1
|
Bai Y, Ma H, Zhang Y, Li J, Hou X, Yang Y, Wang G, Li Y. Hypidone hydrochloride (YL-0919) ameliorates functional deficits after traumatic brain injury in mice by activating the sigma-1 receptor for antioxidation. Neural Regen Res 2025; 20:2325-2336. [PMID: 39359091 PMCID: PMC11759037 DOI: 10.4103/nrr.nrr-d-23-01424] [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: 08/24/2023] [Revised: 12/13/2023] [Accepted: 02/02/2024] [Indexed: 10/04/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202508000-00023/figure1/v/2024-09-30T120553Z/r/image-tiff Traumatic brain injury involves complex pathophysiological mechanisms, among which oxidative stress significantly contributes to the occurrence of secondary injury. In this study, we evaluated hypidone hydrochloride (YL-0919), a self-developed antidepressant with selective sigma-1 receptor agonist properties, and its associated mechanisms and targets in traumatic brain injury. Behavioral experiments to assess functional deficits were followed by assessment of neuronal damage through histological analyses and examination of blood-brain barrier permeability and brain edema. Next, we investigated the antioxidative effects of YL-0919 by assessing the levels of traditional markers of oxidative stress in vivo in mice and in vitro in HT22 cells. Finally, the targeted action of YL-0919 was verified by employing a sigma-1 receptor antagonist (BD-1047). Our findings demonstrated that YL-0919 markedly improved deficits in motor function and spatial cognition on day 3 post traumatic brain injury, while also decreasing neuronal mortality and reversing blood-brain barrier disruption and brain edema. Furthermore, YL-0919 effectively combated oxidative stress both in vivo and in vitro. The protective effects of YL-0919 were partially inhibited by BD-1047. These results indicated that YL-0919 relieved impairments in motor and spatial cognition by restraining oxidative stress, a neuroprotective effect that was partially reversed by the sigma-1 receptor antagonist BD-1047. YL-0919 may have potential as a new treatment for traumatic brain injury.
Collapse
Affiliation(s)
- Yafan Bai
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hui Ma
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yue Zhang
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jinfeng Li
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Xiaojuan Hou
- Hebei North University, Zhangjiakou, Hebei Province, China
| | - Yixin Yang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Guyan Wang
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yunfeng Li
- Beijing Institute of Basic Medical Sciences, Beijing, China
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| |
Collapse
|
2
|
Zhang S, Huang Y, Han C, Chen M, Yang Z, Wang C. Circulating mitochondria carrying cGAS promote endothelial Secreted group IIA phospholipase A2-mediated neuroinflammation through activating astroglial/microglial Integrin-alphavbeta3 in subfornical organ to augment central sympathetic overdrive in heart failure rats. Int Immunopharmacol 2025; 144:113649. [PMID: 39586230 DOI: 10.1016/j.intimp.2024.113649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Sympathoexcitation, a manifestation of heart-brain axis dysregulation, contributes to the progression of heart failure (HF). Our recent study revealed that circulating mitochondria (C-Mito), a newly identified mediator of multi-organ communication, promote sympathoexcitation in HF by aggravating endothelial cell (EC)-derived neuroinflammation in the subfornical organ (SFO), the cardiovascular autonomic neural center. The precise molecular mechanism by which C-Mito promotes SFO-induced endothelial neuroinflammation has not been fully elucidated. OBJECTIVE C-Mito carrying cGAS promote sympathoexcitation by targeting PLA2G2A in ECs of the SFO in HF rats. METHODS Male Sprague-Dawley (SD) rats received a subcutaneous injection of isoprenaline (ISO) at a dosage of 5 mg/kg/day for seven consecutive days to establish a HF model. C-Mito were isolated from HF rats and evaluated. The level of cGAS, a dsDNA sensor recently discovered to be directly localized on the outer membrane of mitochondria, was detected in C-Mito. C-Mito from HF rats (C-MitoHF) or control rats (C-MitoCtrl) were intravenously infused into HF rats. The accumulation of C-Mito in the ECs in the SFO was detected via double immunofluorescence staining. The SFO was processed for RNA sequencing (RNA-Seq) analysis. Secreted group IIA phospholipase A2 (PLA2G2A), the key gene involved in C-MitoHF-associated SFO dysfunction, was identified via bioinformatics analysis. Upregulation of PLA2G2A in the SFO ECs was assessed via immunofluorescence staining and immunoblotting, and PLA2G2A activity was evaluated. The interaction between cGAS and PLA2G2A was detected via co-immunoprecipitation. The dowstream molecular mechanisms of which PLA2G2A induced astroglial/microglial activation were also investigated. AAV9-TIE-shRNA (PLA2G2A) was introduced into the SFO to specifically knockdown endothelial PLA2G2A. Neuronal activation and glial proinflammatory polarization in the SFO were also evaluated. Renal sympathetic nerve activity (RSNA) was measured to evaluate central sympathetic output. Cardiac sympathetic hyperinnervation, myocardial remodeling, and left ventricular systolic function were assessed in C-Mito-treated HF rats. RESULTS Respiratory functional incompetence and oxidative damage were observed in C-MitoHF compared with C-MitoCtrl. Surprisingly, cGAS protein levels in C-MitoHF were significantly higher than those in C-MitoCtrl, while blocking cGAS with its specific inhibitor, RU.521, mitigated respiratory dysfunction and oxidative injury in C-MitoHF. C-Mito entered the ECs of the SFO in HF rats. RNA sequencing revealed that PLA2G2A is a key molecule for the induction of SFO dysfunction by C-MitoHF. The immunoblotting and immunofluorescence results confirmed that, compared with C-MitoCtrl, C-MitoHF increased endothelial PLA2G2A expression in the SFO of HF rats, which could be alleviated by attenuating C-MitoHF-localized cGAS. Furthermore, we found that cGAS directly interacts with PLA2G2A, increased the activity of PLA2AG2, which produced arachidonic acid, and also promoted PLA2G2A secretion in brain ECs. In addition, the inhibition of PLA2G2A in brain ECs significantly mitigated the proinflammatory effect of conditioned cell culture medium from C-MitoHF-treated ECs on astroglia and microglia. Also, we found that PLA2G2A secreted from ECs insulted by C-Mito induced neuroinflammation through activating astriglial/microglial Integrin-alphavbeta3 in the SFO, which further promote central sympathetic overdrive in HF rats. Specific knockdown of endothelial PLA2G2A in the SFO mitigated C-MitoHF-induced presympathetic neuronal sensitization, cardiac sympathetic hyperinnervation, RSNA activation, myocardial remodeling, and systolic dysfunction in HF rats. CONCLUSION C-Mito carrying cGAS promoted cardiac sympathoexcitation by directly targeting PLA2G2A in the ECs of the SFO in HF rats. Secreted PLA2G2A derived from ECs insulted by C-Mito induced neuroinflammation through activating astriglial/microglial Integrin-alphavbeta3 in the SFO, which further promote central sympathetic overdrive in HF rats. Our study indicated that inhibiting cGAS in C-Mito might be a potential treatment for central sympathetic overdrive in HF.
Collapse
Affiliation(s)
- Shutian Zhang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China.
| | - Yijun Huang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Chengzhi Han
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Maoxiang Chen
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Zhaohua Yang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China.
| | - Chunsheng Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China.
| |
Collapse
|
3
|
Kawakami-Mori F. Esaxerenone, organ protection without sympathetic activation. Hypertens Res 2024; 47:2923-2925. [PMID: 39117949 DOI: 10.1038/s41440-024-01809-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 06/29/2024] [Indexed: 08/10/2024]
|
4
|
Lu Y, Wang YD, Xu TQ, Zhao XH, Zhou J, Jin LH, Liu JJ. Pyridostigmine attenuates hypertension by inhibiting activation of the renin-angiotensin system in the hypothalamic paraventricular nucleus. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7995-8007. [PMID: 38767671 DOI: 10.1007/s00210-024-03156-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/10/2024] [Indexed: 05/22/2024]
Abstract
Activation of the renin-angiotensin system (RAS) triggers oxidative stress and an inflammatory response in the hypothalamic paraventricular nucleus (PVN), in turn increasing the sympathetic hyperactivity that is a major cause of hypertension. Pyridostigmine has cardioprotective effects by suppressing the RAS of myocardial tissue. However, whether pyridostigmine attenuates hypertension by inhibiting the RAS of the PVN remains unclear. We thus investigated the effect and mechanism of pyridostigmine on two-kidney one-clip (2K1C)-induced hypertension. 2K1C rats received pyridostigmine, or not, for 8 weeks. Cardiovascular function, hemodynamic parameters, and autonomic activity were measured. The PVN levels of pro-/anti-inflammatory cytokines, oxidative stress, and RAS signaling molecules were evaluated. Our results showed that hypertension was accompanied by cardiovascular dysfunction and an autonomic imbalance characterized by enhanced sympathetic but diminished vagal activity. The PVN levels of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), reactive oxygen species (ROS), NOX-2, and malondialdehyde (MDA) increased; those of IL-10 and superoxide dismutase (SOD) decreased. Moreover, the RAS signaling pathway was activated, as evidenced by increased levels of the angiotensin-converting enzyme (ACE), angiotensin II (Ang II), and the Ang II type 1 receptor (AT1R) and a decreased AT2R level. Pyridostigmine lowered blood pressure and improved cardiovascular function, associated with restoration of the autonomic balance. Meanwhile, pyridostigmine decreased PVN IL-6, TNF-α, ROS, NOX-2, and MDA levels and increased IL-10 and SOD levels. Additionally, pyridostigmine suppressed PVN ACE, Ang II, and AT1R levels and increased AT2R expression. Pyridostigmine attenuated hypertension by inhibiting PVN oxidative stress and inflammation induced by the RAS.
Collapse
Affiliation(s)
- Yi Lu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Pharmacy, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Yi-Dong Wang
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tian-Qi Xu
- Department of Physiology and Pathophysiology, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University School of Basic Medical Sciences, Xi'an, China
| | - Xu-He Zhao
- Department of Physiology and Pathophysiology, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University School of Basic Medical Sciences, Xi'an, China
| | - Jun Zhou
- Department of Pharmacology, Xi'an Jiaotong University School of Basic Medical Sciences, Xi'an, China
| | - Lian-Hai Jin
- Low Pressure and Low Oxygen Environment and Health Intervention Innovation Center, Jilin Medical University, Jilin, China
| | - Jin-Jun Liu
- Department of Physiology and Pathophysiology, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University School of Basic Medical Sciences, Xi'an, China.
| |
Collapse
|
5
|
Luo Y, Zhang J, Jiao Y, Huang H, Ming L, Song Y, Niu Y, Tang X, Liu L, Li Y, Jiang Y. Dihydroartemisinin abolishes cisplatin-induced nephrotoxicity in vivo. J Nat Med 2024; 78:439-454. [PMID: 38351420 DOI: 10.1007/s11418-024-01783-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 01/15/2024] [Indexed: 02/29/2024]
Abstract
Dihydroartemisinin (DHA), a derivative of artemisinin which is primarily used to treat malaria in clinic, also confers protective effect on lipopolysaccharide-induced nephrotoxicity. While, the activities of DHA in cisplatin (CDDP)-caused nephrotoxicity are elusive. To investigate the role and underlying mechanism of DHA in CDDP-induced nephrotoxicity. Mice were randomly separated into four groups: normal, CDDP, and DHA (25 and 50 mg/kg were orally injected 1 h before CDDP for consecutive 10 days). All mice except the normal were single injected intraperitoneally with CDDP (22 mg/kg) for once on the 7th day. Combined with quantitative proteomics and bioinformatics analysis, the impact of DHA on renal cell apoptosis, oxidative stress, biochemical indexes, and inflammation in mice were investigated. Moreover, a human hepatocellular carcinoma cells xenograft model was established to elucidate the impact of DHA on tumor-related effects of CDDP. DHA reduced the levels of creatinine (CREA) (p < 0.01) and blood urea nitrogen (BUN) (p < 0.01), reversed CDDP-induced oxidative, inflammatory, and apoptosis indexes (p < 0.01). Mechanistically, DHA attenuated CDDP-induced inflammation by inhibiting nuclear factor κB p65 (NFκB p65) expression, and suppressed CDDP-induced renal cell apoptosis by inhibiting p63-mediated endogenous and exogenous apoptosis pathways. Additionally, DHA alone significantly decreased the tumor weight and did not destroy the antitumor effect of CDDP, and did not impact AST and ALT. In conclusion, DHA prevents CDDP-triggered nephrotoxicity via reducing inflammation, oxidative stress, and apoptosis. The mechanisms refer to inhibiting NFκB p65-regulated inflammation and alleviating p63-mediated mitochondrial endogenous and Fas death receptor exogenous apoptosis pathway.
Collapse
Affiliation(s)
- Yan Luo
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, School of Pharmacy, Scientific Research Center, Gannan Medical University, Ganzhou, 341000, China
| | - Jiaxing Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yue Jiao
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research On Prevention and Treatment of Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hao Huang
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, Gannan Medical University, Jiangxi, China
| | - Liangshan Ming
- Institute for Advanced Study, Jiangxi University of Chinese Medicine, Jiangxi, China
| | - Yunlei Song
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, School of Pharmacy, Scientific Research Center, Gannan Medical University, Ganzhou, 341000, China
| | - Yanlong Niu
- National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, Gannan Medical University, Jiangxi, China
| | - Xiaolu Tang
- Department of Human Anatomy, School of Basic Medical Science, Gannan Medical University, Jiangxi, China
| | - Liwei Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yi Li
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China.
| | - Yumao Jiang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, School of Pharmacy, Scientific Research Center, Gannan Medical University, Ganzhou, 341000, China.
- Artemisinin Research Center, and Institute of Chinese Materia Medical, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
6
|
Zhang S, Zhao D, Yang Z, Wang F, Yang S, Wang C. Circulating mitochondria promoted endothelial cGAS-derived neuroinflammation in subfornical organ to aggravate sympathetic overdrive in heart failure mice. J Neuroinflammation 2024; 21:27. [PMID: 38243316 PMCID: PMC10799549 DOI: 10.1186/s12974-024-03013-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 01/03/2024] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND Sympathoexcitation contributes to myocardial remodeling in heart failure (HF). Increased circulating pro-inflammatory mediators directly act on the Subfornical organ (SFO), the cardiovascular autonomic center, to increase sympathetic outflow. Circulating mitochondria (C-Mito) are the novel discovered mediators for inter-organ communication. Cyclic GMP-AMP synthase (cGAS) is the pro-inflammatory sensor of damaged mitochondria. OBJECTIVES This study aimed to assess the sympathoexcitation effect of C-Mito in HF mice via promoting endothelial cGAS-derived neuroinflammation in the SFO. METHODS C-Mito were isolated from HF mice established by isoprenaline (0.0125 mg/kg) infusion via osmotic mini-pumps for 2 weeks. Structural and functional analyses of C-Mito were conducted. Pre-stained C-Mito were intravenously injected every day for 2 weeks. Specific cGAS knockdown (cGAS KD) in the SFO endothelial cells (ECs) was achieved via the administration of AAV9-TIE-shRNA (cGAS) into the SFO. The activation of cGAS in the SFO ECs was assessed. The expression of the mitochondrial redox regulator Dihydroorotate dehydrogenase (DHODH) and its interaction with cGAS were also explored. Neuroinflammation and neuronal activation in the SFO were evaluated. Sympathetic activity, myocardial remodeling, and cardiac systolic dysfunction were measured. RESULTS C-Mito were successfully isolated, which showed typical structural characteristics of mitochondria with double-membrane and inner crista. Further analysis showed impaired respiratory complexes activities of C-Mito from HF mice (C-MitoHF) accompanied by oxidative damage. C-Mito entered ECs, instead of glial cells and neurons in the SFO of HF mice. C-MitoHF increased the level of ROS and cytosolic free double-strand DNA (dsDNA), and activated cGAS in cultured brain endothelial cells. Furthermore, C-MitoHF highly expressed DHODH, which interacted with cGAS to facilitate endothelial cGAS activation. C-MitoHF aggravated endothelial inflammation, microglial/astroglial activation, and neuronal sensitization in the SFO of HF mice, which could be ameliorated by cGAS KD in the ECs of the SFO. Further analysis showed C-MitoHF failed to exacerbate sympathoexcitation and myocardial sympathetic hyperinnervation in cGAS KD HF mice. C-MitoHF promoted myocardial fibrosis and hypertrophy, and cardiac systolic dysfunction in HF mice, which could be ameliorated by cGAS KD. CONCLUSION Collectively, we demonstrated that damaged C-MitoHF highly expressed DHODH, which promoted endothelial cGAS activation in the SFO, hence aggravating the sympathoexcitation and myocardial injury in HF mice, suggesting that C-Mito might be the novel therapeutic target for sympathoexcitation in HF.
