Road, Beijing 100853, China
| | - Hao Li
- Department of Anesthesiology, The First Medical Center of the Chinese People's Liberation Army (PLA) General Hospital, No. 28 Fuxing Road, Beijing 100853, China
| | - Lifeng Zhang
- Department of Anesthesiology, Hainan Hospital of the Chinese People's Liberation Army (PLA) General Hospital, Jianglin Road, Haitang District, Sanya, Hainan 572013, China
| | - Li Tian
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, Tongji University, No. 1878 Sichuanbei Road, Shanghai 200081,China.
| | - Weidong Mi
- Department of Anesthesiology, The First Medical Center of the Chinese People's Liberation Army (PLA) General Hospital, No. 28 Fuxing Road, Beijing 100853, China.
| |
Collapse
|
25
|
Abstract
The active hormone of the renin-angiotensin system (RAS), angiotensin II (Ang II), is involved in several human diseases, driving the development and clinical use of several therapeutic drugs, mostly angiotensin I converting enzyme (ACE) inhibitors and angiotensin receptor type I (AT1R) antagonists. However, angiotensin peptides can also bind to receptors different from AT1R, in particular, angiotensin receptor type II (AT2R), resulting in biological and physiological effects different, and sometimes antagonistic, of their binding to AT1R. In the present Perspective, the components of the RAS and the therapeutic tools developed to control it will be reviewed. In particular, the characteristics of AT2R and tools to modulate its functions will be discussed. Agonists or antagonists to AT2R are potential therapeutics in cardiovascular diseases, for agonists, and in the control of pain, for antagonists, respectively. However, controlling their binding properties and their targeting to the target tissues must be optimized.
Collapse
Affiliation(s)
- Lucienne Juillerat-Jeanneret
- Transplantation Center, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Chemin des Boveresses 155, CH1011 Lausanne, Switzerland
| |
Collapse
|
26
|
Chávez-Castillo M, Nava M, Ortega Á, Rojas M, Núñez V, Salazar J, Bermúdez V, Rojas-Quintero J. Depression as an Immunometabolic Disorder: Exploring Shared Pharmacotherapeutics with Cardiovascular Disease. Curr Neuropharmacol 2020; 18:1138-1153. [PMID: 32282306 PMCID: PMC7709154 DOI: 10.2174/1570159x18666200413144401] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/04/2020] [Accepted: 04/16/2020] [Indexed: 12/12/2022] Open
Abstract
Modern times have seen depression and cardiovascular disease (CVD) become notorious public health concerns, corresponding to alarming proportions of morbidity, mortality, decreased quality of life, and economic costs. Expanding comprehension of the pathogenesis of depression as an immunometabolic disorder has identified numerous pathophysiologic phenomena in common with CVD, including chronic inflammation, insulin resistance, and oxidative stress. These shared components could be exploited to offer improved alternatives in the joint management of these conditions. Abundant preclinical and clinical data on the impact of established treatments for CVD in the management of depression have allowed for potential candidates to be proposed for the joint management of depression and CVD as immunometabolic disorders. However, a large proportion of the clinical investigation currently available exhibits marked methodological flaws which preclude the formulation of concrete recommendations in many cases. This situation may be a reflection of pervasive problems present in clinical research in psychiatry, especially pertaining to study homogeneity. Therefore, further high-quality research is essential in the future in this regard.
Collapse
Affiliation(s)
| | | | | | | | | | - Juan Salazar
- Address correspondence to this author at the Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 20th Avenue 4004, Venezuela; Tel/Fax: ++582617597279; E-mail:
| | | | | |
Collapse
|
27
|
Jackson L, Dong G, Althomali W, Sayed MA, Eldahshan W, Baban B, Johnson MH, Filosa J, Fagan SC, Ergul A. Delayed Administration of Angiotensin II Type 2 Receptor (AT2R) Agonist Compound 21 Prevents the Development of Post-stroke Cognitive Impairment in Diabetes Through the Modulation of Microglia Polarization. Transl Stroke Res 2019; 11:762-775. [PMID: 31792796 DOI: 10.1007/s12975-019-00752-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 10/11/2019] [Accepted: 10/30/2019] [Indexed: 12/18/2022]
Abstract
A disabling consequence of stroke is cognitive impairment, occurring in 12%-48% of patients, for which there is no therapy. A critical barrier is the lack of understanding of how post-stroke cognitive impairment (PSCI) develops. While 70% of stroke victims present with comorbid diseases such as diabetes and hypertension, the limited use of comorbid disease models in preclinical research further contributes to this lack of progress. To this end, we used a translational model of diabetes to study the development of PSCI. In addition, we evaluated the application of compound 21 (C21), an angiotensin II Type 2 receptor agonist, for the treatment of PSCI by blinding the treatment assignment, setting strict inclusion criteria, and implementing a delayed administration time point. Diabetes was induced by a high-fat diet (HFD) and low-dose streptozotocin (STZ) combination. Control and diabetic rats were subjected to 1 h middle cerebral artery occlusion (MCAO) or sham surgery. Adhesive removal task (ART) and two-trial Y-maze were utilized to test sensorimotor and cognitive function. Three days post-stroke, rats that met the inclusion criteria were administered C21 or vehicle in drinking water at a dose of 0.12 mg/kg/day for 8 weeks. Samples from freshly harvested brains were analyzed by flow cytometry and immunohistochemistry (IHC). Diabetes exacerbated the development of PSCI and increased inflammation and demyelination. Delayed administration of C21 3 days post-stroke reduced mortality and improved sensorimotor and cognitive deficits. It also reduced inflammation and demyelination through modulation of the M1:M2 ratio in the diabetic animals.
