201
|
Abstract
Arterial aging engages a plethora of key signalling pathways that act in concert to induce vascular smooth muscle cell (VSMC) phenotypic changes leading to vascular degeneration and extracellular matrix degradation responsible for alterations of the mechanical properties of the vascular wall. This review highlights proof-of-concept examples of components of the extracellular matrix, VSMC receptors which connect extracellular and intracellular structures, and signalling pathways regulating changes in mechanotransduction and vascular homeostasis in aging. Furthermore, it provides a new framework for understanding how VSMC stiffness and adhesion to extracellular matrix contribute to arterial stiffness and how interactions with endothelial cells, platelets, and immune cells can regulate vascular aging. The identification of the key players of VSMC changes operating in large and small-sized arteries in response to increased mechanical load may be useful to better elucidate the causes and consequences of vascular aging and associated progression of hypertension, arteriosclerosis, and atherosclerosis.
Collapse
Affiliation(s)
- Patrick Lacolley
- INSERM, U1116, Faculte de Medecine, 9 Avenue de la forêt de Haye, CS 50184, 54505 Vandœuvre-lès-Nancy, France.,Université de Lorraine, Nancy, France
| | - Veronique Regnault
- INSERM, U1116, Faculte de Medecine, 9 Avenue de la forêt de Haye, CS 50184, 54505 Vandœuvre-lès-Nancy, France.,Université de Lorraine, Nancy, France
| | - Alberto P Avolio
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, 2 Technology Place, Macquarie University, Sydney, NSW 2109, Australia
| |
Collapse
|
202
|
T Cell-Derived IL-17A Induces Vascular Dysfunction via Perivascular Fibrosis Formation and Dysregulation of ·NO/cGMP Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6721531. [PMID: 31396305 PMCID: PMC6668561 DOI: 10.1155/2019/6721531] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 05/27/2019] [Indexed: 02/07/2023]
Abstract
Aims The neutrophil recruiting cytokine Interleukin-17A (IL-17A) is a key component in vascular dysfunction and arterial hypertension. Moreover, IL-17A has a central role for the vascular infiltration of myeloid cells into the arterial wall in Angiotensin II-induced vascular inflammation. The intention of our study was to analyze the impact of T cell-derived IL-17A on hypertension, vascular function, and inflammation. Methods and Results Chronic IL-17A overexpression in T cells (CD4-IL-17Aind/+ mice) resulted in elevated reactive oxygen species in the peripheral blood and a significant vascular dysfunction compared to control mice. The vascular dysfunction seen in the CD4-IL-17Aind/+ mice was only accompanied by a modest and nonsignificant accumulation of inflammatory cells within the vessel wall. Therefore, infiltrating myeloid cells did not serve as an explanation of the vascular dysfunction seen in a chronic IL-17A-driven mouse model. In addition to vascular dysfunction, CD4-IL-17Aind/+ mice displayed vascular fibrosis with highly proliferative fibroblasts. This fibroblast proliferation was induced by exposure to IL-17A as confirmed by in vitro experiments with primary murine fibroblastic cells. We also found that the ·NO/cGMP pathway was downregulated in the vasculature of the CD4-IL-17Aind/+ mice, while levels of protein tyrosine kinase 2 (PYK2), an oxidative stress-triggered process associated with T cell activation, were upregulated in the perivascular fat tissue (PVAT). Conclusions Our data demonstrate that T cell-derived IL-17A elicits vascular dysfunction by mediating proliferation of fibroblasts and subsequent vascular fibrosis associated with PYK2 upregulation.
Collapse
|
203
|
Verma D, Kumar R, Pereira RS, Karantanou C, Zanetti C, Minciacchi VR, Fulzele K, Kunz K, Hoelper S, Zia-Chahabi S, Jabagi MJ, Emmerich J, Dray-Spira R, Kuhlee F, Hackmann K, Schroeck E, Wenzel P, Müller S, Filmann N, Fontenay M, Pajevic PD, Krause DS. Vitamin K antagonism impairs the bone marrow microenvironment and hematopoiesis. Blood 2019; 134:227-238. [PMID: 31003999 PMCID: PMC7022447 DOI: 10.1182/blood.2018874214] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 04/04/2019] [Indexed: 12/28/2022] Open
Abstract
Vitamin K antagonists (VKAs) have been used in 1% of the world's population for prophylaxis or treatment of thromboembolic events for 64 years. Impairment of osteoblast function and osteoporosis has been described in patients receiving VKAs. Given the involvement of cells of the bone marrow microenvironment (BMM), such as mesenchymal stem cells (MSCs) and macrophages, as well as other factors such as the extracellular matrix for the maintenance of normal hematopoietic stem cells (HSCs), we investigated a possible effect of VKAs on hematopoiesis via the BMM. Using various transplantation and in vitro assays, we show here that VKAs alter parameters of bone physiology and reduce functional HSCs 8-fold. We implicate impairment of the functional, secreted, vitamin K-dependent, γ-carboxylated form of periostin by macrophages and, to a lesser extent, MSCs of the BMM and integrin β3-AKT signaling in HSCs as at least partly causative of this effect, with VKAs not being directly toxic to HSCs. In patients, VKA use associates with modestly reduced leukocyte and monocyte counts, albeit within the normal reference range. VKAs decrease human HSC engraftment in immunosuppressed mice. Following published examples that alteration of the BMM can lead to hematological malignancies in mice, we describe, without providing a causal link, that the odds of VKA use are higher in patients with vs without a diagnosis of myelodysplastic syndrome (MDS). These results demonstrate that VKA treatment impairs HSC function via impairment of the BMM and the periostin/integrin β3 axis, possibly associating with increased MDS risk.
Collapse
Affiliation(s)
- Divij Verma
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Rahul Kumar
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Raquel S Pereira
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Christina Karantanou
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Costanza Zanetti
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Valentina R Minciacchi
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | | | - Kathrin Kunz
- Institute of Biochemistry II, Goethe University, Frankfurt am Main, Germany
| | - Soraya Hoelper
- Institute of Biochemistry II, Goethe University, Frankfurt am Main, Germany
| | - Sara Zia-Chahabi
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre-Cochin, Laboratory of Hematology, Institut Cochin, Université Paris Descartes, Paris, France
| | - Marie-Joëlle Jabagi
- Department of Epidemiology of Health Products, French National Agency for Medicines and Health Products Safety, Saint-Denis Cedex, France
| | - Joseph Emmerich
- Department of Epidemiology of Health Products, French National Agency for Medicines and Health Products Safety, Saint-Denis Cedex, France
- Vascular Medicine and Cardiology, University Paris Descartes and Hotel Dieu Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Rosemary Dray-Spira
- Department of Epidemiology of Health Products, French National Agency for Medicines and Health Products Safety, Saint-Denis Cedex, France
| | - Franziska Kuhlee
- Institut für Klinische Genetik, Medizinische Fakultät Carl Gustav Carus, Dresden, Germany
| | - Karl Hackmann
- Institut für Klinische Genetik, Medizinische Fakultät Carl Gustav Carus, Dresden, Germany
| | - Evelin Schroeck
- Institut für Klinische Genetik, Medizinische Fakultät Carl Gustav Carus, Dresden, Germany
| | - Philip Wenzel
- University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Stefan Müller
- Institute of Biochemistry II, Goethe University, Frankfurt am Main, Germany
| | - Natalie Filmann
- Institute of Biostatistics and Mathematical Modeling, Goethe University, Frankfurt, Germany
| | - Michaela Fontenay
- Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre-Cochin, Laboratory of Hematology, Institut Cochin, Université Paris Descartes, Paris, France
| | | | - Daniela S Krause
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
- German Cancer Consortium, Heidelberg, Germany
- German Cancer Research Center, Heidelberg, Germany; and
- Faculty of Medicine, Johann Wolfgang Goethe University, Frankfurt, Germany
| |
Collapse
|
204
|
McCarthy CG, Wenceslau CF, Webb RC, Joe B. Novel Contributors and Mechanisms of Cellular Senescence in Hypertension-Associated Premature Vascular Aging. Am J Hypertens 2019; 32:709-719. [PMID: 30982879 DOI: 10.1093/ajh/hpz052] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 03/25/2019] [Accepted: 04/10/2019] [Indexed: 01/08/2023] Open
Abstract
Hypertension has been described as a condition of premature vascular aging, relative to actual chronological age. In fact, many factors that contribute to the deterioration of vascular function as we age are accelerated in hypertension. Nonetheless, the precise mechanisms that underlie the aged phenotype of arteries from hypertensive patients and animals remain elusive. Cellular senescence is an age-related physiologic process in which cells undergo irreversible growth arrest. Although controlled senescence negatively regulates cell proliferation and promotes tissue regeneration, uncontrolled senescence can contribute to disease pathogenesis by presenting the senescence-associated secretory phenotype, in which molecules such as proinflammatory cytokines, matrix metalloproteases, and reactive oxygen species are released into tissue microenvironments. This review will address and critically evaluate the current literature on the role of cellular senescence in hypertension, with particular emphasis on cells types that mediate and modulate vascular function and structure.
Collapse
Affiliation(s)
- Cameron G McCarthy
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Camilla F Wenceslau
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - R Clinton Webb
- Department of Physiology, Augusta University, Augusta, Georgia, USA
| | - Bina Joe
- Center for Hypertension and Precision Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| |
Collapse
|
205
|
Nephropathy in Hypertensive Animals Is Linked to M2 Macrophages and Increased Expression of the YM1/Chi3l3 Protein. Mediators Inflamm 2019; 2019:9086758. [PMID: 31360120 PMCID: PMC6652056 DOI: 10.1155/2019/9086758] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 06/03/2019] [Accepted: 06/16/2019] [Indexed: 12/21/2022] Open
Abstract
Macrophages contribute to a continuous increase in blood pressure and kidney damage in hypertension, but their polarization status and the underlying mechanisms have not been clarified. This study revealed an important role for M2 macrophages and the YM1/Chi3l3 protein in hypertensive nephropathy in a mouse model of hypertension. Bone marrow cells were isolated from the femurs and tibia of male FVB/N (control) and transgenic hypertensive animals that overexpressed the rat form of angiotensinogen (TGM(rAOGEN)123, TGM123-FVB/N). The cells were treated with murine M-CSF and subsequently with LPS+IFN-γ to promote their polarization into M1 macrophages and IL-4+IL-13 to trigger the M2 phenotype. We examined the kidneys of TGM123-FVB/N animals to assess macrophage polarization and end-organ damage. mRNA expression was evaluated using real-time PCR, and protein levels were assessed through ELISA, CBA, Western blot, and immunofluorescence. Histology confirmed high levels of renal collagen. Cells stimulated with LPS+IFN-γ in vitro showed no significant difference in the expression of CD86, an M1 marker, compared to cells from the controls or the hypertensive mice. When stimulated with IL-4+IL-13, however, macrophages of the hypertensive group showed a significant increase in CD206 expression, an M2 marker. The M2/M1 ratio reached 288%. Our results indicate that when stimulated in vitro, macrophages from hypertensive mice are predisposed toward polarization to an M2 phenotype. These data support results from the kidneys where we found an increased infiltration of macrophages predominantly polarized to M2 associated with high levels of YM1/Chi3l3 (91,89%), suggesting that YM1/Chi3l3 may be a biomarker of hypertensive nephropathy.
Collapse
|
206
|
Yin X, Cao H, Wei Y, Li HH. Alteration of the IL-33-sST2 pathway in hypertensive patients and a mouse model. Hypertens Res 2019; 42:1664-1671. [PMID: 31235844 PMCID: PMC8075887 DOI: 10.1038/s41440-019-0291-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 04/28/2019] [Accepted: 05/25/2019] [Indexed: 01/11/2023]
Abstract
Inflammatory cells play an important role in the occurrence of hypertension. Recent studies have demonstrated that interleukin-33/suppression of tumorigenicity 2 (IL-33/ST2) signaling plays a critical role in the pathogenesis of several cardiovascular diseases. We aimed to evaluate the association of IL-33 and its receptor levels with the occurrence of hypertension in angiotensin II (Ang II)-infused mice using microarray analysis and validated our results in human specimens. Male wild-type mice were infused with Ang II (1500 ng/kg/min) for 1, 3 and 7 days. Patients with essential hypertension (EH) (n = 166) and healthy control subjects (n = 306) were enrolled. Levels of IL-33 and ST2 mRNAs in serum and peripheral blood mononuclear cells (PBMCs) were analyzed by Luminex assay or ELISA and qPCR analysis. We found that IL-33 expression was significantly increased in the aortas of mice receiving Ang II infusion compared with that of control mice. In contrast, the levels of IL-33 in serum and PBMCs were not significantly different between hypertensive patients and normal controls. However, the levels of soluble ST2 (sST2) in serum and PBMCs were markedly higher in hypertensive patients than in controls (P < 0.001 and P = 0.014, respectively). In addition, the ST2L level in PBMCs was also significantly decreased in hypertensive patients (P = 0.028). Further, logistic analysis showed that the odds ratios of having hypertension based on sST2 levels in serum and PBMCs were 9.714 and 2.244 (P = 0.013 and P = 0.024, respectively) compared with the control group. Above all, sST2 acted as a risk factor for the occurrence of hypertension and may be a promising novel predictive marker for EH.