Collapse
Affiliation(s)
- Shutian Zhang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Dajun Zhao
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Zhaohua Yang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Fanshun Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Shouguo Yang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
| | - Chunsheng Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
| |
Collapse
|
7
|
Sotozawa M, Kinguchi S, Wakui H, Azushima K, Funakoshi K, Nakajima W, Miyazaki T, Takahashi T, Tamura K. Enhancement of angiotensin II type 1 receptor-associated protein in the paraventricular nucleus suppresses angiotensin II-dependent hypertension. Hypertens Res 2024; 47:67-77. [PMID: 37884662 DOI: 10.1038/s41440-023-01480-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/29/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023]
Abstract
The renin-angiotensin system in the brain plays a pivotal role in modulating sympathetic nerve activity and contributes to the pathogenesis of hypertension. Angiotensin II (Ang II) type 1 receptor (AT1R)-associated protein (ATRAP) promotes internalization of AT1R while suppressing pathological overactivation of AT1R signaling. However, the pathophysiological function of ATRAP in the brain remains unknown. Therefore, this study aims to investigate whether ATRAP in the paraventricular nucleus (PVN) is involved in neurogenic hypertension pathogenesis in Ang II-infused rats. The ATRAP/AT1R ratio, which serves as an indicator of tissue AT1R hyperactivity, tended to decrease within the PVN in the Ang II group than in the vehicle group. This suggests an Ang II-induced hyperactivation of the AT1R signaling pathway in the PVN. Lentiviral vectors were generated to stimulate ATRAP expression. At 6 weeks of age, rats were microinjected with LV-Venus (Venus-expressing lentivirus) or LV-ATRAP (Venus-ATRAP-expressing lentivirus). The rats were then randomly divided into four groups: (1) Vehicle/LV-Venus, (2) Vehicle/LV-ATRAP, (3) Ang II/LV-Venus, and (4) Ang II/LV-ATRAP. Two weeks after microinjection, vehicle or Ang II was administered systemically for 2 weeks. In the Ang II/LV-ATRAP group, systolic blood pressure at 1 and 2 weeks following administration was significantly lower than that in the Ang II/LV-Venus group. Furthermore, urinary adrenaline levels tended to decrease in the Ang II/LV-ATRAP group than in the Ang II/LV-Venus group. These findings suggest that enhanced ATRAP expression in the PVN suppresses Ang II-induced hypertension, potentially by suppressing hyperactivation of the tissue AT1R signaling pathway and, subsequently, sympathetic nerve activity.
Collapse
Affiliation(s)
- Mari Sotozawa
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Sho Kinguchi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiromichi Wakui
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Kengo Azushima
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Kengo Funakoshi
- Department of Neuroanatomy, Yokohama City University School of Medicine, Yokohama, Japan
| | - Waki Nakajima
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tomoyuki Miyazaki
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takuya Takahashi
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
8
|
Iftikhar N, Hussain AI, Fatima T, Alsuwayt B, Althaiban AK. Bioactivity-Guided Isolation and Antihypertensive Activity of Citrullus colocynthis Polyphenols in Rats with Genetic Model of Hypertension. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1880. [PMID: 37893598 PMCID: PMC10608828 DOI: 10.3390/medicina59101880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023]
Abstract
Background and Objectives: Citrullus colocynthis belongs to the Cucurbitaceae family and is a wild medicinal plant used in folk literature to treat various diseases. The purpose of the current study was to explore the antihypertensive and antioxidant potentials of Citrullus colocynthis (CC) polyphenol-rich fractions using a spontaneous hypertensive rat (SHR) model. Materials and Methods: The concentrated aqueous ethanol extract of CC fruit was successively fractioned using solvents of increasing polarity, i.e., hexane, chloroform, ethyl acetate and n-butanol. The obtained extracts were analyzed for total phenolic content (TPC), total flavonoid content (TFC) and total flavonol content (TOF). Moreover, the CC extracts were further evaluated for radical scavenging capacity using 2,2-diphenyl-1-picrylhydrazyl (DPPH) and 2,2-azino-bis-3-ethylbenzothiazoline-6-sulfonic acid (ABTS) assays and antioxidant activity using inhibition of linoleic acid peroxidation and determination of reducing potential protocols. The phytochemical components were characterized by HPLC-MWD-ESI-MS in positive ionization mode. Results: The results showed that ethyl acetate fraction (EAF) exhibited a higher content of phenolic compounds in term of TPC (289 mg/g), TFC (7.6 mg/g) and TOF (35.7 mg/g). EAF showed higher antioxidant and DPPH and ABTS scavenging activities with SC50 values of 6.2 and 79.5 µg/mL, respectively. LCMS analysis revealed that twenty polyphenol compounds were identified in the EAF, including phenolic acids and flavonoids, mainly myricetin and quercetin derivatives. The in vivo antihypertensive activity of EAF of CC on SHR revealed that it significantly decreased the mean arterial pressure (MAP), systolic blood pressure (SBP), diastolic blood pressures (DBP) and pulse pressure (PP) as compared to normal and hypertensive control groups. Moreover, EAF of CC significantly reduced the oxidative stress in the animals in a dose-dependent manner by normalizing the levels of superoxide dismutase (SOD), malondialdehyde (MDA), reduced glutathione (GSH), nitric oxide (NOx) and total antioxidant capacity (TAC). Furthermore, the treatment groups, especially the 500 mg of EAF per kg body weight (EA-500) group, significantly (p ≤ 0.05) improved the electrocardiogram (ECG) pattern and pulse wave velocity (PWV). Conclusion: It was concluded that the EAF of CC is a rich source of polyphenols and showed the best antioxidant activity and antihypertensive potential in SHR.
Collapse
Affiliation(s)
- Neelam Iftikhar
- Natural Product and Synthetic Chemistry Laboratory, Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan;
| | - Abdullah Ijaz Hussain
- Natural Product and Synthetic Chemistry Laboratory, Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan;
- Central Hi-Tech Laboratory, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Tabinda Fatima
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hafr Al Batin, Hafar Al Batin 39524, Saudi Arabia;
| | - Bader Alsuwayt
- Department of Pharmacy Practice, College of Pharmacy, University of Hafr Al Batin, Hafar Al Batin 39524, Saudi Arabia; (B.A.); (A.K.A.)
| | - Abdullah K. Althaiban
- Department of Pharmacy Practice, College of Pharmacy, University of Hafr Al Batin, Hafar Al Batin 39524, Saudi Arabia; (B.A.); (A.K.A.)
| |
Collapse
|
9
|
Cao W, Yang Z, Liu X, Ren S, Su H, Yang B, Liu Y, Wilcox CS, Hou FF. A kidney-brain neural circuit drives progressive kidney damage and heart failure. Signal Transduct Target Ther 2023; 8:184. [PMID: 37169751 PMCID: PMC10175540 DOI: 10.1038/s41392-023-01402-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/16/2023] [Accepted: 02/24/2023] [Indexed: 05/13/2023] Open
Abstract
Chronic kidney disease (CKD) and heart failure (HF) are highly prevalent, aggravate each other, and account for substantial mortality. However, the mechanisms underlying cardiorenal interaction and the role of kidney afferent nerves and their precise central pathway remain limited. Here, we combined virus tracing techniques with optogenetic techniques to map a polysynaptic central pathway linking kidney afferent nerves to subfornical organ (SFO) and thereby to paraventricular nucleus (PVN) and rostral ventrolateral medulla that modulates sympathetic outflow. This kidney-brain neural circuit was overactivated in mouse models of CKD or HF and subsequently enhanced the sympathetic discharge to both the kidney and the heart in each model. Interruption of the pathway by kidney deafferentation, selective deletion of angiotensin II type 1a receptor (AT1a) in SFO, or optogenetic silence of the kidney-SFO or SFO-PVN projection decreased the sympathetic discharge and lessened structural damage and dysfunction of both kidney and heart in models of CKD and HF. Thus, kidney afferent nerves activate a kidney-brain neural circuit in CKD and HF that drives the sympathetic nervous system to accelerate disease progression in both organs. These results demonstrate the crucial role of kidney afferent nerves and their central connections in engaging cardiorenal interactions under both physiological and disease conditions. This suggests novel therapies for CKD or HF targeting this kidney-brain neural circuit.
Collapse
Affiliation(s)
- Wei Cao
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, PR China
| | - Zhichen Yang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, PR China
| | - Xiaoting Liu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, PR China
| | - Siqiang Ren
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence; Key Laboratory of Mental Health of the Ministry of Education; Guangdong Province Key Laboratory of Psychiatric Disorders, Southern Medical University, Guangzhou, Guangdong, China
| | - Huanjuan Su
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, PR China
| | - Bihui Yang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, PR China
| | - Youhua Liu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, PR China
| | - Christopher S Wilcox
- Division of Nephrology and Hypertension, Georgetown University Medical Central, Washington, DC, USA
| | - Fan Fan Hou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, PR China.
| |
Collapse
|
10
|
Yang K, Wu B, Wei W, Li C, Li L, Cong Z, Xiang Q. Curdione ameliorates sepsis-induced lung injury by inhibiting platelet-mediated neutrophil extracellular trap formation. Int Immunopharmacol 2023; 118:110082. [PMID: 36989889 DOI: 10.1016/j.intimp.2023.110082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/21/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
Sepsis-associated acute lung injury remains to be a major cause of morbidity and mortality worldwide, and there is a lack of effective therapeutic drugs. Curdione, an activeingredient of Curcuma zedoary, a traditional Chinese medicine (TCM), possesses a variety of pharmacological actions, such as anti-inflammatory, antioxidant and inhibition of platelet aggregation. However, whether curdione protects against sepsis-induced lung injury is still undetermined. In this study, we investigated the effects of curdione on sepsis-induced lung injury. Cecal ligation and puncture (CLP) surgery was performed in mice to establish a model of sepsis. Twenty-four hours after CLP, bronchoalveolar lavage fluid (BALF) and lung tissue samples were harvested for investigation. The protective effects of curdione on acute lung injury and potential mechanisms were explored by detecting pathological sections, exudative proteins, oxidative responses, inflammatory factors, platelet activation, neutrophil infiltration, and neutrophil extracellular trap (NET) formation in the lung and were further verified in vitro. We showed that treatment with curdione clearly relieved histopathological changes, reduced inflammatory cytokine elevation and total protein concentrations in BALF, and decreased oxidative stress responses in lung tissues. In addition, curdione inhibited platelet activation, further blocking the interaction between platelets and neutrophils. Finally, neutrophil infiltration and NET formation was also reduced in mice treated with curdione. In conclusion, curdione alleviates sepsis-induced lung injury by inhibiting platelet-mediated neutrophil recruitment, infiltration, and NET formation as well as its anti-inflammatory and antioxidant properties. Curdione has great therapeutic potential in sepsis.
Collapse
Affiliation(s)
- Kai Yang
- Department of Anesthesiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, No.725, South Wanping Road, Xuhui District, Shanghai 200032, China
| | - Bin Wu
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Wu Wei
- Department of Anesthesiology, the Eighth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, No. A17, Heishanhu Road, Haidian District, Beijing 100091, China
| | - Cuiyu Li
- Department of Anesthesiology, the Eighth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, No. A17, Heishanhu Road, Haidian District, Beijing 100091, China
| | - Lu Li
- Department of Infectious Diseases, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Zhukai Cong
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China.
| | - Qian Xiang
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China.
| |
Collapse
|
11
|
Issotina Zibrila A, Wang Z, Sangaré-Oumar MM, Zeng M, Liu X, Wang X, Zeng Z, Kang YM, Liu J. Role of blood-borne factors in sympathoexcitation-mediated hypertension: Potential neurally mediated hypertension in preeclampsia. Life Sci 2022; 320:121351. [PMID: 36592790 DOI: 10.1016/j.lfs.2022.121351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/19/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023]
Abstract
Hypertension remains a threat for society due to its unknown causes, preventing proper management, for the growing number of patients, for its state as a high-risk factor for stroke, cardiac and renal complication and as cause of disability. Data from clinical and animal researches have suggested the important role of many soluble factors in the pathophysiology of hypertension through their neuro-stimulating effects. Central targets of these factors are of molecular, cellular and structural nature. Preeclampsia (PE) is characterized by high level of soluble factors with strong pro-hypertensive activity and includes immune factors such as proinflammatory cytokines (PICs). The potential neural effect of those factors in PE is still poorly understood. Shedding light into the potential central effect of the soluble factors in PE may advance our current comprehension of the pathophysiology of hypertension in PE, which will contribute to better management of the disease. In this paper, we summarized existing data in respect of hypothesis of this review, that is, the existence of the neural component in the pathophysiology of the hypertension in PE. Future studies would address this hypothesis to broaden our understanding of the pathophysiology of hypertension in PE.
Collapse
Affiliation(s)
- Abdoulaye Issotina Zibrila
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an 710061, Shaanxi, PR China; Department of Animal Physiology, Faculty of science and Technology, University of Abomey-Calavi, 06 BP 2584 Cotonou, Benin
| | - Zheng Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, PR China
| | - Machioud Maxime Sangaré-Oumar
- Department of Animal Physiology, Faculty of science and Technology, University of Abomey-Calavi, 06 BP 2584 Cotonou, Benin
| | - Ming Zeng
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an 710061, Shaanxi, PR China
| | - Xiaoxu Liu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an 710061, Shaanxi, PR China
| | - Xiaomin Wang
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an 710061, Shaanxi, PR China
| | - Zhaoshu Zeng
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an 710061, Shaanxi, PR China
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an 710061, Shaanxi, PR China.
| | - Jinjun Liu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases, Xi'an 710061, Shaanxi, PR China.
| |
Collapse
|
12
|
Renin-a in the Subfornical Organ Plays a Critical Role in the Maintenance of Salt-Sensitive Hypertension. Biomolecules 2022; 12:biom12091169. [PMID: 36139008 PMCID: PMC9496084 DOI: 10.3390/biom12091169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022] Open
Abstract
The brain renin-angiotensin system plays important roles in blood pressure and cardiovascular regulation. There are two isoforms of prorenin in the brain: the classic secreted form (prorenin/sREN) encoded by renin-a, and an intracellular form (icREN) encoded by renin-b. Emerging evidence indicates the importance of renin-b in cardiovascular and metabolic regulation. However, the role of endogenous brain prorenin in the development of salt-sensitive hypertension remains undefined. In this study, we test the hypothesis that renin-a produced locally in the brain contributes to the pathogenesis of hypertension. Using RNAscope, we report for the first time that renin mRNA is expressed in several regions of the brain, including the subfornical organ (SFO), the paraventricular nucleus of the hypothalamus (PVN), and the brainstem, where it is found in glutamatergic, GABAergic, cholinergic, and tyrosine hydroxylase-positive neurons. Notably, we found that renin mRNA was significantly elevated in the SFO and PVN in a mouse model of DOCA-salt–induced hypertension. To examine the functional importance of renin-a in the SFO, we selectively ablated renin-a in the SFO in renin-a–floxed mice using a Cre-lox strategy. Importantly, renin-a ablation in the SFO attenuated the maintenance of DOCA-salt–induced hypertension and improved autonomic function without affecting fluid or sodium intake. Molecularly, ablation of renin-a prevented the DOCA-salt–induced elevation in NADPH oxidase 2 (NOX2) in the SFO without affecting NOX4 or angiotensin II type 1 and 2 receptors. Collectively, our findings demonstrate that endogenous renin-a within the SFO is important for the pathogenesis of salt-sensitive hypertension.