Collapse
Affiliation(s)
- Ladonya Jackson
- Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, USA.,Charlie Norwood Veterans Affairs Medical Center, Augusta, USA.,Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Guangkuo Dong
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Waleed Althomali
- Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, USA.,Charlie Norwood Veterans Affairs Medical Center, Augusta, USA
| | - Mohammed A Sayed
- Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, USA.,Charlie Norwood Veterans Affairs Medical Center, Augusta, USA
| | - Wael Eldahshan
- Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, USA.,Charlie Norwood Veterans Affairs Medical Center, Augusta, USA.,Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Babak Baban
- Oral Biology, Dental College of Georgia, Augusta, GA, USA
| | - Maribeth H Johnson
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Jessica Filosa
- Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Susan C Fagan
- Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, USA.,Charlie Norwood Veterans Affairs Medical Center, Augusta, USA
| | - Adviye Ergul
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 171 Ashley Ave. MSC, Charleston, SC, 908, USA. .,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, USA.
| |
Collapse
|
28
|
Garcia-Garrote M, Perez-Villalba A, Garrido-Gil P, Belenguer G, Parga JA, Perez-Sanchez F, Labandeira-Garcia JL, Fariñas I, Rodriguez-Pallares J. Interaction between Angiotensin Type 1, Type 2, and Mas Receptors to Regulate Adult Neurogenesis in the Brain Ventricular-Subventricular Zone. Cells 2019; 8:E1551. [PMID: 31801296 PMCID: PMC6952803 DOI: 10.3390/cells8121551] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/21/2019] [Accepted: 11/28/2019] [Indexed: 12/30/2022] Open
Abstract
The renin-angiotensin system (RAS), and particularly its angiotensin type-2 receptors (AT2), have been classically involved in processes of cell proliferation and maturation during development. However, the potential role of RAS in adult neurogenesis in the ventricular-subventricular zone (V-SVZ) and its aging-related alterations have not been investigated. In the present study, we analyzed the role of major RAS receptors on neurogenesis in the V-SVZ of adult mice and rats. In mice, we showed that the increase in proliferation of cells in this neurogenic niche was induced by activation of AT2 receptors but depended partially on the AT2-dependent antagonism of AT1 receptor expression, which restricted proliferation. Furthermore, we observed a functional dependence of AT2 receptor actions on Mas receptors. In rats, where the levels of the AT1 relative to those of AT2 receptor are much lower, pharmacological inhibition of the AT1 receptor alone was sufficient in increasing AT2 receptor levels and proliferation in the V-SVZ. Our data revealed that interactions between RAS receptors play a major role in the regulation of V-SVZ neurogenesis, particularly in proliferation, generation of neuroblasts, and migration to the olfactory bulb, both in young and aged brains, and suggest potential beneficial effects of RAS modulators on neurogenesis.
Collapse
MESH Headings
- Age Factors
- Angiotensin II/metabolism
- Animals
- Immunohistochemistry
- Lateral Ventricles/metabolism
- Male
- Mice
- Mice, Knockout
- Models, Biological
- Neural Stem Cells/metabolism
- Neurogenesis/genetics
- Protein Binding
- Rats
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 2/genetics
- Receptor, Angiotensin, Type 2/metabolism
Collapse
Affiliation(s)
- Maria Garcia-Garrote
- Laboratorio de Neurobiología Celular y Molecular de la Enfermedad de Parkinson, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Dpto. Ciencias Morfolóxicas, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Ana Perez-Villalba
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Faculty of Psychology, Universidad Católica de Valencia, Valencia, 46100 Burjassot, Spain
| | - Pablo Garrido-Gil
- Laboratorio de Neurobiología Celular y Molecular de la Enfermedad de Parkinson, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Dpto. Ciencias Morfolóxicas, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - German Belenguer
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Departamento de Biología Celular, Biología Funcional y Antropología Física and Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain
| | - Juan A Parga
- Laboratorio de Neurobiología Celular y Molecular de la Enfermedad de Parkinson, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Dpto. Ciencias Morfolóxicas, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Francisco Perez-Sanchez
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Departamento de Biología Celular, Biología Funcional y Antropología Física and Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain
| | - Jose Luis Labandeira-Garcia
- Laboratorio de Neurobiología Celular y Molecular de la Enfermedad de Parkinson, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Dpto. Ciencias Morfolóxicas, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Isabel Fariñas
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Departamento de Biología Celular, Biología Funcional y Antropología Física and Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain
| | - Jannette Rodriguez-Pallares
- Laboratorio de Neurobiología Celular y Molecular de la Enfermedad de Parkinson, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Dpto. Ciencias Morfolóxicas, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| |
Collapse
|
29
|