Collapse
Affiliation(s)
- Xiaoyun Yin
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China
| | - Huajun Cao
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| | - Yingjie Wei
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China.
| | - Hui-Hua Li
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China. .,School of Public Health, Dalian Medical University, Dalian, 116044, People's Republic of China.
| |
Collapse
|
207
|
Abstract
Sodium intake is undoubtedly indispensable for normal body functions but can be detrimental when taken in excess of dietary requirements. The consequences of excessive salt intake are becoming increasingly clear as high salt consumption persists across the globe. Salt has long been suspected to promote the development of hypertension and cardiovascular diseases and is now also recognized as a potential modulator of inflammatory and autoimmune diseases through its direct and indirect effects on immune cells. The finding that, in addition to the kidneys, other organs such as the skin regulate sodium levels in the body prompted new hypotheses, including the concept that skin-resident macrophages might participate in tissue sodium regulation through their interactions with lymphatic vessels. Moreover, immune cells such as macrophages and different T cell subsets are found in sodium-rich interstitial microenvironments, where sodium levels modulate their function. Alterations to the intestinal bacterial community induced by excess dietary salt represent another relevant axis whereby salt indirectly modulates immune cell function. Depending on the inflammatory context, sodium might either contribute to protective immunity (for example, by enhancing host responses against cutaneous pathogens) or it might contribute to immune dysregulation and promote the development of cardiovascular and autoimmune diseases.
Collapse
|
208
|
Vascular Inflammation and Oxidative Stress: Major Triggers for Cardiovascular Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7092151. [PMID: 31341533 PMCID: PMC6612399 DOI: 10.1155/2019/7092151] [Citation(s) in RCA: 419] [Impact Index Per Article: 69.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/20/2019] [Indexed: 02/08/2023]
Abstract
Cardiovascular disease is a leading cause of death and reduced quality of life, proven by the latest data of the Global Burden of Disease Study, and is only gaining in prevalence worldwide. Clinical trials have identified chronic inflammatory disorders as cardiovascular risks, and recent research has revealed a contribution by various inflammatory cells to vascular oxidative stress. Atherosclerosis and cardiovascular disease are closely associated with inflammation, probably due to the close interaction of inflammation with oxidative stress. Classical therapies for inflammatory disorders have demonstrated protective effects in various models of cardiovascular disease; especially established drugs with pleiotropic immunomodulatory properties have proven beneficial cardiovascular effects; normalization of oxidative stress seems to be a common feature of these therapies. The close link between inflammation and redox balance was also supported by reports on aggravated inflammatory phenotype in the absence of antioxidant defense proteins (e.g., superoxide dismutases, heme oxygenase-1, and glutathione peroxidases) or overexpression of reactive oxygen species producing enzymes (e.g., NADPH oxidases). The value of immunomodulation for the treatment of cardiovascular disease was recently supported by large-scale clinical trials demonstrating reduced cardiovascular mortality in patients with established atherosclerotic disease when treated by highly specific anti-inflammatory therapies (e.g., using monoclonal antibodies against cytokines). Modern antidiabetic cardiovascular drugs (e.g., SGLT2 inhibitors, DPP-4 inhibitors, and GLP-1 analogs) seem to share these immunomodulatory properties and display potent antioxidant effects, all of which may explain their successful lowering of cardiovascular risk.
Collapse
|
209
|
Wenzel P. Monocytes as immune targets in arterial hypertension. Br J Pharmacol 2019; 176:1966-1977. [PMID: 29885051 PMCID: PMC6534790 DOI: 10.1111/bph.14389] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/25/2018] [Accepted: 05/30/2018] [Indexed: 12/11/2022] Open
Abstract
The role of myelomonocytic cells appears to be critical for the initiation, progression and manifestation of arterial hypertension. Monocytes can induce vascular inflammation as well as tissue remodelling and (mal)adaptation by secreting chemokines and cytokines, producing ROS, expressing coagulation factors and transforming into macrophages. A multitude of adhesion molecules promote the infiltration and accumulation of monocytes into the kidney, heart, brain and vasculature in hypertension. All these facets offer the possibility to pharmacologically target monocytes and may represent novel therapeutic ways to treat hypertension, attenuate hypertension-associated end organ damage or prevent the development or worsening of high blood pressure. LINKED ARTICLES: This article is part of a themed section on Immune Targets in Hypertension. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.12/issuetoc.
Collapse
Affiliation(s)
- Philip Wenzel
- Center for Cardiology ‐ Cardiology IUniversity Medical Center MainzMainzGermany
- Center for Thrombosis and HemostasisUniversity Medical Center MainzMainzGermany
- German Center for Cardiovascular Research (DZHK), partner site Rhine‐Main
| |
Collapse
|
210
|
Carnagarin R, Matthews V, Zaldivia MTK, Peter K, Schlaich MP. The bidirectional interaction between the sympathetic nervous system and immune mechanisms in the pathogenesis of hypertension. Br J Pharmacol 2019; 176:1839-1852. [PMID: 30129037 PMCID: PMC6534787 DOI: 10.1111/bph.14481] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 07/26/2018] [Accepted: 08/05/2018] [Indexed: 12/14/2022] Open
Abstract
Over the last few years, evidence has accumulated to suggest that hypertension is, at least in part, an immune-mediated inflammatory disorder. Many links between immunity and hypertension have been established and provide a complex framework of mechanistic interactions contributing to the rise in BP. These include immune-mediated inflammatory processes affecting regulatory brain nuclei and interactions with other mediators of cardiovascular regulation such as the sympathetic nervous system. Sympathoexcitation differentially regulates T-cells based upon activation status of the immune cell as well as the resident organ. Exogenous and endogenous triggers activate signalling pathways in innate and adaptive immune cells resulting in pro-inflammatory cytokine production and activation of T-lymphocytes in the cardiovascular and renal regions, now considered major factors in the development of essential hypertension. The inflammatory cascade is sustained and exacerbated by the immune flow via the brain-bone marrow-spleen-gastrointestinal axis and thereby further aggravating immune-mediated pathways resulting in a vicious cycle of established hypertension and target organ damage. This review summarizes the evidence and recent advances in linking immune-mediated inflammation, sympathetic activation and their bidirectional interactions with the development of hypertension. LINKED ARTICLES: This article is part of a themed section on Immune Targets in Hypertension. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.12/issuetoc.
Collapse
Affiliation(s)
- Revathy Carnagarin
- Dobney Hypertension Centre, School of Medicine – Royal Perth Hospital UnitThe University of Western AustraliaPerthWAAustralia
| | - Vance Matthews
- Dobney Hypertension Centre, School of Medicine – Royal Perth Hospital UnitThe University of Western AustraliaPerthWAAustralia
| | - Maria T K Zaldivia
- Atherothrombosis and Vascular BiologyBaker Heart and Diabetes InstituteMelbourneVicAustralia
- Department of MedicineMonash University, Royal Perth HospitalPerthWAAustralia
| | - Karlheinz Peter
- Atherothrombosis and Vascular BiologyBaker Heart and Diabetes InstituteMelbourneVicAustralia
- Department of MedicineMonash University, Royal Perth HospitalPerthWAAustralia
| | - Markus P Schlaich
- Dobney Hypertension Centre, School of Medicine – Royal Perth Hospital UnitThe University of Western AustraliaPerthWAAustralia
- Department of CardiologyRoyal Perth HospitalPerthWAAustralia
- Department of NephrologyRoyal Perth HospitalPerthWAAustralia
| |
Collapse
|
211
|
Chuong P, Wysoczynski M, Hellmann J. Do Changes in Innate Immunity Underlie the Cardiovascular Benefits of Exercise? Front Cardiovasc Med 2019; 6:70. [PMID: 31192231 PMCID: PMC6549037 DOI: 10.3389/fcvm.2019.00070] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/10/2019] [Indexed: 12/29/2022] Open
Affiliation(s)
- Phillip Chuong
- Division of Cardiovascular Medicine, Diabetes and Obesity Center, University of Louisville School of Medicine, Louisville, KY, United States
| | - Marcin Wysoczynski
- Division of Cardiovascular Medicine, Diabetes and Obesity Center, University of Louisville School of Medicine, Louisville, KY, United States
| | - Jason Hellmann
- Division of Cardiovascular Medicine, Diabetes and Obesity Center, University of Louisville School of Medicine, Louisville, KY, United States
| |
Collapse
|
212
|
Steven S, Dib M, Hausding M, Kashani F, Oelze M, Kröller-Schön S, Hanf A, Daub S, Roohani S, Gramlich Y, Lutgens E, Schulz E, Becker C, Lackner KJ, Kleinert H, Knosalla C, Niesler B, Wild PS, Münzel T, Daiber A. CD40L controls obesity-associated vascular inflammation, oxidative stress, and endothelial dysfunction in high fat diet-treated and db/db mice. Cardiovasc Res 2019; 114:312-323. [PMID: 29036612 DOI: 10.1093/cvr/cvx197] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 09/25/2017] [Indexed: 01/29/2023] Open
Abstract
Aims CD40 ligand (CD40L) signaling controls vascular oxidative stress and related dysfunction in angiotensin-II-induced arterial hypertension by regulating vascular immune cell recruitment and platelet activation. Here we investigated the role of CD40L in experimental hyperlipidemia. Methods and results Male wild type and CD40L-/- mice (C57BL/6 background) were subjected to high fat diet for sixteen weeks. Weight, cholesterol, HDL, and LDL levels, endothelial function (isometric tension recording), oxidative stress (NADPH oxidase expression, dihydroethidium fluorescence) and inflammatory parameters (inducible nitric oxide synthase, interleukin-6 expression) were assessed. CD40L expression, weight, leptin and lipids were increased, and endothelial dysfunction, oxidative stress and inflammation were more pronounced in wild type mice on a high fat diet, all of which was almost normalized by CD40L deficiency. Similar results were obtained in diabetic db/db mice with CD40/TRAF6 inhibitor (6877002) therapy. In a small human study higher serum sCD40L levels and an inflammatory phenotype were detected in the blood and Aorta ascendens of obese patients (body mass index > 35) that underwent by-pass surgery. Conclusion CD40L controls obesity-associated vascular inflammation, oxidative stress and endothelial dysfunction in mice and potentially humans. Thus, CD40L represents a therapeutic target in lipid metabolic disorders which is a leading cause in cardiovascular disease.