Collapse
|
13
|
Schwarz KG, Pereyra KV, Toledo C, Andrade DC, Díaz HS, Díaz-Jara E, Ortolani D, Rios-Gallardo A, Arias P, Las Heras A, Vera I, Ortiz FC, Inestrosa NC, Vio CP, Del Rio R. Effects of enriched-potassium diet on cardiorespiratory outcomes in experimental non-ischemic chronic heart failure. Biol Res 2021; 54:43. [PMID: 34952651 PMCID: PMC8710008 DOI: 10.1186/s40659-021-00365-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/09/2021] [Indexed: 12/11/2022] Open
Abstract
Background Chronic heart failure (CHF) is a global health problem. Increased sympathetic outflow, cardiac arrhythmogenesis and irregular breathing patterns have all been associated with poor outcomes in CHF. Several studies showed that activation of the renin-angiotensin system (RAS) play a key role in CHF pathophysiology. Interestingly, potassium (K+) supplemented diets showed promising results in normalizing RAS axis and autonomic dysfunction in vascular diseases, lowering cardiovascular risk. Whether subtle increases in dietary K+ consumption may exert similar effects in CHF has not been previously tested. Accordingly, we aimed to evaluate the effects of dietary K+ supplementation on cardiorespiratory alterations in rats with CHF. Methods Adult male Sprague–Dawley rats underwent volume overload to induce non-ischemic CHF. Animals were randomly allocated to normal chow diet (CHF group) or supplemented K+ diet (CHF+K+ group) for 6 weeks. Cardiac arrhythmogenesis, sympathetic outflow, baroreflex sensitivity, breathing disorders, chemoreflex function, respiratory–cardiovascular coupling and cardiac function were evaluated. Results Compared to normal chow diet, K+ supplemented diet in CHF significantly reduced arrhythmia incidence (67.8 ± 15.1 vs. 31.0 ± 3.7 events/hour, CHF vs. CHF+K+), decreased cardiac sympathetic tone (ΔHR to propranolol: − 97.4 ± 9.4 vs. − 60.8 ± 8.3 bpm, CHF vs. CHF+K+), restored baroreflex function and attenuated irregular breathing patterns. Additionally, supplementation of the diet with K+ restores normal central respiratory chemoreflex drive and abrogates pathological cardio-respiratory coupling in CHF rats being the outcome an improved cardiac function. Conclusion Our findings support that dietary K+ supplementation in non-ischemic CHF alleviate cardiorespiratory dysfunction. Supplementary Information The online version contains supplementary material available at 10.1186/s40659-021-00365-z.
Collapse
Affiliation(s)
- Karla G Schwarz
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Katherin V Pereyra
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Camilo Toledo
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - David C Andrade
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Fisiología y Medicina de Altura, Departamento Biomédico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
| | - Hugo S Díaz
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Esteban Díaz-Jara
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Domiziana Ortolani
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Angélica Rios-Gallardo
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - Paulina Arias
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexandra Las Heras
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ignacio Vera
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fernando C Ortiz
- Mechanisms of Myelin Formation and Repair Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Nibaldo C Inestrosa
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile.,Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carlos P Vio
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.,Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile. .,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile. .,Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
14
|
Zhang S, Hu L, Han C, Huang R, Ooi K, Qian X, Ren X, Chu D, Zhang H, Du D, Xia C. PLIN2 Mediates Neuroinflammation and Oxidative/Nitrosative Stress via Downregulating Phosphatidylethanolamine in the Rostral Ventrolateral Medulla of Stressed Hypertensive Rats. J Inflamm Res 2021; 14:6331-6348. [PMID: 34880641 PMCID: PMC8646230 DOI: 10.2147/jir.s329230] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/03/2021] [Indexed: 12/22/2022] Open
Abstract
Purpose Oxidative/nitrosative stress, neuroinflammation and their intimate interactions mediate sympathetic overactivation in hypertension. An immoderate inflammatory response is characterized not only by elevated proinflammatory cytokines (PICs) but by increases in mitochondrial dysfunction, reactive oxygen species (ROS), and nitric oxide (NO). Recent data pinpoint that both the phospholipid and lipid droplets (LDs) are potent modulators of microglia physiology. Methods Stress rats underwent compound stressors for 15 days with PLIN2-siRNA or scrambled-siRNA (SC-siRNA) administrated into the rostral ventrolateral medulla (RVLM). Lipids were analyzed by mass spectroscopy-based quantitative lipidomics. The phenotypes and proliferation of microglia, LDs, in the RVLM of rats were detected; blood pressure (BP) and myocardial injury in rats were evaluated. The anti-oxidative/nitrosative stress effect of phosphatidylethanolamine (PE) was explored in cultured primary microglia. Results Lipidomics analysis showed that 75 individual lipids in RVLM were significantly dysregulated by stress [PE was the most one], demonstrating that lipid composition changed with stress. In vitro, prorenin stress induced the accumulation of LDs, increased PICs, which could be blocked by siRNA-PLIN2 in microglia. PLIN2 knockdown upregulated the PE synthesis in microglia. Anti-oxidative/nitrosative stress effect of PE delivery was confirmed by the decrease of ROS and decrease in 3-NT and MDA in prorenin-treated microglia. PLIN2 knockdown in the RVLM blocked the number of iNOS+ and PCNA+ microglia, decreased BP, alleviated cardiac fibrosis and hypertrophy in stressed rats. Conclusion PLIN2 mediates microglial polarization/proliferation via downregulating PE in the RVLM of stressed rats. Delivery of PE is a promising strategy for combating neuroinflammation and oxidative/nitrosative stress in stress-induced hypertension.
Collapse
Affiliation(s)
- Shutian Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Li Hu
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200240, People's Republic of China
| | - Chengzhi Han
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Renhui Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Kokwin Ooi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xinyi Qian
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xiaorong Ren
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| | - Dechang Chu
- College of Agriculture and Bioengineering, Heze University, Heze, 274000, People's Republic of China
| | - Haili Zhang
- College of Agriculture and Bioengineering, Heze University, Heze, 274000, People's Republic of China
| | - Dongshu Du
- School of Life Science, Shanghai University, Shanghai, 200444, People's Republic of China
| | - Chunmei Xia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, People's Republic of China
| |
Collapse
|
15
|
Bian R, Gong J, Li J, Li P. Relaxin increased blood pressure and sympathetic activity in paraventricular nucleus of hypertensive rats via enhancing oxidative stress. Peptides 2021; 141:170550. [PMID: 33839220 DOI: 10.1016/j.peptides.2021.170550] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 02/08/2023]
Abstract
Relaxin, an ovarian polypeptide hormone, is found in the hypothalamic paraventricular nucleus (PVN) which is an important central integrative site for the control of blood pressure and sympathetic outflow. The aim of this study was to determine if superoxide anions modulate the effects of relaxin in the PVN. Experiments were performed in normotensive Wistar-Kyoto (WKY) rats and spontaneously hypertensive rats (SHRs). Relaxin mRNA and protein, and its receptor, relaxin family peptide receptor 1 (RXFP1) levels in PVN were 3.24, 3.17, and 3.64 times higher in SHRs than in WKY rats, respectively. Microinjection of relaxin-2 into the PVN dose-dependently increased mean arterial pressure (MAP), renal sympathetic nerve activity (RSNA) and heart rate (HR) in both WKY rats and SHRs, although the effects on MAP (16.87 ± 1.99 vs. 8.97 ± 1.48 mm Hg in 100 nmol), RSNA (22.60 ± 2.15 vs. 11.77 ± 1.43 % in 100 nmol) and HR (22.85 ± 3.13 vs. 12.62 ± 2.83 beats/min in 100 nmol) were greater in SHRs. Oxidative stress level was enhanced after relaxin-2 microinjection into the PVN. Pretreatment with superoxide anion scavengers or NADPH oxidase inhibitor blocked, and superoxide dismutase inhibitor potentiated the effects of relaxin-2 on MAP, RSNA and HR. RXFP1 knockdown significantly attenuated the blood pressure of SHRs, and inhibited the increases of atrial natriuretic peptide, brain natriuretic peptide, collagen I, collagen III and fibronectin in the heart of SHRs. These results demonstrated that relaxin is expressed in the PVN, and contributes to hypertension and sympathetic overdrive via oxidative stress. Down-regulation of RXFP1 in the PVN could attenuate hypertension and cardiac remodeling.
Collapse
Affiliation(s)
- Rong Bian
- Center of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Juexiao Gong
- Department of Cardiology, the Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jianan Li
- Center of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Peng Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
16
|
Abstract
Dr Irvine Page proposed the Mosaic Theory of Hypertension in the 1940s advocating that hypertension is the result of many factors that interact to raise blood pressure and cause end-organ damage. Over the years, Dr Page modified his paradigm, and new concepts regarding oxidative stress, inflammation, genetics, sodium homeostasis, and the microbiome have arisen that allow further refinements of the Mosaic Theory. A constant feature of this approach to understanding hypertension is that the various nodes are interdependent and that these almost certainly vary between experimental models and between individuals with hypertension. This review discusses these new concepts and provides an introduction to other reviews in this compendium of Circulation Research.
Collapse
Affiliation(s)
- David G. Harrison
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center
| | - Thomas M. Coffman
- Cardiovascular and Metabolic Disorders Research Program, Duke-National University of Singapore Medical School
| | | |
Collapse
|
17
|
Burke SL, Barzel B, Jackson KL, Gueguen C, Young MJ, Head GA. Role of Mineralocorticoid and Angiotensin Type 1 Receptors in the Paraventricular Nucleus in Angiotensin-Induced Hypertension. Front Physiol 2021; 12:640373. [PMID: 33762970 PMCID: PMC7982587 DOI: 10.3389/fphys.2021.640373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/16/2021] [Indexed: 11/25/2022] Open
Abstract
The hypothalamic paraventricular nucleus (PVN) is an important site where an interaction between circulating angiotensin (Ang) and mineralocorticoid receptor (MR) activity may modify sympathetic nerve activity (SNA) to influence long-term elevation of blood pressure. We examined in conscious Ang II-treated rabbits, the effects on blood pressure and tonic and reflex renal SNA (RSNA) of microinjecting into the PVN either RU28318 to block MR, losartan to block Ang (AT1) receptors or muscimol to inhibit GABA A receptor agonist actions. Male rabbits received a moderate dose of Ang II (24 ng/kg/min subcutaneously) for 3 months (n = 13) or sham treatment (n = 13). At 3 months, blood pressure increased by +19% in the Ang II group compared to 10% in the sham (P = 0.022) but RSNA was similar. RU28318 lowered blood pressure in both Ang II and shams but had a greater effect on RSNA and heart rate in the Ang II-treated group (P < 0.05). Losartan also lowered RSNA, while muscimol produced sympatho-excitation in both groups. In Ang II-treated rabbits, RU28318 attenuated the blood pressure increase following chemoreceptor stimulation but did not affect responses to air jet stress. In contrast losartan and muscimol reduced blood pressure and RSNA responses to both hypoxia and air jet. While neither RU28318 nor losartan changed the RSNA baroreflex, RU28318 augmented the range of the heart rate baroreflex by 10% in Ang II-treated rabbits. Muscimol, however, augmented the RSNA baroreflex by 11% in sham animals and none of the treatments altered baroreflex sensitivity. In conclusion, 3 months of moderate Ang II treatment promotes activation of reflex RSNA principally via MR activation in the PVN, rather than via activation of AT1 receptors. However, the onset of hypertension is independent of both. Interestingly, the sympatho-excitatory effects of muscimol in both groups suggest that overall, the PVN regulates a tonic sympatho-inhibitory influence on blood pressure control.
Collapse
Affiliation(s)
- Sandra L. Burke
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Benjamin Barzel
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Kristy L. Jackson
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Cindy Gueguen
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Morag J. Young
- Cardiovascular Endocrinology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Geoffrey A. Head
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Pharmacology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
18
|
Hirooka Y. Sympathetic Activation in Hypertension: Importance of the Central Nervous System. Am J Hypertens 2020; 33:914-926. [PMID: 32374869 DOI: 10.1093/ajh/hpaa074] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 04/18/2020] [Accepted: 05/01/2020] [Indexed: 12/20/2022] Open
Abstract
The sympathetic nervous system plays a critical role in the pathogenesis of hypertension. The central nervous system (CNS) organizes the sympathetic outflow and various inputs from the periphery. The brain renin-angiotensin system has been studied in various regions involved in controlling sympathetic outflow. Recent progress in cardiovascular research, particularly in vascular biology and neuroscience, as well as in traditional physiological approaches, has advanced the field of the neural control of hypertension in which the CNS plays a vital role. Cardiovascular research relating to hypertension has focused on the roles of nitric oxide, oxidative stress, inflammation, and immunity, and the network among various organs, including the heart, kidney, spleen, gut, and vasculature. The CNS mechanisms are similarly networked with these factors and are widely studied in neuroscience. In this review, I describe the development of the conceptual flow of this network in the field of hypertension on the basis of several important original research articles and discuss potential future breakthroughs leading to clinical precision medicine.
Collapse
Affiliation(s)
- Yoshitaka Hirooka
- Department of Medical Technology and Sciences, School of Health Sciences at Fukuoka, International University of Health and Welfare, Okawa City, Fukuoka, Japan
- Department of Cardiovascular Medicine, Hypertension and Heart Failure Center, Takagi Hospital, Okawa City, Fukuoka, Japan
| |
Collapse
|
19
|
Shaqura M, Li L, Mohamed DM, Li X, Treskatsch S, Buhrmann C, Shakibaei M, Beyer A, Mousa SA, Schäfer M. Neuronal aldosterone elicits a distinct genomic response in pain signaling molecules contributing to inflammatory pain. J Neuroinflammation 2020; 17:183. [PMID: 32532285 PMCID: PMC7291517 DOI: 10.1186/s12974-020-01864-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 06/02/2020] [Indexed: 11/10/2022] Open
Abstract
Background Recently, mineralocorticoid receptors (MR) were identified in peripheral nociceptive neurons, and their acute antagonism was responsible for immediate and short-lasting (non-genomic) antinociceptive effects. The same neurons were shown to produce the endogenous ligand aldosterone by the enzyme aldosterone synthase. Methods Here, we investigate whether endogenous aldosterone contributes to inflammation-induced hyperalgesia via the distinct genomic regulation of specific pain signaling molecules in an animal model of Freund’s complete adjuvant (FCA)-induced hindpaw inflammation. Results Chronic intrathecal application of MR antagonist canrenoate-K (over 4 days) attenuated nociceptive behavior in rats with FCA hindpaw inflammation suggesting a tonic activation of neuronal MR by endogenous aldosterone. Consistently, double immunofluorescence confocal microscopy showed abundant co-localization of MR with several pain signaling molecules such as TRPV1, CGRP, Nav1.8, and trkA whose enhanced expression of mRNA and proteins during inflammation was downregulated following i.t. canrenoate-K. More importantly, inhibition of endogenous aldosterone production in peripheral sensory neurons by continuous intrathecal delivery of a specific aldosterone synthase inhibitor prevented the inflammation-induced enhanced transcriptional expression of TRPV1, CGRP, Nav1.8, and trkA and subsequently attenuated nociceptive behavior. Evidence for such a genomic effect of endogenous aldosterone was supported by the demonstration of an enhanced nuclear translocation of MR in peripheral sensory dorsal root ganglia (DRG) neurons. Conclusion Taken together, chronic inhibition of local production of aldosterone by its processing enzyme aldosterone synthase within peripheral sensory neurons may contribute to long-lasting downregulation of specific pain signaling molecules and may, thus, persistently reduce inflammation-induced hyperalgesia.
