101
|
Jiang L, Li Y, Wang L, Guo J, Liu W, Meng G, Zhang L, Li M, Cong L, Sun M. Recent Insights Into the Prognostic and Therapeutic Applications of Lysozymes. Front Pharmacol 2021; 12:767642. [PMID: 34925025 PMCID: PMC8678502 DOI: 10.3389/fphar.2021.767642] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/10/2021] [Indexed: 01/15/2023] Open
Abstract
Lysozymes are naturally occurring enzymes present in a variety of biological organisms, such as bacteria, fungi, and animal bodily secretions and tissues. It is also the main ingredient of many ethnomedicines. It is well known that lysozymes and lysozyme-like enzymes can be used as anti-bacterial agents by degrading bacterial cell wall peptidoglycan that leads to cell death, and can also inhibit fungi, yeasts, and viruses. In addition to its direct antimicrobial activity, lysozyme is also an important component of the innate immune system in most mammals. Increasing evidence has shown the immune-modulatory effects of lysozymes against infection and inflammation. More recently, studies have revealed the anti-cancer activities of lysozyme in multiple types of tumors, potentially through its immune-modulatory activities. In this review, we summarized the major functions and underlying mechanisms of lysozymes derived from animal and plant sources. We highlighted the therapeutic applications and recent advances of lysozymes in cancers, hypertension, and viral diseases, aiming toseeking alternative therapies for standard medical treatment bypassing side effects. We also evaluated the role of lysozyme as a promising cancer marker for prognosis to indicate the outcomes recurrence for patients.
Collapse
Affiliation(s)
- Lin Jiang
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
| | - Yunhe Li
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
| | - Liye Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Jian Guo
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
| | - Wei Liu
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
| | - Guixian Meng
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
| | - Lei Zhang
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
| | - Miao Li
- Department of Neurosurgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Lina Cong
- School of Biological Engineering, Dalian Polytechnic University, Dalian, China
| | - Meiyan Sun
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
| |
Collapse
|
102
|
Perrotta M, Carnevale D. Brain Areas Involved in Modulating the Immune Response Participating in Hypertension and Its Target Organ Damage. Antioxid Redox Signal 2021; 35:1515-1530. [PMID: 34269604 DOI: 10.1089/ars.2021.0142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Significance: Hypertension is a multifactorial disease ensuing from the continuous challenge imposed by several risk factors on the cardiovascular system. Classically known pathophysiological alterations associated with hypertension comprise neurogenic mechanisms dysregulating the autonomic nervous system (ANS), vascular dysfunction, and excessive activation of the renin angiotensin system. During the past few years, a considerable number of studies indicated that immune activation and inflammation also have an important role in the onset and maintenance of hypertension. Critical Issues: On these premises, it has been necessary to reconsider the pathophysiological mechanisms underlying hypertension development, taking into account the potential interactions established between classically known determinants of high blood pressure and the immune system. Recent Advances: Interestingly, central nervous system areas controlling cardiovascular functions are enriched with Angiotensin II receptors. Observations showing that these brain areas are crucial for mediating peripheral ANS and immune responses were suggestive of a critical role of neuroimmune interactions in hypertension. In fact, the ANS, characterized by an intricate network of afferent and efferent fibers, represents an intermediate between the brain and peripheral responses that are essential for blood pressure regulation. Future Directions: In this review, we will summarize studies showing how specific brain areas can modulate immune responses that are involved in hypertension. Antioxid. Redox Signal. 35, 1515-1530.
Collapse
Affiliation(s)
- Marialuisa Perrotta
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Daniela Carnevale
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy.,Research Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
103
|
Frenis K, Kuntic M, Hahad O, Bayo Jimenez MT, Oelze M, Daub S, Steven S, Münzel T, Daiber A. Redox Switches in Noise-Induced Cardiovascular and Neuronal Dysregulation. Front Mol Biosci 2021; 8:784910. [PMID: 34869603 PMCID: PMC8637611 DOI: 10.3389/fmolb.2021.784910] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022] Open
Abstract
Environmental exposures represent a significant health hazard, which cumulatively may be responsible for up to 2/3 of all chronic non-communicable disease and associated mortality (Global Burden of Disease Study and The Lancet Commission on Pollution and Health), which has given rise to a new concept of the exposome: the sum of environmental factors in every individual’s experience. Noise is part of the exposome and is increasingly being investigated as a health risk factor impacting neurological, cardiometabolic, endocrine, and immune health. Beyond the well-characterized effects of high-intensity noise on cochlear damage, noise is relatively well-studied in the cardiovascular field, where evidence is emerging from both human and translational experiments that noise from traffic-related sources could represent a risk factor for hypertension, ischemic heart disease, diabetes, and atherosclerosis. In the present review, we comprehensively discuss the current state of knowledge in the field of noise research. We give a brief survey of the literature documenting experiments in noise exposure in both humans and animals with a focus on cardiovascular disease. We also discuss the mechanisms that have been uncovered in recent years that describe how exposure to noise affects physiological homeostasis, leading to aberrant redox signaling resulting in metabolic and immune consequences, both of which have considerable impact on cardiovascular health. Additionally, we discuss the molecular pathways of redox involvement in the stress responses to noise and how they manifest in disruptions of the circadian rhythm, inflammatory signaling, gut microbiome composition, epigenetic landscape and vessel function.
Collapse
Affiliation(s)
- Katie Frenis
- Department of Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany.,Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
| | - Marin Kuntic
- Department of Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany
| | - Omar Hahad
- Department of Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | | | - Matthias Oelze
- Department of Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany
| | - Steffen Daub
- Department of Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany
| | - Sebastian Steven
- Department of Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| |
Collapse
|
104
|
Tian Y, Luo J, Xu Q, Liu Y, Cai R, Zhou MS. Macrophage depletion protects against endothelial dysfunction and cardiac remodeling in angiotensin II hypertensive mice. Clin Exp Hypertens 2021; 43:699-706. [PMID: 34176379 DOI: 10.1080/10641963.2021.1945075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Objective: Hypertension is associated with a low-grade systemic inflammation in cardiovascular system. Macrophage infiltration may initiate an inflammatory process that contributes to vascular and ventricular remodeling in hypertensive human and mice. The present study investigated the effect of chemical depletion of macrophage using liposome encapsulated clodronate (LEC) on cardiac hypertrophy and remodeling in angiotensin (Ang) II hypertensive mice.Methods: C57BL/6 mice received an Ang II (1.1 mg/kg/day with a minipump) infusion for 2 weeks to induce hypertension. Endothelium-dependent relaxation (ED) was examined by organ bath, hematoxylin and staining and Masson-Trichrome staining were used to evaluate aorta and cardiac hypertrophy and fibrosis.Results: Ang II infusion significantly increased systolic blood pressure (SBP), cardiac hypertrophy and fibrosis, and impaired EDR accompanied by increased macrophage infiltration in the heart. Treatment with LEC significantly lowered Ang II-induced cardiac hypertrophy and fibrosis and cardiac macrophage infiltration, and improved EDR with a mild reduction in SBP. Ang II increased the expression of inflammatory cytokines tumor necross factor alpha and interleukin 1 beta and profibrotic factors transforming growth factor beta 1 and fibronectin in the heart, with was reduced by LEC treatment. Treatment with LEC prevented Ang II-induced the phosphorphorylation of ERK1/2 and c-Jun-N-terminal kinase.Conclusions: Our study suggests that cardiac macrophage may be critical for hypertensive cardiac hypertrophy and remodeling, the underlying mechanisms may involve initial heart inflammation and the activation of hypertrophic MAPKs pathway.
Collapse
Affiliation(s)
- Yuantong Tian
- The Open Project of Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, P.R. of China
| | - Jun Luo
- Department of Cardiology, Affiliated Ganzhou City Hospital, Nanchang Medical University, Ganzhou, P.R. of China
| | - Qian Xu
- Department of Physiology, Shenyang Medical University, Shenyang, P.R. of China
| | - Yueyang Liu
- Department of Physiology, Shenyang Medical University, Shenyang, P.R. of China
| | - Ruiping Cai
- Department of Physiology, Shenyang Medical University, Shenyang, P.R. of China
| | - Ming-Sheng Zhou
- Department of Physiology, Shenyang Medical University, Shenyang, P.R. Of China & the Open Project of Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University. Ganzhou, P.R. of China
| |
Collapse
|
105
|
Van Beusecum JP, Barbaro NR, Smart CD, Patrick DM, Loperena R, Zhao S, Ao M, Xiao L, Shibao CA, Harrison DG. Growth Arrest Specific-6 and Axl Coordinate Inflammation and Hypertension. Circ Res 2021; 129:975-991. [PMID: 34565181 PMCID: PMC9125747 DOI: 10.1161/circresaha.121.319643] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/24/2021] [Indexed: 01/08/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Justin P. Van Beusecum
- Divison of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Natalia R. Barbaro
- Divison of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Charles D. Smart
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - David M. Patrick
- Divison of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Divison of Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Roxana Loperena
- Vanderbilt Institute of Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN
| | - Shilin Zhao
- Vanderbilt Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN
| | - Mingfang Ao
- Divison of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Liang Xiao
- Divison of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Cyndya A. Shibao
- Divison of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - David G. Harrison
- Divison of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| |
Collapse
|
106
|
Peng M, He J, Xue Y, Yang X, Liu S, Gong Z. Role of Hypertension on the Severity of COVID-19: A Review. J Cardiovasc Pharmacol 2021; 78:e648-e655. [PMID: 34321401 PMCID: PMC8562915 DOI: 10.1097/fjc.0000000000001116] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 07/04/2021] [Indexed: 12/15/2022]
Abstract
ABSTRACT The novel coronavirus disease (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rapidly evolved into a global pandemic. The substantial morbidity and mortality associated with the infection has prompted us to understand potential risk factors that can predict patient outcomes. Hypertension has been identified as the most prevalent cardiovascular comorbidity in patients infected with COVID-19 that demonstrably increases the risk of hospitalization and death. Initial studies implied that renin-angiotensin-aldosterone system inhibitors might increase the risk of viral infection and aggravate disease severity, thereby causing panic given the high global prevalence of hypertension. Nonetheless, subsequent evidence supported the administration of antihypertensive drugs and noted that they do not increase the severity of COVID-19 infection in patients with hypertension, rather may have a beneficial effect. To date, the precise mechanism by which hypertension predisposes to unfavorable outcomes in patients infected with COVID-19 remains unknown. In this mini review, we elaborate on the pathology of SARS-CoV-2 infection coexisting with hypertension and summarize potential mechanisms, focusing on the dual roles of angiotensin-converting enzyme 2 and the disorders of renin-angiotensin-aldosterone system in COVID-19 and hypertension. The effects of proinflammatory factors released because of immune response and gastrointestinal dysfunction in COVID-19 are also discussed.
Collapse
Affiliation(s)
- Mei Peng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China; and
- Institute of Hospital Pharmacy, Central South University, Changsha, China.
| | - Jia He
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China; and
- Institute of Hospital Pharmacy, Central South University, Changsha, China.
| | - Ying Xue
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China; and
- Institute of Hospital Pharmacy, Central South University, Changsha, China.
| | - Xue Yang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China; and
- Institute of Hospital Pharmacy, Central South University, Changsha, China.
| | - Shao Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China; and
- Institute of Hospital Pharmacy, Central South University, Changsha, China.
| | - Zhicheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China; and
- Institute of Hospital Pharmacy, Central South University, Changsha, China.
| |
Collapse
|
107
|
Innate immunity and clinical hypertension. J Hum Hypertens 2021; 36:503-509. [PMID: 34689174 DOI: 10.1038/s41371-021-00627-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/27/2021] [Accepted: 10/06/2021] [Indexed: 01/10/2023]
Abstract
Emerging evidence has supported a role of inflammation and immunity in the genesis of hypertension. In humans and experimental models of hypertension, cells of the innate and adaptive immune system enter target tissues, including vessels and the kidney, and release powerful mediators including cytokines, matrix metalloproteinases and reactive oxygen species that cause tissue damage, fibrosis and dysfunction. These events augment the blood pressure elevations in hypertension and promote end-organ damage. Factors that activate immune cells include sympathetic outflow, increased sodium within microenvironments where these cells reside, and signals received from the vasculature. In particular, the activated endothelium releases reactive oxygen species and interleukin (IL)-6 which in turn stimulate transformation of monocytes to become antigen presenting cells and produce cytokines like IL-1β and IL-23, which further affect T cell function to produce IL-17A. Genetic deletion or neutralization of these cytokines ameliorates hypertension and end-organ damage. In this review, we will consider in depth features of the hypertensive milieu that lead to these events and consider new treatment approaches to limit the untoward effects of inflammation in hypertension.
Collapse
|
108
|
Zaman R, Hamidzada H, Kantores C, Wong A, Dick SA, Wang Y, Momen A, Aronoff L, Lin J, Razani B, Mital S, Billia F, Lavine KJ, Nejat S, Epelman S. Selective loss of resident macrophage-derived insulin-like growth factor-1 abolishes adaptive cardiac growth to stress. Immunity 2021; 54:2057-2071.e6. [PMID: 34363749 DOI: 10.1016/j.immuni.2021.07.006] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 02/20/2021] [Accepted: 07/09/2021] [Indexed: 12/24/2022]
Abstract
Hypertension affects one-third of the world's population, leading to cardiac dysfunction that is modulated by resident and recruited immune cells. Cardiomyocyte growth and increased cardiac mass are essential to withstand hypertensive stress; however, whether immune cells are involved in this compensatory cardioprotective process is unclear. In normotensive animals, single-cell transcriptomics of fate-mapped self-renewing cardiac resident macrophages (RMs) revealed transcriptionally diverse cell states with a core repertoire of reparative gene programs, including high expression of insulin-like growth factor-1 (Igf1). Hypertension drove selective in situ proliferation and transcriptional activation of some cardiac RM states, directly correlating with increased cardiomyocyte growth. During hypertension, inducible ablation of RMs or selective deletion of RM-derived Igf1 prevented adaptive cardiomyocyte growth, and cardiac mass failed to increase, which led to cardiac dysfunction. Single-cell transcriptomics identified a conserved IGF1-expressing macrophage subpopulation in human cardiomyopathy. Here we defined the absolute requirement of RM-produced IGF-1 in cardiac adaptation to hypertension.