Collapse
Affiliation(s)
- Sebastian Steven
- Center for Cardiology 1, Molecular Cardiology; Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.,Center for Thrombosis and Hemostasis (CTH), Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Mobin Dib
- Center for Cardiology 1, Molecular Cardiology; Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Michael Hausding
- Center for Cardiology 1, Molecular Cardiology; Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Fatemeh Kashani
- Center for Cardiology 1, Molecular Cardiology; Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Matthias Oelze
- Center for Cardiology 1, Molecular Cardiology; Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Swenja Kröller-Schön
- Center for Cardiology 1, Molecular Cardiology; Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Alina Hanf
- Center for Cardiology 1, Molecular Cardiology; Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Steffen Daub
- Center for Cardiology 1, Molecular Cardiology; Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Siyer Roohani
- Center for Cardiology 1, Molecular Cardiology; Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Yves Gramlich
- Center for Cardiology 1, Molecular Cardiology; Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Esther Lutgens
- Department of Medical Biochemistry, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands.,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian's University (LMU), Munich, Germany
| | - Eberhard Schulz
- Center for Cardiology 1, Molecular Cardiology; Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Christian Becker
- Department of Dermatology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Karl J Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Hartmut Kleinert
- Department of Pharmacology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Christoph Knosalla
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Beate Niesler
- nCounter Core Facility, Institute of Human Genetics, University of Heidelberg, Heidelberg, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
| | - Philipp S Wild
- Center for Cardiology 1, Molecular Cardiology; Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.,Center for Thrombosis and Hemostasis (CTH), Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Thomas Münzel
- Center for Cardiology 1, Molecular Cardiology; Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.,Center for Thrombosis and Hemostasis (CTH), Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Andreas Daiber
- Center for Cardiology 1, Molecular Cardiology; Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.,Center for Thrombosis and Hemostasis (CTH), Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| |
Collapse
|
213
|
Ganguly A, Sharma K, Majumder K. Food-derived bioactive peptides and their role in ameliorating hypertension and associated cardiovascular diseases. ADVANCES IN FOOD AND NUTRITION RESEARCH 2019; 89:165-207. [PMID: 31351525 DOI: 10.1016/bs.afnr.2019.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Non-communicable diseases including cardiovascular diseases (CVDs) and associated metabolic disorders are responsible for nearly 40 million deaths globally per year. Hypertension or high blood pressure (BP) is one of the primary reasons for the development of CVDs. A healthy nutritional strategy complementing with physical activity can substantially reduce high BP and prevent the occurrence of CVD-associated morbidity and mortality. Bioactive peptides currently are the next wave of the promising bench to clinic options for potential targeting chronic and acute health issues including hypertension. Peptides demonstrating anti-inflammatory, anti-oxidant, and angiotensin-converting enzyme-I inhibitory activity are widely studied for the amelioration of hypertension and associated CVDs. Isolating these potent bioactive peptides from different food sources is a promising endeavor toward nutraceutical based dietary management and prevention of hypertension. Understanding the pathophysiology of hypertension and the action mechanisms of the bioactive peptides would complement in designing and characterizing more potent peptides and suitable comprehensive dietary plans for the prevention of hypertension and associated CVDs.
Collapse
Affiliation(s)
- Advaita Ganguly
- Comprehensive Tissue Centre, UAH Transplant Services, Alberta Health Services, Edmonton, AB, Canada
| | - Kumakshi Sharma
- Health, Safety and Environment Branch, National Research Council Canada, Edmonton, AB, Canada
| | - Kaustav Majumder
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, United States.
| |
Collapse
|
214
|
Yu H, Kalogeris T, Korthuis RJ. Reactive species-induced microvascular dysfunction in ischemia/reperfusion. Free Radic Biol Med 2019; 135:182-197. [PMID: 30849489 PMCID: PMC6503659 DOI: 10.1016/j.freeradbiomed.2019.02.031] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 02/26/2019] [Accepted: 02/26/2019] [Indexed: 12/13/2022]
Abstract
Vascular endothelial cells line the inner surface of the entire cardiovascular system as a single layer and are involved in an impressive array of functions, ranging from the regulation of vascular tone in resistance arteries and arterioles, modulation of microvascular barrier function in capillaries and postcapillary venules, and control of proinflammatory and prothrombotic processes, which occur in all segments of the vascular tree but can be especially prominent in postcapillary venules. When tissues are subjected to ischemia/reperfusion (I/R), the endothelium of resistance arteries and arterioles, capillaries, and postcapillary venules become dysfunctional, resulting in impaired endothelium-dependent vasodilator and enhanced endothelium-dependent vasoconstrictor responses along with increased vulnerability to thrombus formation, enhanced fluid filtration and protein extravasation, and increased blood-to-interstitium trafficking of leukocytes in these functionally distinct segments of the microcirculation. The number of capillaries open to flow upon reperfusion also declines as a result of I/R, which impairs nutritive perfusion. All of these pathologic microvascular events involve the formation of reactive species (RS) derived from molecular oxygen and/or nitric oxide. In addition to these effects, I/R-induced RS activate NLRP3 inflammasomes, alter connexin/pannexin signaling, provoke mitochondrial fission, and cause release of microvesicles in endothelial cells, resulting in deranged function in arterioles, capillaries, and venules. It is now apparent that this microvascular dysfunction is an important determinant of the severity of injury sustained by parenchymal cells in ischemic tissues, as well as being predictive of clinical outcome after reperfusion therapy. On the other hand, RS production at signaling levels promotes ischemic angiogenesis, mediates flow-induced dilation in patients with coronary artery disease, and instigates the activation of cell survival programs by conditioning stimuli that render tissues resistant to the deleterious effects of prolonged I/R. These topics will be reviewed in this article.
Collapse
Affiliation(s)
- Hong Yu
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, 1 Hospital Drive, Columbia, MO 65212, USA
| | - Ted Kalogeris
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, 1 Hospital Drive, Columbia, MO 65212, USA
| | - Ronald J Korthuis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, 1 Hospital Drive, Columbia, MO 65212, USA; Dalton Cardiovascular Research Center, University of Missouri, 134 Research Park Drive, Columbia, MO 65211, USA.
| |
Collapse
|
215
|
Taylor EB, Wolf VL, Dent E, Ryan MJ. Mechanisms of hypertension in autoimmune rheumatic diseases. Br J Pharmacol 2019; 176:1897-1913. [PMID: 30714094 PMCID: PMC6534791 DOI: 10.1111/bph.14604] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 12/12/2018] [Accepted: 01/07/2019] [Indexed: 02/06/2023] Open
Abstract
Patients with autoimmune rheumatic diseases including rheumatoid arthritis and systemic lupus erythematosus have an increased prevalence of hypertension. There is now a large body of evidence showing that the immune system is a key mediator in both human primary hypertension and experimental models. Many of the proposed immunological mechanisms leading to primary hypertension are paralleled in autoimmune rheumatic disorders. Therefore, examining the link between autoimmunity and hypertension can be informative for understanding primary hypertension. This review examines the prevalent hypertension, the immune mediators that contribute to the prevalent hypertension and their impact on renal function and how the risk of hypertension is potentially influenced by common hormonal changes that are associated with autoimmune rheumatic diseases. Linked Articles This article is part of a themed section on Immune Targets in Hypertension. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.12/issuetoc
Collapse
Affiliation(s)
- Erin B Taylor
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Victoria L Wolf
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Elena Dent
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Michael J Ryan
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi, USA.,G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
216
|
Mikolajczyk TP, Nosalski R, Skiba DS, Koziol J, Mazur M, Justo-Junior AS, Kowalczyk P, Kusmierczyk Z, Schramm-Luc A, Luc K, Maffia P, Graham D, Kiss AK, Naruszewicz M, Guzik TJ. 1,2,3,4,6-Penta-O-galloyl-β-d-glucose modulates perivascular inflammation and prevents vascular dysfunction in angiotensin II-induced hypertension. Br J Pharmacol 2019; 176:1951-1965. [PMID: 30658013 PMCID: PMC6534792 DOI: 10.1111/bph.14583] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 10/26/2018] [Accepted: 12/02/2018] [Indexed: 12/16/2022] Open
Abstract
Background and Purpose Hypertension is a multifactorial disease, manifested by vascular dysfunction, increased superoxide production, and perivascular inflammation. In this study, we have hypothesized that 1,2,3,4,6‐penta‐O‐galloyl‐β‐d‐glucose (PGG) would inhibit vascular inflammation and protect from vascular dysfunction in an experimental model of hypertension. Experimental Approach PGG was administered to mice every 2 days at a dose of 10 mg·kg−1 i.p during 14 days of Ang II infusion. It was used at a final concentration of 20 μM for in vitro studies in cultured cells. Key Results Ang II administration increased leukocyte and T‐cell content in perivascular adipose tissue (pVAT), and administration of PGG significantly decreased total leukocyte and T‐cell infiltration in pVAT. This effect was observed in relation to all T‐cell subsets. PGG also decreased the content of T‐cells bearing CD25, CCR5, and CD44 receptors and the expression of both monocyte chemoattractant protein 1 (CCL2) in aorta and RANTES (CCL5) in pVAT. PGG administration decreased the content of TNF+ and IFN‐γ+ CD8 T‐cells and IL‐17A+ CD4+ and CD3+CD4−CD8− cells. Importantly, these effects of PGG were associated with improved vascular function and decreased ROS production in the aortas of Ang II‐infused animals independently of the BP increase. Mechanistically, PGG (20 μM) directly inhibited CD25 and CCR5 expression in cultured T‐cells. It also decreased the content of IFN‐γ+ CD8+ and CD3+CD4−CD8− cells and IL‐17A+ CD3+CD4−CD8− cells. Conclusion and Implication PGG may constitute an interesting immunomodulating strategy in the regulation of vascular dysfunction and hypertension. Linked Articles This article is part of a themed section on Immune Targets in Hypertension. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.12/issuetoc
Collapse
Affiliation(s)
- Tomasz P Mikolajczyk
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK.,Department of Internal and Agricultural Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Ryszard Nosalski
- Department of Internal and Agricultural Medicine, Jagiellonian University Medical College, Krakow, Poland.,Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Dominik S Skiba
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Joanna Koziol
- Department of Internal and Agricultural Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Magdalena Mazur
- Department of Internal and Agricultural Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Amauri S Justo-Junior
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Paulina Kowalczyk
- Department of Pharmacognosy and Molecular Basis of Phytotherapy, Medical University of Warsaw, Warsaw, Poland
| | - Zofia Kusmierczyk
- Department of Pharmacognosy and Molecular Basis of Phytotherapy, Medical University of Warsaw, Warsaw, Poland
| | - Agata Schramm-Luc
- Department of Internal and Agricultural Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Kevin Luc
- Department of Internal and Agricultural Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Pasquale Maffia
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK.,Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK.,Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Delyth Graham
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Anna K Kiss
- Department of Pharmacognosy and Molecular Basis of Phytotherapy, Medical University of Warsaw, Warsaw, Poland
| | - Marek Naruszewicz
- Department of Pharmacognosy and Molecular Basis of Phytotherapy, Medical University of Warsaw, Warsaw, Poland
| | - Tomasz J Guzik
- Department of Internal and Agricultural Medicine, Jagiellonian University Medical College, Krakow, Poland.,Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
217
|
Schloss MJ, Nahrendorf M. Some Macrophages Are Softies. Immunity 2019; 49:199-201. [PMID: 30134196 DOI: 10.1016/j.immuni.2018.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Adequate maintenance of the arterial extracellular matrix is essential for steady-state vascular functions. In this issue of Immunity, Lim et al. (2018) describe that aortic LYVE-1+ macrophages regulate steady-state arterial matrix content by interacting with smooth muscle cells and collagen.
Collapse
Affiliation(s)
- Maximilian J Schloss
- Center for Systems Biology and Department of Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Matthias Nahrendorf
- Center for Systems Biology and Department of Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
218
|
Barrows IR, Ramezani A, Raj DS. Inflammation, Immunity, and Oxidative Stress in Hypertension-Partners in Crime? Adv Chronic Kidney Dis 2019; 26:122-130. [PMID: 31023446 DOI: 10.1053/j.ackd.2019.03.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 02/26/2019] [Accepted: 03/01/2019] [Indexed: 02/07/2023]
Abstract
Hypertension is considered as the most common risk factor for cardiovascular disease. Inflammatory processes link hypertension and cardiovascular disease, and participate in their pathophysiology. In recent years, there has been an increase in research focused on unraveling the role of inflammation and immune activation in development and maintenance of hypertension. Although inflammation is known to be associated with hypertension, whether inflammation is a cause or effect of hypertension remains to be elucidated. This review describes the recent studies that link inflammation and hypertension and demonstrate the involvement of oxidative stress and endothelial dysfunction-two of the key processes in the development of hypertension. Etiology of hypertension, including novel immune cell subtypes, cytokines, toll-like receptors, inflammasomes, and gut microbiome, found to be associated with inflammation and hypertension are summarized and discussed. Most recent findings in this field are presented with special emphasis on potential of anti-inflammatory drugs and statins for treatment of hypertension.