Collapse
Affiliation(s)
- Mohammed Shaqura
- Department of Anaesthesiology and Intensive Care Medicine, Charité - University Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
| | - Li Li
- Department of Anaesthesiology and Intensive Care Medicine, Charité - University Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
| | - Doaa M Mohamed
- Department of Anaesthesiology and Intensive Care Medicine, Charité - University Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany.,Department of Zoology, Faculty of Science, Aswan University, Tingar, Egypt
| | - Xiongjuan Li
- Department of Anesthesiology, Second Affiliated Hospital of Guangzhou Medical University, No. 250, Hai'zhu District, Guangzhou, 510260, China
| | - Sascha Treskatsch
- Department of Anaesthesiology and Intensive Care Medicine, Charité - University Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
| | - Constanze Buhrmann
- Department of Anatomy, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Mehdi Shakibaei
- Department of Anatomy, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Antje Beyer
- Department of Anaesthesiology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Shaaban A Mousa
- Department of Anaesthesiology and Intensive Care Medicine, Charité - University Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany.
| | - Michael Schäfer
- Department of Anaesthesiology and Intensive Care Medicine, Charité - University Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Benjamin Franklin, Berlin, Germany
| |
Collapse
|
20
|
Brown CV, Boulet LM, Vermeulen TD, Sands SA, Wilson RJA, Ayas NT, Floras JS, Foster GE. Angiotensin II-Type I Receptor Antagonism Does Not Influence the Chemoreceptor Reflex or Hypoxia-Induced Central Sleep Apnea in Men. Front Neurosci 2020; 14:382. [PMID: 32410951 PMCID: PMC7198907 DOI: 10.3389/fnins.2020.00382] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 03/27/2020] [Indexed: 12/15/2022] Open
Abstract
Components of the renin-angiotensin system (RAS) situated within the carotid body or central nervous system may promote hypoxia-induced chemoreceptor reflex sensitization or central sleep apnea (CSA). We determined if losartan, an angiotensin-II type-I receptor (AT1R) antagonist, would attenuate chemoreceptor reflex sensitivity before or after 8 h of nocturnal hypoxia, and consequently CSA severity. In a double-blind, randomized, placebo-controlled, crossover protocol, 14 men (age: 25 ± 2 years; BMI: 24.6 ± 1.1 kg/m2; means ± SEM) ingested 3 doses of either losartan (50 mg) or placebo every 8 h. Chemoreceptor reflex sensitivity was assessed during hypoxic and hyperoxic hypercapnic ventilatory response (HCVR) tests and during six-20s hypoxic apneas before and after 8 h of sleep in normobaric hypoxia (FIO2 = 0.135). Loop gain was assessed from a ventilatory control model fitted to the ventilatory pattern of CSA recorded during polysomnography. Prior to nocturnal hypoxia, losartan had no effect on either the hyperoxic (losartan: 3.6 ± 1.1, placebo: 4.0 ± 0.6 l/min/mmHg; P = 0.9) or hypoxic HCVR (losartan: 5.3 ± 1.4, placebo: 5.7 ± 0.68 l/min/mmHg; P = 1.0). Likewise, losartan did not influence either the hyperoxic (losartan: 4.2 ± 1.3, placebo: 3.8 ± 1.1 l/min/mmHg; P = 0.5) or hypoxic HCVR (losartan: 6.6 ± 1.8, placebo: 6.3 ± 1.5 l/min/mmHg; P = 0.9) after nocturnal hypoxia. Cardiorespiratory responses to apnea and participants’ apnea hypopnea indexes during placebo and losartan were similar (73 ± 15 vs. 75 ± 14 events/h; P = 0.9). Loop gain, which correlated with CSA severity (r = 0.94, P < 0.001), was similar between treatments. In summary, in young healthy men, hypoxia-induced CSA severity is strongly associated with loop gain, but the AT1R does not modulate chemoreceptor reflex sensitivity before or after 8 h of nocturnal hypoxia.
Collapse
Affiliation(s)
- Courtney V Brown
- Centre for Heart, Lung, and Vascular Health, School of Health and Exercise Science, University of British Columbia - Okanagan, Kelowna, BC, Canada
| | - Lindsey M Boulet
- Centre for Heart, Lung, and Vascular Health, School of Health and Exercise Science, University of British Columbia - Okanagan, Kelowna, BC, Canada
| | - Tyler D Vermeulen
- Centre for Heart, Lung, and Vascular Health, School of Health and Exercise Science, University of British Columbia - Okanagan, Kelowna, BC, Canada
| | - Scott A Sands
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Richard J A Wilson
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Najib T Ayas
- Sleep Disorders Program, University of British Columbia, Vancouver, BC, Canada.,Respiratory and Critical Care Divisions, University of British Columbia, Vancouver, BC, Canada
| | - John S Floras
- University Health Network and Sinai Health System Division of Cardiology, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Glen E Foster
- Centre for Heart, Lung, and Vascular Health, School of Health and Exercise Science, University of British Columbia - Okanagan, Kelowna, BC, Canada
| |
Collapse
|
21
|
Aldosterone Synthase in Peripheral Sensory Neurons Contributes to Mechanical Hypersensitivity during Local Inflammation in Rats. Anesthesiology 2020; 132:867-880. [DOI: 10.1097/aln.0000000000003127] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Abstract
Background
Recent emerging evidence suggests that extra-adrenal synthesis of aldosterone occurs (e.g., within the failing heart and in certain brain areas). In this study, the authors investigated evidence for a local endogenous aldosterone production through its key processing enzyme aldosterone synthase within peripheral nociceptive neurons.
Methods
In male Wistar rats (n = 5 to 8 per group) with Freund’s complete adjuvant hind paw inflammation, the authors examined aldosterone, aldosterone synthase, and mineralocorticoid receptor expression in peripheral sensory neurons using quantitative reverse transcriptase–polymerase chain reaction, Western blot, immunohistochemistry, and immunoprecipitation. Moreover, the authors explored the nociceptive behavioral changes after selective mineralocorticoid receptor antagonist, canrenoate-K, or specific aldosterone synthase inhibitor application.
Results
In rats with Freund’s complete adjuvant–induced hind paw inflammation subcutaneous and intrathecal application of mineralocorticoid receptor antagonist, canrenoate-K, rapidly and dose-dependently attenuated nociceptive behavior (94 and 48% reduction in mean paw pressure thresholds, respectively), suggesting a tonic activation of neuronal mineralocorticoid receptors by an endogenous ligand. Indeed, aldosterone immunoreactivity was abundant in peptidergic nociceptive neurons of dorsal root ganglia and colocalized predominantly with its processing enzyme aldosterone synthase and mineralocorticoid receptors. Moreover, aldosterone and its synthesizing enzyme were significantly upregulated in peripheral sensory neurons under inflammatory conditions. The membrane mineralocorticoid receptor consistently coimmunoprecipitated with endogenous aldosterone, confirming a functional link between mineralocorticoid receptors and its endogenous ligand. Importantly, inhibition of endogenous aldosterone production in peripheral sensory neurons by a specific aldosterone synthase inhibitor attenuated nociceptive behavior after hind paw inflammation (a 32% reduction in paw pressure thresholds; inflammation, 47 ± 2 [mean ± SD] vs. inflammation + aldosterone synthase inhibitor, 62 ± 2).
Conclusions
Local production of aldosterone by its processing enzyme aldosterone synthase within peripheral sensory neurons contributes to ongoing mechanical hypersensitivity during local inflammation via intrinsic activation of neuronal mineralocorticoid receptors.
Editor’s Perspective
What We Already Know about This Topic
What This Article Tells Us That Is New
Collapse
|
22
|
Ezetimibe Attenuates Oxidative Stress and Neuroinflammation via the AMPK/Nrf2/TXNIP Pathway after MCAO in Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4717258. [PMID: 31998437 PMCID: PMC6964721 DOI: 10.1155/2020/4717258] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 10/18/2019] [Accepted: 10/31/2019] [Indexed: 12/05/2022]
Abstract
Oxidative stress and neuroinflammation play essential roles in ischemic stroke-induced brain injury. Previous studies have reported that Ezetimibe (Eze) exerts antioxidative stress and anti-inflammatory properties in hepatocytes. In the present study, we investigated the effects of Eze on oxidative stress and neuroinflammation in a rat middle cerebral artery occlusion (MCAO) model. One hundred and ninety-eight male Sprague-Dawley rats were used. Animals assigned to MCAO were given either Eze or its control. To explore the downstream signaling of Eze, the following interventions were given: AMPK inhibitor dorsomorphin and nuclear factor erythroid 2-related factor 2 (Nrf2) siRNA. Intranasal administration of Eze, 1 h post-MCAO, further increased the endogenous p-AMPK expression, reducing brain infarction, neurologic deficits, neutrophil infiltration, microglia/macrophage activation, number of dihydroethidium- (DHE-) positive cells, and malonaldehyde (MDA) levels. Specifically, treatment with Eze increased the expression of p-AMPK, Nrf2, and HO-1; Romo-1, thioredoxin-interacting protein (TXNIP), NOD-like receptor protein 3 (NLRP3), Cleaved Caspase-1, and IL-1β were reduced. Dorsomorphin and Nrf2 siRNA reversed the protective effects of Eze. In summary, Eze decreases oxidative stress and subsequent neuroinflammation via activation of the AMPK/Nrf2/TXNIP pathway after MCAO in rats. Therefore, Eze may be a potential therapeutic approach for ischemic stroke patients.
Collapse
|
23
|
Qi J, Yu XJ, Fu LY, Liu KL, Gao TT, Tu JW, Kang KB, Shi XL, Li HB, Li Y, Kang YM. Exercise Training Attenuates Hypertension Through TLR4/MyD88/NF-κB Signaling in the Hypothalamic Paraventricular Nucleus. Front Neurosci 2019; 13:1138. [PMID: 31708733 PMCID: PMC6821652 DOI: 10.3389/fnins.2019.01138] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 10/09/2019] [Indexed: 12/20/2022] Open
Abstract
Exercise training (ExT) is beneficial for cardiovascular health, yet the central mechanism by which aerobic ExT attenuates the hypertensive responses remains unclear. Activation of pro-inflammatory cytokines (PICs) in the hypothalamic paraventricular nucleus (PVN) is important for the sympathoexcitation and hypertensive response. We thus hypothesized that aerobic ExT can decrease the blood pressure of hypertensive rats by reducing the levels of PICs through TLR4/MyD88/NF-κB signaling within the PVN. To examine this hypothesis, two-kidney-one-clip (2K1C) renovascular hypertensive rats were assigned to two groups: sedentary or exercise training and examined for 8 weeks. At the same time, bilateral PVN infusion of vehicle or TAK242, a TLR4 inhibitor, was performed on both groups. As a result, the systolic blood pressure (SBP), renal sympathetic nerve activity (RSNA) and plasma levels of norepinephrine (NE), epinephrine (EPI) were found significantly increased in 2K1C hypertensive rats. These rats also had higher levels of Fra-like activity, NF-κB p65 activity, TLR4, MyD88, IL-1β and TNF-α in the PVN than SHAM rats. Eight weeks of ExT attenuated the RSNA and SBP, repressed the NF-κB p65 activity, and reduced the increase of plasma levels of NE, EPI, and the expression of Fra-like, TLR4, MyD88, IL-1β and TNF-α in the PVN of 2K1C rats. These findings are highly similar to the results in 2K1C rats with bilateral PVN infusions of TLR4 inhibitor (TAK242). This suggests that 8 weeks of aerobic ExT may decrease blood pressure in hypertensive rats by reducing the PICs activation through TLR4/MyD88/NF-κB signaling within the PVN, and thus delays the progression of 2K1C renovascular hypertension.
Collapse
Affiliation(s)
- Jie Qi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University, Xi'an, China
| | - Xiao-Jing Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University, Xi'an, China
| | - Li-Yan Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University, Xi'an, China
| | - Kai-Li Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University, Xi'an, China
| | - Tian-Tian Gao
- School of Clinical Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Jia-Wei Tu
- School of Clinical Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Kai B Kang
- Department of Ophthalmology and Visual Sciences, The University of Illinois at Chicago, Chicago, IL, United States
| | - Xiao-Lian Shi
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Hong-Bao Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University, Xi'an, China
| | - Ying Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University, Xi'an, China
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
24
|
Zemskov EA, Lu Q, Ornatowski W, Klinger CN, Desai AA, Maltepe E, Yuan JXJ, Wang T, Fineman JR, Black SM. Biomechanical Forces and Oxidative Stress: Implications for Pulmonary Vascular Disease. Antioxid Redox Signal 2019; 31:819-842. [PMID: 30623676 PMCID: PMC6751394 DOI: 10.1089/ars.2018.7720] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Oxidative stress in the cell is characterized by excessive generation of reactive oxygen species (ROS). Superoxide (O2-) and hydrogen peroxide (H2O2) are the main ROS involved in the regulation of cellular metabolism. As our fundamental understanding of the underlying causes of lung disease has increased it has become evident that oxidative stress plays a critical role. Recent Advances: A number of cells in the lung both produce, and respond to, ROS. These include vascular endothelial and smooth muscle cells, fibroblasts, and epithelial cells as well as the cells involved in the inflammatory response, including macrophages, neutrophils, eosinophils. The redox system is involved in multiple aspects of cell metabolism and cell homeostasis. Critical Issues: Dysregulation of the cellular redox system has consequential effects on cell signaling pathways that are intimately involved in disease progression. The lung is exposed to biomechanical forces (fluid shear stress, cyclic stretch, and pressure) due to the passage of blood through the pulmonary vessels and the distension of the lungs during the breathing cycle. Cells within the lung respond to these forces by activating signal transduction pathways that alter their redox state with both physiologic and pathologic consequences. Future Directions: Here, we will discuss the intimate relationship between biomechanical forces and redox signaling and its role in the development of pulmonary disease. An understanding of the molecular mechanisms induced by biomechanical forces in the pulmonary vasculature is necessary for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Evgeny A Zemskov
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Qing Lu
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Wojciech Ornatowski
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Christina N Klinger
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Ankit A Desai
- Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Emin Maltepe
- Department of Pediatrics, University of California, San Francisco, San Francisco, California
| | - Jason X-J Yuan
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Ting Wang
- Department of Internal Medicine, The University of Arizona Health Sciences, Phoenix, Arizona
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California, San Francisco, San Francisco, California
| | - Stephen M Black
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| |
Collapse
|
25
|
Kang Y, Ding L, Dai H, Wang F, Zhou H, Gao Q, Xiong X, Zhang F, Song T, Yuan Y, Zhu G, Zhou Y. Intermedin in Paraventricular Nucleus Attenuates Ang II-Induced Sympathoexcitation through the Inhibition of NADPH Oxidase-Dependent ROS Generation in Obese Rats with Hypertension. Int J Mol Sci 2019; 20:ijms20174217. [PMID: 31466304 PMCID: PMC6747263 DOI: 10.3390/ijms20174217] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/19/2019] [Accepted: 08/26/2019] [Indexed: 12/19/2022] Open
Abstract
Increased reactive oxygen species (ROS) induced by angiotensin II (Ang II) in the paraventricular nucleus (PVN) play a critical role in sympathetic overdrive in hypertension (OH). Intermedin (IMD), a bioactive peptide, has extensive clinically prospects in preventing and treating cardiovascular diseases. The study was designed to test the hypothesis that IMD in the PVN can inhibit the generation of ROS caused by Ang II for attenuating sympathetic nerve activity (SNA) and blood pressure (BP) in rats with obesity-related hypertension (OH). Male Sprague-Dawley rats (160-180 g) were used to induce OH by feeding of a high-fat diet (42% kcal as fat) for 12 weeks. The dynamic changes of sympathetic outflow were evaluated as the alterations of renal sympathetic nerve activity (RSNA) and mean arterial pressure (MAP) responses to certain chemicals. The results showed that the protein expressions of Ang II type 1 receptor (AT1R), calcitonin receptor-like receptor (CRLR) and receptor activity-modifying protein 2 (RAMP2) and RAMP3 were markedly increased, but IMD was much lower in OH rats when compared to control rats. IMD itself microinjection into PVN not only lowered SNA, NADPH oxidase activity and ROS level, but also decreased Ang II-caused sympathetic overdrive, and increased NADPH oxidase activity, ROS levels and mitogen-activated protein kinase/extracellular signal regulated kinase (MAPK/ERK) activation in OH rats. However, those effects were mostly blocked by the adrenomedullin (AM) receptor antagonist AM22-52 pretreatment. The enhancement of SNA caused by Ang II can be significantly attenuated by the pretreatment of AT1R antagonist lorsatan, superoxide scavenger Tempol and NADPH oxidase inhibitor apocynin (Apo) in OH rats. ERK activation inhibitor U0126 in the PVN reversed Ang II-induced enhancement of SNA, and Apo and IMD pretreatment in the PVN decreased Ang II-induced ERK activation. Chronic IMD administration in the PVN resulted in significant reductions in basal SNA and BP in OH rats. Moreover, IMD lowered NADPH oxidase activity and ROS level in the PVN; reduced the protein expressions of AT1R and NADPH oxidase subunits NOX2 and NOX4, and ERK activation in the PVN; and decreased Ang II levels-inducing sympathetic overactivation. These results indicated that IMD via AM receptors in the PVN attenuates SNA and hypertension, and decreases Ang II-induced enhancement of SNA through the inhibition of NADPH oxidase activity and ERK activation.