Collapse
Affiliation(s)
- Rysa Zaman
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Ted Rogers Centre for Heart Research, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Homaira Hamidzada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Ted Rogers Centre for Heart Research, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Crystal Kantores
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Anthony Wong
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Ted Rogers Centre for Heart Research, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Sarah A Dick
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Yiming Wang
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Abdul Momen
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Laura Aronoff
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Ted Rogers Centre for Heart Research, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Julia Lin
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Ted Rogers Centre for Heart Research, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Babak Razani
- Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Seema Mital
- Ted Rogers Centre for Heart Research, Toronto, ON, Canada; Division of Cardiology, Hospital for Sick Children, Toronto, ON, Canada; Department of Paediatrics, University of Toronto, Toronto, ON, Canada
| | - Filio Billia
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Ted Rogers Centre for Heart Research, Toronto, ON, Canada; Peter Munk Cardiac Centre, Toronto, ON, Canada
| | - Kory J Lavine
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Sara Nejat
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Ted Rogers Centre for Heart Research, Toronto, ON, Canada; Peter Munk Cardiac Centre, Toronto, ON, Canada
| | - Slava Epelman
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada; Ted Rogers Centre for Heart Research, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Peter Munk Cardiac Centre, Toronto, ON, Canada.
| |
Collapse
|
109
|
Kij A, Bar A, Przyborowski K, Proniewski B, Mateuszuk L, Jasztal A, Kieronska-Rudek A, Marczyk B, Matyjaszczyk-Gwarda K, Tworzydlo A, Enggaard C, Hansen PBL, Jensen B, Walczak M, Chlopicki S. Thrombin Inhibition Prevents Endothelial Dysfunction and Reverses 20-HETE Overproduction without Affecting Blood Pressure in Angiotensin II-Induced Hypertension in Mice. Int J Mol Sci 2021; 22:ijms22168664. [PMID: 34445374 PMCID: PMC8395447 DOI: 10.3390/ijms22168664] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 08/05/2021] [Accepted: 08/07/2021] [Indexed: 02/02/2023] Open
Abstract
Angiotensin II (Ang II) induces hypertension and endothelial dysfunction, but the involvement of thrombin in these responses is not clear. Here, we assessed the effects of the inhibition of thrombin activity by dabigatran on Ang II-induced hypertension and endothelial dysfunction in mice with a particular focus on NO- and 20-HETE-dependent pathways. As expected, dabigatran administration significantly delayed thrombin generation (CAT assay) in Ang II-treated hypertensive mice, and interestingly, it prevented endothelial dysfunction development, but it did not affect elevated blood pressure nor excessive aortic wall thickening. Dabigatran’s effects on endothelial function in Ang II-treated mice were evidenced by improved NO-dependent relaxation in the aorta in response to acetylcholine in vivo (MRI measurements) and increased systemic NO bioavailability (NO2− quantification) with a concomitant increased ex vivo production of endothelium-derived NO (EPR analysis). Dabigatran treatment also contributed to the reduction in the endothelial expression of pro-inflammatory vWF and ICAM-1. Interestingly, the fall in systemic NO bioavailability in Ang II-treated mice was associated with increased 20-HETE concentration in plasma (UPLC-MS/MS analysis), which was normalised by dabigatran treatment. Taking together, the inhibition of thrombin activity in Ang II-induced hypertension in mice improves the NO-dependent function of vascular endothelium and normalises the 20-HETE-depedent pathway without affecting the blood pressure and vascular remodelling.
Collapse
Affiliation(s)
- Agnieszka Kij
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; (A.K.); (A.B.); (K.P.); (B.P.); (L.M.); (A.J.); (A.K.-R.); (B.M.); (K.M.-G.); (A.T.); (M.W.)
| | - Anna Bar
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; (A.K.); (A.B.); (K.P.); (B.P.); (L.M.); (A.J.); (A.K.-R.); (B.M.); (K.M.-G.); (A.T.); (M.W.)
| | - Kamil Przyborowski
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; (A.K.); (A.B.); (K.P.); (B.P.); (L.M.); (A.J.); (A.K.-R.); (B.M.); (K.M.-G.); (A.T.); (M.W.)
| | - Bartosz Proniewski
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; (A.K.); (A.B.); (K.P.); (B.P.); (L.M.); (A.J.); (A.K.-R.); (B.M.); (K.M.-G.); (A.T.); (M.W.)
| | - Lukasz Mateuszuk
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; (A.K.); (A.B.); (K.P.); (B.P.); (L.M.); (A.J.); (A.K.-R.); (B.M.); (K.M.-G.); (A.T.); (M.W.)
| | - Agnieszka Jasztal
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; (A.K.); (A.B.); (K.P.); (B.P.); (L.M.); (A.J.); (A.K.-R.); (B.M.); (K.M.-G.); (A.T.); (M.W.)
| | - Anna Kieronska-Rudek
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; (A.K.); (A.B.); (K.P.); (B.P.); (L.M.); (A.J.); (A.K.-R.); (B.M.); (K.M.-G.); (A.T.); (M.W.)
| | - Brygida Marczyk
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; (A.K.); (A.B.); (K.P.); (B.P.); (L.M.); (A.J.); (A.K.-R.); (B.M.); (K.M.-G.); (A.T.); (M.W.)
| | - Karolina Matyjaszczyk-Gwarda
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; (A.K.); (A.B.); (K.P.); (B.P.); (L.M.); (A.J.); (A.K.-R.); (B.M.); (K.M.-G.); (A.T.); (M.W.)
| | - Anna Tworzydlo
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; (A.K.); (A.B.); (K.P.); (B.P.); (L.M.); (A.J.); (A.K.-R.); (B.M.); (K.M.-G.); (A.T.); (M.W.)
| | - Camilla Enggaard
- Department of Cardiovascular and Renal Research, University of Southern Denmark, J.B. Winsløws Vej 21, 5000 Odense, Denmark; (C.E.); (P.B.L.H.); (B.J.)
| | - Pernille B. Lærkegaard Hansen
- Department of Cardiovascular and Renal Research, University of Southern Denmark, J.B. Winsløws Vej 21, 5000 Odense, Denmark; (C.E.); (P.B.L.H.); (B.J.)
| | - Boye Jensen
- Department of Cardiovascular and Renal Research, University of Southern Denmark, J.B. Winsløws Vej 21, 5000 Odense, Denmark; (C.E.); (P.B.L.H.); (B.J.)
| | - Maria Walczak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; (A.K.); (A.B.); (K.P.); (B.P.); (L.M.); (A.J.); (A.K.-R.); (B.M.); (K.M.-G.); (A.T.); (M.W.)
- Chair and Department of Toxicology, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; (A.K.); (A.B.); (K.P.); (B.P.); (L.M.); (A.J.); (A.K.-R.); (B.M.); (K.M.-G.); (A.T.); (M.W.)
- Chair of Pharmacology, Jagiellonian University Medical College, Grzegorzecka 16, 31-531 Krakow, Poland
- Correspondence:
| |
Collapse
|
110
|
Jama HA, Muralitharan RR, Xu C, O'Donnell JA, Bertagnolli M, Broughton BRS, Head GA, Marques FZ. Rodent models of hypertension. Br J Pharmacol 2021; 179:918-937. [PMID: 34363610 DOI: 10.1111/bph.15650] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 01/03/2023] Open
Abstract
Elevated blood pressure (BP), or hypertension, is the main risk factor for cardiovascular disease. As a multifactorial and systemic disease that involves multiple organs and systems, hypertension remains a challenging disease to study. Models of hypertension are invaluable to support the discovery of the specific genetic, cellular and molecular mechanisms underlying essential hypertension, as well as to test new possible treatments to lower BP. Rodent models have proven to be an invaluable tool for advancing the field. In this review, we discuss the strengths and weaknesses of rodent models of hypertension through a systems approach. We highlight the ways how target organs and systems including the kidneys, vasculature, the sympathetic nervous system (SNS), immune system and the gut microbiota influence BP in each rodent model. We also discuss often overlooked hypertensive conditions such as pulmonary hypertension and hypertensive-pregnancy disorders, providing an important resource for researchers.
Collapse
Affiliation(s)
- Hamdi A Jama
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Australia.,Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Rikeish R Muralitharan
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Australia.,Institute for Medical Research, Ministry of Health Malaysia, Kuala Lumpur, Malaysia
| | - Chudan Xu
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Australia
| | - Joanne A O'Donnell
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Australia
| | - Mariane Bertagnolli
- Laboratory of Maternal-child Health, Hospital Sacre-Coeur Research Center, CIUSSS Nord-de-l'Île-de-Montréal, Montreal, Canada.,School of Physical and Occupational Therapy, Faculty of Medicine, McGill University, Montreal, Canada
| | - Bradley R S Broughton
- Department of Pharmacology, Biomedicine Discovery Institute, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, Australia
| | - Geoffrey A Head
- Department of Pharmacology, Biomedicine Discovery Institute, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, Australia.,Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash University, Melbourne, Australia.,Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
| |
Collapse
|
111
|
Birk M, Baum E, Zadeh JK, Manicam C, Pfeiffer N, Patzak A, Helmstädter J, Steven S, Kuntic M, Daiber A, Gericke A. Angiotensin II Induces Oxidative Stress and Endothelial Dysfunction in Mouse Ophthalmic Arteries via Involvement of AT1 Receptors and NOX2. Antioxidants (Basel) 2021; 10:antiox10081238. [PMID: 34439486 PMCID: PMC8389243 DOI: 10.3390/antiox10081238] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 12/13/2022] Open
Abstract
Angiotensin II (Ang II) has been implicated in the pathophysiology of various age-dependent ocular diseases. The purpose of this study was to test the hypothesis that Ang II induces endothelial dysfunction in mouse ophthalmic arteries and to identify the underlying mechanisms. Ophthalmic arteries were exposed to Ang II in vivo and in vitro to determine vascular function by video microscopy. Moreover, the formation of reactive oxygen species (ROS) was quantified and the expression of prooxidant redox genes and proteins was determined. The endothelium-dependent artery responses were blunted after both in vivo and in vitro exposure to Ang II. The Ang II type 1 receptor (AT1R) blocker, candesartan, and the ROS scavenger, Tiron, prevented Ang II-induced endothelial dysfunction. ROS levels and NOX2 expression were increased following Ang II incubation. Remarkably, Ang II failed to induce endothelial dysfunction in ophthalmic arteries from NOX2-deficient mice. Following Ang II incubation, endothelium-dependent vasodilation was mainly mediated by cytochrome P450 oxygenase (CYP450) metabolites, while the contribution of nitric oxide synthase (NOS) and 12/15-lipoxygenase (12/15-LOX) pathways became negligible. These findings provide evidence that Ang II induces endothelial dysfunction in mouse ophthalmic arteries via AT1R activation and NOX2-dependent ROS formation. From a clinical point of view, the blockade of AT1R signaling and/or NOX2 may be helpful to retain or restore endothelial function in ocular blood vessels in certain ocular diseases.
Collapse
Affiliation(s)
- Michael Birk
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (M.B.); (E.B.); (J.K.Z.); (C.M.); (N.P.)
- Department of Ophthalmology, University Eye Hospital Tübingen, Elfriede-Aulhorn-Straße 7, 72076 Tübingen, Germany
| | - Ewa Baum
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (M.B.); (E.B.); (J.K.Z.); (C.M.); (N.P.)
- Department of Social Sciences and the Humanities, Poznan University of Medical Sciences, ul. Rokietnicka 7, 60-806 Poznań, Poland
| | - Jenia Kouchek Zadeh
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (M.B.); (E.B.); (J.K.Z.); (C.M.); (N.P.)
| | - Caroline Manicam
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (M.B.); (E.B.); (J.K.Z.); (C.M.); (N.P.)
| | - Norbert Pfeiffer
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (M.B.); (E.B.); (J.K.Z.); (C.M.); (N.P.)
| | - Andreas Patzak
- Institute of Vegetative Physiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany;
| | - Johanna Helmstädter
- Department of Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center, Johannes Gutenberg University, Building 605, Langenbeckstr. 1, 55131 Mainz, Germany; (J.H.); (S.S.); (M.K.); (A.D.)
| | - Sebastian Steven
- Department of Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center, Johannes Gutenberg University, Building 605, Langenbeckstr. 1, 55131 Mainz, Germany; (J.H.); (S.S.); (M.K.); (A.D.)
| | - Marin Kuntic
- Department of Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center, Johannes Gutenberg University, Building 605, Langenbeckstr. 1, 55131 Mainz, Germany; (J.H.); (S.S.); (M.K.); (A.D.)
| | - Andreas Daiber
- Department of Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center, Johannes Gutenberg University, Building 605, Langenbeckstr. 1, 55131 Mainz, Germany; (J.H.); (S.S.); (M.K.); (A.D.)
| | - Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (M.B.); (E.B.); (J.K.Z.); (C.M.); (N.P.)
- Correspondence: ; Tel.: +49-613-117-8276
| |
Collapse
|
112
|
Wenzel UO, Ehmke H, Bode M. Immune mechanisms in arterial hypertension. Recent advances. Cell Tissue Res 2021; 385:393-404. [PMID: 33394136 PMCID: PMC8523494 DOI: 10.1007/s00441-020-03409-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/15/2020] [Indexed: 02/06/2023]
Abstract
Increasing evidence indicates that hypertension and hypertensive end-organ damage are not only mediated by hemodynamic injury. Inflammation also plays an important role in the pathophysiology and contributes to the deleterious consequences of this disease. Cells of the innate immune system including monocyte/macrophages and dendritic cells can promote blood pressure elevation via effects mostly on kidney and vascular function. Moreover, convincing evidence shows that T and B cells from the adaptive immune system are involved in hypertension and hypertensive end-organ damage. Skin monocyte/macrophages, regulatory T cells, natural killer T cells, and myeloid-derived suppressor cells have been shown to exert blood pressure controlling effects. Sodium intake is undoubtedly indispensable for normal body function but can be detrimental when taken in excess of dietary requirements. Sodium levels also modulate the function of monocyte/macrophages, dendritic cells, and different T cell subsets. Some of these effects are mediated by changes in the microbiome and metabolome that can be found after high salt intake. Modulation of the immune response can reduce severity of blood pressure elevation and hypertensive end-organ damage in several animal models. The purpose of this review is to briefly summarize recent advances in immunity and hypertension as well as hypertensive end-organ damage.
Collapse
Affiliation(s)
- Ulrich O Wenzel
- III. Department of Medicine, University Hospital Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
| | - Heimo Ehmke
- Department of Cellular and Integrative Physiology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Marlies Bode
- III. Department of Medicine, University Hospital Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| |
Collapse
|
113
|
Trott DW, Machin DR, Phuong TTT, Adeyemo AO, Bloom SI, Bramwell RC, Sorensen ES, Lesniewski LA, Donato AJ. T cells mediate cell non-autonomous arterial ageing in mice. J Physiol 2021; 599:3973-3991. [PMID: 34164826 PMCID: PMC8425389 DOI: 10.1113/jp281698] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/21/2021] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Increased large artery stiffness and impaired endothelium-dependent dilatation occur with advanced age. We sought to determine whether T cells mechanistically contribute to age-related arterial dysfunction. We found that old mice exhibited greater proinflammatory T cell accumulation around both the aorta and mesenteric arteries. Pharmacologic depletion or genetic deletion of T cells in old mice resulted in ameliorated large artery stiffness and greater endothelium-dependent dilatation compared with mice with T cells intact. ABSTRACT Ageing of the arteries is characterized by increased large artery stiffness and impaired endothelium-dependent dilatation. T cells contribute to hypertension in acute rodent models but whether they contribute to chronic age-related arterial dysfunction is unknown. To determine whether T cells directly mediate age-related arterial dysfunction, we examined large elastic artery and resistance artery function in young (4-6 months) and old (22-24 months) wild-type mice treated with anti-CD3 F(ab'2) fragments to deplete T cells (150 μg, i.p. every 7 days for 28 days) or isotype control fragments. Old mice exhibited greater numbers of T cells in both aorta and mesenteric vasculature when compared with young mice. Old mice treated with anti-CD3 fragments exhibited depletion of T cells in blood, spleen, aorta and mesenteric vasculature. Old mice also exhibited greater numbers of aortic and mesenteric IFN-γ and TNF-α-producing T cells when compared with young mice. Old control mice exhibited greater large artery stiffness and impaired resistance artery endothelium-dependent dilatation in comparison with young mice. In old mice, large artery stiffness was ameliorated with anti-CD3 treatment. Anti-CD3-treated old mice also exhibited greater endothelium-dependent dilatation than age-matched controls. We also examined arterial function in young and old Rag-1-/- mice, which lack lymphocytes. Rag-1-/- mice exhibited blunted increases in large artery stiffness with age compared with wild-type mice. Old Rag-1-/- mice also exhibited greater endothelium-dependent dilatation compared with old wild-type mice. Collectively, these results demonstrate that T cells play an important role in age-related arterial dysfunction.