Collapse
|
219
|
Kondo M, Imanishi M, Fukushima K, Ikuto R, Murai Y, Horinouchi Y, Izawa-Ishizawa Y, Goda M, Zamami Y, Takechi K, Chuma M, Ikeda Y, Fujino H, Tsuchiya K, Ishizawa K. Xanthine Oxidase Inhibition by Febuxostat in Macrophages Suppresses Angiotensin II-Induced Aortic Fibrosis. Am J Hypertens 2019; 32:249-256. [PMID: 30351343 PMCID: PMC7110082 DOI: 10.1093/ajh/hpy157] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/19/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Several reports from basic researches and clinical studies have suggested that xanthine oxidase (XO) inhibitors have suppressive effects on cardiovascular diseases. However, the roles of a XO inhibitor, febuxostat (FEB), in the pathogenesis of vascular remodeling and hypertension independent of the serum uric acid level remain unclear. METHODS To induce vascular remodeling in mice, angiotensin II (Ang II) was infused for 2 weeks with a subcutaneously implanted osmotic minipump. FEB was administered every day during Ang II infusion. Aortic fibrosis was assessed by elastica van Gieson staining. Mouse macrophage RAW264.7 cells (RAW) and mouse embryonic fibroblasts were used for in vitro studies. RESULTS FEB suppressed Ang II-induced blood pressure elevation and aortic fibrosis. Immunostaining showed that Ang II-induced macrophage infiltration in the aorta tended to be suppressed by FEB, and XO was mainly colocalized in macrophages, not in fibroblasts. Transforming growth factor-β1 (TGF-β1) mRNA expression was induced in the aorta in the Ang II alone group, but not in the Ang II + FEB group. Ang II induced α-smooth muscle actin-positive fibroblasts in the aortic wall, but FEB suppressed them. XO expression and activity were induced by Ang II stimulation alone but not by Ang II + FEB in RAW. FEB suppressed Ang II-induced TGF-β1 mRNA expression in RAW. CONCLUSIONS Our results suggested that FEB ameliorates Ang II-induced aortic fibrosis via suppressing macrophage-derived TGF-β1 expression.
Collapse
Affiliation(s)
- Masateru Kondo
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
- Department of Clinical Pharmacology and Therapeutics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Masaki Imanishi
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Keijo Fukushima
- Department of Pharmacology for Life Sciences, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Raiki Ikuto
- Department of Clinical Pharmacology and Therapeutics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yoichi Murai
- Department of Clinical Pharmacology and Therapeutics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yuya Horinouchi
- Department of Pharmacology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yuki Izawa-Ishizawa
- Department of Pharmacology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Mitsuhiro Goda
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Yoshito Zamami
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
- Department of Clinical Pharmacology and Therapeutics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Kenshi Takechi
- Clinical Trial Center for Developmental Therapeutics, Tokushima University Hospital, Tokushima, Japan
| | - Masayuki Chuma
- Clinical Trial Center for Developmental Therapeutics, Tokushima University Hospital, Tokushima, Japan
| | - Yasumasa Ikeda
- Department of Pharmacology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Hiromichi Fujino
- Department of Pharmacology for Life Sciences, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Koichiro Tsuchiya
- Department of Medical Pharmacology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Keisuke Ishizawa
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
- Department of Clinical Pharmacology and Therapeutics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| |
Collapse
|
220
|
Cicalese S, Scalia R, Eguchi S. Xanthine Oxidase Inhibition as a Potential Treatment for Aortic Stiffness in Hypertension. Am J Hypertens 2019; 32:234-236. [PMID: 30561498 DOI: 10.1093/ajh/hpy197] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 12/14/2018] [Indexed: 12/15/2022] Open
Affiliation(s)
- Stephanie Cicalese
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University
| |
Collapse
|
221
|
|
222
|
Kröller-Schön S, Jansen T, Tran TLP, Kvandová M, Kalinovic S, Oelze M, Keaney JF, Foretz M, Viollet B, Daiber A, Kossmann S, Lagrange J, Frenis K, Wenzel P, Münzel T, Schulz E. Endothelial α1AMPK modulates angiotensin II-mediated vascular inflammation and dysfunction. Basic Res Cardiol 2019; 114:8. [DOI: 10.1007/s00395-019-0717-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 12/18/2018] [Accepted: 01/09/2019] [Indexed: 12/11/2022]
|
223
|
Daiber A, Xia N, Steven S, Oelze M, Hanf A, Kröller-Schön S, Münzel T, Li H. New Therapeutic Implications of Endothelial Nitric Oxide Synthase (eNOS) Function/Dysfunction in Cardiovascular Disease. Int J Mol Sci 2019; 20:ijms20010187. [PMID: 30621010 PMCID: PMC6337296 DOI: 10.3390/ijms20010187] [Citation(s) in RCA: 186] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 12/27/2018] [Accepted: 12/28/2018] [Indexed: 02/07/2023] Open
Abstract
The Global Burden of Disease Study identified cardiovascular risk factors as leading causes of global deaths and life years lost. Endothelial dysfunction represents a pathomechanism that is associated with most of these risk factors and stressors, and represents an early (subclinical) marker/predictor of atherosclerosis. Oxidative stress is a trigger of endothelial dysfunction and it is a hall-mark of cardiovascular diseases and of the risk factors/stressors that are responsible for their initiation. Endothelial function is largely based on endothelial nitric oxide synthase (eNOS) function and activity. Likewise, oxidative stress can lead to the loss of eNOS activity or even “uncoupling” of the enzyme by adverse regulation of well-defined “redox switches” in eNOS itself or up-/down-stream signaling molecules. Of note, not only eNOS function and activity in the endothelium are essential for vascular integrity and homeostasis, but also eNOS in perivascular adipose tissue plays an important role for these processes. Accordingly, eNOS protein represents an attractive therapeutic target that, so far, was not pharmacologically exploited. With our present work, we want to provide an overview on recent advances and future therapeutic strategies that could be used to target eNOS activity and function in cardiovascular (and other) diseases, including life style changes and epigenetic modulations. We highlight the redox-regulatory mechanisms in eNOS function and up- and down-stream signaling pathways (e.g., tetrahydrobiopterin metabolism and soluble guanylyl cyclase/cGMP pathway) and their potential pharmacological exploitation.
Collapse
Affiliation(s)
- Andreas Daiber
- Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany.
| | - Ning Xia
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
| | - Sebastian Steven
- Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
| | - Matthias Oelze
- Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
| | - Alina Hanf
- Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
| | - Swenja Kröller-Schön
- Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
| | - Thomas Münzel
- Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany.
| | - Huige Li
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
| |
Collapse
|
224
|
Abstract
Research during the last decade has generated numerous insights on the presence, phenotype, and function of myeloid cells in cardiovascular organs. Newer tools with improved detection sensitivities revealed sizable populations of tissue-resident macrophages in all major healthy tissues. The heart and blood vessels contain robust numbers of these cells; for instance, 8% of noncardiomyocytes in the heart are macrophages. This number and the cell's phenotype change dramatically in disease conditions. While steady-state macrophages are mostly monocyte independent, macrophages residing in the inflamed vascular wall and the diseased heart derive from hematopoietic organs. In this review, we will highlight signals that regulate macrophage supply and function, imaging applications that can detect changes in cell numbers and phenotype, and opportunities to modulate cardiovascular inflammation by targeting macrophage biology. We strive to provide a systems-wide picture, i.e., to focus not only on cardiovascular organs but also on tissues involved in regulating cell supply and phenotype, as well as comorbidities that promote cardiovascular disease. We will summarize current developments at the intersection of immunology, detection technology, and cardiovascular health.
Collapse
Affiliation(s)
- Vanessa Frodermann
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School , Boston, Massachusetts ; and Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School , Boston, Massachusetts
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School , Boston, Massachusetts ; and Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School , Boston, Massachusetts
| |
Collapse
|
225
|
Wen Y, Crowley SD. Renal Effects of Cytokines in Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1165:443-454. [PMID: 31399978 DOI: 10.1007/978-981-13-8871-2_21] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Preclinical studies point to a key role for immune cells in hypertension via augmenting renal injury and/or hypertensive responses. Blood pressure elevation in rheumatologic patients is attenuated by anti-inflammatory therapies. Both the innate and adaptive immune systems contribute to the pathogenesis of hypertension by modulating renal sodium balance, blood flow, and functions of the vasculature and epithelial cells in the kidney. Monocytes/macrophages and T lymphocytes are pivotal mediators of hypertensive responses, while dendritic cells and B lymphocytes can regulate blood pressure indirectly by promoting T lymphocytes activation. Pro-inflammatory cytokines, such as tumor necrosis factor-α (TNF), interleukin-1 (IL-1), interleukin-17 (IL-17), and interferon-γ (IFN), amplify blood pressure elevation and/or renal injury. By contrast, interleukin-10 (IL-10) protects against renal and vascular function when produced by T helper 2 cells (Th2) and regulatory T cells (Treg). Thus, understanding the renal effects of cytokines in hypertension will provide targets for precise immunotherapies to inhibit targeted organ damage while preserving necessary immunity.
Collapse
Affiliation(s)
- Yi Wen
- Division of Nephrology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China.,Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, NC, USA
| | - Steven D Crowley
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, NC, USA.
| |
Collapse
|
226
|
Phillips EH, Lorch AH, Durkes AC, Goergen CJ. Early pathological characterization of murine dissecting abdominal aortic aneurysms. APL Bioeng 2018; 2:046106. [PMID: 31069328 PMCID: PMC6481730 DOI: 10.1063/1.5053708] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 12/10/2018] [Indexed: 12/11/2022] Open
Abstract
We report here on the early pathology of a well-established murine model of dissecting abdominal aortic aneurysms (AAAs). Continuous infusion of angiotensin II (AngII) into apolipoprotein E-deficient mice induces the formation of aortic dissection and expansion at some point after implantation of miniosmotic pumps containing AngII. While this model has been studied extensively at a chronic stage, we investigated the early pathology of dissecting AAA formation at multiple scales. Using high-frequency ultrasound, we screened 12-week-old male mice daily for initial formation of these aneurysmal lesions between days 3 and 10 post-implantation. We euthanized animals on the day of diagnosis of a dissecting AAA or at day 10 if no aneurysmal lesion developed. Aortic expansion and reduced vessel wall strain occurred in animals regardless of whether a dissecting AAA developed by day 10. The aortas of mice that did not develop dissecting AAAs showed intermediate changes in morphology and biomechanical properties. RNA sequencing and gene expression analysis revealed multiple proinflammatory and matrix remodeling genes to be upregulated in the suprarenal aorta of AngII-infused mice as compared to saline-infused controls. Histology and immunohistochemistry confirmed that extracellular matrix remodeling and inflammatory cell infiltration, notably neutrophils and macrophages, occurred in AngII-infused mice with and without dissecting AAAs but not saline-infused controls. Understanding early disease processes is a critical step forward in translating experimental results in cardiovascular disease research. This work advances our understanding of this well-established murine model with applications for improving early diagnosis and therapy of acute aortic syndrome in humans.
Collapse
Affiliation(s)
- Evan H Phillips
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| | - Adam H Lorch
- Department of Biology, Purdue University, West Lafayette, Indiana 47907, USA
| | - Abigail C Durkes
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana 47907, USA
| | | |
Collapse
|
227
|
Oxidative Stress in Cardiac Tissue of Patients Undergoing Coronary Artery Bypass Graft Surgery: The Effects of Overweight and Obesity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6598326. [PMID: 30647815 PMCID: PMC6311809 DOI: 10.1155/2018/6598326] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 10/14/2018] [Indexed: 12/11/2022]
Abstract
Background Obesity is one of the major cardiovascular risk factors and is associated with oxidative stress and myocardial dysfunction. We hypothesized that obesity affects cardiac function and morbidity by causing alterations in enzymatic redox patterns. Methods Sixty-one patients undergoing coronary artery bypass grafting (CABG) were included in the study. Excessive right atrial myocardial tissue emerging from the operative connection to the extracorporeal circulation was harvested. Patients were assigned to control (n = 19, body mass index (BMI): <25 kg/m2), overweight (n = 25, 25 kg/m2 < BMI < 30 kg/m2), or obese (n = 17, BMI: >30 kg/m2) groups. Oxidative enzyme systems were studied directly in the cardiac muscles of patients undergoing CABG who were grouped according to BMI. Molecular biological methods and high-performance liquid chromatography were used to detect the expression and activity of oxidative enzymes and the formation of reactive oxygen species (ROS). Results We found increased levels of ROS and increased expression of ROS-producing enzymes (i.e., p47phox, xanthine oxidase) and decreased antioxidant defense mechanisms (mitochondrial aldehyde dehydrogenase, heme oxygenase-1, and eNOS) in line with elevated inflammatory markers (vascular cell adhesion molecule-1) in the right atrial myocardial tissue and by trend also in serum (sVCAM-1 and CCL5/RANTES). Conclusion Increasing BMI in patients undergoing CABG is related to altered myocardial redox patterns, which indicates increased oxidative stress with inadequate antioxidant compensation. These changes suggest that the myocardium of obese patients suffering from coronary artery disease is more susceptible to cardiomyopathy and possible damage by ischemia and reperfusion, for example, during cardiac surgery.