Collapse
Affiliation(s)
- Ying Kang
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Lei Ding
- Department of Pathophysiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Hangbing Dai
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Fangzheng Wang
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Hong Zhou
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Qing Gao
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Xiaoqing Xiong
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Feng Zhang
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Tianrun Song
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Yan Yuan
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Guoqing Zhu
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Yebo Zhou
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
26
|
Gong J, Shen Y, Li P, Zhao K, Chen X, Li Y, Sheng Y, Zhou B, Kong X. Superoxide anions mediate the effects of angiotensin (1-7) analog, alamandine, on blood pressure and sympathetic activity in the paraventricular nucleus. Peptides 2019; 118:170101. [PMID: 31199949 DOI: 10.1016/j.peptides.2019.170101] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 06/07/2019] [Accepted: 06/07/2019] [Indexed: 12/19/2022]
Abstract
Microinjection of alamandine into the hypothalamic paraventricular nucleus (PVN) increased blood pressure and enhanced sympathetic activity. The aim of this study was to determine if superoxide anions modulate alamandine's effects in the PVN. Mean arterial pressure (MAP) and renal sympathetic nerve activity (RSNA) were recorded in anaesthetized normotensive Wistar-Kyoto (WKY) rats and spontaneously hypertensive rats (SHRs). Microinjection of alamandine into the PVN increased MAP and RSNA in both WKY rats and SHRs, although to a greater extent in SHRs. These effects were blocked by pretreatment with an alamandine receptor (MrgD) antagonist D-Pro7-Ang-(1-7). Pretreatment with superoxide anion scavengers, tempol and tiron, and NADPH oxidase inhibitor apocynin (APO), also blocked the effects of alamandine on MAP and RSNA. In addition, pretreatment in the PVN with a superoxide dismutase (SOD) inhibitor diethyldithiocarbamic acid (DETC) potentiated the increases of MAP and RSNA induced by alamandine administration, with a greater response observed in SHRs. Superoxide anions and NADPH oxidase levels in the PVN were higher in SHRs than that in WKY rats. Alamandine treatment increased the levels of superoxide anions and NADPH oxidase in WKY and SHRs, however, with greater effect in SHRs. These alamandine-induced increases were inhibited by D-Pro7-Ang-(1-7) pretreatment in the PVN of both rats. These results demonstrate that superoxide anions in the PVN modulate alamandine-induced increases in blood pressure and sympathetic activity in both normotensive and hypertensive rats. Alamandine increases NADPH oxidase activity to induce superoxide anion production, which is mediated by the alamandine receptor.
Collapse
Affiliation(s)
- Juexiao Gong
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Department of Cardiology, the Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yihui Shen
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Peng Li
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kun Zhao
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xuguan Chen
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yong Li
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yanhui Sheng
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Bin Zhou
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Xiangqing Kong
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
27
|
Lu J, Wang HW, Ahmad M, Keshtkar-Jahromi M, Blaustein MP, Hamlyn JM, Leenen FHH. Central and peripheral slow-pressor mechanisms contributing to Angiotensin II-salt hypertension in rats. Cardiovasc Res 2019; 114:233-246. [PMID: 29126194 DOI: 10.1093/cvr/cvx214] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 11/07/2017] [Indexed: 12/17/2022] Open
Abstract
Aims High salt intake markedly enhances hypertension induced by angiotensin II (Ang II). We explored central and peripheral slow-pressor mechanisms which may be activated by Ang II and salt. Methods and results In protocol I, Wistar rats were infused subcutaneously with low-dose Ang II (150 ng/kg/min) and fed regular (0.4%) or high salt (2%) diet for 14 days. In protocol II, Ang II-high salt was combined with intracerebroventricular infusion of mineralocorticoid receptor (MR) blockers (eplerenone, spironolactone), epithelial sodium channel (ENaC) blocker (benzamil), angiotensin II type 1 receptor (AT1R) blocker (losartan) or vehicles. Ang II alone raised mean arterial pressure (MAP) ∼10 mmHg, but Ang II-high salt increased MAP ∼50 mmHg. Ang II-high salt elevated plasma corticosterone, aldosterone and endogenous ouabain but not Ang II alone. Both Ang II alone and Ang II-high salt increased mRNA and protein expression of CYP11B2 (aldosterone synthase gene) in the adrenal cortex but not of CYP11B1 (11-β-hydroxylase gene). In the aorta, Ang II-high salt increased sodium-calcium exchanger-1 (NCX1) protein. The Ang II-high salt induced increase in MAP was largely prevented by central infusion of MR blockers, benzamil or losartan. Central blockades significantly lowered plasma aldosterone and endogenous ouabain and markedly decreased Ang II-high salt induced CYP11B2 mRNA expression in the adrenal cortex and NCX1 protein in the aorta. Conclusion These results suggest that in Ang II-high salt hypertension, MR-ENaC-AT1R signalling in the brain increases circulating aldosterone and endogenous ouabain, and arterial NCX1. These factors can amplify blood pressure responses to centrally-induced sympatho-excitation and thereby contribute to severe hypertension.
Collapse
Affiliation(s)
- Jiao Lu
- Brain and Heart Research Group, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y 4W7, Canada
| | - Hong-Wei Wang
- Brain and Heart Research Group, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y 4W7, Canada
| | - Monir Ahmad
- Brain and Heart Research Group, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y 4W7, Canada
| | - Marzieh Keshtkar-Jahromi
- Brain and Heart Research Group, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y 4W7, Canada.,Department of Physiology, University of Maryland School of Medicine, 655 West Baltimore S, Baltimore, MD 21201, USA
| | - Mordecai P Blaustein
- Department of Physiology, University of Maryland School of Medicine, 655 West Baltimore S, Baltimore, MD 21201, USA.,Department of Medicine, University of Maryland School of Medicine, 655 West Baltimore S, Baltimore, MD 21201, USA
| | - John M Hamlyn
- Department of Physiology, University of Maryland School of Medicine, 655 West Baltimore S, Baltimore, MD 21201, USA
| | - Frans H H Leenen
- Brain and Heart Research Group, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y 4W7, Canada
| |
Collapse
|
28
|
Mowry FE, Biancardi VC. Neuroinflammation in hypertension: the renin-angiotensin system versus pro-resolution pathways. Pharmacol Res 2019; 144:279-291. [PMID: 31039397 DOI: 10.1016/j.phrs.2019.04.029] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/22/2019] [Accepted: 04/23/2019] [Indexed: 12/31/2022]
Abstract
Overstimulation of the pro-inflammatory pathways within brain areas responsible for sympathetic outflow is well evidenced as a primary contributing factor to the establishment and maintenance of neurogenic hypertension. However, the precise mechanisms and stimuli responsible for promoting a pro-inflammatory state are not fully elucidated. Recent work has unveiled novel compounds derived from omega-3 polyunsaturated fatty acids (ω-3 PUFAs), termed specialized pro-resolving mediators (SPMs), which actively regulate the resolution of inflammation. Failure or dysregulation of the resolution process has been linked to a variety of chronic inflammatory and neurodegenerative diseases. Given the pathologic role of neuroinflammation in the hypertensive state, SPMs and their associated pathways may provide a link between hypertension and the long-standing association of dietary ω-3 PUFAs with cardioprotection. Herein, we review recent progress in understanding the RAS-driven pathophysiology of neurogenic hypertension, particularly in regards to the chronic low-grade neuroinflammatory response. In addition, we examine the potential for an impaired resolution of inflammation process in the context of hypertension.
Collapse
Affiliation(s)
- Francesca Elisabeth Mowry
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Alabama, USA
| | - Vinicia Campana Biancardi
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Alabama, USA; Center for Neurosciences Research Initiative, Auburn University, Alabama, USA.
| |
Collapse
|
29
|
Abstract
Purpose of Review Although an independent brain renin-angiotensin system is often assumed to exist, evidence for this concept is weak. Most importantly, renin is lacking in the brain, and both brain angiotensinogen and angiotensin (Ang) II levels are exceptionally low. In fact, brain Ang II levels may well represent uptake of circulating Ang II via Ang II type 1 (AT1) receptors. Recent Findings Nevertheless, novel drugs are now aimed at the brain RAS, i.e., aminopeptidase A inhibitors should block Ang III formation from Ang II, and hence diminish AT1 receptor stimulation by Ang III, while AT2 and Mas receptor agonists are reported to induce neuroprotection after stroke. The endogenous agonists of these receptors and their origin remain unknown. Summary This review addresses the questions whether independent angiotensin generation truly occurs in the brain, what its relationship with the kidney is, and how centrally acting RAS blockers/agonists might work.
Collapse
Affiliation(s)
- Liwei Ren
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands
- AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, China
| | - Xifeng Lu
- AstraZeneca-Shenzhen University Joint Institute of Nephrology, Department of Physiology, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, China
| | - A H Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands.
| |
Collapse
|
30
|
Guimarães DD, Cruz JC, Carvalho-Galvão A, Zhuge Z, Marques SM, Naves LM, Persson AEG, Weitzberg E, Lundberg JO, Balarini CM, Pedrino GR, Braga VA, Carlström M. Dietary Nitrate Reduces Blood Pressure in Rats With Angiotensin II–Induced Hypertension via Mechanisms That Involve Reduction of Sympathetic Hyperactivity. Hypertension 2019; 73:839-848. [DOI: 10.1161/hypertensionaha.118.12425] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Drielle D. Guimarães
- From the Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (D.D.G., J.C.C., Z.Z., A.E.G.P., E.W., J.O.L., M.C.)
- Biotechnology Center (D.D.G., J.C.C., A.C.-G., C.M.B., V.A.B.), Federal University of Paraiba, Joao Pessoa, Brazil
| | - Josiane C. Cruz
- From the Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (D.D.G., J.C.C., Z.Z., A.E.G.P., E.W., J.O.L., M.C.)
- Biotechnology Center (D.D.G., J.C.C., A.C.-G., C.M.B., V.A.B.), Federal University of Paraiba, Joao Pessoa, Brazil
| | - Alynne Carvalho-Galvão
- Biotechnology Center (D.D.G., J.C.C., A.C.-G., C.M.B., V.A.B.), Federal University of Paraiba, Joao Pessoa, Brazil
| | - Zhengbing Zhuge
- From the Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (D.D.G., J.C.C., Z.Z., A.E.G.P., E.W., J.O.L., M.C.)
| | - Stefanne M. Marques
- Department of Physiological Sciences, Federal University of Goias, Goiania, Brazil (S.M.M., L.M.N., G.R.P.)
| | - Lara M. Naves
- Department of Physiological Sciences, Federal University of Goias, Goiania, Brazil (S.M.M., L.M.N., G.R.P.)
| | - A. Erik G. Persson
- From the Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (D.D.G., J.C.C., Z.Z., A.E.G.P., E.W., J.O.L., M.C.)
- Department of Medical Cell Biology, Uppsala University, Sweden (A.E.G.P.)
| | - Eddie Weitzberg
- From the Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (D.D.G., J.C.C., Z.Z., A.E.G.P., E.W., J.O.L., M.C.)
| | - Jon O. Lundberg
- From the Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (D.D.G., J.C.C., Z.Z., A.E.G.P., E.W., J.O.L., M.C.)
| | - Camille M. Balarini
- Biotechnology Center (D.D.G., J.C.C., A.C.-G., C.M.B., V.A.B.), Federal University of Paraiba, Joao Pessoa, Brazil
- Health Sciences Center (C.M.B.), Federal University of Paraiba, Joao Pessoa, Brazil
| | - Gustavo R. Pedrino
- Department of Physiological Sciences, Federal University of Goias, Goiania, Brazil (S.M.M., L.M.N., G.R.P.)
| | - Valdir A. Braga
- Biotechnology Center (D.D.G., J.C.C., A.C.-G., C.M.B., V.A.B.), Federal University of Paraiba, Joao Pessoa, Brazil
| | - Mattias Carlström
- From the Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (D.D.G., J.C.C., Z.Z., A.E.G.P., E.W., J.O.L., M.C.)
| |
Collapse
|
31
|
Souza LAC, Worker CJ, Li W, Trebak F, Watkins T, Gayban AJB, Yamasaki E, Cooper SG, Drumm BT, Feng Y. (Pro)renin receptor knockdown in the paraventricular nucleus of the hypothalamus attenuates hypertension development and AT 1 receptor-mediated calcium events. Am J Physiol Heart Circ Physiol 2019; 316:H1389-H1405. [PMID: 30925093 DOI: 10.1152/ajpheart.00780.2018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Activation of the brain renin-angiotensin system (RAS) is a pivotal step in the pathogenesis of hypertension. The paraventricular nucleus (PVN) of the hypothalamus is a critical part of the angiotensinergic sympatho-excitatory neuronal network involved in neural control of blood pressure and hypertension. However, the importance of the PVN (pro)renin receptor (PVN-PRR)-a key component of the brain RAS-in hypertension development has not been examined. In this study, we investigated the involvement and mechanisms of the PVN-PRR in DOCA-salt-induced hypertension, a mouse model of hypertension. Using nanoinjection of adeno-associated virus-mediated Cre recombinase expression to knock down the PRR specifically in the PVN, we report here that PVN-PRR knockdown attenuated the enhanced blood pressure and sympathetic tone associated with hypertension. Mechanistically, we found that PVN-PRR knockdown was associated with reduced activation of ERK (extracellular signal-regulated kinase)-1/2 in the PVN and rostral ventrolateral medulla during hypertension. In addition, using the genetically encoded Ca2+ biosensor GCaMP6 to monitor Ca2+-signaling events in the neurons of PVN brain slices, we identified a reduction in angiotensin II type 1 receptor-mediated Ca2+ activity as part of the mechanism by which PVN-PRR knockdown attenuates hypertension. Our study demonstrates an essential role of the PRR in PVN neurons in hypertension through regulation of ERK1/2 activation and angiotensin II type 1 receptor-mediated Ca2+ activity. NEW & NOTEWORTHY PRR knockdown in PVN neurons attenuates the development of DOCA-salt hypertension and autonomic dysfunction through a decrease in ERK1/2 activation in the PVN and RVLM during hypertension. In addition, PRR knockdown reduced AT1aR expression and AT1R-mediated calcium activity during hypertension. Furthermore, we characterized the neuronal targeting specificity of AAV serotype 2 in the mouse PVN and validated the advantages of the genetically encoded calcium biosensor GCaMP6 in visualizing neuronal calcium activity in the PVN.
Collapse
Affiliation(s)
- Lucas A C Souza
- Department of Pharmacology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Center for Cardiovascular Research, University of Nevada, Reno, School of Medicine , Reno, Nevada
| | - Caleb J Worker
- Department of Pharmacology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Center for Cardiovascular Research, University of Nevada, Reno, School of Medicine , Reno, Nevada
| | - Wencheng Li
- Department of Pathology, Wake Forest University , Winston-Salem, North Carolina
| | - Fatima Trebak
- Department of Pharmacology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Center for Cardiovascular Research, University of Nevada, Reno, School of Medicine , Reno, Nevada
| | - Trevor Watkins
- Department of Pharmacology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Center for Cardiovascular Research, University of Nevada, Reno, School of Medicine , Reno, Nevada
| | - Ariana Julia B Gayban
- Department of Pharmacology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Center for Cardiovascular Research, University of Nevada, Reno, School of Medicine , Reno, Nevada
| | - Evan Yamasaki
- Department of Pharmacology, University of Nevada, Reno, School of Medicine , Reno, Nevada
| | - Silvana G Cooper
- Department of Pharmacology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Center for Cardiovascular Research, University of Nevada, Reno, School of Medicine , Reno, Nevada
| | - Bernard T Drumm
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine , Reno, Nevada
| | - Yumei Feng
- Department of Pharmacology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine , Reno, Nevada.,Center for Cardiovascular Research, University of Nevada, Reno, School of Medicine , Reno, Nevada
| |
Collapse
|
32
|
Li C, Li Y, Zhao Z, Lv Y, Gu B, Zhao L. Aerobic exercise regulates synaptic transmission and reactive oxygen species production in the paraventricular nucleus of spontaneously hypertensive rats. Brain Res 2019; 1712:82-92. [PMID: 30735639 DOI: 10.1016/j.brainres.2019.02.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 01/29/2019] [Accepted: 02/04/2019] [Indexed: 02/07/2023]
Abstract
Aerobic exercise lowers blood pressure in patients with hypertension, but the underlying mechanisms remain incompletely understood. The hypothalamic paraventricular nucleus (PVN) plays a key role in the control of sympathetic outflow and cardiovascular tone. We examined whether chronic aerobic exercise altered synaptic transmission and reactive oxygen species (ROS) production in the PVN. In the present study, spontaneously hypertensive rats (SHRs) were subjected to exercise training for 8 weeks, five times per week, with Wistar Kyoto (WKY) rats as the cohort control. Miniature excitatory and inhibitory postsynaptic currents (mEPSCs and mIPSCs) were recorded from the PVN in ex vivo hypothalamic slice preparations obtained after the last training, and biomarkers of oxidative stress and physical indexes were observed. The mean frequency and amplitude, as well as the rise time and the decay time constant of mIPSCs, significantly decreased in 20-wk-old SHRs compared to WKY 20-wk-old controls. In contrast to mIPSCs, only the mean mEPSC frequency was higher, and there were no other changes in mEPSCs in comparison to the control group. SHRs exhibited higher ROS, 8-OHdG, and MDA; and lower SOD1, SOD2, CAT, Ogg1, and SOD and CAT activity in the PVN. These SHRs also had a significant increase in heart rate, blood pressure and sympathetic nerve activity, and higher levels of norepinephrine (NE). Exercise training ameliorated all these abnormalities, resulting in an increase in the mean frequency, amplitude and kinetics of mIPSCs, accompanied by a decrease in the mean frequency of mEPSCs in the PVN. This study demonstrates that moderate intensity, high frequency exercise training induces a selective enhancement of inhibitory synaptic transmission in the PVN, which may dampen sympathetic activity and reduce blood pressure in hypertension. These changes may be due to antioxidant-related adaptations in the PVNs of SHRs.