Collapse
Affiliation(s)
- Daniel W Trott
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- Department of Kinesiology, University of Texas at Arlington, Texas, USA
| | - Daniel R Machin
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Tam T T Phuong
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - AdeLola O Adeyemo
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Samuel I Bloom
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA
| | - R Colton Bramwell
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Eric S Sorensen
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Lisa A Lesniewski
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA
- Geriatrics Research Education and Clinical Center, Veteran's Affairs Medical Center, Salt Lake City, Utah, USA
| | - Anthony J Donato
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA
- Geriatrics Research Education and Clinical Center, Veteran's Affairs Medical Center, Salt Lake City, Utah, USA
- Department of Biochemistry, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
114
|
Lu W, Meng Z, Hernandez R, Zhou C. Fibroblast-specific IKKβ deficiency ameliorates angiotensin II-induced adverse cardiac remodeling in mice. JCI Insight 2021; 6:e150161. [PMID: 34324438 PMCID: PMC8492299 DOI: 10.1172/jci.insight.150161] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 07/28/2021] [Indexed: 12/03/2022] Open
Abstract
Cardiac inflammation and fibrosis contribute significantly to hypertension-related adverse cardiac remodeling. IκB kinase β (IKK-β), a central coordinator of inflammation through activation of NF-κB, has been demonstrated as a key molecular link between inflammation and cardiovascular disease. However, the cell-specific contribution of IKK-β signaling toward adverse cardiac remodeling remains elusive. Cardiac fibroblasts are one of the most populous nonmyocyte cell types in the heart that play a key role in mediating cardiac fibrosis and remodeling. To investigate the function of fibroblast IKK-β, we generated inducible fibroblast-specific IKK-β–deficient mice. Here, we report an important role of IKK-β in the regulation of fibroblast functions and cardiac remodeling. Fibroblast-specific IKK-β–deficient male mice were protected from angiotensin II–induced cardiac hypertrophy, fibrosis, and macrophage infiltration. Ablation of fibroblast IKK-β inhibited angiotensin II–stimulated fibroblast proinflammatory and profibrogenic responses, leading to ameliorated cardiac remodeling and improved cardiac function in IKK-β–deficient mice. Findings from this study establish fibroblast IKK-β as a key factor regulating cardiac fibrosis and function in hypertension-related cardiac remodeling.
Collapse
Affiliation(s)
- Weiwei Lu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, United States of America
| | - Zhaojie Meng
- Division of Biomedical Sciences, University of California, Riverside, United States of America
| | - Rebecca Hernandez
- Division of Biomedical Sciences, University of California, Riverside, United States of America
| | - Changcheng Zhou
- Division of Biomedical Sciences, University of California, Riverside, United States of America
| |
Collapse
|
115
|
Wu M, Wu X, Cheng Y, Shen Z, Chen X, Xie Q, Chu J, Li J, Liu L, Wei L, Long L, Cai Q, Peng J, Shen A. Qingda Granule Attenuates Angiotensin II-Induced Blood Pressure and Inhibits Ca 2+/ERK Signaling Pathway. Front Pharmacol 2021; 12:688877. [PMID: 34393778 PMCID: PMC8358933 DOI: 10.3389/fphar.2021.688877] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/28/2021] [Indexed: 11/30/2022] Open
Abstract
Objective: As a well-known traditional Chinese medicine formula prescribed by academician Ke-ji Chen, Qingda granule (QDG) lowered the blood pressure of spontaneously hypertensive rats and attenuated hypertensive cardiac remodeling and inflammation. However, its functional role and underlying mechanisms on hypertensive vascular function remain largely unclear. This study aims to assess the effects of QDG treatment on Angiotensin II- (AngII-) induced hypertension and vascular function and explore its underlying mechanisms both in vitro and in vivo. Methods: In an in vivo study, 25 male C57BL/6 mice were randomly divided into five groups, including Control, AngII, AngII + QDG-L, AngII + QDG-M, and AngII + QDG-H groups (n = 5 for each group). Mice in AngII and AngII + QDG-L/-M/-H groups were infused with AngII (500 ng/kg/min), while in the Control group, they were infused with saline. Mice in AngII + QDG were intragastrically given different concentrations of QDG (0.5725, 1.145, or 2.29 g/kg/day), while in Control and AngII groups, they were intragastrically given equal volumes of double distilled water for 2 weeks. Blood pressure was determined at 0, 1, and 2 weeks of treatment. Ultrasound was used to detect the pulse wave velocity (PWV) and HE staining to detect the pathological change of the abdominal aorta. RNA sequencing (RNA-seq) was performed to identify the differentially expressed transcripts (DETs) and related signaling pathways. IHC was used to detect the expression of p-ERK in the abdominal aorta. Primary isolated rat vascular smooth muscle cells (VSMCs) were used to assess the cellular Ca2+ release and activation of the ERK pathway by confocal microscope and western blotting analysis, respectively. Results: QDG treatment significantly alleviated the elevated blood pressure, the PWV, and the thickness of the abdominal aorta in AngII-induced hypertensive mice. RNA-seq and KEGG analyses identified 1,505 DETs and multiple enriched pathways (including vascular contraction and calcium signaling pathway) after QDG treatment. Furthermore, confocal microscope showed that QDG treatment partially attenuated the increase of Ca2+ release with the stimulation of AngII in cultured VSMCs. In addition, IHC and western blotting indicated that QDG treatment also partially alleviated the increase of phospho-ERK levels in abdominal aorta tissues of mice and cultured VSMCs after the infusion or stimulation of AngII. Conclusion: QDG treatment attenuated the elevation of blood pressure, abdominal aorta dysfunction, pathological changes, Ca2+ release, and activation of the ERK signaling pathway.
Collapse
Affiliation(s)
- Meizhu Wu
- Academy of Integrative Medicine, Fuzhou, China.,Chen Keji Academic Thought Inheritance Studio, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xiangyan Wu
- Academy of Integrative Medicine, Fuzhou, China.,Chen Keji Academic Thought Inheritance Studio, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Ying Cheng
- Academy of Integrative Medicine, Fuzhou, China.,Chen Keji Academic Thought Inheritance Studio, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Zhiqing Shen
- Academy of Integrative Medicine, Fuzhou, China.,Chen Keji Academic Thought Inheritance Studio, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xiaoping Chen
- Academy of Integrative Medicine, Fuzhou, China.,Chen Keji Academic Thought Inheritance Studio, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Qiurong Xie
- Academy of Integrative Medicine, Fuzhou, China.,Chen Keji Academic Thought Inheritance Studio, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jianfeng Chu
- Academy of Integrative Medicine, Fuzhou, China.,Chen Keji Academic Thought Inheritance Studio, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jiapeng Li
- Department of Physical Education, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Liya Liu
- Academy of Integrative Medicine, Fuzhou, China.,Chen Keji Academic Thought Inheritance Studio, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Lihui Wei
- Academy of Integrative Medicine, Fuzhou, China.,Chen Keji Academic Thought Inheritance Studio, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Linzi Long
- Academy of Integrative Medicine, Fuzhou, China.,Chen Keji Academic Thought Inheritance Studio, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qiaoyan Cai
- Academy of Integrative Medicine, Fuzhou, China.,Chen Keji Academic Thought Inheritance Studio, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jun Peng
- Academy of Integrative Medicine, Fuzhou, China.,Chen Keji Academic Thought Inheritance Studio, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Aling Shen
- Academy of Integrative Medicine, Fuzhou, China.,Chen Keji Academic Thought Inheritance Studio, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
116
|
Helmstädter J, Keppeler K, Aust F, Küster L, Frenis K, Filippou K, Vujacic-Mirski K, Tsohataridis S, Kalinovic S, Kröller-Schön S, Oelze M, Bosmann M, Münzel T, Daiber A, Steven S. GLP-1 Analog Liraglutide Improves Vascular Function in Polymicrobial Sepsis by Reduction of Oxidative Stress and Inflammation. Antioxidants (Basel) 2021; 10:antiox10081175. [PMID: 34439423 PMCID: PMC8388926 DOI: 10.3390/antiox10081175] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 12/12/2022] Open
Abstract
Sepsis causes high mortality in the setting of septic shock. LEADER and other trials revealed cardioprotective and anti-inflammatory properties of glucagon-like peptide-1 (GLP-1) analogs like liraglutide (Lira). We previously demonstrated improved survival in lipopolysaccharide (LPS)-induced endotoxemia by inhibition of GLP-1 degradation. Here we investigate the effects of Lira in the polymicrobial sepsis model of cecal ligation and puncture (CLP). C57BL/6J mice were intraperitoneally injected with Lira (200 µg/kg/d; 3 days) and sepsis induced by CLP after one day of GLP-1 analog treatment. Survival and body temperature were monitored. Aortic vascular function (isometric tension recording), protein expression (immunohistochemistry and dot blot) and gene expression (qRT-PCR) were determined. Endothelium-dependent relaxation in the aorta was impaired by CLP and correlated with markers of inflammation (e.g., interleukin 6 and inducible nitric oxide synthase) and oxidative stress (e.g., 3-nitrotyrosine) was higher in septic mice, all of which was almost completely normalized by Lira therapy. We demonstrate that the GLP-1 analog Lira ameliorates sepsis-induced endothelial dysfunction by the reduction of vascular inflammation and oxidative stress. Accordingly, the findings suggest that the antioxidant and anti-inflammatory effects of GLP-1 analogs may be a valuable tool to protect the cardiovascular system from dysbalanced inflammation in polymicrobial sepsis.
Collapse
Affiliation(s)
- Johanna Helmstädter
- Center for Cardiology, Department of Cardiology 1–Molecular Cardiology, University Medical Center, 55131 Mainz, Germany; (J.H.); (K.K.); (F.A.); (L.K.); (K.F.); (K.F.); (K.V.-M.); (S.T.); (S.K.); (S.K.-S.); (M.O.); (T.M.); (A.D.)
| | - Karin Keppeler
- Center for Cardiology, Department of Cardiology 1–Molecular Cardiology, University Medical Center, 55131 Mainz, Germany; (J.H.); (K.K.); (F.A.); (L.K.); (K.F.); (K.F.); (K.V.-M.); (S.T.); (S.K.); (S.K.-S.); (M.O.); (T.M.); (A.D.)
| | - Franziska Aust
- Center for Cardiology, Department of Cardiology 1–Molecular Cardiology, University Medical Center, 55131 Mainz, Germany; (J.H.); (K.K.); (F.A.); (L.K.); (K.F.); (K.F.); (K.V.-M.); (S.T.); (S.K.); (S.K.-S.); (M.O.); (T.M.); (A.D.)
| | - Leonie Küster
- Center for Cardiology, Department of Cardiology 1–Molecular Cardiology, University Medical Center, 55131 Mainz, Germany; (J.H.); (K.K.); (F.A.); (L.K.); (K.F.); (K.F.); (K.V.-M.); (S.T.); (S.K.); (S.K.-S.); (M.O.); (T.M.); (A.D.)
| | - Katie Frenis
- Center for Cardiology, Department of Cardiology 1–Molecular Cardiology, University Medical Center, 55131 Mainz, Germany; (J.H.); (K.K.); (F.A.); (L.K.); (K.F.); (K.F.); (K.V.-M.); (S.T.); (S.K.); (S.K.-S.); (M.O.); (T.M.); (A.D.)
| | - Konstantina Filippou
- Center for Cardiology, Department of Cardiology 1–Molecular Cardiology, University Medical Center, 55131 Mainz, Germany; (J.H.); (K.K.); (F.A.); (L.K.); (K.F.); (K.F.); (K.V.-M.); (S.T.); (S.K.); (S.K.-S.); (M.O.); (T.M.); (A.D.)
| | - Ksenija Vujacic-Mirski
- Center for Cardiology, Department of Cardiology 1–Molecular Cardiology, University Medical Center, 55131 Mainz, Germany; (J.H.); (K.K.); (F.A.); (L.K.); (K.F.); (K.F.); (K.V.-M.); (S.T.); (S.K.); (S.K.-S.); (M.O.); (T.M.); (A.D.)
| | - Simeon Tsohataridis
- Center for Cardiology, Department of Cardiology 1–Molecular Cardiology, University Medical Center, 55131 Mainz, Germany; (J.H.); (K.K.); (F.A.); (L.K.); (K.F.); (K.F.); (K.V.-M.); (S.T.); (S.K.); (S.K.-S.); (M.O.); (T.M.); (A.D.)
| | - Sanela Kalinovic
- Center for Cardiology, Department of Cardiology 1–Molecular Cardiology, University Medical Center, 55131 Mainz, Germany; (J.H.); (K.K.); (F.A.); (L.K.); (K.F.); (K.F.); (K.V.-M.); (S.T.); (S.K.); (S.K.-S.); (M.O.); (T.M.); (A.D.)
| | - Swenja Kröller-Schön
- Center for Cardiology, Department of Cardiology 1–Molecular Cardiology, University Medical Center, 55131 Mainz, Germany; (J.H.); (K.K.); (F.A.); (L.K.); (K.F.); (K.F.); (K.V.-M.); (S.T.); (S.K.); (S.K.-S.); (M.O.); (T.M.); (A.D.)
| | - Matthias Oelze
- Center for Cardiology, Department of Cardiology 1–Molecular Cardiology, University Medical Center, 55131 Mainz, Germany; (J.H.); (K.K.); (F.A.); (L.K.); (K.F.); (K.F.); (K.V.-M.); (S.T.); (S.K.); (S.K.-S.); (M.O.); (T.M.); (A.D.)
| | - Markus Bosmann
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany;
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Thomas Münzel
- Center for Cardiology, Department of Cardiology 1–Molecular Cardiology, University Medical Center, 55131 Mainz, Germany; (J.H.); (K.K.); (F.A.); (L.K.); (K.F.); (K.F.); (K.V.-M.); (S.T.); (S.K.); (S.K.-S.); (M.O.); (T.M.); (A.D.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Andreas Daiber
- Center for Cardiology, Department of Cardiology 1–Molecular Cardiology, University Medical Center, 55131 Mainz, Germany; (J.H.); (K.K.); (F.A.); (L.K.); (K.F.); (K.F.); (K.V.-M.); (S.T.); (S.K.); (S.K.-S.); (M.O.); (T.M.); (A.D.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Sebastian Steven
- Center for Cardiology, Department of Cardiology 1–Molecular Cardiology, University Medical Center, 55131 Mainz, Germany; (J.H.); (K.K.); (F.A.); (L.K.); (K.F.); (K.F.); (K.V.-M.); (S.T.); (S.K.); (S.K.-S.); (M.O.); (T.M.); (A.D.)
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Langenbeckstr. 1, 55131 Mainz, Germany;
- Correspondence: ; Tel.: +49-(0)6131-176-948
| |
Collapse
|
117
|
Li C, Li L, Sun M, Sun J, Shao L, Xu M, Hou Y, Peng J, Wang L, Hou M. Predictive Value of High ICAM-1 Level for Poor Treatment Response to Low-Dose Decitabine in Adult Corticosteroid Resistant ITP Patients. Front Immunol 2021; 12:689663. [PMID: 34326842 PMCID: PMC8313967 DOI: 10.3389/fimmu.2021.689663] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/15/2021] [Indexed: 11/13/2022] Open
Abstract
Primary immune thrombocytopenia (ITP) is an autoimmune hemorrhagic disease. Endothelial cell activation/injury has been found in some autoimmune diseases including SLE, systemic sclerosis, and rheumatoid arthritis, but its role in ITP pathogenesis remains unclear. This study attempted to elucidate the correlation between endothelial dysfunction and disease severity of ITP and find related markers to predict response to low-dose decitabine treatment. Compared with healthy volunteers, higher plasma levels of soluble intercellular adhesion molecule-1 (ICAM-1), vascular endothelial growth factor (VEGF), and Angiopoietin-2 were found in adult corticosteroid resistant ITP patients. Notably, ICAM-1 levels were negatively correlated with the platelet count, and positively associated with the bleeding score. Recently, we have reported the efficacy and safety of low-dose decitabine in adult patients with ITP who failed for the first line therapies. Here, we evaluated the correlation of plasma ICAM-1 level with the efficacy of low-dose decitabine therapy for corticosteroid resistant ITP. A total of 29 adult corticosteroid resistant ITP patients who received consecutive treatments of low-dose decitabine were enrolled in this study. Fourteen patients showed response (nine showed complete response and five showed partial response). The levels of ICAM-1 before and after treatment were significantly higher in the non-responsive ITP patients than in the responsive patients. As shown in the multivariable logistic regression model, the odds of developing no-response to low-dose decitabine increased by 36.8% for per 5 ng/ml increase in plasma ICAM-1 level [odds ratio (OR) 1.368, 95% confidence interval (CI): 1.060 to 1.764]. In summary, this was the first study to elucidate the relationship between endothelial dysfunction and corticosteroid resistant ITP and identify the potential predictive value of ICAM-1 level for response to low-dose decitabine.