Collapse
|
228
|
Forrester SJ, Booz GW, Sigmund CD, Coffman TM, Kawai T, Rizzo V, Scalia R, Eguchi S. Angiotensin II Signal Transduction: An Update on Mechanisms of Physiology and Pathophysiology. Physiol Rev 2018; 98:1627-1738. [PMID: 29873596 DOI: 10.1152/physrev.00038.2017] [Citation(s) in RCA: 723] [Impact Index Per Article: 103.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The renin-angiotensin-aldosterone system plays crucial roles in cardiovascular physiology and pathophysiology. However, many of the signaling mechanisms have been unclear. The angiotensin II (ANG II) type 1 receptor (AT1R) is believed to mediate most functions of ANG II in the system. AT1R utilizes various signal transduction cascades causing hypertension, cardiovascular remodeling, and end organ damage. Moreover, functional cross-talk between AT1R signaling pathways and other signaling pathways have been recognized. Accumulating evidence reveals the complexity of ANG II signal transduction in pathophysiology of the vasculature, heart, kidney, and brain, as well as several pathophysiological features, including inflammation, metabolic dysfunction, and aging. In this review, we provide a comprehensive update of the ANG II receptor signaling events and their functional significances for potential translation into therapeutic strategies. AT1R remains central to the system in mediating physiological and pathophysiological functions of ANG II, and participation of specific signaling pathways becomes much clearer. There are still certain limitations and many controversies, and several noteworthy new concepts require further support. However, it is expected that rigorous translational research of the ANG II signaling pathways including those in large animals and humans will contribute to establishing effective new therapies against various diseases.
Collapse
Affiliation(s)
- Steven J Forrester
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - George W Booz
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Curt D Sigmund
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Thomas M Coffman
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| |
Collapse
|
229
|
Santisteban MM, Iadecola C. Hypertension, dietary salt and cognitive impairment. J Cereb Blood Flow Metab 2018; 38:2112-2128. [PMID: 30295560 PMCID: PMC6282225 DOI: 10.1177/0271678x18803374] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/31/2018] [Indexed: 12/12/2022]
Abstract
Dementia is growing at an alarming rate worldwide. Although Alzheimer disease is the leading cause, over 50% of individuals diagnosed with Alzheimer disease have vascular lesions at autopsy. There has been an increasing appreciation of the pathogenic role of vascular risk factors in cognitive impairment caused by neurodegeneration. Midlife hypertension is a leading risk factor for late-life dementia. Hypertension alters cerebrovascular structure, impairs the major factors regulating the cerebral microcirculation, and promotes Alzheimer pathology. Experimental studies have identified brain perivascular macrophages as the major free radical source mediating neurovascular dysfunction of hypertension. Recent evidence indicates that high dietary salt may also induce cognitive impairment. Contrary to previous belief, the effect is not necessarily associated with hypertension and is mediated by a deficit in endothelial nitric oxide. Collectively, the evidence suggests a remarkable cellular diversity of the impact of vascular risk factors on the cerebral vasculature and cognition. Whereas long-term longitudinal epidemiological studies are needed to resolve the temporal relationships between vascular risk factors and cognitive dysfunction, single-cell molecular studies of the vasculature in animal models will provide a fuller mechanistic understanding. This knowledge is critical for developing new preventive, diagnostic, and therapeutic approaches for these devastating diseases of the mind.
Collapse
Affiliation(s)
- Monica M Santisteban
- Feil Family Brain and Mind Research Institute Weill Cornell Medicine, New York, NY, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
230
|
Abstract
PURPOSE OF REVIEW Low-grade inflammation drives elevations in blood pressure (BP) and consequent target organ damage in diverse experimental models of hypertension. Here, we discuss recent advances elucidating immune-mediated mechanisms of BP elevation and associated target organ damage. RECENT FINDINGS Inflammatory mediators produced by immune cells or target organs act on the kidney, vasculature, skin, and nervous system to modulate hypertension. For example, cells of the innate immune system, including monocytes, neutrophils, and dendritic cells (DCs), can all promote BP elevation via actions in the vasculature and kidney. Macrophages expressing VEGF-C impact non-osmotic sodium storage in the skin that in turn regulates salt sensitivity. Within the adaptive immune system, activated T cells can secrete tumor necrosis factor-alpha (TNF-α), interleukin-17a (IL-17a), and interferon-gamma (IFN-γ), each of which has augmented BP and renal damage in pre-clinical models. Inversely, deficiency of IL-17a in mice blunts the hypertensive response and attenuates renal sodium retention via a serum- and glucocorticoid-regulated kinase 1 (SGK1)-dependent pathway. Linking innate and adaptive immune responses, dendritic cells activated by augmented extracellular sodium concentrations stimulate T lymphocytes to produce pro-hypertensive cytokines. By contrast, regulatory T cells (Tregs) can protect against hypertension and associated kidney injury. Rodent studies reveal diverse mechanisms via which cells of the innate and adaptive immune systems drive blood pressure elevation by altering the inflammatory milieu in the kidney, vasculature, and brain.
Collapse
|
231
|
Schüler R, Brand A, Klebow S, Wild J, Veras FP, Ullmann E, Roohani S, Kolbinger F, Kossmann S, Wohn C, Daiber A, Münzel T, Wenzel P, Waisman A, Clausen BE, Karbach S. Antagonization of IL-17A Attenuates Skin Inflammation and Vascular Dysfunction in Mouse Models of Psoriasis. J Invest Dermatol 2018; 139:638-647. [PMID: 30367871 DOI: 10.1016/j.jid.2018.09.021] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 08/31/2018] [Accepted: 09/24/2018] [Indexed: 11/17/2022]
Abstract
Besides skin inflammation, patients with severe psoriasis suffer from an increased risk of cardiovascular mortality. IL-17A plays a central role in the development of psoriasis and might connect skin and vascular disease. The aim of this study was to clarify whether anti-IL-17A therapy could also ameliorate the vascular dysfunction associated with severe psoriasis. We analyzed three murine models with varying severities of psoriasis-like skin disease concerning their vascular function and inflammation: (i) K14-IL-17Aind/+ mice with keratinocyte-specific IL-17A overexpression and an early-onset severe psoriasis-like phenotype; (ii) homozygous CD11c-IL-17Aind/ind and heterozygous CD11c-IL-17Aind/+ mice overexpressing IL-17A in CD11c+ cells, leading to a delayed onset of moderate psoriasis-like skin disease; and (iii) the acute model of imiquimod-induced psoriasis-like skin inflammation. Similar to the severity of skin disease, vascular dysfunction correlated with peripheral IL-17A levels and neutrophil infiltration into the aortic vessel wall. Successful anti-IL-17A treatment of psoriatic skin lesions diminished peripheral oxidative stress levels, proinflammatory cytokines, and vascular inflammation. These data highlight the pivotal role of IL-17A linking the development of skin lesions and vascular disease in psoriasis. Anti-IL-17A therapy might thus represent a useful approach to attenuate and prevent vascular disease in psoriasis patients.
Collapse
Affiliation(s)
- Rebecca Schüler
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; Center of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; Center of Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Anna Brand
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Sabrina Klebow
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Johannes Wild
- Center of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; Center of Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; German Center of Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Rhine-Main, Germany
| | - Flávio P Veras
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Elisabeth Ullmann
- Center of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Siyer Roohani
- Center of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Frank Kolbinger
- Novartis Institutes for BioMedical Research and Novartis Pharma, Basel, Switzerland
| | - Sabine Kossmann
- Center of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; Center of Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Christian Wohn
- Aix Marseille University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Andreas Daiber
- Center of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; German Center of Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Rhine-Main, Germany
| | - Thomas Münzel
- Center of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; German Center of Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Rhine-Main, Germany
| | - Philip Wenzel
- Center of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; Center of Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; German Center of Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Rhine-Main, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Björn E Clausen
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
| | - Susanne Karbach
- Center of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; Center of Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; German Center of Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Rhine-Main, Germany.
| |
Collapse
|
232
|
Satou R, Penrose H, Navar LG. Inflammation as a Regulator of the Renin-Angiotensin System and Blood Pressure. Curr Hypertens Rep 2018; 20:100. [PMID: 30291560 DOI: 10.1007/s11906-018-0900-0] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSE OF REVIEW Mechanisms facilitating progression of hypertension via cross stimulation of the renin-angiotensin system (RAS) and inflammation have been proposed. Accordingly, we review and update evidence for regulation of RAS components by pro-inflammatory factors. RECENT FINDINGS Angiotensin II (Ang II), which is produced by RAS, induces vasoconstriction and consequent blood pressure elevation. In addition to this direct action, chronically elevated Ang II stimulates several pathophysiological mechanisms including generation of oxidative stress, stimulation of the nervous system, alterations in renal hemodynamics, and activation of the immune system. In particular, an activated immune system has been shown to contribute to the development of hypertension. Recent studies have demonstrated that immune cell-derived pro-inflammatory cytokines regulate RAS components, further accelerating systemic and local Ang II formation. Specifically, regulation of angiotensinogen (AGT) production by pro-inflammatory cytokines in the liver and kidney is proposed as a key mechanism underlying the progression of Ang II-dependent hypertension.
Collapse
Affiliation(s)
- Ryousuke Satou
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, 1430 Tulane Avenue, SL39, New Orleans, LA, 70112-2699, USA.
| | - Harrison Penrose
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, 1430 Tulane Avenue, SL39, New Orleans, LA, 70112-2699, USA
| | - L Gabriel Navar
- Department of Physiology and Hypertension and Renal Center of Excellence, Tulane University School of Medicine, 1430 Tulane Avenue, SL39, New Orleans, LA, 70112-2699, USA
| |
Collapse
|
233
|
Gordon JH, LaMonte MJ, Genco RJ, Zhao J, Cimato TR, Hovey KM, Wactawski-Wende J. Association of clinical measures of periodontal disease with blood pressure and hypertension among postmenopausal women. J Periodontol 2018; 89:1193-1202. [PMID: 29802640 PMCID: PMC6170702 DOI: 10.1002/jper.17-0562] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/08/2017] [Accepted: 12/29/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Hypertension and periodontal disease are common conditions among postmenopausal women. Periodontal disease has been found associated with hypertension in previous studies, but data in postmenopausal women is limited. METHODS We assessed the cross-sectional associations of clinically measured periodontal disease with prevalent hypertension and measured systolic blood pressure (SBP) among 1341 postmenopausal women enrolled in the Buffalo Osteoporosis and Periodontal Disease (OsteoPerio) study, an ancillary study of the Women's Health Initiative-Observational Study. RESULTS Clinical attachment level (CAL) and number of teeth missing were positively associated with SBP among those not taking antihypertensive medication in crude and multivariable adjusted linear regression models (both P < 0.05). Alveolar crestal height (ACH) and gingival bleeding on probing were associated with higher SBP in crude but not multivariable adjusted models. Neither probing pocket depth (PPD) nor severity categories of periodontitis were associated with SBP. Number of teeth missing was significantly associated with prevalent hypertension in crude and multivariable adjusted models (OR = 1.14, per 5 teeth; P = 0.04). ACH was associated with prevalent hypertension in crude but not adjusted models. CAL, PPD, gingival bleeding, and severity of periodontitis were not significantly associated with prevalent hypertension. CONCLUSIONS These results suggest that measures of oral health including CAL and number of teeth missing are associated with blood pressure in postmenopausal women. Prospective studies are needed to further investigate these associations and the potential underlying mechanisms for these relationships.