Chow BSM, Kocan M, Shen M, Wang Y, Han L, Chew JY, Wang C, Bosnyak S, Mirabito-Colafella KM, Barsha G, Wigg B, Johnstone EKM, Hossain MA, Pfleger KDG, Denton KM, Widdop RE, Summers RJ, Bathgate RAD, Hewitson TD, Samuel CS. AT1R-AT2R-RXFP1 Functional Crosstalk in Myofibroblasts: Impact on the Therapeutic Targeting of Renal and Cardiac Fibrosis. J Am Soc Nephrol 2019; 30:2191-2207. [PMID: 31511361 DOI: 10.1681/asn.2019060597] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 07/29/2019] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Recombinant human relaxin-2 (serelaxin), which has organ-protective actions mediated via its cognate G protein-coupled receptor relaxin family peptide receptor 1 (RXFP1), has emerged as a potential agent to treat fibrosis. Studies have shown that serelaxin requires the angiotensin II (AngII) type 2 receptor (AT2R) to ameliorate renal fibrogenesis in vitro and in vivo. Whether its antifibrotic actions are affected by modulation of the AngII type 1 receptor (AT1R), which is expressed on myofibroblasts along with RXFP1 and AT2R, is unknown. METHODS We examined the signal transduction mechanisms of serelaxin when applied to primary rat renal and human cardiac myofibroblasts in vitro, and in three models of renal- or cardiomyopathy-induced fibrosis in vivo. RESULTS The AT1R blockers irbesartan and candesartan abrogated antifibrotic signal transduction of serelaxin via RXFP1 in vitro and in vivo. Candesartan also ameliorated serelaxin's antifibrotic actions in the left ventricle of mice with cardiomyopathy, indicating that candesartan's inhibitory effects were not confined to the kidney. We also demonstrated in a transfected cell system that serelaxin did not directly bind to AT1Rs but that constitutive AT1R-RXFP1 interactions could form. To potentially explain these findings, we also demonstrated that renal and cardiac myofibroblasts expressed all three receptors and that antagonists acting at each receptor directly or allosterically blocked the antifibrotic effects of either serelaxin or an AT2R agonist (compound 21). CONCLUSIONS These findings have significant implications for the concomitant use of RXFP1 or AT2R agonists with AT1R blockers, and suggest that functional interactions between the three receptors on myofibroblasts may represent new targets for controlling fibrosis progression.
Collapse
Affiliation(s)
- Bryna S M Chow
- Florey Institute of Neuroscience and Mental Health.,Department of Biochemistry and Molecular Biology, and
| | - Martina Kocan
- Florey Institute of Neuroscience and Mental Health.,Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| | - Matthew Shen
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Yan Wang
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Lei Han
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Jacqueline Y Chew
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Chao Wang
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Sanja Bosnyak
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia.,Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Katrina M Mirabito-Colafella
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Giannie Barsha
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Belinda Wigg
- Department of Nephrology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Elizabeth K M Johnstone
- Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia, Australia
| | | | - Kevin D G Pfleger
- Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia, Australia.,Department of Pharmacology and Therapeutics, ARC Centre for Personalised Therapeutic Technologies, Melbourne, Australia; and.,Dimerix Limited, Nedlands, Western Australia, Australia
| | - Kate M Denton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Robert E Widdop
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Roger J Summers
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia.,Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Ross A D Bathgate
- Florey Institute of Neuroscience and Mental Health.,Department of Biochemistry and Molecular Biology, and
| | - Tim D Hewitson
- Department of Nephrology, Royal Melbourne Hospital, Parkville, Victoria, Australia.,Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Chrishan S Samuel
- Department of Biochemistry and Molecular Biology, and .,Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| |
Collapse
|
30
|
Ishrat T, Fouda AY, Pillai B, Eldahshan W, Ahmed H, Waller JL, Ergul A, Fagan SC. Dose-response, therapeutic time-window and tPA-combinatorial efficacy of compound 21: A randomized, blinded preclinical trial in a rat model of thromboembolic stroke. J Cereb Blood Flow Metab 2019; 39. [PMID: 29537907 PMCID: PMC6681526 DOI: 10.1177/0271678x18764773] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The aim of this translational, randomized, controlled, blinded preclinical trial was to determine the effect of compound 21 (C21) in embolic stroke. Rats were subjected to embolic-middle cerebral artery occlusion (eMCAO). They received C21 (0.01, 0.03 and 0.06 mg/kg/d) or saline (orally) for five days, with the first-dose given IV at 3 h post-eMCAO. For the time-window study, the optimal-dose of C21 was initiated at 3, 6 or 24 h post-eMCAO and continued for five days. For the combinatorial study, animals received IV-tissue plasminogen activator (tPA) at either 2 or 4 h, with IV-C21 (0.01 mg/kg) or saline at 3 h post-eMCAO and daily thereafter for five days. After performing the behavior tests, brains were collected for analyses. The dose-response study showed significant motor improvements with the lowest-dose (0.01 mg/kg) of C21. In the time-window study, this same dose resulted in improvements when given 6 h and 24 h post-eMCAO. Moreover, C21-treated animals performed better on the novel object recognition test. Neither the single treatment with C21 or tPA (4 h) nor the combination therapy was effective in reducing the hemorrhage or infarct size, although C21 alone lowered sensorimotor deficit scores post-eMCAO. Future studies should focus on the long-term cognitive benefits of C21, rather than acute neuroprotection.