Collapse
Affiliation(s)
- Chaoyang Li
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lizhen Li
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Meng Sun
- Jinan Vocational College of Nursing, Jinan, China
| | - Jianzhi Sun
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Linlin Shao
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Miao Xu
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yu Hou
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jun Peng
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Leading Research Group of Scientific Innovation, Department of Science and Technology of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lin Wang
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ming Hou
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Leading Research Group of Scientific Innovation, Department of Science and Technology of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
118
|
Schäfer K, Wenzel P. When big eaters stop feasting: loss of metabolic control in macrophages exacerbates hypertension in obesity. Cardiovasc Res 2021; 117:351-353. [PMID: 32298411 DOI: 10.1093/cvr/cvaa103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Katrin Schäfer
- Center for Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK e. V.; RheinMain site), Mainz, Germany
| | - Philip Wenzel
- Center for Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK e. V.; RheinMain site), Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
119
|
Bruno AS, Lopes PDD, de Oliveira KCM, de Oliveira AK, de Assis Cau SB. Vascular Inflammation in Hypertension: Targeting Lipid Mediators Unbalance and Nitrosative Stress. Curr Hypertens Rev 2021; 17:35-46. [PMID: 31858899 DOI: 10.2174/1573402116666191220122332] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/24/2019] [Accepted: 11/28/2019] [Indexed: 11/22/2022]
Abstract
Arterial hypertension is a worldwide public health threat. High Blood Pressure (BP) is commonly associated with endothelial dysfunction, nitric oxide synthases (NOS) unbalance and high peripheral vascular resistance. In addition to those, inflammation has also been designated as one of the major components of BP increase and organ damage in hypertension. This minireview discusses vascular inflammatory triggers of high BP and aims to fill the existing gaps of antiinflammatory therapy of hypertension. Among the reasons discussed, enhanced prostaglandins rather than resolvins lipid mediators, immune cell infiltration and oxidative/nitrosative stress are pivotal players of BP increase within the inflammatory hypothesis. To address these inflammatory targets, this review also proposes new concepts in hypertension treatment with non-steroidal antiinflammatory drugs (NSAIDs), nitric oxide-releasing NSAIDs (NO-NSAIDs) and specialized proresolving mediators (SPM). In this context, the failure of NSAIDs in hypertension treatment seems to be associated with the reduction of endogenous NO bioavailability, which is not necessarily an effect of all drug members of this pharmacological class. For this reason, NO-releasing NSAIDs seem to be safer and more specific therapy to treat vascular inflammation in hypertension than regular NSAIDs.
Collapse
Affiliation(s)
- Alexandre S Bruno
- Department of Pharmacology, Institute of Biological Science, Federal University of Minas Gerais, MG, Brazil
| | - Patricia das Dores Lopes
- Department of Pharmacology, Institute of Biological Science, Federal University of Minas Gerais, MG, Brazil
| | - Karla C M de Oliveira
- Department of Pharmacology, Institute of Biological Science, Federal University of Minas Gerais, MG, Brazil
| | - Anizia K de Oliveira
- Department of Pharmacology, Institute of Biological Science, Federal University of Minas Gerais, MG, Brazil
| | - Stefany B de Assis Cau
- Department of Pharmacology, Institute of Biological Science, Federal University of Minas Gerais, MG, Brazil
| |
Collapse
|
120
|
Stamm P, Oelze M, Steven S, Kröller-Schön S, Kvandova M, Kalinovic S, Jasztal A, Kij A, Kuntic M, Bayo Jimenez MT, Proniewski B, Li H, Schulz E, Chlopicki S, Daiber A, Münzel T. Direct comparison of inorganic nitrite and nitrate on vascular dysfunction and oxidative damage in experimental arterial hypertension. Nitric Oxide 2021; 113-114:57-69. [PMID: 34091009 DOI: 10.1016/j.niox.2021.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 05/24/2021] [Accepted: 06/01/2021] [Indexed: 01/17/2023]
Abstract
Arterial hypertension is one of the major health risk factors leading to coronary artery disease, stroke or peripheral artery disease. Dietary uptake of inorganic nitrite (NO2-) and nitrate (NO3-) via vegetables leads to enhanced vascular NO bioavailability and provides antihypertensive effects. The present study aims to understand the underlying vasoprotective effects of nutritional NO2- and NO3- co-therapy in mice with angiotensin-II (AT-II)-induced arterial hypertension. High-dose AT-II (1 mg/kg/d, 1w, s. c.) was used to induce arterial hypertension in male C57BL/6 mice. Additional inorganic nitrite (7.5 mg/kg/d, p. o.) or nitrate (150 mg/kg/d, p. o.) were administered via the drinking water. Blood pressure (tail-cuff method) and endothelial function (isometric tension) were determined. Oxidative stress and inflammation markers were quantified in aorta, heart, kidney and blood. Co-treatment with inorganic nitrite, but not with nitrate, normalized vascular function, oxidative stress markers and inflammatory pathways in AT-II treated mice. Of note, the highly beneficial effects of nitrite on all parameters and the less pronounced protection by nitrate, as seen by improvement of some parameters, were observed despite no significant increase in plasma nitrite levels by both therapies. Methemoglobin levels tended to be higher upon nitrite/nitrate treatment. Nutritional nitric oxide precursors represent a non-pharmacological treatment option for hypertension that could be applied to the general population (e.g. by eating certain vegetables). The more beneficial effects of inorganic nitrite may rely on superior NO bioactivation and stronger blood pressure lowering effects. Future large-scale clinical studies should investigate whether hypertension and cardiovascular outcome in general can be influenced by dietary inorganic nitrite therapy.
Collapse
Affiliation(s)
- Paul Stamm
- Department of Cardiology, Cardiology I, Laboratory of Molecular Cardiology, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Matthias Oelze
- Department of Cardiology, Cardiology I, Laboratory of Molecular Cardiology, University Medical Center Mainz, Mainz, Germany
| | - Sebastian Steven
- Department of Cardiology, Cardiology I, Laboratory of Molecular Cardiology, University Medical Center Mainz, Mainz, Germany
| | - Swenja Kröller-Schön
- Department of Cardiology, Cardiology I, Laboratory of Molecular Cardiology, University Medical Center Mainz, Mainz, Germany
| | - Miroslava Kvandova
- Department of Cardiology, Cardiology I, Laboratory of Molecular Cardiology, University Medical Center Mainz, Mainz, Germany
| | - Sanela Kalinovic
- Department of Cardiology, Cardiology I, Laboratory of Molecular Cardiology, University Medical Center Mainz, Mainz, Germany
| | - Agnieszka Jasztal
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Agnieszka Kij
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Marin Kuntic
- Department of Cardiology, Cardiology I, Laboratory of Molecular Cardiology, University Medical Center Mainz, Mainz, Germany
| | - Maria Teresa Bayo Jimenez
- Department of Cardiology, Cardiology I, Laboratory of Molecular Cardiology, University Medical Center Mainz, Mainz, Germany
| | - Bartosz Proniewski
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Huige Li
- Department of Pharmacology, University Medical Center Mainz, Mainz, Germany
| | - Eberhard Schulz
- Department of Cardiology, Cardiology I, Laboratory of Molecular Cardiology, University Medical Center Mainz, Mainz, Germany; Department of Cardiology, Celle General Hospital, Celle, Germany
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland; Department of Pharmacology, Medical College of the Jagiellonian University, Krakow, Poland
| | - Andreas Daiber
- Department of Cardiology, Cardiology I, Laboratory of Molecular Cardiology, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany.
| | - Thomas Münzel
- Department of Cardiology, Cardiology I, Laboratory of Molecular Cardiology, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany.
| |
Collapse
|
121
|
Perrotta S, Carnevale D. A neurohumoral activation of renin-angiotensin-aldosterone system in endothelial dysfunction modulating immunity in heart failure. Cardiovasc Res 2021; 117:9-10. [PMID: 32750101 DOI: 10.1093/cvr/cvaa243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Sara Perrotta
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Daniela Carnevale
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy.,Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, 86077 Pozzilli (IS), Italy
| |
Collapse
|
122
|
Münzel T, Templin C, Cammann VL, Hahad O. Takotsubo Syndrome: Impact of endothelial dysfunction and oxidative stress. Free Radic Biol Med 2021; 169:216-223. [PMID: 33864955 DOI: 10.1016/j.freeradbiomed.2021.03.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/17/2021] [Accepted: 03/25/2021] [Indexed: 12/30/2022]
Abstract
Takotsubo Syndrome (TTS) is characterized by a transient left ventricular dysfunction recovering spontaneously within days or weeks. Although the pathophysiology of TTS remains obscure, there is growing evidence suggesting TTS to be associated with increased production of reactive oxygen species (ROS), which may be involved in causing transient coronary and peripheral endothelial dysfunction leading to a transient impairment of myocardial contraction due to stunning (apical ballooning). Endothelial dysfunction is mainly caused by decreased vascular and myocardial nitric oxide bioavailability in response to increased ROS production. Accordingly, studies in humans and animal models demonstrated increased myocardial dihydroethidium staining of the myocardium in endomyocardial biopsy specimens, increased levels of hydrogen peroxide and malondialdehyde as well as reduced glutathione levels compatible with increased oxidative stress. As significant superoxide sources the mitochondria and the NADPH oxidase isoform NOX-4 and the NOX-2 regulating cytosolic subunit p67phox have been identified. Treatment with antioxidants such as sodium hydrosulfide reduced superoxide production in mitochondria and reduced expression of NOX-4 and p67phox, respectively. The presence of superoxide and nitric oxide also provides the basis for the concept of nitro-oxidative as well as nitrosative stress in TTS.
Collapse
Affiliation(s)
- Thomas Münzel
- Department of Cardiology, University Medical Center of Johannes Gutenberg University Mainz, Mainz, Germany.
| | | | | | - Omar Hahad
- Department of Cardiology, University Medical Center of Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
123
|
Diabetic Kidney Disease, Cardiovascular Disease and Non-Alcoholic Fatty Liver Disease: A New Triumvirate? J Clin Med 2021; 10:jcm10092040. [PMID: 34068699 PMCID: PMC8126096 DOI: 10.3390/jcm10092040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/03/2021] [Accepted: 05/06/2021] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease is a highly prevalent disease worldwide with a renowned relation to cardiovascular disease and chronic kidney disease. These diseases share a common pathophysiology including insulin resistance, oxidative stress, chronic inflammation, dysbiosis and genetic susceptibilities. Non-alcoholic fatty liver disease is especially prevalent and more severe in type 2 diabetes. Patients with non-alcoholic fatty liver disease should have liver fibrosis assessment in order to identify those at the highest risk of adverse outcomes so that appropriate management strategies can be implemented. Early diagnosis and treatment of non-alcoholic fatty liver disease could ameliorate the burden of cardiovascular disease and chronic kidney disease.
Collapse
|
124
|
Frenis K, Helmstädter J, Ruan Y, Schramm E, Kalinovic S, Kröller-Schön S, Bayo Jimenez MT, Hahad O, Oelze M, Jiang S, Wenzel P, Sommer CJ, Frauenknecht KBM, Waisman A, Gericke A, Daiber A, Münzel T, Steven S. Ablation of lysozyme M-positive cells prevents aircraft noise-induced vascular damage without improving cerebral side effects. Basic Res Cardiol 2021; 116:31. [PMID: 33929610 PMCID: PMC8087569 DOI: 10.1007/s00395-021-00869-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 04/13/2021] [Indexed: 12/17/2022]
Abstract
Aircraft noise induces vascular and cerebral inflammation and oxidative stress causing hypertension and cardiovascular/cerebral dysfunction. With the present studies, we sought to determine the role of myeloid cells in the vascular vs. cerebral consequences of exposure to aircraft noise. Toxin-mediated ablation of lysozyme M+ (LysM+) myeloid cells was performed in LysMCreiDTR mice carrying a cre-inducible diphtheria toxin receptor. In the last 4d of toxin treatment, the animals were exposed to noise at maximum and mean sound pressure levels of 85 and 72 dB(A), respectively. Flow cytometry analysis revealed accumulation of CD45+, CD11b+, F4/80+, and Ly6G-Ly6C+ cells in the aortas of noise-exposed mice, which was prevented by LysM+ cell ablation in the periphery, whereas brain infiltrates were even exacerbated upon ablation. Aircraft noise-induced increases in blood pressure and endothelial dysfunction of the aorta and retinal/mesenteric arterioles were almost completely normalized by ablation. Correspondingly, reactive oxygen species in the aorta, heart, and retinal/mesenteric vessels were attenuated in ablated noise-exposed mice, while microglial activation and abundance in the brain was greatly increased. Expression of phagocytic NADPH oxidase (NOX-2) and vascular cell adhesion molecule-1 (VCAM-1) mRNA in the aorta was reduced, while NFκB signaling appeared to be activated in the brain upon ablation. In sum, we show dissociation of cerebral and peripheral inflammatory reactions in response to aircraft noise after LysM+ cell ablation, wherein peripheral myeloid inflammatory cells represent a dominant part of the pathomechanism for noise stress-induced cardiovascular effects and their central nervous counterparts, microglia, as key mediators in stress responses.
Collapse
Affiliation(s)
- Katie Frenis
- Department of Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Building 605, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Johanna Helmstädter
- Department of Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Building 605, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Yue Ruan
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Eva Schramm
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Sanela Kalinovic
- Department of Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Building 605, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Swenja Kröller-Schön
- Department of Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Building 605, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Maria Teresa Bayo Jimenez
- Department of Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Building 605, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Omar Hahad
- Department of Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Building 605, Langenbeckstr. 1, 55131, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Matthias Oelze
- Department of Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Building 605, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Subao Jiang
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Philip Wenzel
- Department of Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Building 605, Langenbeckstr. 1, 55131, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Clemens J Sommer
- Institute of Neuropathology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Katrin B M Frauenknecht
- Institute of Neuropathology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Adrian Gericke
- Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Building 605, Langenbeckstr. 1, 55131, Mainz, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany.
| | - Thomas Münzel
- Department of Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Building 605, Langenbeckstr. 1, 55131, Mainz, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany.
| | - Sebastian Steven
- Department of Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University, Building 605, Langenbeckstr. 1, 55131, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
125
|
Ruggeri Barbaro N, Van Beusecum J, Xiao L, do Carmo L, Pitzer A, Loperena R, Foss JD, Elijovich F, Laffer CL, Montaniel KR, Galindo CL, Chen W, Ao M, Mernaugh RL, Alsouqi A, Ikizler TA, Fogo AB, Moreno H, Zhao S, Davies SS, Harrison DG, Kirabo A. Sodium activates human monocytes via the NADPH oxidase and isolevuglandin formation. Cardiovasc Res 2021; 117:1358-1371. [PMID: 33038226 PMCID: PMC8064439 DOI: 10.1093/cvr/cvaa207] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/11/2020] [Accepted: 07/09/2020] [Indexed: 12/12/2022] Open
Abstract
AIMS Prior studies have focused on the role of the kidney and vasculature in salt-induced modulation of blood pressure; however, recent data indicate that sodium accumulates in tissues and can activate immune cells. We sought to examine mechanisms by which salt causes activation of human monocytes both in vivo and in vitro. METHODS AND RESULTS To study the effect of salt in human monocytes, monocytes were isolated from volunteers to perform several in vitro experiments. Exposure of human monocytes to elevated Na+ex vivo caused a co-ordinated response involving isolevuglandin (IsoLG)-adduct formation, acquisition of a dendritic cell (DC)-like morphology, expression of activation markers CD83 and CD16, and increased production of pro-inflammatory cytokines tumour necrosis factor-α, interleukin (IL)-6, and IL-1β. High salt also caused a marked change in monocyte gene expression as detected by RNA sequencing and enhanced monocyte migration to the chemokine CC motif chemokine ligand 5. NADPH-oxidase inhibition attenuated monocyte activation and IsoLG-adduct formation. The increase in IsoLG-adducts correlated with risk factors including body mass index, pulse pressure. Monocytes exposed to high salt stimulated IL-17A production from autologous CD4+ and CD8+ T cells. In addition, to evaluate the effect of salt in vivo, monocytes and T cells isolated from humans were adoptively transferred to immunodeficient NSG mice. Salt feeding of humanized mice caused monocyte-dependent activation of human T cells reflected by proliferation and accumulation of T cells in the bone marrow. Moreover, we performed a cross-sectional study in 70 prehypertensive subjects. Blood was collected for flow cytometric analysis and 23Na magnetic resonance imaging was performed for tissue sodium measurements. Monocytes from humans with high skin Na+ exhibited increased IsoLG-adduct accumulation and CD83 expression. CONCLUSION Human monocytes exhibit co-ordinated increases in parameters of activation, conversion to a DC-like phenotype and ability to activate T cells upon both in vitro and in vivo sodium exposure. The ability of monocytes to be activated by sodium is related to in vivo cardiovascular disease risk factors. We therefore propose that in addition to the kidney and vasculature, immune cells like monocytes convey salt-induced cardiovascular risk in humans.