Collapse
Affiliation(s)
- Joshua H Gordon
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, State University of New York, Buffalo, NY
| | - Michael J LaMonte
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, State University of New York, Buffalo, NY
| | - Robert J Genco
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, State University of New York, Buffalo, NY
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY
| | - Jiwei Zhao
- Department of Biostatistics, School of Public Health and Health Professions, University at Buffalo, State University of New York, Buffalo, NY
| | - Thomas R Cimato
- Department of Medicine/Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY
| | - Kathleen M Hovey
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, State University of New York, Buffalo, NY
| | - Jean Wactawski-Wende
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, State University of New York, Buffalo, NY
| |
Collapse
|
234
|
Affiliation(s)
- Steven D Crowley
- Division of Nephrology, Department of Medicine at Duke University and Durham VA Medical Centers, DUMC Durham, NC, USA
| |
Collapse
|
235
|
Ahadzadeh E, Rosendahl A, Czesla D, Steffens P, Prüßner L, Meyer-Schwesinger C, Wanner N, Paust HJ, Huber TB, Stahl RAK, Wiech T, Kurts C, Seniuk A, Ehmke H, Wenzel UO. The chemokine receptor CX 3CR1 reduces renal injury in mice with angiotensin II-induced hypertension. Am J Physiol Renal Physiol 2018; 315:F1526-F1535. [PMID: 30207169 DOI: 10.1152/ajprenal.00149.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The role of CX3CR1, also known as fractalkine receptor, in hypertension is unknown. The present study determined the role of the fractalkine receptor CX3CR1 in hypertensive renal and cardiac injury. Expression of CX3CR1 was determined using CX3CR1GFP/+ mice that express a green fluorescent protein (GFP) reporter in CX3CR1+ cells. FACS analysis of leukocytes isolated from the kidney showed that 34% of CD45+ cells expressed CX3CR1. Dendritic cells were the majority of positive cells (67%) followed by macrophages (10%), NK cells (6%), and T cells (10%). With the use of confocal microscopy, the receptor was detected in the kidney only on infiltrating cells but not on resident renal cells. To evaluate the role of CX3CR1 in hypertensive end-organ injury, an aggravated model of hypertension was used. Unilateral nephrectomy was performed followed by infusion of angiotensin II (ANG II, 1.5 ng·g-1·min-1) and a high-salt diet in wild-type ( n = 15) and CX3CR1-deficient mice ( n = 18). CX3CR1 deficiency reduced the number of renal dendritic cells and increased the numbers of renal CD11b/F4/80+ macrophages and CD11b/Ly6G+ neutrophils in ANG II-infused mice. Surprisingly, CX3CR1-deficient mice exhibited increased albuminuria, glomerular injury, and reduced podocyte density in spite of similar levels of arterial hypertension. In contrast, cardiac damage as assessed by increased heart weight, cardiac fibrosis, and expression of fetal genes, and matrix components were not different between both genotypes. Our findings suggest that CX3CR1 exerts protective properties by modulating the invasion of inflammatory cells in hypertensive renal injury. CX3CR1 inhibition should be avoided in hypertension because it may promote hypertensive renal injury.
Collapse
Affiliation(s)
- Erfan Ahadzadeh
- III. Department of Medicine, University Hospital Hamburg-Eppendorf , Hamburg , Germany
| | - Alva Rosendahl
- III. Department of Medicine, University Hospital Hamburg-Eppendorf , Hamburg , Germany
| | - Daniel Czesla
- III. Department of Medicine, University Hospital Hamburg-Eppendorf , Hamburg , Germany
| | - Paula Steffens
- III. Department of Medicine, University Hospital Hamburg-Eppendorf , Hamburg , Germany
| | - Lennard Prüßner
- III. Department of Medicine, University Hospital Hamburg-Eppendorf , Hamburg , Germany
| | | | - Nicola Wanner
- III. Department of Medicine, University Hospital Hamburg-Eppendorf , Hamburg , Germany
| | - Hans Joachim Paust
- III. Department of Medicine, University Hospital Hamburg-Eppendorf , Hamburg , Germany
| | - Tobias B Huber
- III. Department of Medicine, University Hospital Hamburg-Eppendorf , Hamburg , Germany
| | - Rolf A K Stahl
- III. Department of Medicine, University Hospital Hamburg-Eppendorf , Hamburg , Germany
| | - Thorsten Wiech
- Department of Nephropathology University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Kurts
- Institutes of Molecular Medicine and Experimental Immunology, Rheinische Friedrich-Wilhelms University , Bonn , Germany
| | - Anika Seniuk
- Department of Cellular and Integrative Physiology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Heimo Ehmke
- Department of Cellular and Integrative Physiology, University Hospital Hamburg-Eppendorf, Hamburg, Germany.,German Centre for Cardiovascular Research, partner site Hamburg/Kiel/Lübeck, Germany
| | - Ulrich O Wenzel
- III. Department of Medicine, University Hospital Hamburg-Eppendorf , Hamburg , Germany.,German Centre for Cardiovascular Research, partner site Hamburg/Kiel/Lübeck, Germany
| |
Collapse
|
236
|
Eggebrecht L, Prochaska JH, Schulz A, Arnold N, Jünger C, Göbel S, Laubert‐Reh D, Binder H, Beutel ME, Pfeiffer N, Blankenberg S, Lackner KJ, Spronk HM, ten Cate H, Münzel T, Wild PS. Intake of Vitamin K Antagonists and Worsening of Cardiac and Vascular Disease: Results From the Population-Based Gutenberg Health Study. J Am Heart Assoc 2018; 7:e008650. [PMID: 30371151 PMCID: PMC6201416 DOI: 10.1161/jaha.118.008650] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 07/10/2018] [Indexed: 11/30/2022]
Abstract
Background Preclinical data have indicated a link between use of vitamin K antagonists ( VKA ) and detrimental effects on vascular structure and function. The objective of the present study was to determine the relationship between VKA intake and different phenotypes of subclinical cardiovascular disease in the population. Methods and Results Clinical and laboratory data, as well as medical-technical examinations were assessed from 15 010 individuals aged 35 to 74 years during a highly standardized 5-hour visit at the study center of the population-based Gutenberg Health Study. In total, the study sample comprised 287 VKA users and 14 564 VKA nonusers. Multivariable analysis revealed an independent association between VKA intake and stiffness index (β=+2.54 m/s; [0.41/4.66]; P=0.019), ankle-brachial index (β=-0.03; [-0.04/-0.01]; P<0.0001), intima-media thickness (β=+0.03 mm [0.01/0.05]; P=0.0098), left ventricular ejection fraction (β=-4.02% [-4.70/-3.33]; P<0.0001), E/E' (β=+0.04 [0.01/0.08]; P=0.014) left ventricular mass (β=+5.34 g/m2.7 [4.26/6.44]; P<0.0001), and humoral markers of cardiac function and inflammation (midregional pro-atrial natriuretic peptide: β=+0.58 pmol/L [0.50/0.65]; P<0.0001; midregional pro-adrenomedullin: β=+0.18 nmol/L [0.14/0.22]; P<0.0001; N-terminal pro B-type natriuretic peptide: β=+1.90 pg/mL [1.63/2.17]; P<0.0001; fibrinogen: β=+143 mg/dL [132/153]; P<0.0001; C-reactive protein: β=+0.31 mg/L [0.20/0.43]; P<0.0001). Sensitivity analysis in the subsample of participants with atrial fibrillation stratified by intake of VKA demonstrated consistent and robust results. Genetic variants in CYP 2C9, CYP 4F2, and VKORC 1 were modulating effects of VKA on subclinical markers of cardiovascular disease. Conclusions These data demonstrate negative effects of VKA on vascular and cardiac phenotypes of subclinical cardiovascular disease, indicating a possible influence on long-term disease development. These findings may be clinically relevant for the provision of individually tailored antithrombotic therapy.
Collapse
Affiliation(s)
- Lisa Eggebrecht
- Preventive Cardiology and Preventive MedicineCenter for CardiologyUniversity Medical Center MainzMainzGermany
- Center for Translational Vascular Biology (CTVB)University Medical Center MainzMainzGermany
| | - Jürgen H. Prochaska
- Preventive Cardiology and Preventive MedicineCenter for CardiologyUniversity Medical Center MainzMainzGermany
- Center for Translational Vascular Biology (CTVB)University Medical Center MainzMainzGermany
- Center for Thrombosis and HemostasisUniversity Medical Center MainzMainzGermany
- DZHK (German Center for Cardiovascular Research)Partner Site Rhine‐MainMainzGermany
| | - Andreas Schulz
- Preventive Cardiology and Preventive MedicineCenter for CardiologyUniversity Medical Center MainzMainzGermany
- Center for Translational Vascular Biology (CTVB)University Medical Center MainzMainzGermany
| | - Natalie Arnold
- Preventive Cardiology and Preventive MedicineCenter for CardiologyUniversity Medical Center MainzMainzGermany
- Center for Translational Vascular Biology (CTVB)University Medical Center MainzMainzGermany
| | - Claus Jünger
- Preventive Cardiology and Preventive MedicineCenter for CardiologyUniversity Medical Center MainzMainzGermany
- Center for Translational Vascular Biology (CTVB)University Medical Center MainzMainzGermany
| | - Sebastian Göbel
- Center for Translational Vascular Biology (CTVB)University Medical Center MainzMainzGermany
- DZHK (German Center for Cardiovascular Research)Partner Site Rhine‐MainMainzGermany
- Center for Cardiology – Cardiology IUniversity Medical Center MainzMainzGermany
| | - Dagmar Laubert‐Reh
- Preventive Cardiology and Preventive MedicineCenter for CardiologyUniversity Medical Center MainzMainzGermany
- Center for Translational Vascular Biology (CTVB)University Medical Center MainzMainzGermany
| | - Harald Binder
- Center for Translational Vascular Biology (CTVB)University Medical Center MainzMainzGermany
- Institute for Medical Biometry and StatisticsUniversity of FreiburgGermany
| | - Manfred E. Beutel
- Center for Translational Vascular Biology (CTVB)University Medical Center MainzMainzGermany
- Department of Psychosomatic Medicine and PsychotherapyUniversity Medical Center MainzMainzGermany
| | - Nobert Pfeiffer
- Center for Translational Vascular Biology (CTVB)University Medical Center MainzMainzGermany
- Department of OphthalmologyUniversity Medical Center MainzMainzGermany
| | - Stefan Blankenberg
- Clinic for General and Interventional CardiologyUniversity Heart Centre HamburgHamburgGermany
- DZHK (German Center for Cardiovascular Research)Partner Site Hamburg/Kiel/LübeckHamburgGermany
| | - Karl J. Lackner
- Center for Translational Vascular Biology (CTVB)University Medical Center MainzMainzGermany
- DZHK (German Center for Cardiovascular Research)Partner Site Rhine‐MainMainzGermany
- Institute for Clinical Chemistry and Laboratory MedicineUniversity Medical Center MainzMainzGermany
| | - Henri M. Spronk
- Laboratory for Clinical Thrombosis and HemostasisDepartment of Internal MedicineCardiovascular Research Institute Maastricht (CARIM)Maastricht University Medical CenterMaastrichtThe Netherlands
| | - Hugo ten Cate
- Center for Thrombosis and HemostasisUniversity Medical Center MainzMainzGermany
- Laboratory for Clinical Thrombosis and HemostasisDepartment of Internal MedicineCardiovascular Research Institute Maastricht (CARIM)Maastricht University Medical CenterMaastrichtThe Netherlands
| | - Thomas Münzel
- Center for Translational Vascular Biology (CTVB)University Medical Center MainzMainzGermany
- DZHK (German Center for Cardiovascular Research)Partner Site Rhine‐MainMainzGermany
- Center for Cardiology – Cardiology IUniversity Medical Center MainzMainzGermany
| | - Philipp S. Wild
- Preventive Cardiology and Preventive MedicineCenter for CardiologyUniversity Medical Center MainzMainzGermany
- Center for Translational Vascular Biology (CTVB)University Medical Center MainzMainzGermany
- Center for Thrombosis and HemostasisUniversity Medical Center MainzMainzGermany
- DZHK (German Center for Cardiovascular Research)Partner Site Rhine‐MainMainzGermany
| |
Collapse
|
237
|
Small HY, Migliarino S, Czesnikiewicz-Guzik M, Guzik TJ. Hypertension: Focus on autoimmunity and oxidative stress. Free Radic Biol Med 2018; 125:104-115. [PMID: 29857140 DOI: 10.1016/j.freeradbiomed.2018.05.085] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 05/22/2018] [Accepted: 05/28/2018] [Indexed: 12/25/2022]
Abstract
Understanding the causal role of the immune and inflammatory responses in hypertension has led to questions regarding the links between hypertension and autoimmunity. Immune pathology in primary hypertension mimics several autoimmune mechanisms observed in the pathogenesis of systemic lupus erythematosus, psoriasis, systemic sclerosis, rheumatoid arthritis and periodontitis. More importantly, the prevalence of hypertension in patients with these autoimmune diseases is significantly increased, when compared to control populations. Clinical and epidemiological evidence is reviewed along with possible mechanisms linking hypertension and autoimmunity. Inflammation and oxidative stress are linked in a self-perpetuating cycle that significantly contributes to the vascular dysfunction and renal damage associated with hypertension. T cell, B cell, macrophage and NK cell infiltration into these organs is essential for this pathology. Effector cytokines such as IFN-γ, TNF-α and IL-17 affect Na+/H+ exchangers in the kidney. In blood vessels, they lead to endothelial dysfunction and loss of nitric oxide bioavailability and cause vasoconstriction. Both renal and vascular effects are, in part, mediated through induction of reactive oxygen species-producing enzymes such as superoxide anion generating NADPH oxidases and dysfunction of anti-oxidant systems. These mechanisms have recently become important therapeutic targets of novel therapies focused on scavenging oxidative (isolevuglandin) modification of neo-antigenic peptides. Effects of classical immune targeted therapies focused on immunosuppression and anti-cytokine treatments are also reviewed.