Collapse
Affiliation(s)
- Tauheed Ishrat
- 1 Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis TN, USA
| | - Abdelrahman Y Fouda
- 2 Charlie Norwood VA Medical Center, and Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Athens, GA, USA
| | - Bindu Pillai
- 2 Charlie Norwood VA Medical Center, and Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Athens, GA, USA
| | - Wael Eldahshan
- 2 Charlie Norwood VA Medical Center, and Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Athens, GA, USA
| | - Heba Ahmed
- 2 Charlie Norwood VA Medical Center, and Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Athens, GA, USA
| | - Jennifer L Waller
- 3 Department of Biostatistics and Epidemiology, Augusta University, Augusta, GA, USA
| | - Adviye Ergul
- 2 Charlie Norwood VA Medical Center, and Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Athens, GA, USA.,4 Department of Physiology, Augusta University, Augusta, GA, USA
| | - Susan C Fagan
- 2 Charlie Norwood VA Medical Center, and Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Athens, GA, USA.,5 Department of Neurology, Augusta University, Augusta, GA, USA
| |
Collapse
|
31
|
Khorooshi R, Tofte-Hansen EU, Tygesen C, Montanana-Rosell R, Limburg HL, Marczynska J, Asgari N, Steckelings UM, Owens T. Angiotensin AT2 receptor–induced interleukin-10 attenuates neuromyelitis optica spectrum disorder–like pathology. Mult Scler 2019; 26:1187-1196. [DOI: 10.1177/1352458519860327] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background: Neuromyelitis optica spectrum disorder (NMOSD) is a relapsing inflammatory central nervous system (CNS) disease for which there is no cure. Immunoglobulin G autoantibodies specific for the water channel aquaporin-4 are a serum biomarker, believed to induce complement-dependent astrocyte damage with secondary demyelination. Objective: To investigate the effect of angiotensin AT2 receptor (AT2R) stimulation on NMOSD-like pathology and its underlying mechanism. Methods: NMOSD-like pathology was induced in mice by intracerebral injection of immunoglobulin-G isolated from NMOSD patient serum, with complement. This mouse model produces the characteristic histological features of NMOSD. A specific AT2R agonist, Compound 21 (C21), was given intracerebrally at day 0 and by intrathecal injection at day 2. Results: Loss of aquaporin-4 and glial fibrillary acidic protein was attenuated by treatment with C21. Administration of C21 induced mRNA for interleukin-10 in the brain. NMOSD-like pathology was exacerbated in interleukin-10-deficient mice, suggesting a protective role. C21 treatment did not attenuate NMOSD-like pathology in interleukin-10-deficient mice, indicating that the protective effect of AT2R stimulation was dependent on interleukin-10. Conclusion: Our findings identify AT2R as a novel potential therapeutic target for the treatment of NMOSD. Interleukin-10 signaling is an essential part of the protective mechanism counteracting NMOSD pathology.
Collapse
Affiliation(s)
- Reza Khorooshi
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Emil Ulrikkaholm Tofte-Hansen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Camilla Tygesen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Roser Montanana-Rosell
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Hannah Liska Limburg
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Joanna Marczynska
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Nasrin Asgari
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark/ Department of Neurology, Slagelse Hospital, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Ulrike Muscha Steckelings
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Trevor Owens
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
32
|
Luo Y, Tang H, Li H, Zhao R, Huang Q, Liu J. Recent advances in the development of neuroprotective agents and therapeutic targets in the treatment of cerebral ischemia. Eur J Med Chem 2019; 162:132-146. [DOI: 10.1016/j.ejmech.2018.11.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/30/2018] [Accepted: 11/06/2018] [Indexed: 11/25/2022]
|
33
|
Mattina GF, Van Lieshout RJ, Steiner M. Inflammation, depression and cardiovascular disease in women: the role of the immune system across critical reproductive events. Ther Adv Cardiovasc Dis 2019; 13:1753944719851950. [PMID: 31144599 PMCID: PMC6545651 DOI: 10.1177/1753944719851950] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 04/29/2019] [Indexed: 12/18/2022] Open
Abstract
Women are at increased risk for developing depression and cardiovascular disease (CVD) across the lifespan and their comorbidity is associated with adverse outcomes that contribute significantly to rates of morbidity and mortality in women worldwide. Immune-system activity has been implicated in the etiology of both depression and CVD, but it is unclear how inflammation contributes to sex differences in this comorbidity. This narrative review provides an updated synthesis of research examining the association of inflammation with depression and CVD, and their comorbidity in women. Recent research provides evidence of pro-inflammatory states and sex differences associated with alterations in the hypothalamic-pituitary-adrenal axis, the renin-angiotensin-aldosterone system and the serotonin/kynurenine pathway, that likely contribute to the development of depression and CVD. Changes to inflammatory cytokines in relation to reproductive periods of hormonal fluctuation (i.e. the menstrual cycle, perinatal period and menopause) are highlighted and provide a greater understanding of the unique vulnerability women experience in developing both depressed mood and adverse cardiovascular events. Inflammatory biomarkers hold substantial promise when combined with a patient's reproductive and mental health history to aid in the prediction, identification and treatment of the women most at risk for CVD and depression. However, more research is needed to improve our understanding of the mechanisms underlying inflammation in relation to their comorbidity, and how these findings can be translated to improve women's health.