Collapse
Affiliation(s)
- Natalia Ruggeri Barbaro
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN 37232-6602, USA
| | - Justin Van Beusecum
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN 37232-6602, USA
| | - Liang Xiao
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN 37232-6602, USA
| | - Luciana do Carmo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN 37232-6602, USA
| | - Ashley Pitzer
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN 37232-6602, USA
| | - Roxana Loperena
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN 37232-6602, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Jason D Foss
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN 37232-6602, USA
| | - Fernando Elijovich
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN 37232-6602, USA
| | - Cheryl L Laffer
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN 37232-6602, USA
| | - Kim R Montaniel
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN 37232-6602, USA
| | - Cristi L Galindo
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wei Chen
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN 37232-6602, USA
| | - Mingfang Ao
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN 37232-6602, USA
| | | | - Aseel Alsouqi
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Talat A Ikizler
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Agnes B Fogo
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Heitor Moreno
- Department of Intern Medicine, Faculty of Medical Sciences, Cardiovascular Pharmacology Laboratory, University of Campinas, Campinas, Brazil
| | - Shilin Zhao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sean S Davies
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN 37232-6602, USA
| | - David G Harrison
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN 37232-6602, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Room 536 Robinson Research Building, Nashville, TN 37232-6602, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
126
|
Bayo Jimenez MT, Frenis K, Kröller-Schön S, Kuntic M, Stamm P, Kvandová M, Oelze M, Li H, Steven S, Münzel T, Daiber A. Noise-Induced Vascular Dysfunction, Oxidative Stress, and Inflammation Are Improved by Pharmacological Modulation of the NRF2/HO-1 Axis. Antioxidants (Basel) 2021; 10:antiox10040625. [PMID: 33921821 PMCID: PMC8073373 DOI: 10.3390/antiox10040625] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/10/2021] [Accepted: 04/12/2021] [Indexed: 12/24/2022] Open
Abstract
Vascular oxidative stress, inflammation, and subsequent endothelial dysfunction are consequences of traditional cardiovascular risk factors, all of which contribute to cardiovascular disease. Environmental stressors, such as traffic noise and air pollution, may also facilitate the development and progression of cardiovascular and metabolic diseases. In our previous studies, we investigated the influence of aircraft noise exposure on molecular mechanisms, identifying oxidative stress and inflammation as central players in mediating vascular function. The present study investigates the role of heme oxygenase-1 (HO-1) as an antioxidant response preventing vascular consequences following exposure to aircraft noise. C57BL/6J mice were treated with the HO-1 inducer hemin (25 mg/kg i.p.) or the NRF2 activator dimethyl fumarate (DMF, 20 mg/kg p.o.). During therapy, the animals were exposed to noise at a maximum sound pressure level of 85 dB(A) and a mean sound pressure level of 72 dB(A). Our data showed a marked protective effect of both treatments on animals exposed to noise for 4 days by normalization of arterial hypertension and vascular dysfunction in the noise-exposed groups. We observed a partial normalization of noise-triggered oxidative stress and inflammation by hemin and DMF therapy, which was associated with HO-1 induction. The present study identifies possible new targets for the mitigation of the adverse health effects caused by environmental noise exposure. Since natural dietary constituents can achieve HO-1 and NRF2 induction, these pathways represent promising targets for preventive measures.
Collapse
Affiliation(s)
- Maria Teresa Bayo Jimenez
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131 Mainz, Germany; (M.T.B.J.); (K.F.); (S.K.-S.); (M.K.); (P.S.); (M.K.); (M.O.); (T.M.)
| | - Katie Frenis
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131 Mainz, Germany; (M.T.B.J.); (K.F.); (S.K.-S.); (M.K.); (P.S.); (M.K.); (M.O.); (T.M.)
| | - Swenja Kröller-Schön
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131 Mainz, Germany; (M.T.B.J.); (K.F.); (S.K.-S.); (M.K.); (P.S.); (M.K.); (M.O.); (T.M.)
| | - Marin Kuntic
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131 Mainz, Germany; (M.T.B.J.); (K.F.); (S.K.-S.); (M.K.); (P.S.); (M.K.); (M.O.); (T.M.)
| | - Paul Stamm
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131 Mainz, Germany; (M.T.B.J.); (K.F.); (S.K.-S.); (M.K.); (P.S.); (M.K.); (M.O.); (T.M.)
| | - Miroslava Kvandová
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131 Mainz, Germany; (M.T.B.J.); (K.F.); (S.K.-S.); (M.K.); (P.S.); (M.K.); (M.O.); (T.M.)
| | - Matthias Oelze
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131 Mainz, Germany; (M.T.B.J.); (K.F.); (S.K.-S.); (M.K.); (P.S.); (M.K.); (M.O.); (T.M.)
| | - Huige Li
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131 Mainz, Germany;
| | - Sebastian Steven
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131 Mainz, Germany; (M.T.B.J.); (K.F.); (S.K.-S.); (M.K.); (P.S.); (M.K.); (M.O.); (T.M.)
- Correspondence: (S.S.); (A.D.)
| | - Thomas Münzel
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131 Mainz, Germany; (M.T.B.J.); (K.F.); (S.K.-S.); (M.K.); (P.S.); (M.K.); (M.O.); (T.M.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, Langenbeckstraße 1, 55131 Mainz, Germany; (M.T.B.J.); (K.F.); (S.K.-S.); (M.K.); (P.S.); (M.K.); (M.O.); (T.M.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany
- Correspondence: (S.S.); (A.D.)
| |
Collapse
|
127
|
CRIP1 expression in monocytes related to hypertension. Clin Sci (Lond) 2021; 135:911-924. [PMID: 33782695 DOI: 10.1042/cs20201372] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 03/23/2021] [Accepted: 03/29/2021] [Indexed: 12/21/2022]
Abstract
Hypertension is a complex and multifactorial disorder caused by lifestyle and environmental factors, inflammation and disease-related genetic factors and is a risk factor for stroke, ischemic heart disease and renal failure. Although circulating monocytes and tissue macrophages contribute to the pathogenesis of hypertension, the underlying mechanisms are poorly understood. Cysteine rich protein 1 (CRIP1) is highly expressed in immune cells, and CRIP1 mRNA expression in monocytes associates with blood pressure (BP) and is up-regulated by proinflammatory modulation suggesting a link between CRIP1 and BP regulation through the immune system. To address this functional link, we studied CRIP1 expression in immune cells in relation to BP using a human cohort study and hypertensive mouse models. CRIP1 expression in splenic monocytes/macrophages and in circulating monocytes was significantly affected by angiotensin II (Ang II) in a BP-elevating dose (2 mg/kg/day). In the human cohort study, monocytic CRIP1 expression levels were associated with elevated BP, whereas upon differentiation of monocytes to macrophages this association along with the CRIP1 expression level was diminished. In conclusion, CRIP1-positive circulating and splenic monocytes seem to play an important role in hypertension related inflammatory processes through endogenous hormones such as Ang II. These findings suggest that CRIP1 may affect the interaction between the immune system, in particular monocytes, and the pathogenesis of hypertension.
Collapse
|
128
|
Abstract
A link between oxidative stress and hypertension has been firmly established in multiple animal models of hypertension but remains elusive in humans. While initial studies focused on inactivation of nitric oxide by superoxide, our understanding of relevant reactive oxygen species (superoxide, hydrogen peroxide, and peroxynitrite) and how they modify complex signaling pathways to promote hypertension has expanded significantly. In this review, we summarize recent advances in delineating the primary and secondary sources of reactive oxygen species (nicotinamide adenine dinucleotide phosphate oxidases, uncoupled endothelial nitric oxide synthase, endoplasmic reticulum, and mitochondria), the posttranslational oxidative modifications they induce on protein targets important for redox signaling, their interplay with endogenous antioxidant systems, and the role of inflammasome activation and endoplasmic reticular stress in the development of hypertension. We highlight how oxidative stress in different organ systems contributes to hypertension, describe new animal models that have clarified the importance of specific proteins, and discuss clinical studies that shed light on how these processes and pathways are altered in human hypertension. Finally, we focus on the promise of redox proteomics and systems biology to help us fully understand the relationship between ROS and hypertension and their potential for designing and evaluating novel antihypertensive therapies.
Collapse
Affiliation(s)
- Kathy K Griendling
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, USA
| | - Livia L Camargo
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow
| | - Francisco Rios
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow
| | - Rhéure Alves-Lopes
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow
| |
Collapse
|
129
|
Madhur MS, Elijovich F, Alexander MR, Pitzer A, Ishimwe J, Van Beusecum JP, Patrick DM, Smart CD, Kleyman TR, Kingery J, Peck RN, Laffer CL, Kirabo A. Hypertension: Do Inflammation and Immunity Hold the Key to Solving this Epidemic? Circ Res 2021; 128:908-933. [PMID: 33793336 PMCID: PMC8023750 DOI: 10.1161/circresaha.121.318052] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Elevated cardiovascular risk including stroke, heart failure, and heart attack is present even after normalization of blood pressure in patients with hypertension. Underlying immune cell activation is a likely culprit. Although immune cells are important for protection against invading pathogens, their chronic overactivation may lead to tissue damage and high blood pressure. Triggers that may initiate immune activation include viral infections, autoimmunity, and lifestyle factors such as excess dietary salt. These conditions activate the immune system either directly or through their impact on the gut microbiome, which ultimately produces chronic inflammation and hypertension. T cells are central to the immune responses contributing to hypertension. They are activated in part by binding specific antigens that are presented in major histocompatibility complex molecules on professional antigen-presenting cells, and they generate repertoires of rearranged T-cell receptors. Activated T cells infiltrate tissues and produce cytokines including interleukin 17A, which promote renal and vascular dysfunction and end-organ damage leading to hypertension. In this comprehensive review, we highlight environmental, genetic, and microbial associated mechanisms contributing to both innate and adaptive immune cell activation leading to hypertension. Targeting the underlying chronic immune cell activation in hypertension has the potential to mitigate the excess cardiovascular risk associated with this common and deadly disease.
Collapse
Affiliation(s)
- Meena S. Madhur
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center
- Department of Molecular Physiology and Biophysics, Vanderbilt University
| | - Fernando Elijovich
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Matthew R. Alexander
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center
| | - Ashley Pitzer
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jeanne Ishimwe
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Justin P. Van Beusecum
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - David M. Patrick
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center
| | - Charles D. Smart
- Department of Molecular Physiology and Biophysics, Vanderbilt University
| | - Thomas R. Kleyman
- Departments of Medicine, Cell Biology, Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Justin Kingery
- Center for Global Health, Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Weill Bugando School of Medicine, Mwanza, Tanzania
| | - Robert N. Peck
- Center for Global Health, Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Weill Bugando School of Medicine, Mwanza, Tanzania
- Mwanza Intervention Trials Unit (MITU), Mwanza, Tanzania
| | - Cheryl L. Laffer
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University
| |
Collapse
|
130
|
Fang S, Livergood MC, Nakagawa P, Wu J, Sigmund CD. Role of the Peroxisome Proliferator Activated Receptors in Hypertension. Circ Res 2021; 128:1021-1039. [PMID: 33793338 DOI: 10.1161/circresaha.120.318062] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nuclear receptors represent a large family of ligand-activated transcription factors which sense the physiological environment and make long-term adaptations by mediating changes in gene expression. In this review, we will first discuss the fundamental mechanisms by which nuclear receptors mediate their transcriptional responses. We will focus on the PPAR (peroxisome proliferator-activated receptor) family of adopted orphan receptors paying special attention to PPARγ, the isoform with the most compelling evidence as an important regulator of arterial blood pressure. We will review genetic data showing that rare mutations in PPARγ cause severe hypertension and clinical trial data which show that PPARγ activators have beneficial effects on blood pressure. We will detail the tissue- and cell-specific molecular mechanisms by which PPARs in the brain, kidney, vasculature, and immune system modulate blood pressure and related phenotypes, such as endothelial function. Finally, we will discuss the role of placental PPARs in preeclampsia, a life threatening form of hypertension during pregnancy. We will close with a viewpoint on future research directions and implications for developing novel therapies.
Collapse
Affiliation(s)
- Shi Fang
- Department of Physiology, Cardiovascular Center (S.F., P.N., J.W., C.D.S.), Medical College of Wisconsin, Milwaukee.,Department of Neuroscience and Pharmacology, University of Iowa (S.F.)
| | - M Christine Livergood
- Department of Obstetrics and Gynecology (M.C.L.), Medical College of Wisconsin, Milwaukee
| | - Pablo Nakagawa
- Department of Physiology, Cardiovascular Center (S.F., P.N., J.W., C.D.S.), Medical College of Wisconsin, Milwaukee
| | - Jing Wu
- Department of Physiology, Cardiovascular Center (S.F., P.N., J.W., C.D.S.), Medical College of Wisconsin, Milwaukee
| | - Curt D Sigmund
- Department of Physiology, Cardiovascular Center (S.F., P.N., J.W., C.D.S.), Medical College of Wisconsin, Milwaukee
| |
Collapse
|
131
|
Wenstedt EFE, van Croonenburg TJ, van den Born BJH, Van den Bossche J, Hooijmans CR, Vogt L. The effect of macrophage-targeted interventions on blood pressure - a systematic review and meta-analysis of preclinical studies. Transl Res 2021; 230:123-138. [PMID: 33166696 DOI: 10.1016/j.trsl.2020.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/08/2020] [Accepted: 11/03/2020] [Indexed: 01/01/2023]
Abstract
An increasing body of evidence shows a role for macrophages and monocytes (as their precursors) in hypertension, but with conflicting results with regard to whether they are protective or harmful. Therefore, we systematically reviewed the effect of macrophage interventions on blood pressure in animal models, to explore which factors determine the blood pressure increasing vs. decreasing effect. A search in PubMED and EMBASE yielded 9620 records, 26 of which were included. Eighteen studies (involving 22 different experiments (k = 22)) performed macrophage depletion, whereas 12 studies specifically deleted certain macrophage proteins. The blood pressure effects of macrophage depletion were highly various and directed toward both directions, as expected, which could not be reduced to differences in animal species or methods of hypertension induction. Prespecified subgroup analysis did reveal a potential role for the route in which the macrophage-depleting agent is being administrated (intraperitoneal vs intravenous subgroup difference of P = 0.07 (k = 22), or P < 0.001 in studies achieving considerable (ie, >50%) depletion (k = 18)). Along with findings from specific macrophage protein deletion studies-showing that deletion of one single macrophage protein (like TonEBP, endothelin-B, EP4, NOX-2 and the angiotensin II type 1 receptor) can alter blood pressure responses to hypertensive stimuli-the indication that each route has its specific depletion pattern regarding targeted tissues and macrophage phenotypes suggests a determinative role for these features. These hypothesis-generating results encourage more detailed depletion characterization of each technique by direct experimental comparisons, providing a chance to obtain more knowledge on which macrophages are beneficial versus detrimental in hypertension development.