Collapse
Affiliation(s)
- Heather Y Small
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Serena Migliarino
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Marta Czesnikiewicz-Guzik
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK; Department of Dental Prophylaxis and Experimental Dentistry, Dental School of Jagiellonian University, Krakow, Poland
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK; Department of Internal and Agricultural Medicine, Jagiellonian University Collegium Medicum, Krakow, Poland.
| |
Collapse
|
238
|
Loperena R, Van Beusecum JP, Itani HA, Engel N, Laroumanie F, Xiao L, Elijovich F, Laffer CL, Gnecco JS, Noonan J, Maffia P, Jasiewicz-Honkisz B, Czesnikiewicz-Guzik M, Mikolajczyk T, Sliwa T, Dikalov S, Weyand CM, Guzik TJ, Harrison DG. Hypertension and increased endothelial mechanical stretch promote monocyte differentiation and activation: roles of STAT3, interleukin 6 and hydrogen peroxide. Cardiovasc Res 2018; 114:1547-1563. [PMID: 29800237 PMCID: PMC6106108 DOI: 10.1093/cvr/cvy112] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/11/2018] [Accepted: 05/16/2018] [Indexed: 01/05/2023] Open
Abstract
Aims Monocytes play an important role in hypertension. Circulating monocytes in humans exist as classical, intermediate, and non-classical forms. Monocyte differentiation can be influenced by the endothelium, which in turn is activated in hypertension by mechanical stretch. We sought to examine the role of increased endothelial stretch and hypertension on monocyte phenotype and function. Methods and results Human monocytes were cultured with confluent human aortic endothelial cells undergoing either 5% or 10% cyclical stretch. We also characterized circulating monocytes in normotensive and hypertensive humans. In addition, we quantified accumulation of activated monocytes and monocyte-derived cells in aortas and kidneys of mice with Angiotensin II-induced hypertension. Increased endothelial stretch enhanced monocyte conversion to CD14++CD16+ intermediate monocytes and monocytes bearing the CD209 marker and markedly stimulated monocyte mRNA expression of interleukin (IL)-6, IL-1β, IL-23, chemokine (C-C motif) ligand 4, and tumour necrosis factor α. STAT3 in monocytes was activated by increased endothelial stretch. Inhibition of STAT3, neutralization of IL-6 and scavenging of hydrogen peroxide prevented formation of intermediate monocytes in response to increased endothelial stretch. We also found evidence that nitric oxide (NO) inhibits formation of intermediate monocytes and STAT3 activation. In vivo studies demonstrated that humans with hypertension have increased intermediate and non-classical monocytes and that intermediate monocytes demonstrate evidence of STAT3 activation. Mice with experimental hypertension exhibit increased aortic and renal infiltration of monocytes, dendritic cells, and macrophages with activated STAT3. Conclusions These findings provide insight into how monocytes are activated by the vascular endothelium during hypertension. This is likely in part due to a loss of NO signalling and increased release of IL-6 and hydrogen peroxide by the dysfunctional endothelium and a parallel increase in STAT activation in adjacent monocytes. Interventions to enhance bioavailable NO, reduce IL-6 or hydrogen peroxide production or to inhibit STAT3 may have anti-inflammatory roles in hypertension and related conditions.
Collapse
Affiliation(s)
- Roxana Loperena
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Justin P Van Beusecum
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hana A Itani
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Noah Engel
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Fanny Laroumanie
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Liang Xiao
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Fernando Elijovich
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cheryl L Laffer
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Juan S Gnecco
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, USA
| | - Jonathan Noonan
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK
| | - Pasquale Maffia
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Barbara Jasiewicz-Honkisz
- Department of Internal Medicine, Jagiellonian University School of Medicine, Cracow, Poland
- Department of Immunology, Jagiellonian University School of Medicine, Cracow, Poland
| | | | - Tomasz Mikolajczyk
- Department of Internal Medicine, Jagiellonian University School of Medicine, Cracow, Poland
- Department of Immunology, Jagiellonian University School of Medicine, Cracow, Poland
| | - Tomasz Sliwa
- Department of Internal Medicine, Jagiellonian University School of Medicine, Cracow, Poland
- Department of Immunology, Jagiellonian University School of Medicine, Cracow, Poland
| | - Sergey Dikalov
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cornelia M Weyand
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - David G Harrison
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
239
|
Pandey KN. Molecular and genetic aspects of guanylyl cyclase natriuretic peptide receptor-A in regulation of blood pressure and renal function. Physiol Genomics 2018; 50:913-928. [PMID: 30169131 DOI: 10.1152/physiolgenomics.00083.2018] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Natriuretic peptides (NPs) exert diverse effects on several biological and physiological systems, such as kidney function, neural and endocrine signaling, energy metabolism, and cardiovascular function, playing pivotal roles in the regulation of blood pressure (BP) and cardiac and vascular homeostasis. NPs are collectively known as anti-hypertensive hormones and their main functions are directed toward eliciting natriuretic/diuretic, vasorelaxant, anti-proliferative, anti-inflammatory, and anti-hypertrophic effects, thereby, regulating the fluid volume, BP, and renal and cardiovascular conditions. Interactions of NPs with their cognate receptors display a central role in all aspects of cellular, biochemical, and molecular mechanisms that govern physiology and pathophysiology of BP and cardiovascular events. Among the NPs atrial and brain natriuretic peptides (ANP and BNP) activate guanylyl cyclase/natriuretic peptide receptor-A (GC-A/NPRA) and initiate intracellular signaling. The genetic disruption of Npr1 (encoding GC-A/NPRA) in mice exhibits high BP and hypertensive heart disease that is seen in untreated hypertensive subjects, including high BP and heart failure. There has been a surge of interest in the NPs and their receptors and a wealth of information have emerged in the last four decades, including molecular structure, signaling mechanisms, altered phenotypic characterization of transgenic and gene-targeted animal models, and genetic analyses in humans. The major goal of the present review is to emphasize and summarize the critical findings and recent discoveries regarding the molecular and genetic regulation of NPs, physiological metabolic functions, and the signaling of receptor GC-A/NPRA with emphasis on the BP regulation and renal and cardiovascular disorders.
Collapse
Affiliation(s)
- Kailash N Pandey
- Department of Physiology, Tulane University Health Sciences Center, School of Medicine , New Orleans, Louisiana
| |
Collapse
|
240
|
Abstract
PURPOSE OF REVIEW Inflammatory cytokines contribute to the pathogenesis of hypertension through effects on renal blood flow and sodium handling. This review will update recent advances that explore the renal actions of immune cells and cytokines in the pathogenesis of hypertension. RECENT FINDINGS Populations of cells from both the innate and adaptive immune systems contribute to hypertension by modulating functions of the vasculature and epithelial cells in the kidney. Macrophages and T lymphocytes can directly regulate the hypertensive response and consequent target organ damage. Dendritic cells and B lymphocytes can alter blood pressure (BP) indirectly by facilitating T-cell activation. Proinflammatory cytokines, including tumor necrosis factor-α, interleukin 17, interleukin 1, and interferon-γ augment BP and/or renal injury when produced by T helper 1 cells, T helper 17 cells, and macrophages. In contrast, interleukin 10 improves vascular and renal functions in preclinical hypertension studies. The effects of transforming growth factor-β are complex because of its profibrotic and immunosuppressive functions that also depend on the localization and concentration of this pleiotropic cytokine. SUMMARY Preclinical studies point to a key role for cytokines in hypertension via their actions in the kidney. Consistent with this notion, anti-inflammatory therapies can attenuate BP elevation in human patients with rheumatologic disease. Conversely, impaired natriuresis may further polarize both T lymphocytes and macrophages toward a proinflammatory state, in a pathogenic, feed-forward loop of immune activation and BP elevation. Understanding the precise renal actions of cytokines in hypertension will be necessary to inhibit cytokine-dependent hypertensive responses while preserving systemic immunity and tumor surveillance.
Collapse
|
241
|
Trott DW, Henson GD, Ho MHT, Allison SA, Lesniewski LA, Donato AJ. Age-related arterial immune cell infiltration in mice is attenuated by caloric restriction or voluntary exercise. Exp Gerontol 2018; 109:99-107. [PMID: 28012941 PMCID: PMC5481497 DOI: 10.1016/j.exger.2016.12.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 12/12/2016] [Accepted: 12/16/2016] [Indexed: 12/12/2022]
Abstract
Age-related arterial inflammation is associated with dysfunction of the arteries and increased risk for cardiovascular disease. To determine if aging increases arterial immune cell infiltration as well as the populations of immune cells principally involved, we tested the hypothesis that large elastic and resistance arteries in old mice would exhibit increased immune cell infiltration compared to young controls. Additionally, we hypothesized that vasoprotective lifestyle interventions such as lifelong caloric restriction or 8weeks of voluntary wheel running would attenuate age-related arterial immune cell infiltration. The aorta and mesenteric vasculature with surrounding perivascular adipose was excised from young normal chow (YNC, 4-6months, n=10), old normal chow (ONC, 28-29months, n=11), old caloric restricted (OCR, 28-29months, n=9), and old voluntary running (OVR, 28-29months, n=5) mice and digested to a single cell suspension. The cells were then labeled with antibodies against CD45 (total leukocytes), CD3 (pan T cells), CD4 (T helper cells), CD8 (cytotoxic T cells), CD19 (B cells), CD11b, and F4/80 (macrophages) and analyzed by flow cytometry. Total leukocytes, T cells (both CD4+ and CD8+ subsets), B cells, and macrophages in both aorta and mesentery were all 5- to 6-fold greater in ONC compared to YNC. Age-related increases in T cell (both CD4+ and CD8+), B cell, and macrophage infiltration in aorta were abolished in OCR mice. OVR mice exhibited 50% lower aortic T cell and normalized macrophage infiltration. B cell infiltration was not affected by VR. Age-related mesenteric CD8+ T cell and macrophage infiltration was normalized in OCR and OVR mice compared to young mice, whereas B cell infiltration was normalized by CR but not VR. Splenic CD4+ T cells from ONC mice exhibited a 3-fold increase in gene expression for the T helper (Th) 1 transcription factor, Tbet, and a 4-fold increase in FoxP3, a T regulatory cell transcription factor, compared to YNC. Splenic B cells and mesenteric macrophages from old mice exhibited decreased proinflammatory cytokine gene expression regardless of treatment group. These results demonstrate that aging is associated with infiltration of immune cells around both the large-elastic and resistance arteries and that the vasoprotective lifestyle interventions, CR and VR, can ameliorate age-related arterial immune cell infiltration.
Collapse
Affiliation(s)
- Daniel W Trott
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, UT, USA.
| | - Grant D Henson
- Department of Exercise and Sport Science, University of Utah, Salt Lake City, UT, USA
| | - Mi H T Ho
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, UT, USA
| | - Sheilah A Allison
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, UT, USA
| | - Lisa A Lesniewski
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, UT, USA; Department of Exercise and Sport Science, University of Utah, Salt Lake City, UT, USA; Geriatric Research, Education, and Clinical Center,Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Anthony J Donato
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, UT, USA; Department of Exercise and Sport Science, University of Utah, Salt Lake City, UT, USA; Geriatric Research, Education, and Clinical Center,Veterans Affairs Medical Center, Salt Lake City, UT, USA; Department of Biochemistry,University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
242
|
Role of NADPH oxidase pathway in renal protection induced by procyanidin B2: In L-NAME induced rat hypertension model. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.04.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
243
|
Abstract
The development of stress drives a host of biological responses that include the overproduction of a family of proteins named heat shock proteins (HSPs), because they were initially studied after heat exposure. HSPs are evolutionarily preserved proteins with a high degree of interspecies homology. HSPs are intracellular proteins that also have extracellular expression. The primary role of HSPs is to protect cell function by preventing irreversible protein damage and facilitating molecular traffic through intracellular pathways. However, in addition to their chaperone role, HSPs are immunodominant molecules that stimulate natural as well as disease-related immune reactivity. The latter may be a consequence of molecular mimicry, generating cross-reactivity between human HSPs and the HSPs of infectious agents. Autoimmune reactivity driven by HSPs could also be the result of enhancement of the immune response to peptides generated during cellular injury and of their role in the delivery of peptides to the major histocompatibility complex in antigen-presenting cells. In humans, HSPs have been found to participate in the pathogenesis of a large number of diseases. This review is focused on the role of HSPs in atherosclerosis and essential hypertension.