Collapse
Affiliation(s)
- Gabriella F. Mattina
- Neuroscience Graduate Program, McMaster University, 1280 Main Street West, ON L8S 4L8, Canada
| | - Ryan J. Van Lieshout
- Neuroscience Graduate Program, McMaster University, ON, Canada
- Department of Psychiatry and Behavioral Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Meir Steiner
- Women’s Health Concerns Clinic, St. Joseph’s Healthcare, Hamilton, ON, Canada
- Department of Psychiatry and Behavioral Neurosciences, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
34
|
Activation of PD-1 Protects Intestinal Immune Defense Through IL-10/miR-155 Pathway After Intestinal Ischemia Reperfusion. Dig Dis Sci 2018; 63:3307-3316. [PMID: 30206757 DOI: 10.1007/s10620-018-5282-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 09/08/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND To date, mechanisms of intestinal immunoglobulin (Ig) dysfunction following intestinal ischemia/reperfusion (I/R) remain unclear. Programmed death 1 (PD-1) is associated with immune responses of lymphocytes. AIM We aimed to verify the hypothesis that activation of PD-1 may improve intestinal immune dysfunction by regulating IL-10/miR-155 production after intestinal IR injury. METHODS Intestinal I/R injury was induced in mice by clamping the superior mesenteric artery for 1 h followed by 2-h reperfusion. PD-L1 fusion Ig, anti-interleukin (IL)-10 monoclonal antibody (mAb), and microRNA (miR)-155 agomir were administered. PD-1 expression, IL-10 mRNA, and protein expression in Peyer's patches (PP) CD4+ cells were measured. MiR-155 levels, tumor necrosis factor (TNF)-α and IL-1β concentration, and activation-induced cytidine deaminase (AID), a key enzyme for intestinal immune antibodies, in PP tissues were measured, respectively. Importantly, the production and cecal bacteria-binding capacity of IgA and IgM were detected. RESULTS Intestinal I/R led to decreased PD-1 expression, imbalanced production, and impaired bacteria-binding capacity of IgA and IgM. Activating PD-1 by PD-L1 Ig facilitated IL-10 synthesis, then decreased miR-155 levels, and subsequently promoted AID expression and reduced TNF-α, IL-1β concentration. Upregulation of AID improved the disruptions of intestinal immune barrier caused by IgA and IgM dysfunction. Anti-IL-10 mAb and miR-155 agomir abolished the protective effects of PD-L1 Ig on the intestinal immune defense. CONCLUSION Activation of PD-1 with PD-L1 Ig relieves intestinal immune defensive injury through IL-10/miR-155 pathway following intestinal I/R attack. PD-1, IL-10, and miR-155 may be potential targets for the damages of intestinal barrier and immunity.
Collapse
|
35
|
Li Z, Zeng G, Zheng X, Wang W, Ling Y, Tang H, Zhang J. Neuroprotective effect of formononetin against TBI in rats via suppressing inflammatory reaction in cortical neurons. Biomed Pharmacother 2018; 106:349-354. [DOI: 10.1016/j.biopha.2018.06.041] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 06/06/2018] [Accepted: 06/12/2018] [Indexed: 12/11/2022] Open
|
36
|
Ye L, Gao L, Cheng H. Inflammatory Profiles of the Interleukin Family and Network in Cerebral Hemorrhage. Cell Mol Neurobiol 2018; 38:1321-1333. [PMID: 30027390 DOI: 10.1007/s10571-018-0601-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 07/06/2018] [Indexed: 12/19/2022]
Abstract
Cerebral hemorrhage is a series of devastating cerebrovascular diseases with high mortality, morbidity and recurrence rate. Localized and systemic immuno-reactions are involved. Aggregation of immunocytes, which were both recruited from the peripheral circulation and resident in the central nervous system, is induced and activated by hematoma-related blood components. Subsequently, various cytokines, chemokines, free radicals and toxic chemicals are secreted to participant host defense responses. Among these, neuro-inflammation plays critical roles in both the pathologic processes of secondary injuries and recovery of neural damages. Numerous treatment strategies have been proposed, aiming at controlling the balance between anti- and proinflammation. Here, we summarized our current understanding and potential clinical applications for cytokines of the interleukin family in the pathogenesis of hemorrhagic stroke. In addition, we conducted protein-protein network, gene ontology and KEGG analysis on the interleukins using online bioinformatic tools to further elaborate the comprehensive mechanisms of interleukins in cerebral hemorrhage.