Collapse
Affiliation(s)
- Eliane F E Wenstedt
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Thirza J van Croonenburg
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Bert-Jan H van den Born
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Jan Van den Bossche
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Carlijn R Hooijmans
- Systematic Review Centre for Laboratory Animal Experimentation (SYRCLE), Department of Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Liffert Vogt
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
| |
Collapse
|
132
|
Andreadou I, Daiber A, Baxter GF, Brizzi MF, Di Lisa F, Kaludercic N, Lazou A, Varga ZV, Zuurbier CJ, Schulz R, Ferdinandy P. Influence of cardiometabolic comorbidities on myocardial function, infarction, and cardioprotection: Role of cardiac redox signaling. Free Radic Biol Med 2021; 166:33-52. [PMID: 33588049 DOI: 10.1016/j.freeradbiomed.2021.02.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/03/2021] [Accepted: 02/06/2021] [Indexed: 02/06/2023]
Abstract
The morbidity and mortality from cardiovascular diseases (CVD) remain high. Metabolic diseases such as obesity, hyperlipidemia, diabetes mellitus (DM), non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) as well as hypertension are the most common comorbidities in patients with CVD. These comorbidities result in increased myocardial oxidative stress, mainly from increased activity of nicotinamide adenine dinucleotide phosphate oxidases, uncoupled endothelial nitric oxide synthase, mitochondria as well as downregulation of antioxidant defense systems. Oxidative and nitrosative stress play an important role in ischemia/reperfusion injury and may account for increased susceptibility of the myocardium to infarction and myocardial dysfunction in the presence of the comorbidities. Thus, while early reperfusion represents the most favorable therapeutic strategy to prevent ischemia/reperfusion injury, redox therapeutic strategies may provide additive benefits, especially in patients with heart failure. While oxidative and nitrosative stress are harmful, controlled release of reactive oxygen species is however important for cardioprotective signaling. In this review we summarize the current data on the effect of hypertension and major cardiometabolic comorbidities such as obesity, hyperlipidemia, DM, NAFLD/NASH on cardiac redox homeostasis as well as on ischemia/reperfusion injury and cardioprotection. We also review and discuss the therapeutic interventions that may restore the redox imbalance in the diseased myocardium in the presence of these comorbidities.
Collapse
Affiliation(s)
- Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece.
| | - Andreas Daiber
- Department of Cardiology 1, Molecular Cardiology, University Medical Center, Langenbeckstr. 1, 55131, Mainz, Germany; Partner Site Rhine-Main, German Center for Cardiovascular Research (DZHK), Langenbeckstr, Germany.
| | - Gary F Baxter
- Division of Pharmacology, School of Pharmacy and Pharmaceutical Sciences, Cardiff University, United Kingdom
| | | | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, Italy; Neuroscience Institute, National Research Council of Italy (CNR), Padova, Italy
| | - Nina Kaludercic
- Neuroscience Institute, National Research Council of Italy (CNR), Padova, Italy
| | - Antigone Lazou
- Laboratory of Animal Physiology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
| | - Coert J Zuurbier
- Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany.
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| |
Collapse
|
133
|
Herster F, Karbach S, Chatterjee M, Weber ANR. Platelets: Underestimated Regulators of Autoinflammation in Psoriasis. J Invest Dermatol 2021; 141:1395-1403. [PMID: 33810836 DOI: 10.1016/j.jid.2020.12.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/02/2020] [Accepted: 12/02/2020] [Indexed: 01/01/2023]
Abstract
Platelets have long been known as mediators of hemostasis and, more recently, as mediators of thromboinflammation, although their physiopathological role has mostly been investigated in the context of disease of internal organs, such as liver and kidney, or systemic disorders. Of late, exciting recent data suggest that platelets may also play a role in inflammation at distal sites such as the skin: recent studies show that platelets, by engaging polymorphonuclear neutrophils (PMNs), contribute to local inflammation in the frequent skin disorder, psoriasis. In an experimental model, systemic depletion of platelets drastically attenuated skin inflammation by preventing PMN infiltration of the skin. A broader role of platelets in different types of skin inflammation is therefore likely, and in this paper, we specifically review recent advances in psoriasis. Special emphasis is given to the crosstalk with systemic platelet effects, which may be of interest in psoriasis-related cardiovascular comorbidities. Furthermore, we discuss the potential for platelet-centered interventions in the therapy for psoriasis.
Collapse
Affiliation(s)
- Franziska Herster
- Department of Immunology, Interfaculty Institute of Cell Biology, University of Tübingen, Tübingen, Germany; Department of Molecular Oncology, Robert Bosch Centrum für Tumorerkrankungen (RBCT), Robert-Bosch-Krankenhaus, Stuttgart, Germany
| | - Susanne Karbach
- Center for Cardiology - Cardiology I, University Medical Center Mainz and Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Madhumita Chatterjee
- Department of Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | - Alexander N R Weber
- Department of Immunology, Interfaculty Institute of Cell Biology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
134
|
Wang T, Gao L, Yang Z, Wang F, Guo Y, Wang B, Hua R, Shang H, Xu J. Restraint Stress in Hypertensive Rats Activates the Intestinal Macrophages and Reduces Intestinal Barrier Accompanied by Intestinal Flora Dysbiosis. J Inflamm Res 2021; 14:1085-1110. [PMID: 33790622 PMCID: PMC8007621 DOI: 10.2147/jir.s294630] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/22/2021] [Indexed: 12/14/2022] Open
Abstract
Purpose Hypertension (HTN) is a major risk factor for cardiovascular disease. In recent years, there were numerous studies on the function of stress in HTN. However, the gut dysbiosis linked to hypertension in animal models under stress is still incompletely understood. Purpose of this study is to use multiple determination method to determine the juvenile stage intestinal bacteria, cytokines and changes in hormone levels. Methods Four groups of juvenile male spontaneously hypertensive rats (SHRs) and age-matched male Wistar-Kyoto (WKY) rats were randomly selected as control and experimental groups. Rats in the two stress groups were exposed to restraint stress for 3 hours per day for 7 consecutive days. In one day three times in the method of non-invasive type tail-cuff monitoring blood pressure. The detailed mechanism was illuminated based on the intestinal change using immunohistochemical and immunofluorescence staining and the stress-related hormone and inflammation factors were analyzed via ELISA method. The integrity of the epithelial barrier was assessed using FITC/HRP and the expression levels of proteins associated with the tight junction was detected by Western blot. The alteration of stress-related intestinal flora from ileocecal junction and distal colon were also analyzed using its 16S rDNA sequencing. Results The results indicate that acute stress rapidly increases mean arterial pressure which is positive correlation to hormone concentration, especially in SHR-stress group. Meanwhile, stress promoted the enhancement of epithelial permeability accompanied with a reduced expression of the tight junction-related protein and the macrophages (Mφ) aggregation to the lamina propria. There were remarkable significant increase of stress-related hormones and pro-inflammatory factor interleukin (IL)-6 along with a decrease in the diversity of intestinal flora and an imbalance in the F/B ratio. Conclusion Our results reveal that stress accompanied with HTN could significantly disrupt the domino effect between intestinal flora and homeostasis.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of Physiology and Pathophysiology, Basic Medical College, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Lei Gao
- Department of Biomedical Informatics, School of Biomedical Engineering, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Zejun Yang
- Department of Clinical Medicine, Basic Medical College, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Feifei Wang
- Department of Clinical Medicine, Basic Medical College, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Yuexin Guo
- Department of Oral Medicine, Basic Medical College, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Boya Wang
- Eight Program of Clinical Medicine, Peking University Health Science Center, Beijing, 100081, People's Republic of China
| | - Rongxuan Hua
- Department of Clinical Medicine, Basic Medical College, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Hongwei Shang
- Experimental Center for Morphological Research Platform, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Jingdong Xu
- Department of Physiology and Pathophysiology, Basic Medical College, Capital Medical University, Beijing, 100069, People's Republic of China
| |
Collapse
|
135
|
Michell DL, Shihata WA, Andrews KL, Abidin NAZ, Jefferis AM, Sampson AK, Lumsden NG, Huet O, Parat MO, Jennings GL, Parton RG, Woollard KJ, Kaye DM, Chin-Dusting JPF, Murphy AJ. High intraluminal pressure promotes vascular inflammation via caveolin-1. Sci Rep 2021; 11:5894. [PMID: 33723357 PMCID: PMC7960707 DOI: 10.1038/s41598-021-85476-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 02/19/2021] [Indexed: 01/18/2023] Open
Abstract
The aetiology and progression of hypertension involves various endogenous systems, such as the renin angiotensin system, the sympathetic nervous system, and endothelial dysfunction. Recent data suggest that vascular inflammation may also play a key role in the pathogenesis of hypertension. This study sought to determine whether high intraluminal pressure results in vascular inflammation. Leukocyte adhesion was assessed in rat carotid arteries exposed to 1 h of high intraluminal pressure. The effect of intraluminal pressure on signaling mechanisms including reactive oxygen species production (ROS), arginase expression, and NFĸB translocation was monitored. 1 h exposure to high intraluminal pressure (120 mmHg) resulted in increased leukocyte adhesion and inflammatory gene expression in rat carotid arteries. High intraluminal pressure also resulted in a downstream signaling cascade of ROS production, arginase expression, and NFĸB translocation. This process was found to be angiotensin II-independent and mediated by the mechanosensor caveolae, as caveolin-1 (Cav1)-deficient endothelial cells and mice were protected from pressure-induced vascular inflammatory signaling and leukocyte adhesion. Cav1 deficiency also resulted in a reduction in pressure-induced glomerular macrophage infiltration in vivo. These findings demonstrate Cav1 is an important mechanosensor in pressure-induced vascular and renal inflammation.
Collapse
Affiliation(s)
- Danielle L Michell
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Medicine, Monash University, Clayton, VIC, Australia
| | - Waled A Shihata
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.
- Department of Medicine, Monash University, Clayton, VIC, Australia.
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia.
| | - Karen L Andrews
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Department of Pharmacology, Monash University, Clayton, VIC, Australia
| | - Nurul Aisha Zainal Abidin
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Department of Pharmacology, Monash University, Clayton, VIC, Australia
| | | | | | | | - Olivier Huet
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Marie-Odile Parat
- School of Pharmacy, University of Queensland, St Lucia, QLD, Australia
| | | | - Robert G Parton
- Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis, University of Queensland, St Lucia, QLD, Australia
| | - Kevin J Woollard
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - David M Kaye
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Jaye P F Chin-Dusting
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Medicine, Monash University, Clayton, VIC, Australia
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Department of Pharmacology, Monash University, Clayton, VIC, Australia
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| |
Collapse
|
136
|
Jung R, Wild J, Ringen J, Karbach S, Wenzel P. Innate Immune Mechanisms of Arterial Hypertension and Autoimmune Disease. Am J Hypertens 2021; 34:143-153. [PMID: 32930786 DOI: 10.1093/ajh/hpaa145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/15/2020] [Accepted: 09/14/2020] [Indexed: 12/16/2022] Open
Abstract
The immune system is indispensable in the development of vascular dysfunction and hypertension. The interplay between immune cells and the vasculature, kidneys, heart, and blood pressure regulating nuclei in the central nervous system results in a complex and closely interwoven relationship of the immune system with arterial hypertension. A better understanding of this interplay is necessary for optimized and individualized antihypertensive therapy. Our review article focuses on innate cells in hypertension and to what extent they impact on development and preservation of elevated blood pressure. Moreover, we address the association of hypertension with chronic autoimmune diseases. The latter are ideally suited to learn about immune-mediated mechanisms in cardiovascular disease leading to high blood pressure.
Collapse
Affiliation(s)
- Rebecca Jung
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Johannes Wild
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
- Department of Cardiology, University Medical Center Mainz, Mainz, Germany
| | - Julia Ringen
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Susanne Karbach
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
- Department of Cardiology, University Medical Center Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), partner site Rhine-Main, Germany
| | - Philip Wenzel
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
- Department of Cardiology, University Medical Center Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), partner site Rhine-Main, Germany
| |
Collapse
|
137
|
Topchieva LV, Korneva VA, Kurbatova IV. The relationship of the carriership of allelic variations in rs2228145 (A > C) of the IL6R gene with the levels of VCAM1 and ICAM1 gene transcripts in patients with essential hypertension. Vavilovskii Zhurnal Genet Selektsii 2021; 24:96-101. [PMID: 33659786 PMCID: PMC7716534 DOI: 10.18699/vj20.600] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The levels of plasma interleukin 6 and its soluble receptors were found to be elevated in subjects with cardiovascular diseases, which points to amplification of the IL-6-mediated trans-signaling pathway in cells and the development of chronic inflammation. The allelic variation in the rs2228145 IL6R gene is associated with a change in the contents of the soluble and membrane-bound receptor forms mediating the biological activity of IL-6. Cytokine IL-6 is involved in the development of endothelial dysfunction by regulating the expression of the VCAM1 and ICAM1 genes, encoding intercellular adhesion molecules. Prior to this work, no data on the association of essential arterial hypertension (EAH) with rs2228145 allelic variations of the IL6R gene have been reported. The aim of our work was to study the relationship of the carriership of rs2228145 (A > C) allelic variations with the development of EAH and the VCAM1 and ICAM1 transcript levels. We analyzed samples of DNA isolated from the whole blood of 148 healthy donors and 152 patients with EAH (stages I–II). The genotyping was performed by PCR-RFLP. The level of transcripts in peripheral blood leukocytes (PBL) was assessed by real-time PCR. Differences in the frequency distributions of rs2228145 (A > C) genotypes between the control group and the group of patients with EAH (χ2 = 9.303) were found. The frequency of the CC genotype in EAH patients was higher than in healthy people (0.191 and 0.095, respectively). The risk of EAH (I–II stages) development was shown to be 2.3 times higher in CC genotype carriers as compared to individuals with other genotypes (OR = 2.257, 95 % confidence interval 1.100–4.468).
The levels of VCAM1 and ICAM1 gene transcripts in PBL of patients with EAH were significantly higher than in healthy people. The level of ICAM1 gene transcripts was almost 4 times higher in patients with CC genotype. The Kruskal–Wallis analysis of variance revealed an effect of rs2228145 (A > C) genotype on the transcriptional activity of ICAM1, which argues for its role in the pathogenesis of endothelial dysfunction and essential hypertension.