Collapse
Affiliation(s)
- Cui Li
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Yan Li
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
| | - Ziqi Zhao
- College of Life Science, University of Chinese Academy of Science, Beijing, China
| | - Yuanyuan Lv
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing, China
| | - Boya Gu
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing, China
| | - Li Zhao
- Department of Exercise Physiology, Beijing Sport University, Beijing, China; Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing, China.
| |
Collapse
|
33
|
Rossi NF, Zenner Z, Rishi AK, Levi E, Maliszewska-Scislo M. AT 1 receptors in the subfornical organ modulate arterial pressure and the baroreflex in two-kidney, one-clip hypertensive rats. Am J Physiol Regul Integr Comp Physiol 2019; 316:R172-R185. [PMID: 30624974 DOI: 10.1152/ajpregu.00289.2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The subfornical organ (SFO), a forebrain circumventricular organ that lies outside the blood-brain barrier, has been implicated in arterial pressure and baroreflex responses to angiotensin II (ANG II). We tested whether pharmacological inhibition or selective silencing of SFO ANG II type 1 receptors (AT1R) of two-kidney, one-clip rats with elevated plasma ANG II decreases resting arterial pressure and renal sympathetic nerve activity (RSNA) and/or modulates arterial baroreflex responses of heart rate (HR) and RSNA. Male Sprague-Dawley rats underwent renal artery clipping [2-kidney, 1-clip (2K,1C)] or sham clipping (sham). After 6 wk, conscious rats instrumented with vascular catheters, renal nerve electrodes, and a cannula directed to the SFO were studied. In another set of experiments, rats were instrumented with hemodynamic and nerve radio transmitters and injected with scrambled RNA or silencing RNA targeted against AT1R. Mean arterial pressure (MAP) was significantly higher in 2K,1C rats. Acute SFO injection with the AT1R inhibitor losartan did not change MAP in sham or 2K,1C rats. Baroreflex curves of HR and RSNA were shifted rightward in 2K,1C rats. Losartan exerted no effect. SFO AT1R knockdown did not influence MAP in sham rats but decreased MAP in 2K,1C rats, despite no change in plasma ANG II or resting RSNA. AT1R knockdown prevented the reduction in maximum gain and slope of baroreflex responses of HR and RSNA; the reduced RSNA response to baroreceptor unloading was partially restored in 2K,1C rats. These findings show that AT1R activation within the SFO contributes to hypertension and baroreflex dysfunction in 2K,1C rats and highlight the temporal requirement for reversal of these effects.
Collapse
Affiliation(s)
- Noreen F Rossi
- Departments of Internal Medicine and Physiology, Wayne State University School of Medicine , Detroit, Michigan.,John D. Dingell Veterans Administration Medical Center , Detroit, Michigan
| | - Zachary Zenner
- Departments of Internal Medicine and Physiology, Wayne State University School of Medicine , Detroit, Michigan
| | - Arun K Rishi
- Department of Oncology, Wayne State University School of Medicine , Detroit, Michigan.,John D. Dingell Veterans Administration Medical Center , Detroit, Michigan
| | - Edi Levi
- Department of Pathology, Wayne State University School of Medicine , Detroit, Michigan.,John D. Dingell Veterans Administration Medical Center , Detroit, Michigan
| | - Maria Maliszewska-Scislo
- Departments of Internal Medicine and Physiology, Wayne State University School of Medicine , Detroit, Michigan
| |
Collapse
|
34
|
Forrester SJ, Booz GW, Sigmund CD, Coffman TM, Kawai T, Rizzo V, Scalia R, Eguchi S. Angiotensin II Signal Transduction: An Update on Mechanisms of Physiology and Pathophysiology. Physiol Rev 2018; 98:1627-1738. [PMID: 29873596 DOI: 10.1152/physrev.00038.2017] [Citation(s) in RCA: 720] [Impact Index Per Article: 102.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The renin-angiotensin-aldosterone system plays crucial roles in cardiovascular physiology and pathophysiology. However, many of the signaling mechanisms have been unclear. The angiotensin II (ANG II) type 1 receptor (AT1R) is believed to mediate most functions of ANG II in the system. AT1R utilizes various signal transduction cascades causing hypertension, cardiovascular remodeling, and end organ damage. Moreover, functional cross-talk between AT1R signaling pathways and other signaling pathways have been recognized. Accumulating evidence reveals the complexity of ANG II signal transduction in pathophysiology of the vasculature, heart, kidney, and brain, as well as several pathophysiological features, including inflammation, metabolic dysfunction, and aging. In this review, we provide a comprehensive update of the ANG II receptor signaling events and their functional significances for potential translation into therapeutic strategies. AT1R remains central to the system in mediating physiological and pathophysiological functions of ANG II, and participation of specific signaling pathways becomes much clearer. There are still certain limitations and many controversies, and several noteworthy new concepts require further support. However, it is expected that rigorous translational research of the ANG II signaling pathways including those in large animals and humans will contribute to establishing effective new therapies against various diseases.
Collapse
Affiliation(s)
- Steven J Forrester
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - George W Booz
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Curt D Sigmund
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Thomas M Coffman
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| |
Collapse
|
35
|
Haspula D, Clark MA. Molecular Basis of the Brain Renin Angiotensin System in Cardiovascular and Neurologic Disorders: Uncovering a Key Role for the Astroglial Angiotensin Type 1 Receptor AT1R. J Pharmacol Exp Ther 2018; 366:251-264. [PMID: 29752427 DOI: 10.1124/jpet.118.248831] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/08/2018] [Indexed: 12/13/2022] Open
Abstract
The central renin angiotensin system (RAS) is one of the most widely investigated cardiovascular systems in the brain. It is implicated in a myriad of cardiovascular diseases. However, studies from the last decade have identified its involvement in several neurologic abnormalities. Understanding the molecular functionality of the various RAS components can thus provide considerable insight into the phenotypic differences and mechanistic drivers of not just cardiovascular but also neurologic disorders. Since activation of one of its primary receptors, the angiotensin type 1 receptor (AT1R), results in an augmentation of oxidative stress and inflammatory cytokines, it becomes essential to investigate not just neuronal RAS but glial RAS as well. Glial cells are key homeostatic regulators in the brain and are critical players in the resolution of overt oxidative stress and neuroinflammation. Designing better and effective therapeutic strategies that target the brain RAS could well hinge on understanding the molecular basis of both neuronal and glial RAS. This review provides a comprehensive overview of the major studies that have investigated the mechanisms and regulation of the brain RAS, and it also provides insight into the potential role of glial AT1Rs in the pathophysiology of cardiovascular and neurologic disorders.
Collapse
Affiliation(s)
- Dhanush Haspula
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, Wisconsin (D.H.); and College of Pharmacy, Department of Pharmaceutical Sciences, Nova Southeastern University, Ft. Lauderdale, Florida (M.A.C.)
| | - Michelle A Clark
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, Wisconsin (D.H.); and College of Pharmacy, Department of Pharmaceutical Sciences, Nova Southeastern University, Ft. Lauderdale, Florida (M.A.C.)
| |
Collapse
|
36
|
Chan SHH, Chan JYH. Mitochondria and Reactive Oxygen Species Contribute to Neurogenic Hypertension. Physiology (Bethesda) 2018; 32:308-321. [PMID: 28615314 DOI: 10.1152/physiol.00006.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 04/05/2017] [Accepted: 04/13/2017] [Indexed: 02/07/2023] Open
Abstract
Beyond its primary role as fuel generators, mitochondria are engaged in a variety of cellular processes, including redox homeostasis. Mitochondrial dysfunction, therefore, may have a profound impact on high-energy-demanding organs such as the brain. Here, we review the roles of mitochondrial biogenesis and bioenergetics, and their associated signaling in cellular redox homeostasis, and illustrate their contributions to the oxidative stress-related neural mechanism of hypertension, focusing on specific brain areas that are involved in the generation or modulation of sympathetic outflows to the cardiovascular system. We also highlight future challenges of research on mitochondrial physiology and pathophysiology.
Collapse
Affiliation(s)
- Samuel H H Chan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Julie Y H Chan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| |
Collapse
|
37
|
Sandgren JA, Linggonegoro DW, Zhang SY, Sapouckey SA, Claflin KE, Pearson NA, Leidinger MR, Pierce GL, Santillan MK, Gibson-Corley KN, Sigmund CD, Grobe JL. Angiotensin AT 1A receptors expressed in vasopressin-producing cells of the supraoptic nucleus contribute to osmotic control of vasopressin. Am J Physiol Regul Integr Comp Physiol 2018; 314:R770-R780. [PMID: 29364700 PMCID: PMC6032302 DOI: 10.1152/ajpregu.00435.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/03/2018] [Accepted: 01/17/2018] [Indexed: 11/22/2022]
Abstract
Angiotensin II (ANG) stimulates the release of arginine vasopressin (AVP) from the neurohypophysis through activation of the AT1 receptor within the brain, although it remains unclear whether AT1 receptors expressed on AVP-expressing neurons directly mediate this control. We explored the hypothesis that ANG acts through AT1A receptors expressed directly on AVP-producing cells to regulate AVP secretion. In situ hybridization and transgenic mice demonstrated localization of AVP and AT1A mRNA in the supraoptic nucleus (SON) and the paraventricular nucleus (PVN), but coexpression of both AVP and AT1A mRNA was only observed in the SON. Mice harboring a conditional allele for the gene encoding the AT1A receptor (AT1Aflox) were then crossed with AVP-Cre mice to generate mice that lack AT1A in all cells that express the AVP gene (AT1AAVP-KO). AT1AAVP-KO mice exhibited spontaneously increased plasma and serum osmolality but no changes in fluid or salt-intake behaviors, hematocrit, or total body water. AT1AAVP-KO mice exhibited reduced AVP secretion (estimated by measurement of copeptin) in response to osmotic stimuli such as acute hypertonic saline loading and in response to chronic intracerebroventricular ANG infusion. However, the effects of these receptors on AVP release were masked by complex stimuli such as overnight dehydration and DOCA-salt treatment, which simultaneously induce osmotic, volemic, and pressor stresses. Collectively, these data support the expression of AT1A in AVP-producing cells of the SON but not the PVN, and a role for AT1A receptors in these cells in the osmotic regulation of AVP secretion.
Collapse
MESH Headings
- Angiotensin II/administration & dosage
- Angiotensin II/pharmacology
- Animals
- Body Water
- Feeding Behavior
- Injections, Intraventricular
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Osmosis
- Paraventricular Hypothalamic Nucleus/metabolism
- Receptor, Angiotensin, Type 1/biosynthesis
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/physiology
- Sodium, Dietary
- Supraoptic Nucleus/metabolism
- Supraoptic Nucleus/physiology
- Vasoconstrictor Agents/administration & dosage
- Vasoconstrictor Agents/pharmacology
- Vasopressins/biosynthesis
- Vasopressins/physiology
Collapse
Affiliation(s)
| | | | - Shao Yang Zhang
- Department of Pharmacology, University of Iowa , Iowa City, Iowa
| | | | | | - Nicole A Pearson
- Department of Pharmacology, University of Iowa , Iowa City, Iowa
| | | | - Gary L Pierce
- Department of Health and Human Physiology, University of Iowa , Iowa City, Iowa
- Center for Hypertension Research, University of Iowa , Iowa City, Iowa
- François M. Abboud Cardiovascular Research Center, University of Iowa , Iowa City, Iowa
| | - Mark K Santillan
- Department of Obstetrics and Gynecology, University of Iowa , Iowa City, Iowa
- Center for Hypertension Research, University of Iowa , Iowa City, Iowa
| | - Katherine N Gibson-Corley
- Department of Pathology, University of Iowa , Iowa City, Iowa
- Center for Hypertension Research, University of Iowa , Iowa City, Iowa
- Fraternal Order of Eagles' Diabetes Research Center, University of Iowa , Iowa City, Iowa
| | - Curt D Sigmund
- Department of Pharmacology, University of Iowa , Iowa City, Iowa
- Center for Hypertension Research, University of Iowa , Iowa City, Iowa
- François M. Abboud Cardiovascular Research Center, University of Iowa , Iowa City, Iowa
| | - Justin L Grobe
- Department of Pharmacology, University of Iowa , Iowa City, Iowa
- Center for Hypertension Research, University of Iowa , Iowa City, Iowa
- François M. Abboud Cardiovascular Research Center, University of Iowa , Iowa City, Iowa
- Iowa Neuroscience Institute, University of Iowa , Iowa City, Iowa
- Obesity Research and Education Initiative, University of Iowa , Iowa City, Iowa
- Fraternal Order of Eagles' Diabetes Research Center, University of Iowa , Iowa City, Iowa
| |
Collapse
|
38
|
Shen YH, Chen XR, Yang CX, Liu BX, Li P. Alamandine injected into the paraventricular nucleus increases blood pressure and sympathetic activation in spontaneously hypertensive rats. Peptides 2018; 103:98-102. [PMID: 29580957 DOI: 10.1016/j.peptides.2018.03.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 03/19/2018] [Accepted: 03/22/2018] [Indexed: 11/24/2022]
Abstract
Alamandine is a newly discovered new component of the renin-angiotensin (Ang) system (RAS) that has been shown to exert vasoactive effects in some areas of the nervous system. The present study investigated whether administration of alamandine to the hypothalamic paraventricular nucleus (PVN) modulates blood pressure and sympathetic activity. Mean arterial pressure (MAP) and renal sympathetic nerve activity (RSNA) were recorded in anaesthetized rats. PVN microinjection of alamandine increased MAP and RSNA both in Wistar-Kyoto (WKY) rats and in spontaneously hypertensive rats (SHRs), but to a greater extent in SHRs. Moreover, these effects were blocked by pretreatment with alamandine receptor Mas-related G-protein-coupled receptor, member D (MrgD) antagonist D-Pro7-Ang-(1-7), adenylyl cyclase (AC) inhibitor SQ22536, and protein kinase A (PKA) inhibitor rp-adenosine-3',5'-cyclic monophosphorothionate (Rp-cAMP). Treatment with D-Pro7-Ang-(1-7), SQ22536, or Rp-cAMP alone in PVN decreased MAP and RSNA in the SHRs. Conversely cAMP alone increased MAP and RSNA, and pretreatment with cAMP enhanced alamandine's effects. These results indicate that microinjection of alamandine into the PVN increases blood pressure and sympathetic outflow via MrgD and the cAMP-PKA pathway.
Collapse
Affiliation(s)
- Yi-Hui Shen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xi-Ru Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chun-Xi Yang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Bo-Xun Liu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Peng Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
39
|
Yu Y, Wei SG, Weiss RM, Felder RB. Angiotensin II Type 1a Receptors in the Subfornical Organ Modulate Neuroinflammation in the Hypothalamic Paraventricular Nucleus in Heart Failure Rats. Neuroscience 2018; 381:46-58. [PMID: 29684507 DOI: 10.1016/j.neuroscience.2018.04.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 04/06/2018] [Accepted: 04/11/2018] [Indexed: 11/30/2022]
Abstract
Inflammation in the hypothalamic paraventricular nucleus (PVN) contributes to neurohumoral excitation and its adverse consequences in systolic heart failure (HF). The stimuli that trigger inflammation in the PVN in HF are not well understood. Angiotensin II (AngII) has pro-inflammatory effects, and circulating levels of AngII increase in HF. The subfornical organ (SFO), a circumventricular structure that lacks an effective blood-brain barrier and senses circulating AngII, contains PVN-projecting neurons. We hypothesized that activation of AngII type 1a receptors (AT1aR) in the SFO induces neuroinflammation downstream in the PVN. Male rats received SFO microinjections of an adeno-associated virus carrying shRNA for AT1aR, a scrambled shRNA, or vehicle. One week later, some rats were euthanized to confirm the transfection potential and knockdown efficiency of the shRNA. Others underwent coronary artery ligation to induce HF or a sham coronary artery ligation (Sham). Four weeks later, HF rats that received the scrambled shRNA had increased mRNA in SFO and PVN for AT1aR, inflammatory mediators and indicators of neuronal and glial activation, increased plasma levels of AngII, tumor necrosis factor-α, norepinephrine and arginine vasopressin, and impaired cardiac function, compared with Sham rats that received scrambled shRNA. The central abnormalities were ameliorated in HF rats that received AT1aR shRNA, as were plasma norepinephrine and vasopressin. Sham rats that received AT1aR shRNA had reduced SFO AT1aR mRNA but no other changes compared with Sham rats that received scrambled shRNA. The results suggest that activation of AT1aR in the SFO upregulates the neuroinflammation in the PVN that contributes to neurohumoral excitation in HF.