Collapse
Affiliation(s)
- Lei Ye
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Jixi 218, Hefei, 230022, People's Republic of China
- Institute of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Jixi 218, Hefei, 230022, People's Republic of China
| | - Lu Gao
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Jixi 218, Hefei, 230022, People's Republic of China
- Institute of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Jixi 218, Hefei, 230022, People's Republic of China
| | - Hongwei Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Jixi 218, Hefei, 230022, People's Republic of China.
- Institute of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Jixi 218, Hefei, 230022, People's Republic of China.
| |
Collapse
|
37
|
Toedebusch R, Belenchia A, Pulakat L. Cell-Specific Protective Signaling Induced by the Novel AT2R-Agonist NP-6A4 on Human Endothelial and Smooth Muscle Cells. Front Pharmacol 2018; 9:928. [PMID: 30186168 PMCID: PMC6111462 DOI: 10.3389/fphar.2018.00928] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 07/30/2018] [Indexed: 01/06/2023] Open
Abstract
Cardiovascular disease incidence continues to rise and new treatment paradigms are warranted. We reported previously that activation of Angiotensin II receptor (encoded by the X-linked Agtr2 gene) by a new peptide agonist, NP-6A4, was more effective in protecting mouse cardiomyocyte HL-1 cells and human coronary artery vascular smooth muscle cells (hCAVSMCs) from acute nutrient deficiency than other drugs tested. To elucidate further the protective effects of NP-6A4 in human cells, we studied the effects of NP-6A4 treatment on functions of human coronary artery endothelial cells (hCAECs), and hCAVSMCs. In hCAVSMCs, NP-6A4 (1 μM) increased Agtr2 mRNA (sixfold, p < 0.05) after 12-h exposure, whereas in hCAECs, significant increase in Agtr2 mRNA (hCAECs: eightfold) was observed after prolonged exposure. Interestingly, NP-6A4 treatment (1 μM, 12 h) increased AT2R protein levels in all human cells tested. Pre-treatment with AT2R-antagonist PD123319 (20 μM) and anti-AT2R siRNA (1 μM) suppressed this effect. Thus, NP-6A4 activates a positive feedback loop for AT2R expression and signaling in hCAVSMCs and hCAECs. NP-6A4 (1–20 μM) increased cell index (CI) of hCAVSMCs as determined by real time cell analyzer (RTCA), indicating that high concentrations of NP-6A4 were not cytotoxic for hCAVSMCs, rather promoting better cell attachment and growth. Seahorse Extracellular Flux Assay revealed that NP-6A4 (1 μM) treatment for 7 days increased whole cell-based mitochondrial parameters of hCAVSMCs, specifically maximal respiration (p < 0.05), spare respiratory capacity (p < 0.05) and ATP production (p < 0.05). NP-6A4 (1 μM; 7 days) also suppressed Reactive Oxygen Species (ROS) in hCAVSMCs. Exposure to Doxorubicin (DOXO) (1 μM) increased ROS in hCAVSMCs and this effect was suppressed by NP-6A4 (1 μM). In hCAECs grown in complete medium, NP-6A4 (1 μM) and Ang II (1 μM) exerted similar changes in CI. Additionally, NP-6A4 (5 μM: 12 h) increased expression of eNOS (sixfold, p < 0.05) and generation of nitric oxide (1.3-fold, p < 0.05) in hCAECs and pre-treatment with PD123319 (20 μM) suppressed this effect partially (65%). Finally, NP-6A4 decreased phosphorylation of Jun-N-terminal kinase, implicated in apoptosis of ECs in atherosclerotic sites. Taken together, NP-6A4, through its ability to increase AT2R expression and signaling, exerts different cell-specific protective effects in human VSMCs and ECs.
Collapse
Affiliation(s)
- Ryan Toedebusch
- Department of Medicine, University of Missouri, Columbia, MO, United States.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| | - Anthony Belenchia
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States.,Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Lakshmi Pulakat
- Department of Medicine, University of Missouri, Columbia, MO, United States.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States.,Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| |
Collapse
|
38
|
Wang W, Wang Q, Yu W, Chen L, Li Z. Efficacy of phosphocreatine pre-administration on XIAP and Smac in ischemic penumbra of rats with focal cerebral ischemia reperfusion injury. Acta Cir Bras 2018. [PMID: 29513810 DOI: 10.1590/s0102-865020180020000003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023] Open
Abstract
PURPOSE To observe the efficacy of phosphocreatine pre-administration (PCr-PA) on X-linked inhibitor of apoptosis protein (XIAP), the second mitochondia-derived activator of caspase (Smac) and apoptosis in the ischemic penumbra of rats with focal cerebral ischemia-reperfusion injury (CIRI). METHODS A total of 60 healthy male Sprague Dawley (SD) rats were randomly divided into three groups (n=20): group A (the sham operation group), group B <intraperitoneally injected with 20 mg/kg (10 mg/ml) of saline before preparing the ischemia-reperfusion (IR) model>, and group C <intraperitoneally injected with 20 mg/kg (10 mg/ml) of PCr immediately before preparing the IR model>. After 24 h for reperfusion, the neurological function was evaluated and the tissue was sampled to detect expression of XIAP, Smac and caspase-3 positive cells in the ischemic penumbra so as to observe the apoptosis. RESULTS Compared with group B, neurological deficit scores, numbers of apoptotic cells, expression of Smac,caspase-9 and the numbers of Caspase-3 positive cells were decreased while expression of XIAP were increased in the ischemic penumbra of group C. CONCLUSIONS Phosphocreatine pre-administration may elicit neuroprotective effects in the brain by increasing expression of X-linked inhibitor of apoptosis protein, reducing expression of second mitochondia-derived activator of caspase, and inhibiting the apoptosis in the ischemic penumbra.