Collapse
Affiliation(s)
- L V Topchieva
- Institute of Biology of the Karelian Research Centre of the Russian Academy of Sciences, Petrozavodsk, Russia
| | - V A Korneva
- Petrozavodsk State University, Petrozavodsk, Russia
| | - I V Kurbatova
- Institute of Biology of the Karelian Research Centre of the Russian Academy of Sciences, Petrozavodsk, Russia
| |
Collapse
|
138
|
Olivencia MA, Martínez-Casales M, Peraza DA, García-Redondo AB, Mondéjar-Parreño G, Hernanz R, Salaices M, Cogolludo A, Pennington MW, Valenzuela C, Briones AM. K V 1.3 channels are novel determinants of macrophage-dependent endothelial dysfunction in angiotensin II-induced hypertension in mice. Br J Pharmacol 2021; 178:1836-1854. [PMID: 33556997 DOI: 10.1111/bph.15407] [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: 07/16/2020] [Revised: 01/28/2021] [Accepted: 01/31/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE KV 1.3 channels are expressed in vascular smooth muscle cells (VSMCs), where they contribute to proliferation rather than contraction and participate in vascular remodelling. KV 1.3 channels are also expressed in macrophages, where they assemble with KV 1.5 channels (KV 1.3/KV 1.5), whose activation generates a KV current. In macrophages, the KV 1.3/KV 1.5 ratio is increased by classical activation (M1). Whether these channels are involved in angiotensin II (AngII)-induced vascular remodelling, and whether they can modulate the macrophage phenotype in hypertension, remains unknown. We characterized the role of KV 1.3 channels in vascular damage in hypertension. EXPERIMENTAL APPROACH We used AngII-infused mice treated with two selective KV 1.3 channel inhibitors (HsTX[R14A] and [EWSS]ShK). Vascular function and structure were measured using wire and pressure myography, respectively. VSMC and macrophage electrophysiology were studied using the patch-clamp technique; gene expression was analysed using RT-PCR. KEY RESULTS AngII increased KV 1.3 channel expression in mice aorta and peritoneal macrophages which was abolished by HsTX[R14A] treatment. KV 1.3 inhibition did not prevent hypertension, vascular remodelling, or stiffness but corrected AngII-induced macrophage infiltration and endothelial dysfunction in the small mesenteric arteries and/or aorta, via a mechanism independent of electrophysiological changes in VSMCs. AngII modified the electrophysiological properties of peritoneal macrophages, indicating an M1-like activated state, with enhanced expression of proinflammatory cytokines that induced endothelial dysfunction. These effects were prevented by KV 1.3 blockade. CONCLUSIONS AND IMPLICATIONS We unravelled a new role for KV 1.3 channels in the macrophage-dependent endothelial dysfunction induced by AngII in mice which might be due to modulation of macrophage phenotype.
Collapse
Affiliation(s)
- Miguel A Olivencia
- Departamento de Farmacología, Universidad Autónoma de Madrid, Instituto de Investigación Hospital La Paz, Madrid, Spain.,Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber de Enfermedades Respiratorias (CIBERES), Spain
| | - Marta Martínez-Casales
- Departamento de Farmacología, Universidad Autónoma de Madrid, Instituto de Investigación Hospital La Paz, Madrid, Spain.,Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain
| | - Diego A Peraza
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
| | - Ana B García-Redondo
- Departamento de Farmacología, Universidad Autónoma de Madrid, Instituto de Investigación Hospital La Paz, Madrid, Spain.,Ciber de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Gema Mondéjar-Parreño
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber de Enfermedades Respiratorias (CIBERES), Spain
| | - Raquel Hernanz
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain.,Ciber de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Mercedes Salaices
- Departamento de Farmacología, Universidad Autónoma de Madrid, Instituto de Investigación Hospital La Paz, Madrid, Spain.,Ciber de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Angel Cogolludo
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber de Enfermedades Respiratorias (CIBERES), Spain
| | | | - Carmen Valenzuela
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain.,Ciber de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Ana M Briones
- Departamento de Farmacología, Universidad Autónoma de Madrid, Instituto de Investigación Hospital La Paz, Madrid, Spain.,Ciber de Enfermedades Cardiovasculares (CIBERCV), Spain
| |
Collapse
|
139
|
McCarthy CG, Saha P, Golonka RM, Wenceslau CF, Joe B, Vijay-Kumar M. Innate Immune Cells and Hypertension: Neutrophils and Neutrophil Extracellular Traps (NETs). Compr Physiol 2021; 11:1575-1589. [PMID: 33577121 DOI: 10.1002/cphy.c200020] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Uncontrolled immune system activation amplifies end-organ injury in hypertension. Nonetheless, the exact mechanisms initiating this exacerbated inflammatory response, thereby contributing to further increases in blood pressure (BP), are still being revealed. While participation of lymphoid-derived immune cells has been well described in the hypertension literature, the mechanisms by which myeloid-derived innate immune cells contribute to T cell activation, and subsequent BP elevation, remains an active area of investigation. In this article, we critically analyze the literature to understand how monocytes, macrophages, dendritic cells, and polymorphonuclear leukocytes, including mast cells, eosinophils, basophils, and neutrophils, contribute to hypertension and hypertension-associated end-organ injury. The most abundant leukocytes, neutrophils, are indisputably increased in hypertension. However, it is unknown how (and why) they switch from critical first responders of the innate immune system, and homeostatic regulators of BP, to tissue-damaging, pro-hypertensive mediators. We propose that myeloperoxidase-derived pro-oxidants, neutrophil elastase, neutrophil extracellular traps (NETs), and interactions with other innate and adaptive immune cells are novel mechanisms that could contribute to the inflammatory cascade in hypertension. We further posit that the gut microbiota serves as a set point for neutropoiesis and their function. Finally, given that hypertension appears to be a key risk factor for morbidity and mortality in COVID-19 patients, we put forth evidence that neutrophils and NETs cause cardiovascular injury post-coronavirus infection, and thus may be proposed as an intriguing therapeutic target for high-risk individuals. © 2021 American Physiological Society. Compr Physiol 11:1575-1589, 2021.
Collapse
Affiliation(s)
- Cameron G McCarthy
- Program in Physiological Genomics, UT Microbiome Consortium, Center for Hypertension & Personalized Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Piu Saha
- Program in Physiological Genomics, UT Microbiome Consortium, Center for Hypertension & Personalized Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Rachel M Golonka
- Program in Physiological Genomics, UT Microbiome Consortium, Center for Hypertension & Personalized Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Camilla F Wenceslau
- Program in Physiological Genomics, UT Microbiome Consortium, Center for Hypertension & Personalized Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Bina Joe
- Program in Physiological Genomics, UT Microbiome Consortium, Center for Hypertension & Personalized Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Matam Vijay-Kumar
- Program in Physiological Genomics, UT Microbiome Consortium, Center for Hypertension & Personalized Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| |
Collapse
|
140
|
Zekavat SM, Honigberg M, Pirruccello JP, Kohli P, Karlson EW, Newton-Cheh C, Zhao H, Natarajan P. Elevated Blood Pressure Increases Pneumonia Risk: Epidemiological Association and Mendelian Randomization in the UK Biobank. MED 2021; 2:137-148.e4. [PMID: 33283203 PMCID: PMC7703520 DOI: 10.1016/j.medj.2020.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/22/2020] [Accepted: 11/02/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Small studies have correlated hypertension with pneumonia risk; whether this is recapitulated in larger prospective studies, and represents a causal association, is unclear. METHODS We estimated the risk for prevalent hypertension with incident respiratory diseases over mean follow-up of 8 years among 377,143 British participants in the UK Biobank. Mendelian randomization of blood pressure on pneumonia was implemented using 75 independent, genome-wide significant variants associated with systolic and diastolic blood pressures among 299,024 individuals not in the UK Biobank. Secondary analyses with pulmonary function tests were performed. FINDINGS In total, 107,310 participants (30%) had hypertension at UK Biobank enrollment, and 9,969 (3%) developed pneumonia during follow-up. Prevalent hypertension was independently associated with increased risk for incident pneumonia (HR: 1.36; 95% CI: 1.29-1.43; p < 0.001), as well as other incident respiratory diseases. Genetic predisposition to a 5 mm Hg increase in blood pressure was associated with increased risk for incident pneumonia for systolic blood pressure (HR: 1.08; 95% CI: 1.04-1.13; p < 0.001) and diastolic blood pressure (HR: 1.11; 95% CI: 1.03-1.20; p = 0.005). Additionally, consistent with epidemiologic associations, increased blood pressure genetic risk was significantly associated with reduced performance on pulmonary function tests (p < 0.001). CONCLUSIONS These results suggest that elevated blood pressure increases risk for pneumonia. Maintaining adequate blood pressure control, in addition to other measures, may reduce risk for pneumonia. FUNDING S.M.Z. (1F30HL149180-01), M.H. (T32HL094301-07), and P.N. (R01HL1427, R01HL148565, and R01HL148050) are supported by the National Institutes of Health. J.P. is supported by the John S. LaDue Memorial Fellowship.
Collapse
Affiliation(s)
- Seyedeh M Zekavat
- Yale School of Medicine, New Haven, CT, USA
- Computational Biology & Bioinformatics Program, Yale University, New Haven, CT, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Michael Honigberg
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - James P Pirruccello
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Puja Kohli
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Vertex Pharmaceuticals, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Elizabeth W Karlson
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Allergy, Immunology, and Rheumatology, Brigham and Women's Hospital, Boston, MA, USA
| | - Christopher Newton-Cheh
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Hongyu Zhao
- Computational Biology & Bioinformatics Program, Yale University, New Haven, CT, USA
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Pradeep Natarajan
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
141
|
Abstract
PURPOSE OF REVIEW Macrophages play an important role in regulating homeostasis, kidney injury, repair, and tissue fibrogenesis. The present review will discuss recent advances that explore the novel subsets and functions of macrophage in the pathogenesis of kidney damage and hypertension. RECENT FINDINGS Macrophages differentiate into a variety of subsets in microenvironment-dependent manner. Although the M1/M2 nomenclature is still applied in considering the pro-inflammatory versus anti-inflammatory effects of macrophages in kidney injury, novel, and accurate macrophage phenotypes are defined by flow cytometric markers and single-cell RNA signatures. Studies exploring the crosstalk between macrophages and other cells are rapidly advancing with the additional recognition of exosome trafficking between cells. Using murine conditional mutants, actions of macrophage can be defined more precisely than in bone marrow transfer models. Some studies revealed the opposing effects of the same protein in renal parenchymal cells and macrophages, highlighting a need for the development of cell-specific immune therapies for translation. SUMMARY Macrophage-targeted therapies hold potential for limiting kidney injury and hypertension. To realize this potential, future studies will be required to understand precise mechanisms in macrophage polarization, crosstalk, proliferation, and maturation in the setting of renal disease.
Collapse
|
142
|
Gao Y, Ren C, Li X, Yu W, Li S, Li H, Wang Y, Li D, Ren M, Ji X. Ischemic Conditioning Ameliorated Hypertension and Vascular Remodeling of Spontaneously Hypertensive Rat via Inflammatory Regulation. Aging Dis 2021; 12:116-131. [PMID: 33532132 PMCID: PMC7801289 DOI: 10.14336/ad.2020.0320] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 03/27/2020] [Indexed: 01/03/2023] Open
Abstract
Vascular remodeling is an initial step in the development of hypertension. Limb remote ischemic conditioning (LRIC) is a physiological treatment that induces endogenous protective effect during acute ischemic injury. However, the impact of long-term LRIC on hypertension, a chronic disease, is unknown. In this study, we aimed to investigate the LRIC effect on blood pressure and vascular remodeling in spontaneously hypertensive rat (SHR) model and patients with prehypertension and early-stage hypertension. LRIC of rats was performed once a day for 6-weeks. Blood pressure, vascular remodeling (cross-sectional area, extracellular deposition, and smooth muscle cell area), inflammation (inflammatory factors, and inflammatory cells) were compared among normotensive Wistar-Kyoto rats (WKY), WKY RIC group, SHR control group, and SHR RIC. Long-term LRCI treatment (twice a day for 4-weeks) was performed on patients with prehypertension or early-stage hypertension. Blood pressure and pulse wave velocity (PWV) were analyzed before and after LRIC treatment. LRIC treatment decreased blood pressure in SHR (n = 9-10). LRIC ameliorated vascular remodeling by decreasing cross-sectional area, suppressing deposition of the extracellular matrix, and hypertrophy of smooth muscle cell in conduit artery and small resistance artery (n = 7). LRIC decreased proinflammatory factors while increasing the anti-inflammatory factors in the circulation (n = 5). LRIC decreased circulating monocyte and natural killer T-cell levels (n = 5). Furthermore, LRIC treatment decreased blood pressure and improved vascular stiffness in patients (n = 20). In conclusion, long term LRIC could decrease blood pressure and ameliorate vascular remodeling via inflammation regulation. LRIC could be a preventive treatment for people with blood pressure elevation or prehypertension.
Collapse
Affiliation(s)
- Yu Gao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 10053, China.
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical, Beijing, 10053, China.
| | - Changhong Ren
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical, Beijing, 10053, China.
- Beijing municipal geriatric medical research center, Beijing, 10053, China.
| | - Xiaohua Li
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical, Beijing, 10053, China.
- Beijing municipal geriatric medical research center, Beijing, 10053, China.
| | - Wantong Yu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 10053, China.
| | - Sijie Li
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical, Beijing, 10053, China.
- Beijing municipal geriatric medical research center, Beijing, 10053, China.
| | - Haiyan Li
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical, Beijing, 10053, China.
| | - Yan Wang
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical, Beijing, 10053, China.
| | - Dong Li
- Peking University Care Health Management Center, Beijing, 100080, China.
| | - Ming Ren
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 10053, China.
| | - Xunming Ji
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 10053, China.
- Department of Neurosurgery Xuanwu Hospital, Capital Medical, Beijing 100053, China.
| |
Collapse
|
143
|
Wild J, Wenzel P. Myeloid cells, tissue homeostasis, and anatomical barriers as innate immune effectors in arterial hypertension. J Mol Med (Berl) 2021; 99:315-326. [PMID: 33443617 PMCID: PMC7899956 DOI: 10.1007/s00109-020-02019-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 11/18/2020] [Indexed: 11/29/2022]
Abstract
Although essential hypertension affects a large proportion of the human population and is one of the key drivers of cardiovascular mortality worldwide, we still do not have a complete understanding of its pathophysiology. More than 50 years ago, the immune system has been identified as an important part of the pathogenesis of arterial hypertension. An exceeding variety of recent publications deals with the interplay between the numerous different components of the immune system and mechanisms of arterial hypertension and has substantially contributed to our understanding of the role of immunity and inflammation in the pathogenesis of the disease. In this review, we focus on myeloid cells and anatomical barriers as particular aspects of innate immunity in arterial hypertension. Since it represents a first line of defense protecting against pathogens and maintaining tissue homeostasis, innate immunity provides many mechanistic hinge points in the area of hypertension.
Collapse
Affiliation(s)
- Johannes Wild
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany.,Center for Cardiology - Cardiology I and CTH Professorship "Vascular Inflammation", University Medical Center Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.,German Center for Cardiovascular Research (DZHK) - Partner site RheinMain, Berlin, Germany
| | - Philip Wenzel
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany. .,Center for Cardiology - Cardiology I and CTH Professorship "Vascular Inflammation", University Medical Center Mainz, Langenbeckstr. 1, 55131, Mainz, Germany. .,German Center for Cardiovascular Research (DZHK) - Partner site RheinMain, Berlin, Germany.
| |
Collapse
|
144
|
Pan XX, Wu F, Chen XH, Chen DR, Chen HJ, Kong LR, Ruan CC, Gao PJ. T-cell senescence accelerates angiotensin II-induced target organ damage. Cardiovasc Res 2021; 117:271-283. [PMID: 32049355 DOI: 10.1093/cvr/cvaa032] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 01/04/2020] [Accepted: 02/05/2020] [Indexed: 01/08/2023] Open
Abstract
AIMS Aging is a risk factor for cardiovascular diseases and adaptive immunity has been implicated in angiotensin (Ang) II-induced target organ dysfunction. Herein, we sought to determine the role of T-cell senescence in Ang II-induced target organ impairment and to explore the underlying mechanisms. METHODS AND RESULTS Flow cytometric analysis revealed that T cell derived from aged mice exhibited immunosenescence. Adoptive transfer of aged T cells to immunodeficient RAG1 KO mice accelerates Ang II-induced cardiovascular and renal fibrosis compared with young T-cell transfer. Aged T cells also promote inflammatory factor expression and superoxide production in these target organs. In vivo and in vitro studies revealed that Ang II promotes interferon-gamma (IFN-γ) production in the aged T cells comparing to young T cells. Importantly, transfer of senescent T cell that IFN-γ KO mitigates the impairment. Aged T-cell-conditioned medium stimulates inflammatory factor expression and oxidative stress in Ang II-treated renal epithelial cells compared with young T cells, and these effects of aged T-cell-conditioned medium are blunted after IFN-γ-neutralizing antibody pre-treatment. CONCLUSION These results provide a significant insight into the contribution of senescent T cells to Ang II-induced cardiovascular dysfunction and provide an attractive possibility that targeting T cell specifically might be a potential strategy to treat elderly hypertensive patients with end-organ dysfunction.