Collapse
Affiliation(s)
- Yang Yu
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA, USA.
| | - Shun-Guang Wei
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA, USA.
| | - Robert M Weiss
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA, USA.
| | - Robert B Felder
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA, USA; Research Service, Veterans Affairs Medical Center, 601 Highway 6 West, Iowa City, IA, USA.
| |
Collapse
|
40
|
Sun JC, Liu B, Zhang RW, Jiao PL, Tan X, Wang YK, Wang WZ. Overexpression of ß-Arrestin1 in the Rostral Ventrolateral Medulla Downregulates Angiotensin Receptor and Lowers Blood Pressure in Hypertension. Front Physiol 2018; 9:297. [PMID: 29643817 PMCID: PMC5882868 DOI: 10.3389/fphys.2018.00297] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 03/13/2018] [Indexed: 12/25/2022] Open
Abstract
Background: Hypertension is characterized by sympathetic overactivity, which is associated with an enhancement in angiotensin receptor type I (AT1R) in the rostral ventrolateral medulla (RVLM). β-arrestin1, a canonical scaffold protein, has been suggested to show a negative effect on G protein-coupled receptors via its internalization and desensitization and/or the biased signaling pathway. The major objectives of the present study were to observe the effect of β-arrestin1 overexpression in the RVLM on cardiovascular regulation in spontaneously hypertensive rats (SHR), and further determine the effect of β-arrestin1 on AT1R expression in the RVLM. Methods: The animal model of β-arrestin1 overexpression was induced by bilateral injection of adeno-associated virus containing Arrb1 gene (AAV-Arrb1) into the RVLM of WKY and SHR. Results: β-arrestin1 was expressed on the pre-sympathetic neurons in the RVLM, and its expression in the RVLM was significantly (P < 0.05) downregulated by an average of 64% in SHR than WKY. Overexpression of β-arrestin1 in SHR significantly decreased baseline levels of blood pressure and renal sympathetic nerve activity, and attenuated cardiovascular effects induced by RVLM injection of angiotensin II (100 pmol). Furthermore, β-arrestin1 overexpression in the RVLM significantly reduced the expression of AT1R by 65% and NF-κB p65 phosphorylation by 66% in SHR. It was confirmed that β-arrestin1 overexpression in the RVLM led to an enhancement of interaction between β-arrestin1 and IκB-α. Conclusion: Overexpression of β-arrestin1 in the RVLM reduces BP and sympathetic outflow in hypertension, which may be associated with NFκB-mediated AT1R downregulation.
Collapse
Affiliation(s)
- Jia-Cen Sun
- Department of Physiology and Center of Polar Medical Research, Second Military Medical University, Shanghai, China
| | - Bing Liu
- Department of Physiology and Center of Polar Medical Research, Second Military Medical University, Shanghai, China
| | - Ru-Wen Zhang
- Department of Physiology and Center of Polar Medical Research, Second Military Medical University, Shanghai, China
| | - Pei-Lei Jiao
- Department of Physiology and Center of Polar Medical Research, Second Military Medical University, Shanghai, China
| | - Xing Tan
- Department of Physiology and Center of Polar Medical Research, Second Military Medical University, Shanghai, China
| | - Yang-Kai Wang
- Department of Physiology and Center of Polar Medical Research, Second Military Medical University, Shanghai, China
| | - Wei-Zhong Wang
- Department of Physiology and Center of Polar Medical Research, Second Military Medical University, Shanghai, China
| |
Collapse
|
41
|
Leenen FHH, Blaustein MP, Hamlyn JM. Update on angiotensin II: new endocrine connections between the brain, adrenal glands and the cardiovascular system. Endocr Connect 2017; 6:R131-R145. [PMID: 28855243 PMCID: PMC5613704 DOI: 10.1530/ec-17-0161] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 08/30/2017] [Indexed: 12/11/2022]
Abstract
In the brain, angiotensinergic pathways play a major role in chronic regulation of cardiovascular and electrolyte homeostasis. Increases in plasma angiotensin II (Ang II), aldosterone, [Na+] and cytokines can directly activate these pathways. Chronically, these stimuli also activate a slow neuromodulatory pathway involving local aldosterone, mineralocorticoid receptors (MRs), epithelial sodium channels and endogenous ouabain (EO). This pathway increases AT1R and NADPH oxidase subunits and maintains/further increases the activity of angiotensinergic pathways. These brain pathways not only increase the setpoint of sympathetic activity per se, but also enhance its effectiveness by increasing plasma EO and EO-dependent reprogramming of arterial and cardiac function. Blockade of any step in this slow pathway or of AT1R prevents Ang II-, aldosterone- or salt and renal injury-induced forms of hypertension. MR/AT1R activation in the CNS also contributes to the activation of sympathetic activity, the circulatory and cardiac RAAS and increase in circulating cytokines in HF post MI. Chronic central infusion of an aldosterone synthase inhibitor, MR blocker or AT1R blocker prevents a major part of the structural remodeling of the heart and the decrease in LV function post MI, indicating that MR activation in the CNS post MI depends on aldosterone, locally produced in the CNS. Thus, Ang II, aldosterone and EO are not simply circulating hormones that act on the CNS but rather they are also paracrine neurohormones, locally produced in the CNS, that exert powerful effects in key CNS pathways involved in the long-term control of sympathetic and neuro-endocrine function and cardiovascular homeostasis.
Collapse
Affiliation(s)
- Frans H H Leenen
- Brain and Heart Research GroupUniversity of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Mordecai P Blaustein
- Department of PhysiologyUniversity of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of MedicineUniversity of Maryland School of Medicine, Baltimore, Maryland, USA
| | - John M Hamlyn
- Department of PhysiologyUniversity of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
42
|
Hypothalamic and inflammatory basis of hypertension. Clin Sci (Lond) 2017; 131:211-223. [PMID: 28057892 DOI: 10.1042/cs20160001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 11/07/2016] [Accepted: 11/21/2016] [Indexed: 02/07/2023]
Abstract
Hypertension is a major health problem with great consequences for public health. Despite its role as the primary cause of significant morbidity and mortality associated with cardiovascular disease, the pathogenesis of essential hypertension remains largely unknown. The central nervous system (CNS) in general, and the hypothalamus in particular, are intricately involved in the development and maintenance of hypertension. Over the last several decades, the understanding of the brain's role in the development of hypertension has dramatically increased. This brief review is to summarize the neural mechanisms of hypertension with a focus on neuroendocrine and neurotransmitter involvement, highlighting recent findings that suggest that hypothalamic inflammation disrupts key signalling pathways to affect the central control of blood pressure, and therefore suggesting future development of interventional strategies that exploit recent findings pertaining to the hypothalamic control of blood pressure as well as the inflammatory-sympathetic mechanisms involved in hypertension.
Collapse
|
43
|
Chao Y, Ye P, Zhu L, Kong X, Qu X, Zhang J, Luo J, Yang H, Chen S. Low shear stress induces endothelial reactive oxygen species via the AT1R/eNOS/NO pathway. J Cell Physiol 2017; 233:1384-1395. [PMID: 28518223 DOI: 10.1002/jcp.26016] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 05/17/2017] [Indexed: 12/25/2022]
Abstract
Reactive oxygen species (ROS) contribute to many aspects of physiological and pathological cardiovascular processes. However, the underlying mechanism of ROS induction by low shear stress (LSS) remains unclear. Accumulating evidence has shown that the angiotensin II type 1 receptor (AT1R) is involved in inflammation, apoptosis, and ROS production. Our aim was to explore the role of AT1R in LSS-mediated ROS induction. We exposed human umbilical vein endothelial cells (HUVECs) to LSS (3 dyn/cm2 ) for different periods of time. Western blotting and immunofluorescence showed that LSS significantly induced AT1R expression in a time-dependent manner. Using immunohistochemistry, we also noted a similar increase in AT1R expression in the inner curvature of the aortic arch compared to the descending aorta in C57BL/6 mice. Additionally, HUVECs were cultured with a fluorescent probe, either DCFH, DHE or DAF, after being subjected to LSS. Cell chemiluminescence and flow cytometry results revealed that LSS stimulated ROS levels and suppressed nitric oxide (NO) generation in a time-dependent manner, which was reversed by the AT1R antagonist Losartan. We also found that Losartan markedly increased endothelial NO synthase (eNOS) phosphorylation at Ser(633,1177) and dephosphorylation at Thr(495), which involved AKT and ERK. Moreover, the ROS level was significantly reduced by endogenous and exogenous NO donors (L-arginine, SNP) and increased by the eNOS inhibitor L-NAME. Overall, we conclude that LSS induces ROS via AT1R/eNOS/NO.
Collapse
Affiliation(s)
- Yuelin Chao
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Peng Ye
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Linlin Zhu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiangquan Kong
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xinliang Qu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Junxia Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jie Luo
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Hongfeng Yang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Shaoliang Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
44
|
van Thiel BS, Góes Martini A, Te Riet L, Severs D, Uijl E, Garrelds IM, Leijten FPJ, van der Pluijm I, Essers J, Qadri F, Alenina N, Bader M, Paulis L, Rajkovicova R, Domenig O, Poglitsch M, Danser AHJ. Brain Renin-Angiotensin System: Does It Exist? Hypertension 2017; 69:1136-1144. [PMID: 28396529 DOI: 10.1161/hypertensionaha.116.08922] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 01/12/2017] [Accepted: 01/29/2017] [Indexed: 12/14/2022]
Abstract
Because of the presence of the blood-brain barrier, brain renin-angiotensin system activity should depend on local (pro)renin synthesis. Indeed, an intracellular form of renin has been described in the brain, but whether it displays angiotensin (Ang) I-generating activity (AGA) is unknown. Here, we quantified brain (pro)renin, before and after buffer perfusion of the brain, in wild-type mice, renin knockout mice, deoxycorticosterone acetate salt-treated mice, and Ang II-infused mice. Brain regions were homogenized and incubated with excess angiotensinogen to detect AGA, before and after prorenin activation, using a renin inhibitor to correct for nonrenin-mediated AGA. Renin-dependent AGA was readily detectable in brain regions, the highest AGA being present in brain stem (>thalamus=cerebellum=striatum=midbrain>hippocampus=cortex). Brain AGA increased marginally after prorenin activation, suggesting that brain prorenin is low. Buffer perfusion reduced AGA in all brain areas by >60%. Plasma renin (per mL) was 40× to 800× higher than brain renin (per gram). Renin was undetectable in plasma and brain of renin knockout mice. Deoxycorticosterone acetate salt and Ang II suppressed plasma renin and brain renin in parallel, without upregulating brain prorenin. Finally, Ang I was undetectable in brains of spontaneously hypertensive rats, while their brain/plasma Ang II concentration ratio decreased by 80% after Ang II type 1 receptor blockade. In conclusion, brain renin levels (per gram) correspond with the amount of renin present in 1 to 20 μL of plasma. Brain renin disappears after buffer perfusion and varies in association with plasma renin. This indicates that brain renin represents trapped plasma renin. Brain Ang II represents Ang II taken up from blood rather than locally synthesized Ang II.