Collapse
Affiliation(s)
- Wei Wang
- MD, Department of Anesthesiology, the Jiangning Hospital Affiliated, Nanjing Medical University, China. Acquisition and analysis of data, manuscript writing
| | - Qi Wang
- MD, Department of Anesthesiology, the Jiangning Hospital Affiliated, Nanjing Medical University, China. Acquisition and analysis of data, manuscript writing
| | - Wanyou Yu
- MD, Department of Anesthesiology, the Jiangning Hospital Affiliated, Nanjing Medical University, China. Acquisition and analysis of data, manuscript writing
| | - Lianhua Chen
- MD, Department of Anesthesiology, the First People's Hospital of Shanghai Affiliated, Nanjing Medical University, Shanghai, China. Analysis and interpretation of data
| | - Zhong Li
- PhD, Professor, Key Laboratory of Modern Toxicology (Ministry of Education), School of Public Health, Nanjing Medical University, China. Conception and design of the study, critical revision, final approval the manuscript
| |
Collapse
|
39
|
Saxena N, Won J, Choi S, Singh AK, Singh I. S-nitrosoglutathione reductase (GSNOR) inhibitor as an immune modulator in experimental autoimmune encephalomyelitis. Free Radic Biol Med 2018; 121:57-68. [PMID: 29694854 PMCID: PMC6083447 DOI: 10.1016/j.freeradbiomed.2018.04.558] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/13/2018] [Accepted: 04/17/2018] [Indexed: 12/27/2022]
Abstract
We previously reported that S-nitrosoglutathione (GSNO), an endogenous nitric oxide carrier, attenuated TH17-mediated immune responses in experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis (MS). Cellular GSNO homeostasis is regulated via its synthesis by reaction between nitric oxide and glutathione and its enzymatic catabolism by GSNO reductase (GSNOR). In this study, we evaluated potential of reversible inhibitor of GSNOR (N6022) in comparison with exogenous GSNO in immunopathogenesis of EAE. Daily treatment of EAE mice with N6022 or exogenous GSNO significantly attenuated the clinical disease of EAE, but N6022 treatment showed greater efficacy than GSNO. Both N6022 and exogenous GSNO treatments increased the spleen levels of GSNO, as documented by increased protein-associated S-nitrosothiols, and inhibited polarization and CNS effector function of proinflammatory TH17 cells while inducing the polarization and CNS effector function of anti-inflammatory CD4+ CD25+ FOXP3- regulatory T (Treg) cells. Moreover, N6022 further attenuated TH1 while inducing TH2 and CD4+ CD25+ FOXP3+ Treg in their polarization and CNS effector functions. Similar to GSNO, the N6022 treatment protected against the EAE disease induced demyelination. However, neither exogenous GSNO nor N6022 treatment did not cause significant systemic lymphopenic effect as compared to FTY720. Taken together, these data document that optimization of cellular GSNO homeostasis by GSNOR inhibitor (N6022) in NO metabolizing cells attenuates EAE disease via selective inhibition of pro-inflammatory subsets of CD4+ cells (TH1/TH17) while upregulating anti-inflammatory subsets of CD4+ cells (TH2/Treg) without causing lymphopenic effects and thus offers a potential treatment option for MS/EAE.
Collapse
MESH Headings
- Alcohol Dehydrogenase/antagonists & inhibitors
- Animals
- Benzamides/pharmacology
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/enzymology
- CD4-Positive T-Lymphocytes/immunology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Female
- Mice
- Mice, Inbred C57BL
- Protein S/metabolism
- Pyrroles/pharmacology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/enzymology
- T-Lymphocytes, Regulatory/immunology
- Th1 Cells/drug effects
- Th1 Cells/enzymology
- Th1 Cells/immunology
- Tyrosine/analogs & derivatives
- Tyrosine/metabolism
Collapse
Affiliation(s)
- Nishant Saxena
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Jeseong Won
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA.
| | - Seungho Choi
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Avtar K Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA; Pathology and Laboratory Medicine Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA
| | - Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA; Research Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA.
| |
Collapse
|
40
|
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
|
41
|
Tahsili-Fahadan P, Farrokh S, Geocadin RG. Hypothermia and brain inflammation after cardiac arrest. Brain Circ 2018; 4:1-13. [PMID: 30276330 PMCID: PMC6057700 DOI: 10.4103/bc.bc_4_18] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 03/17/2018] [Accepted: 03/18/2018] [Indexed: 12/14/2022] Open
Abstract
The cessation (ischemia) and restoration (reperfusion) of cerebral blood flow after cardiac arrest (CA) induce inflammatory processes that can result in additional brain injury. Therapeutic hypothermia (TH) has been proven as a brain protective strategy after CA. In this article, the underlying pathophysiology of ischemia-reperfusion brain injury with emphasis on the role of inflammatory mechanisms is reviewed. Potential targets for immunomodulatory treatments and relevant effects of TH are also discussed. Further studies are needed to delineate the complex pathophysiology and interactions among different components of immune response after CA and identify appropriate targets for clinical investigations.