Collapse
Affiliation(s)
- Xiao-Xi Pan
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Fang Wu
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiao-Hui Chen
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Dong-Rui Chen
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Hong-Jin Chen
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Ling-Ran Kong
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Cheng-Chao Ruan
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Ping-Jin Gao
- State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Department of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| |
Collapse
|
145
|
Zhang RM, McNerney KP, Riek AE, Bernal‐Mizrachi C. Immunity and Hypertension. Acta Physiol (Oxf) 2021; 231:e13487. [PMID: 32359222 DOI: 10.1111/apha.13487] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/24/2020] [Accepted: 04/25/2020] [Indexed: 12/15/2022]
Abstract
Hypertension is the primary cause of cardiovascular mortality. Despite multiple existing treatments, only half of those with the disease achieve adequate control. Therefore, understanding the mechanisms causing hypertension is essential for the development of novel therapies. Many studies demonstrate that immune cell infiltration of the vessel wall, kidney and central nervous system, as well as their counterparts of oxidative stress, the renal renin-angiotensin system (RAS) and sympathetic tone play a critical role in the development of hypertension. Genetically modified mice lacking components of innate and/or adaptive immunity confirm the importance of chronic inflammation in hypertension and its complications. Depletion of immune cells improves endothelial function, decreases oxidative stress, reduces vascular tone and prevents renal interstitial infiltrates, sodium retention and kidney damage. Moreover, the ablation of microglia or central nervous system perivascular macrophages reduces RAS-induced inflammation and prevents sympathetic nervous system activation and hypertension. Therefore, understanding immune cell functioning and their interactions with tissues that regulate hypertensive responses may be the future of novel antihypertensive therapies.
Collapse
Affiliation(s)
- Rong M. Zhang
- Department of Medicine Division of Endocrinology, Metabolism, and Lipid Research Washington University School of Medicine St. Louis MO USA
| | - Kyle P. McNerney
- Department of Pediatrics Washington University School of Medicine St. Louis MO USA
| | - Amy E. Riek
- Department of Medicine Division of Endocrinology, Metabolism, and Lipid Research Washington University School of Medicine St. Louis MO USA
| | - Carlos Bernal‐Mizrachi
- Department of Medicine Division of Endocrinology, Metabolism, and Lipid Research Washington University School of Medicine St. Louis MO USA
- Department of Cell Biology and Physiology Washington University School of Medicine St. Louis MO USA
- Department of Medicine VA Medical Center St. Louis MO USA
| |
Collapse
|
146
|
Molitor M, Rudi WS, Garlapati V, Finger S, Schüler R, Kossmann S, Lagrange J, Nguyen TS, Wild J, Knopp T, Karbach SH, Knorr M, Ruf W, Münzel T, Wenzel P. Nox2+ myeloid cells drive vascular inflammation and endothelial dysfunction in heart failure after myocardial infarction via angiotensin II receptor type 1. Cardiovasc Res 2021; 117:162-177. [PMID: 32077922 DOI: 10.1093/cvr/cvaa042] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 01/31/2020] [Accepted: 02/14/2020] [Indexed: 12/20/2022] Open
Abstract
AIMS Heart failure (HF) ensuing myocardial infarction (MI) is characterized by the initiation of a systemic inflammatory response. We aimed to elucidate the impact of myelomonocytic cells and their activation by angiotensin II on vascular endothelial function in a mouse model of HF after MI. METHODS AND RESULTS HF was induced in male C57BL/6J mice by permanent ligation of the left anterior descending coronary artery. Compared to sham, HF mice had significantly impaired endothelial function accompanied by enhanced mobilization of Sca-1+c-Kit+ haematopoietic stem cells and Sca-1-c-Kit+ common myeloid and granulocyte-macrophage progenitors in the bone marrow as well as increased vascular infiltration of CD11b+Ly6G-Ly6Chigh monocytes and accumulation of CD11b+ F4/80+ macrophages, assessed by flow cytometry. Using mice with Cre-inducible expression of diphtheria toxin receptor in myeloid cells, we selectively depleted lysozyme M+ myelomonocytic cells for 10 days starting 28 days after MI. While the cardiac phenotype remained unaltered until 38 days post-MI, myeloid cell depletion attenuated vascular accumulation of Nox2+CD45+ cells, endothelial dysfunction, oxidative stress, and vascular expression of adhesion molecules and angiotensin II receptor type 1 (AT1R). Pharmacological blockade of this receptor for 4 weeks did not significantly alter cardiac function, but mimicked the effects of myeloid cell depletion: telmisartan (20 mg/kg/day, fed to C57BL/6J mice) diminished bone marrow myelopoesis and myeloid reactive oxygen species production, attenuated endothelial leucocyte rolling and vascular accumulation of CD11b+Ly6G-Ly6Chigh monocytes and macrophages, resulting in improved vascular function with less abundance of Nox2+CD45+ cells. CONCLUSION Endothelial dysfunction in HF ensuing MI is mediated by inflammatory Nox2+ myeloid cells infiltrating the vessel wall that can be targeted by AT1R blockade.
Collapse
Affiliation(s)
- Michael Molitor
- Center for Thrombosis and Haemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Germany
| | - Wolf-Stephan Rudi
- Center for Thrombosis and Haemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Germany
| | - Venkata Garlapati
- Center for Thrombosis and Haemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Germany
| | - Stefanie Finger
- Center for Thrombosis and Haemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Rebecca Schüler
- Center for Thrombosis and Haemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- Institute for Molecular Medicine, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Sabine Kossmann
- Center for Thrombosis and Haemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- The Heart Research Institute, 7 Eliza Street, Newtown, NSW 2042, Australia
| | - Jeremy Lagrange
- Center for Thrombosis and Haemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Thanh Son Nguyen
- Center for Thrombosis and Haemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Johannes Wild
- Center for Thrombosis and Haemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Germany
| | - Tanja Knopp
- Center for Thrombosis and Haemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- Institute for Molecular Medicine, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Susanne H Karbach
- Center for Thrombosis and Haemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Germany
| | - Maike Knorr
- Center for Thrombosis and Haemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Wolfram Ruf
- Center for Thrombosis and Haemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Germany
- Scripps Research Institute, La Jolla, CA, USA
| | - Thomas Münzel
- Center for Thrombosis and Haemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Germany
| | - Philip Wenzel
- Center for Thrombosis and Haemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Germany
| |
Collapse
|
147
|
Mikolajczyk TP, Szczepaniak P, Vidler F, Maffia P, Graham GJ, Guzik TJ. Role of inflammatory chemokines in hypertension. Pharmacol Ther 2020; 223:107799. [PMID: 33359600 DOI: 10.1016/j.pharmthera.2020.107799] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
Hypertension is associated with immune cells activation and their migration into the kidney, vasculature, heart and brain. These inflammatory mechanisms are critical for blood pressure regulation and mediate target organ damage, creating unique novel targets for pharmacological modulation. In response to angiotensin II and other pro-hypertensive stimuli, the expression of several inflammatory chemokines and their receptors is increased in the target organs, mediating homing of immune cells. In this review, we summarize the contribution of key inflammatory chemokines and their receptors to increased accumulation of immune cells in target organs and effects on vascular dysfunction, remodeling, oxidative stress and fibrosis, all of which contribute to blood pressure elevation. In particular, the role of CCL2, CCL5, CXCL8, CXCL9, CXCL10, CXCL11, CXCL16, CXCL1, CX3CL1, XCL1 and their receptors in the context of hypertension is discussed. Recent studies have tested the efficacy of pharmacological or genetic targeting of chemokines and their receptors on the development of hypertension. Promising results indicate that some of these pathways may serve as future therapeutic targets to improve blood pressure control and prevent target organ consequences including kidney failure, heart failure, atherosclerosis or cognitive impairment.
Collapse
Affiliation(s)
- Tomasz P Mikolajczyk
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland; Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Piotr Szczepaniak
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Francesca Vidler
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Pasquale Maffia
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK; BHF Centre for Excellence Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK; Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Gerard J Graham
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Tomasz J Guzik
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland; BHF Centre for Excellence Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
148
|
Araos P, Figueroa S, Amador CA. The Role of Neutrophils in Hypertension. Int J Mol Sci 2020; 21:ijms21228536. [PMID: 33198361 PMCID: PMC7697449 DOI: 10.3390/ijms21228536] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023] Open
Abstract
It is well accepted that the immune system and some cells from adaptive and innate immunity are necessary for the initiation/perpetuation of arterial hypertension (AH). However, whether neutrophils are part of this group remains debatable. There is evidence showing that the neutrophil/lymphocyte ratio correlates with AH and is higher in non-dipper patients. On the other hand, the experimental neutrophil depletion in mice reduces basal blood pressure. Nevertheless, their participation in AH is still controversial. Apparently, neutrophils may modulate the microenvironment in blood vessels by increasing oxidative stress, favoring endothelial disfunction. In addition, neutrophils may contribute to the tissue infiltration of immune cells, secreting chemoattractant chemokines/cytokines and promoting the proinflammatory phenotype, leading to AH development. In this work, we discuss the potential role of neutrophils in AH by analyzing different mechanisms proposed from clinical and basic studies, with a perspective on cardiovascular and renal damages relating to the hypertensive phenotype.
Collapse
|
149
|
Hering L, Rahman M, Potthoff SA, Rump LC, Stegbauer J. Role of α2-Adrenoceptors in Hypertension: Focus on Renal Sympathetic Neurotransmitter Release, Inflammation, and Sodium Homeostasis. Front Physiol 2020; 11:566871. [PMID: 33240096 PMCID: PMC7680782 DOI: 10.3389/fphys.2020.566871] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 10/19/2020] [Indexed: 12/19/2022] Open
Abstract
The kidney is extensively innervated by sympathetic nerves playing an important role in the regulation of blood pressure homeostasis. Sympathetic nerve activity is ultimately controlled by the central nervous system (CNS). Norepinephrine, the main sympathetic neurotransmitter, is released at prejunctional neuroeffector junctions in the kidney and modulates renin release, renal vascular resistance, sodium and water handling, and immune cell response. Under physiological conditions, renal sympathetic nerve activity (RSNA) is modulated by peripheral mechanisms such as the renorenal reflex, a complex interaction between efferent sympathetic nerves, central mechanism, and afferent sensory nerves. RSNA is increased in hypertension and, therefore, critical for the perpetuation of hypertension and the development of hypertensive kidney disease. Renal sympathetic neurotransmission is not only regulated by RSNA but also by prejunctional α2-adrenoceptors. Prejunctional α2-adrenoceptors serve as autoreceptors which, when activated by norepinephrine, inhibit the subsequent release of norepinephrine induced by a sympathetic nerve impulse. Deletion of α2-adrenoceptors aggravates hypertension ultimately by modulating renal pressor response and sodium handling. α2-adrenoceptors are also expressed in the vasculature, renal tubules, and immune cells and exert thereby effects related to vascular tone, sodium excretion, and inflammation. In the present review, we highlight the role of α2-adrenoceptors on renal sympathetic neurotransmission and its impact on hypertension. Moreover, we focus on physiological and pathophysiological functions mediated by non-adrenergic α2-adrenoceptors. In detail, we discuss the effects of sympathetic norepinephrine release and α2-adrenoceptor activation on renal sodium transporters, on renal vascular tone, and on immune cells in the context of hypertension and kidney disease.
Collapse
Affiliation(s)
- Lydia Hering
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Masudur Rahman
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Sebastian A Potthoff
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Lars C Rump
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Johannes Stegbauer
- Department of Nephrology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
150
|
Göllner M, Ihrig-Biedert I, Petermann V, Saurin S, Oelze M, Kröller-Schön S, Vujacic-Mirski K, Kuntic M, Pautz A, Daiber A, Kleinert H. NOX2ko Mice Show Largely Increased Expression of a Mutated NOX2 mRNA Encoding an Inactive NOX2 Protein. Antioxidants (Basel) 2020; 9:antiox9111043. [PMID: 33114493 PMCID: PMC7692237 DOI: 10.3390/antiox9111043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 11/16/2022] Open
Abstract
Background: The superoxide-generating enzyme nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX2 or gp91phox, the phagocytic isoform) was reported as a major source of oxidative stress in various human diseases. Genetic deletion is widely used to study the impact of NOX2-derived reactive oxygen species (ROS) on disease development and progression in various animal models. Here, we investigate why NOX2 knockout mice show no NOX2 activity but express NOX2 mRNA and protein. Methods and Results: Oxidative burst (NOX2-dependent formation of ROS) was measured by L-012-based chemiluminescence and was largely absent in whole blood of NOX2 knockout mice. Protein expression was still detectable in different tissues of the NOX2 knockout mice, at the expected and a slightly lower molecular weight (determined by Western blot). The NOX2 gene was even largely enhanced at its expressional level in NOX2 knockout mice. RNA sequencing revealed a modified NOX2 mRNA in the knockout mice that is obviously translated to a truncated inactive mutant enzyme. Conclusion: Although the commercial NOX2 knockout mice display no considerable enzymatic NOX2 activity, expression of the NOX2 gene (when using standard primers) and protein (when using antibodies binding to the carboxy-terminal end) can still be detected, which may lead to confusion among investigators.
Collapse
Affiliation(s)
- Monika Göllner
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.G.); (I.I.-B.); (V.P.); (S.S.); (A.P.)
| | - Irmgard Ihrig-Biedert
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.G.); (I.I.-B.); (V.P.); (S.S.); (A.P.)
| | - Victoria Petermann
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.G.); (I.I.-B.); (V.P.); (S.S.); (A.P.)
| | - Sabrina Saurin
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.G.); (I.I.-B.); (V.P.); (S.S.); (A.P.)
| | - Matthias Oelze
- Laboratory of Molecular Cardiology, Department of Cardiology 1, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.O.); (S.K.-S.); (K.V.-M.); (M.K.)
| | - Swenja Kröller-Schön
- Laboratory of Molecular Cardiology, Department of Cardiology 1, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.O.); (S.K.-S.); (K.V.-M.); (M.K.)
| | - Ksenija Vujacic-Mirski
- Laboratory of Molecular Cardiology, Department of Cardiology 1, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.O.); (S.K.-S.); (K.V.-M.); (M.K.)
| | - Marin Kuntic
- Laboratory of Molecular Cardiology, Department of Cardiology 1, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.O.); (S.K.-S.); (K.V.-M.); (M.K.)
| | - Andrea Pautz
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.G.); (I.I.-B.); (V.P.); (S.S.); (A.P.)
| | - Andreas Daiber
- Laboratory of Molecular Cardiology, Department of Cardiology 1, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.O.); (S.K.-S.); (K.V.-M.); (M.K.)
- Correspondence: (A.D.); (H.K.); Tel.: +49-(6131)-17-6280 (A.D.); +49-(6131)-17-9150 (H.K.)
| | - Hartmut Kleinert
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; (M.G.); (I.I.-B.); (V.P.); (S.S.); (A.P.)
- Correspondence: (A.D.); (H.K.); Tel.: +49-(6131)-17-6280 (A.D.); +49-(6131)-17-9150 (H.K.)
| |
Collapse
|