Collapse
Affiliation(s)
- Bibi S van Thiel
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (B.S.v.T., A.G.M., L.t.R., D.S., E.U., I.M.G., F.P.J.L., A.H.J.D.), Department of Vascular Surgery (B.S.v.T., L.t.R., I.v.d.P., J.E.), Department of Molecular Genetics, Cancer Genomics Center Netherlands (B.S.v.T., I.v.d.P., J.E.), Division of Nephrology and Transplantation, Department of Internal Medicine (D.S., E.U.), Department of Radiation Oncology (J.E.), Erasmus MC, Rotterdam, The Netherlands; Department of Molecular Cardiovascular Endocrinology, Max Delbrück Center, Berlin, Germany (F.Q., N.A., M.B.); DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Germany (N.A., M.B.); Berlin Institute of Health (BIH), Germany (M.B.); Charité-University Medicine, Berlin, Germany (M.B.); Institute for Biology, University of Lübeck, Germany (M.B.); Institute of Pathophysiology, Faculty of Medicine, Comenius University (L.P., R.R.); Institute of Normal and Pathophysiological Physiology, Slovak Academy of Sciences, Bratislava, Slovak Republic (L.P.); and Attoquant Diagnostics (O.D., M.P.) and Department of Internal Medicine III (O.D.), Medical University of Vienna, Austria
| | - Alexandre Góes Martini
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (B.S.v.T., A.G.M., L.t.R., D.S., E.U., I.M.G., F.P.J.L., A.H.J.D.), Department of Vascular Surgery (B.S.v.T., L.t.R., I.v.d.P., J.E.), Department of Molecular Genetics, Cancer Genomics Center Netherlands (B.S.v.T., I.v.d.P., J.E.), Division of Nephrology and Transplantation, Department of Internal Medicine (D.S., E.U.), Department of Radiation Oncology (J.E.), Erasmus MC, Rotterdam, The Netherlands; Department of Molecular Cardiovascular Endocrinology, Max Delbrück Center, Berlin, Germany (F.Q., N.A., M.B.); DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Germany (N.A., M.B.); Berlin Institute of Health (BIH), Germany (M.B.); Charité-University Medicine, Berlin, Germany (M.B.); Institute for Biology, University of Lübeck, Germany (M.B.); Institute of Pathophysiology, Faculty of Medicine, Comenius University (L.P., R.R.); Institute of Normal and Pathophysiological Physiology, Slovak Academy of Sciences, Bratislava, Slovak Republic (L.P.); and Attoquant Diagnostics (O.D., M.P.) and Department of Internal Medicine III (O.D.), Medical University of Vienna, Austria
| | - Luuk Te Riet
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (B.S.v.T., A.G.M., L.t.R., D.S., E.U., I.M.G., F.P.J.L., A.H.J.D.), Department of Vascular Surgery (B.S.v.T., L.t.R., I.v.d.P., J.E.), Department of Molecular Genetics, Cancer Genomics Center Netherlands (B.S.v.T., I.v.d.P., J.E.), Division of Nephrology and Transplantation, Department of Internal Medicine (D.S., E.U.), Department of Radiation Oncology (J.E.), Erasmus MC, Rotterdam, The Netherlands; Department of Molecular Cardiovascular Endocrinology, Max Delbrück Center, Berlin, Germany (F.Q., N.A., M.B.); DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Germany (N.A., M.B.); Berlin Institute of Health (BIH), Germany (M.B.); Charité-University Medicine, Berlin, Germany (M.B.); Institute for Biology, University of Lübeck, Germany (M.B.); Institute of Pathophysiology, Faculty of Medicine, Comenius University (L.P., R.R.); Institute of Normal and Pathophysiological Physiology, Slovak Academy of Sciences, Bratislava, Slovak Republic (L.P.); and Attoquant Diagnostics (O.D., M.P.) and Department of Internal Medicine III (O.D.), Medical University of Vienna, Austria
| | - David Severs
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (B.S.v.T., A.G.M., L.t.R., D.S., E.U., I.M.G., F.P.J.L., A.H.J.D.), Department of Vascular Surgery (B.S.v.T., L.t.R., I.v.d.P., J.E.), Department of Molecular Genetics, Cancer Genomics Center Netherlands (B.S.v.T., I.v.d.P., J.E.), Division of Nephrology and Transplantation, Department of Internal Medicine (D.S., E.U.), Department of Radiation Oncology (J.E.), Erasmus MC, Rotterdam, The Netherlands; Department of Molecular Cardiovascular Endocrinology, Max Delbrück Center, Berlin, Germany (F.Q., N.A., M.B.); DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Germany (N.A., M.B.); Berlin Institute of Health (BIH), Germany (M.B.); Charité-University Medicine, Berlin, Germany (M.B.); Institute for Biology, University of Lübeck, Germany (M.B.); Institute of Pathophysiology, Faculty of Medicine, Comenius University (L.P., R.R.); Institute of Normal and Pathophysiological Physiology, Slovak Academy of Sciences, Bratislava, Slovak Republic (L.P.); and Attoquant Diagnostics (O.D., M.P.) and Department of Internal Medicine III (O.D.), Medical University of Vienna, Austria
| | - Estrellita Uijl
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (B.S.v.T., A.G.M., L.t.R., D.S., E.U., I.M.G., F.P.J.L., A.H.J.D.), Department of Vascular Surgery (B.S.v.T., L.t.R., I.v.d.P., J.E.), Department of Molecular Genetics, Cancer Genomics Center Netherlands (B.S.v.T., I.v.d.P., J.E.), Division of Nephrology and Transplantation, Department of Internal Medicine (D.S., E.U.), Department of Radiation Oncology (J.E.), Erasmus MC, Rotterdam, The Netherlands; Department of Molecular Cardiovascular Endocrinology, Max Delbrück Center, Berlin, Germany (F.Q., N.A., M.B.); DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Germany (N.A., M.B.); Berlin Institute of Health (BIH), Germany (M.B.); Charité-University Medicine, Berlin, Germany (M.B.); Institute for Biology, University of Lübeck, Germany (M.B.); Institute of Pathophysiology, Faculty of Medicine, Comenius University (L.P., R.R.); Institute of Normal and Pathophysiological Physiology, Slovak Academy of Sciences, Bratislava, Slovak Republic (L.P.); and Attoquant Diagnostics (O.D., M.P.) and Department of Internal Medicine III (O.D.), Medical University of Vienna, Austria
| | - Ingrid M Garrelds
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (B.S.v.T., A.G.M., L.t.R., D.S., E.U., I.M.G., F.P.J.L., A.H.J.D.), Department of Vascular Surgery (B.S.v.T., L.t.R., I.v.d.P., J.E.), Department of Molecular Genetics, Cancer Genomics Center Netherlands (B.S.v.T., I.v.d.P., J.E.), Division of Nephrology and Transplantation, Department of Internal Medicine (D.S., E.U.), Department of Radiation Oncology (J.E.), Erasmus MC, Rotterdam, The Netherlands; Department of Molecular Cardiovascular Endocrinology, Max Delbrück Center, Berlin, Germany (F.Q., N.A., M.B.); DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Germany (N.A., M.B.); Berlin Institute of Health (BIH), Germany (M.B.); Charité-University Medicine, Berlin, Germany (M.B.); Institute for Biology, University of Lübeck, Germany (M.B.); Institute of Pathophysiology, Faculty of Medicine, Comenius University (L.P., R.R.); Institute of Normal and Pathophysiological Physiology, Slovak Academy of Sciences, Bratislava, Slovak Republic (L.P.); and Attoquant Diagnostics (O.D., M.P.) and Department of Internal Medicine III (O.D.), Medical University of Vienna, Austria
| | - Frank P J Leijten
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (B.S.v.T., A.G.M., L.t.R., D.S., E.U., I.M.G., F.P.J.L., A.H.J.D.), Department of Vascular Surgery (B.S.v.T., L.t.R., I.v.d.P., J.E.), Department of Molecular Genetics, Cancer Genomics Center Netherlands (B.S.v.T., I.v.d.P., J.E.), Division of Nephrology and Transplantation, Department of Internal Medicine (D.S., E.U.), Department of Radiation Oncology (J.E.), Erasmus MC, Rotterdam, The Netherlands; Department of Molecular Cardiovascular Endocrinology, Max Delbrück Center, Berlin, Germany (F.Q., N.A., M.B.); DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Germany (N.A., M.B.); Berlin Institute of Health (BIH), Germany (M.B.); Charité-University Medicine, Berlin, Germany (M.B.); Institute for Biology, University of Lübeck, Germany (M.B.); Institute of Pathophysiology, Faculty of Medicine, Comenius University (L.P., R.R.); Institute of Normal and Pathophysiological Physiology, Slovak Academy of Sciences, Bratislava, Slovak Republic (L.P.); and Attoquant Diagnostics (O.D., M.P.) and Department of Internal Medicine III (O.D.), Medical University of Vienna, Austria
| | - Ingrid van der Pluijm
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (B.S.v.T., A.G.M., L.t.R., D.S., E.U., I.M.G., F.P.J.L., A.H.J.D.), Department of Vascular Surgery (B.S.v.T., L.t.R., I.v.d.P., J.E.), Department of Molecular Genetics, Cancer Genomics Center Netherlands (B.S.v.T., I.v.d.P., J.E.), Division of Nephrology and Transplantation, Department of Internal Medicine (D.S., E.U.), Department of Radiation Oncology (J.E.), Erasmus MC, Rotterdam, The Netherlands; Department of Molecular Cardiovascular Endocrinology, Max Delbrück Center, Berlin, Germany (F.Q., N.A., M.B.); DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Germany (N.A., M.B.); Berlin Institute of Health (BIH), Germany (M.B.); Charité-University Medicine, Berlin, Germany (M.B.); Institute for Biology, University of Lübeck, Germany (M.B.); Institute of Pathophysiology, Faculty of Medicine, Comenius University (L.P., R.R.); Institute of Normal and Pathophysiological Physiology, Slovak Academy of Sciences, Bratislava, Slovak Republic (L.P.); and Attoquant Diagnostics (O.D., M.P.) and Department of Internal Medicine III (O.D.), Medical University of Vienna, Austria
| | - Jeroen Essers
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (B.S.v.T., A.G.M., L.t.R., D.S., E.U., I.M.G., F.P.J.L., A.H.J.D.), Department of Vascular Surgery (B.S.v.T., L.t.R., I.v.d.P., J.E.), Department of Molecular Genetics, Cancer Genomics Center Netherlands (B.S.v.T., I.v.d.P., J.E.), Division of Nephrology and Transplantation, Department of Internal Medicine (D.S., E.U.), Department of Radiation Oncology (J.E.), Erasmus MC, Rotterdam, The Netherlands; Department of Molecular Cardiovascular Endocrinology, Max Delbrück Center, Berlin, Germany (F.Q., N.A., M.B.); DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Germany (N.A., M.B.); Berlin Institute of Health (BIH), Germany (M.B.); Charité-University Medicine, Berlin, Germany (M.B.); Institute for Biology, University of Lübeck, Germany (M.B.); Institute of Pathophysiology, Faculty of Medicine, Comenius University (L.P., R.R.); Institute of Normal and Pathophysiological Physiology, Slovak Academy of Sciences, Bratislava, Slovak Republic (L.P.); and Attoquant Diagnostics (O.D., M.P.) and Department of Internal Medicine III (O.D.), Medical University of Vienna, Austria
| | - Fatimunnisa Qadri
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (B.S.v.T., A.G.M., L.t.R., D.S., E.U., I.M.G., F.P.J.L., A.H.J.D.), Department of Vascular Surgery (B.S.v.T., L.t.R., I.v.d.P., J.E.), Department of Molecular Genetics, Cancer Genomics Center Netherlands (B.S.v.T., I.v.d.P., J.E.), Division of Nephrology and Transplantation, Department of Internal Medicine (D.S., E.U.), Department of Radiation Oncology (J.E.), Erasmus MC, Rotterdam, The Netherlands; Department of Molecular Cardiovascular Endocrinology, Max Delbrück Center, Berlin, Germany (F.Q., N.A., M.B.); DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Germany (N.A., M.B.); Berlin Institute of Health (BIH), Germany (M.B.); Charité-University Medicine, Berlin, Germany (M.B.); Institute for Biology, University of Lübeck, Germany (M.B.); Institute of Pathophysiology, Faculty of Medicine, Comenius University (L.P., R.R.); Institute of Normal and Pathophysiological Physiology, Slovak Academy of Sciences, Bratislava, Slovak Republic (L.P.); and Attoquant Diagnostics (O.D., M.P.) and Department of Internal Medicine III (O.D.), Medical University of Vienna, Austria
| | - Natalia Alenina
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (B.S.v.T., A.G.M., L.t.R., D.S., E.U., I.M.G., F.P.J.L., A.H.J.D.), Department of Vascular Surgery (B.S.v.T., L.t.R., I.v.d.P., J.E.), Department of Molecular Genetics, Cancer Genomics Center Netherlands (B.S.v.T., I.v.d.P., J.E.), Division of Nephrology and Transplantation, Department of Internal Medicine (D.S., E.U.), Department of Radiation Oncology (J.E.), Erasmus MC, Rotterdam, The Netherlands; Department of Molecular Cardiovascular Endocrinology, Max Delbrück Center, Berlin, Germany (F.Q., N.A., M.B.); DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Germany (N.A., M.B.); Berlin Institute of Health (BIH), Germany (M.B.); Charité-University Medicine, Berlin, Germany (M.B.); Institute for Biology, University of Lübeck, Germany (M.B.); Institute of Pathophysiology, Faculty of Medicine, Comenius University (L.P., R.R.); Institute of Normal and Pathophysiological Physiology, Slovak Academy of Sciences, Bratislava, Slovak Republic (L.P.); and Attoquant Diagnostics (O.D., M.P.) and Department of Internal Medicine III (O.D.), Medical University of Vienna, Austria
| | - Michael Bader
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (B.S.v.T., A.G.M., L.t.R., D.S., E.U., I.M.G., F.P.J.L., A.H.J.D.), Department of Vascular Surgery (B.S.v.T., L.t.R., I.v.d.P., J.E.), Department of Molecular Genetics, Cancer Genomics Center Netherlands (B.S.v.T., I.v.d.P., J.E.), Division of Nephrology and Transplantation, Department of Internal Medicine (D.S., E.U.), Department of Radiation Oncology (J.E.), Erasmus MC, Rotterdam, The Netherlands; Department of Molecular Cardiovascular Endocrinology, Max Delbrück Center, Berlin, Germany (F.Q., N.A., M.B.); DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Germany (N.A., M.B.); Berlin Institute of Health (BIH), Germany (M.B.); Charité-University Medicine, Berlin, Germany (M.B.); Institute for Biology, University of Lübeck, Germany (M.B.); Institute of Pathophysiology, Faculty of Medicine, Comenius University (L.P., R.R.); Institute of Normal and Pathophysiological Physiology, Slovak Academy of Sciences, Bratislava, Slovak Republic (L.P.); and Attoquant Diagnostics (O.D., M.P.) and Department of Internal Medicine III (O.D.), Medical University of Vienna, Austria
| | - Ludovit Paulis
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (B.S.v.T., A.G.M., L.t.R., D.S., E.U., I.M.G., F.P.J.L., A.H.J.D.), Department of Vascular Surgery (B.S.v.T., L.t.R., I.v.d.P., J.E.), Department of Molecular Genetics, Cancer Genomics Center Netherlands (B.S.v.T., I.v.d.P., J.E.), Division of Nephrology and Transplantation, Department of Internal Medicine (D.S., E.U.), Department of Radiation Oncology (J.E.), Erasmus MC, Rotterdam, The Netherlands; Department of Molecular Cardiovascular Endocrinology, Max Delbrück Center, Berlin, Germany (F.Q., N.A., M.B.); DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Germany (N.A., M.B.); Berlin Institute of Health (BIH), Germany (M.B.); Charité-University Medicine, Berlin, Germany (M.B.); Institute for Biology, University of Lübeck, Germany (M.B.); Institute of Pathophysiology, Faculty of Medicine, Comenius University (L.P., R.R.); Institute of Normal and Pathophysiological Physiology, Slovak Academy of Sciences, Bratislava, Slovak Republic (L.P.); and Attoquant Diagnostics (O.D., M.P.) and Department of Internal Medicine III (O.D.), Medical University of Vienna, Austria
| | - Romana Rajkovicova
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (B.S.v.T., A.G.M., L.t.R., D.S., E.U., I.M.G., F.P.J.L., A.H.J.D.), Department of Vascular Surgery (B.S.v.T., L.t.R., I.v.d.P., J.E.), Department of Molecular Genetics, Cancer Genomics Center Netherlands (B.S.v.T., I.v.d.P., J.E.), Division of Nephrology and Transplantation, Department of Internal Medicine (D.S., E.U.), Department of Radiation Oncology (J.E.), Erasmus MC, Rotterdam, The Netherlands; Department of Molecular Cardiovascular Endocrinology, Max Delbrück Center, Berlin, Germany (F.Q., N.A., M.B.); DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Germany (N.A., M.B.); Berlin Institute of Health (BIH), Germany (M.B.); Charité-University Medicine, Berlin, Germany (M.B.); Institute for Biology, University of Lübeck, Germany (M.B.); Institute of Pathophysiology, Faculty of Medicine, Comenius University (L.P., R.R.); Institute of Normal and Pathophysiological Physiology, Slovak Academy of Sciences, Bratislava, Slovak Republic (L.P.); and Attoquant Diagnostics (O.D., M.P.) and Department of Internal Medicine III (O.D.), Medical University of Vienna, Austria
| | - Oliver Domenig
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (B.S.v.T., A.G.M., L.t.R., D.S., E.U., I.M.G., F.P.J.L., A.H.J.D.), Department of Vascular Surgery (B.S.v.T., L.t.R., I.v.d.P., J.E.), Department of Molecular Genetics, Cancer Genomics Center Netherlands (B.S.v.T., I.v.d.P., J.E.), Division of Nephrology and Transplantation, Department of Internal Medicine (D.S., E.U.), Department of Radiation Oncology (J.E.), Erasmus MC, Rotterdam, The Netherlands; Department of Molecular Cardiovascular Endocrinology, Max Delbrück Center, Berlin, Germany (F.Q., N.A., M.B.); DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Germany (N.A., M.B.); Berlin Institute of Health (BIH), Germany (M.B.); Charité-University Medicine, Berlin, Germany (M.B.); Institute for Biology, University of Lübeck, Germany (M.B.); Institute of Pathophysiology, Faculty of Medicine, Comenius University (L.P., R.R.); Institute of Normal and Pathophysiological Physiology, Slovak Academy of Sciences, Bratislava, Slovak Republic (L.P.); and Attoquant Diagnostics (O.D., M.P.) and Department of Internal Medicine III (O.D.), Medical University of Vienna, Austria
| | - Marko Poglitsch
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (B.S.v.T., A.G.M., L.t.R., D.S., E.U., I.M.G., F.P.J.L., A.H.J.D.), Department of Vascular Surgery (B.S.v.T., L.t.R., I.v.d.P., J.E.), Department of Molecular Genetics, Cancer Genomics Center Netherlands (B.S.v.T., I.v.d.P., J.E.), Division of Nephrology and Transplantation, Department of Internal Medicine (D.S., E.U.), Department of Radiation Oncology (J.E.), Erasmus MC, Rotterdam, The Netherlands; Department of Molecular Cardiovascular Endocrinology, Max Delbrück Center, Berlin, Germany (F.Q., N.A., M.B.); DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Germany (N.A., M.B.); Berlin Institute of Health (BIH), Germany (M.B.); Charité-University Medicine, Berlin, Germany (M.B.); Institute for Biology, University of Lübeck, Germany (M.B.); Institute of Pathophysiology, Faculty of Medicine, Comenius University (L.P., R.R.); Institute of Normal and Pathophysiological Physiology, Slovak Academy of Sciences, Bratislava, Slovak Republic (L.P.); and Attoquant Diagnostics (O.D., M.P.) and Department of Internal Medicine III (O.D.), Medical University of Vienna, Austria
| | - A H Jan Danser
- From the Division of Vascular Medicine and Pharmacology, Department of Internal Medicine (B.S.v.T., A.G.M., L.t.R., D.S., E.U., I.M.G., F.P.J.L., A.H.J.D.), Department of Vascular Surgery (B.S.v.T., L.t.R., I.v.d.P., J.E.), Department of Molecular Genetics, Cancer Genomics Center Netherlands (B.S.v.T., I.v.d.P., J.E.), Division of Nephrology and Transplantation, Department of Internal Medicine (D.S., E.U.), Department of Radiation Oncology (J.E.), Erasmus MC, Rotterdam, The Netherlands; Department of Molecular Cardiovascular Endocrinology, Max Delbrück Center, Berlin, Germany (F.Q., N.A., M.B.); DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Germany (N.A., M.B.); Berlin Institute of Health (BIH), Germany (M.B.); Charité-University Medicine, Berlin, Germany (M.B.); Institute for Biology, University of Lübeck, Germany (M.B.); Institute of Pathophysiology, Faculty of Medicine, Comenius University (L.P., R.R.); Institute of Normal and Pathophysiological Physiology, Slovak Academy of Sciences, Bratislava, Slovak Republic (L.P.); and Attoquant Diagnostics (O.D., M.P.) and Department of Internal Medicine III (O.D.), Medical University of Vienna, Austria.
| |
Collapse
|