Collapse
Affiliation(s)
- Pouya Tahsili-Fahadan
- Department of Medicine, Virginia Commonwealth University, Falls Church, Virginia, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Salia Farrokh
- Department of Pharmacy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Romergryko G Geocadin
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
42
|
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: 215] [Impact Index Per Article: 30.7] [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
|
43
|
Saavedra JM, Armando I. Angiotensin II AT2 Receptors Contribute to Regulate the Sympathoadrenal and Hormonal Reaction to Stress Stimuli. Cell Mol Neurobiol 2018; 38:85-108. [PMID: 28884431 PMCID: PMC6668356 DOI: 10.1007/s10571-017-0533-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/01/2017] [Indexed: 12/14/2022]
Abstract
Angiotensin II, through AT1 receptor stimulation, mediates multiple cardiovascular, metabolic, and behavioral functions including the response to stressors. Conversely, the function of Angiotensin II AT2 receptors has not been totally clarified. In adult rodents, AT2 receptor distribution is very limited but it is particularly high in the adrenal medulla. Recent results strongly indicate that AT2 receptors contribute to the regulation of the response to stress stimuli. This occurs in association with AT1 receptors, both receptor types reciprocally influencing their expression and therefore their function. AT2 receptors appear to influence the response to many types of stressors and in all components of the hypothalamic-pituitary-adrenal axis. The molecular mechanisms involved in AT2 receptor activation, the complex interactions with AT1 receptors, and additional factors participating in the control of AT2 receptor regulation and activity in response to stressors are only partially understood. Further research is necessary to close this knowledge gap and to clarify whether AT2 receptor activation may carry the potential of a major translational advance.
Collapse
Affiliation(s)
- J M Saavedra
- Department of Pharmacology and Physiology, Georgetown University Medical Center, 3900 Reservoir Road, Bldg. D, Room 287, Washington, DC, 20007, USA.
| | - I Armando
- The George Washington University School of Medicine and Health Sciences, Ross Hall Suite 738 2300 Eye Street, Washington, DC, USA
| |
Collapse
|
44
|
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]
|
45
|
Angiotensin II type 2 receptor (AT2R) as a novel modulator of inflammation in rheumatoid arthritis synovium. Sci Rep 2017; 7:13293. [PMID: 29038523 PMCID: PMC5643391 DOI: 10.1038/s41598-017-13746-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 10/02/2017] [Indexed: 12/29/2022] Open
Abstract
Despite increasing evidence suggesting that angiotensin II type 2 receptor (AT2R) may regulate tissue inflammation, no study has yet analyzed its possible implication in rheumatoid arthritis (RA) synovitis. In this study, we investigated the expression and function of AT2R in synovial tissue and cultured fibroblast-like synoviocytes (FLS) from RA patients. AT2R expression was strongly increased in RA compared with osteoarthritis (OA) synovium, as well as in in cultured RA-FLS respect to OA-FLS and healthy FLS. Treatment with pro-inflammatory cytokines was able not only to boost AT2R expression in RA-FLS and OA-FLS, but also to induce its de novo expression in healthy FLS. The stimulation of AT2R with the specific agonist CGP42112A significantly reduced gene expression of interleukin (IL)-1β and IL-6 and activation of NF-κB in RA-FLS, while opposite effects were elicited by AT2R small interfering RNA. Moreover, AT2R agonism efficiently decreased RA-FLS proliferation and migration either at baseline or under pro-inflammatory cytokine challenge. In conclusion, AT2R is strongly expressed in key effector cells of rheumatoid synovitis, namely RA-FLS, and the activation of AT2R with a specific agonist may effectively dampen their pro-inflammatory and aggressive behavior. AT2R agonism might represent a novel therapeutic strategy for patients with RA.
Collapse
|
46
|
Abstract
Depression remains a debilitating condition with an uncertain aetiology. Recently, attention has been given to the renin-angiotensin system. In the central nervous system, angiotensin II may be important in multiple pathways related to neurodevelopment and regulation of the stress response. Studies of drugs targeting the renin-angiotensin system have yielded promising results. Here, we review the potential beneficial effects of angiotensin blockers in depression and their mechanisms of action. Drugs blocking the angiotensin system have efficacy in several animal models of depression. While no randomised clinical trials were found, case reports and observational studies showed that angiotensin-converting enzyme inhibitors or angiotensin receptor blockers had positive effects on depression, whereas other antihypertensive agents did not. Drugs targeting the renin-angiotensin system act on inflammatory pathways implicated in depression. Both preclinical and clinical data suggest that these drugs possess antidepressant properties. In light of these results, angiotensin system-blocking agents offer new horizons in mood disorder treatment.
Collapse
|