Collapse
Affiliation(s)
- B Rodríguez-Iturbe
- 1 Instituto Venezolano de Investigaciones Científicas (IVIC-Zulia), Nephrology Service Hospital Universitario, Universidad del Zulia , Maracaibo, Venezuela
| | - R J Johnson
- 2 Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus , Aurora, CO, USA
| |
Collapse
|
244
|
Caillon A, Paradis P, Schiffrin EL. Role of immune cells in hypertension. Br J Pharmacol 2018; 176:1818-1828. [PMID: 29952002 DOI: 10.1111/bph.14427] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/22/2018] [Accepted: 06/18/2018] [Indexed: 12/19/2022] Open
Abstract
Inflammatory processes have been shown to play an important role in the mechanisms involved in the pathogenesis of hypertension. Innate and adaptive immune responses participate in BP elevation and end-organ damage. Here, we discuss recent studies focusing on novel inflammatory and immune mechanisms that play roles in BP elevation. Different subpopulations of cells involved in innate and adaptive immune responses, such as dendritic cells, monocytes/macrophages and NK cells, on the one hand, and B and T lymphocytes, on the other, contribute to the vascular and kidney injury in hypertension. Unconventional innate-like T cells such as γδ T cells also participate in hypertensive mechanisms by priming both innate and adaptive immune cells, contributing to trigger vascular inflammation and BP elevation. These cells exert their effects in part via production of various cytokines including pro-inflammatory IFN-γ and IL-17 and anti-inflammatory IL-10. The present review summarizes some of these immune mechanisms that participate in the pathophysiology of hypertension. LINKED ARTICLES: This article is part of a themed section on Immune Targets in Hypertension. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.12/issuetoc.
Collapse
Affiliation(s)
- Antoine Caillon
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, QC, Canada
| | - Pierre Paradis
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, QC, Canada
| | - Ernesto L Schiffrin
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, QC, Canada.,Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, QC, Canada
| |
Collapse
|
245
|
Maheshwari M, Romero CA, Monu SR, Kumar N, Liao TD, Peterson EL, Carretero OA. Renal Protective Effects of N-Acetyl-Seryl-Aspartyl-Lysyl-Proline (Ac-SDKP) in Obese Rats on a High-Salt Diet. Am J Hypertens 2018; 31:902-909. [PMID: 29722788 DOI: 10.1093/ajh/hpy052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/17/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Obesity is a public health problem, associated with salt sensitive hypertension, kidney inflammation, and fibrosis. N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP) is a tetra peptide with anti-inflammatory and anti-fibrotic properties. However, its effect on preventing kidney damage in obesity is unknown. We hypothesized that Zucker obese (ZO) rats on a high-salt (HS) diet develop renal damage, inflammation, fibrosis, and this is prevented with Ac-SDKP treatment. METHODS Zucker lean (ZL) and ZO rats (8 weeks old) were treated with Ac-SDKP (1.6 mg/kg/day) while maintained on either a normal-salt (NS; 0.4%) or HS (4%) diet for 8 weeks. Systolic blood pressure (SBP), albuminuria, renal inflammation, and fibrosis were evaluated. RESULTS HS diet increased macrophage infiltration in the kidneys of both ZL and ZO rats but was significantly higher in ZO rats receiving the HS diet (ZL + NS, 13.9 ± 1.3 vs. ZL + HS, 19.14 ± 1.5 and ZO + NS, 25.5 ± 1.4 vs. ZO + HS, 87.8 ± 10.8 cells/mm2; P < 0.05). Ac-SDKP prevented macrophage infiltration in ZO rats (ZO + HS + Ac-SDKP, 32.18 ± 2.4 cells/mm2; P < 0.05). Similarly, glomerulosclerosis, cortical, and medullary interstitial fibrosis were increased in ZO rats fed the HS diet, and Ac-SDKP attenuated these alterations (P < 0.05). SBP was increased in ZO rats fed the HS diet (ZO + NS, 121.3 ± 8.9 vs. ZO + HS, 164 ± 6.9 mm Hg; P < 0.05), and it was significantly decreased with Ac-SDKP treatment (ZO + HS + Ac-SDKP, 144.05 ± 14.1 mm Hg; P = 0.004). Albuminuria was higher in ZO rats than in ZL rats; however, neither HS nor Ac-SDKP treatment affected it. CONCLUSIONS Ac-SDKP treatment in ZO rats fed a HS diet prevented renal damage by reducing inflammation, fibrosis, and SBP.
Collapse
Affiliation(s)
- Mani Maheshwari
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Cesar A Romero
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
| | - Sumit R Monu
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
| | - Nitin Kumar
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
| | - Tang-Dong Liao
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
| | - Edward L Peterson
- Department of Public Health Sciences, Henry Ford Hospital, Detroit, MI, USA
| | - Oscar A Carretero
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
| |
Collapse
|
246
|
Dai X, Hua L, Chen Y, Wang J, Li J, Wu F, Zhang Y, Su J, Wu Z, Liang C. Mechanisms in hypertension and target organ damage: Is the role of the thymus key? (Review). Int J Mol Med 2018; 42:3-12. [PMID: 29620247 PMCID: PMC5979885 DOI: 10.3892/ijmm.2018.3605] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 03/27/2018] [Indexed: 12/23/2022] Open
Abstract
A variety of cells and cytokines have been shown to be involved in the whole process of hypertension. Data from experimental and clinical studies on hypertension have confirmed the key roles of immune cells and inflammation in the process. Dysfunction of the thymus, which modulates the development and maturation of lymphocytes, has been shown to be associated with the severity of hypertension. Furthermore, gradual atrophy, functional decline or loss of the thymus has been revealed to be associated with aging. The restoration or enhancement of thymus function via upregulation in the expression of thymus transcription factors forkhead box N1 or thymus transplantation may provide an option to halt or reverse the pathological process of hypertension. Therefore, the thymus may be key in hypertension and associated target organ damage, and may provide a novel treatment strategy for the clinical management of patients with hypertension in addition to different commercial drugs. The purpose of this review is to summarize and discuss the advances in our understanding of the impact of thymus function on hypertension from data from animal and human studies, and the potential mechanisms.
Collapse
Affiliation(s)
| | | | | | - Jiamei Wang
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Jingyi Li
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Feng Wu
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Yanda Zhang
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Jiyuan Su
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Zonggui Wu
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Chun Liang
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| |
Collapse
|
247
|
Ruiz-Miyazawa KW, Pinho-Ribeiro FA, Borghi SM, Staurengo-Ferrari L, Fattori V, Amaral FA, Teixeira MM, Alves-Filho JC, Cunha TM, Cunha FQ, Casagrande R, Verri WA. Hesperidin Methylchalcone Suppresses Experimental Gout Arthritis in Mice by Inhibiting NF-κB Activation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:6269-6280. [PMID: 29852732 DOI: 10.1021/acs.jafc.8b00959] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Gout arthritis is a painful inflammatory disease induced by monosodium urate (MSU) crystals. We evaluate the therapeutic potential of the flavonoid hesperidin methylchalcone (HMC) in a mouse model of gout arthritis induced by intra-articular injection of MSU (100 μg/10 μL). Orally given HMC (3-30 mg/kg, 100 μL) reduced in a dose-dependent manner the MSU-induced hyperalgesia (44%, p < 0.05), edema (54%, p < 0.05), and leukocyte infiltration (70%, p < 0.05). HMC (30 mg/kg) inhibited MSU-induced infiltration of LysM-eGFP+ cells (81%, p < 0.05), synovitis (76%, p < 0.05), and oxidative stress (increased GSH, FRAP, and ABTS by 62, 78, and 73%, respectively; reduced O2- and NO by 89 and 48%, p < 0.05) and modulated cytokine production (reduced IL-1β, TNF-α, IL-6, and IL-10 by 35, 72, 37, and 46%, respectively, and increased TGF-β by 90%, p < 0.05). HMC also inhibited MSU-induced NF-κB activation (41%, p < 0.05), gp91phox (66%, p < 0.05) and NLRP3 inflammasome components mRNA expression in vivo (72, 77, 71, and 73% for NLRP3, ASC, pro-caspase-1, and pro-IL-1 β, respectively, p < 0.05), and induced Nrf2/HO-1 mRNA expression (3.9- and 5.1-fold increase, respectively, p < 0.05). HMC (30, 100, and 300 μM) did not inhibit IL-1β secretion by macrophages primed by LPS and challenged with MSU (450 μg/mL), demonstrating that the anti-inflammatory effect of HMC in gout arthritis depends on inhibiting NF-κB but not on direct inhibition of inflammasome. The pharmacological effects of HMC indicate its therapeutic potential for the treatment of gout.
Collapse
Affiliation(s)
- Kenji W Ruiz-Miyazawa
- Departamento de Ciências Patológicas , Universidade Estadual de Londrina-UEL , Rod. Celso Garcia Cid, Km 380, PR445, Cx. Postal 10.011 , 86057-970 Londrina , Paraná , Brazil
| | - Felipe A Pinho-Ribeiro
- Departamento de Ciências Patológicas , Universidade Estadual de Londrina-UEL , Rod. Celso Garcia Cid, Km 380, PR445, Cx. Postal 10.011 , 86057-970 Londrina , Paraná , Brazil
| | - Sergio M Borghi
- Departamento de Ciências Patológicas , Universidade Estadual de Londrina-UEL , Rod. Celso Garcia Cid, Km 380, PR445, Cx. Postal 10.011 , 86057-970 Londrina , Paraná , Brazil
| | - Larissa Staurengo-Ferrari
- Departamento de Ciências Patológicas , Universidade Estadual de Londrina-UEL , Rod. Celso Garcia Cid, Km 380, PR445, Cx. Postal 10.011 , 86057-970 Londrina , Paraná , Brazil
| | - Victor Fattori
- Departamento de Ciências Patológicas , Universidade Estadual de Londrina-UEL , Rod. Celso Garcia Cid, Km 380, PR445, Cx. Postal 10.011 , 86057-970 Londrina , Paraná , Brazil
| | - Flavio A Amaral
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Laboratório de Imunofarmacologia , Universidade Federal de Minas Gerais , 31270-567 Belo Horizonte , Minas Gerais , Brazil
| | - Mauro M Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Laboratório de Imunofarmacologia , Universidade Federal de Minas Gerais , 31270-567 Belo Horizonte , Minas Gerais , Brazil
| | - Jose C Alves-Filho
- Department of Pharmacology, Ribeirão Preto Medical School , University of São Paulo , Avenida Bandeirantes s/n , 14050-490 Ribeirão Preto , São Paulo , Brazil
| | - Thiago M Cunha
- Department of Pharmacology, Ribeirão Preto Medical School , University of São Paulo , Avenida Bandeirantes s/n , 14050-490 Ribeirão Preto , São Paulo , Brazil
| | - Fernando Q Cunha
- Department of Pharmacology, Ribeirão Preto Medical School , University of São Paulo , Avenida Bandeirantes s/n , 14050-490 Ribeirão Preto , São Paulo , Brazil
| | - Rubia Casagrande
- Departamento de Ciências Farmacêuticas , Universidade Estadual de Londrina-UEL , Avenida Robert Koch, 60, Hospital Universitário , 86038-350 Londrina , Paraná , Brazil
| | - Waldiceu A Verri
- Departamento de Ciências Patológicas , Universidade Estadual de Londrina-UEL , Rod. Celso Garcia Cid, Km 380, PR445, Cx. Postal 10.011 , 86057-970 Londrina , Paraná , Brazil
| |
Collapse
|
248
|
Balasubbramanian D, Lopez Gelston CA, Rutkowski JM, Mitchell BM. Immune cell trafficking, lymphatics and hypertension. Br J Pharmacol 2018; 176:1978-1988. [PMID: 29797446 DOI: 10.1111/bph.14370] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/10/2018] [Accepted: 05/15/2018] [Indexed: 12/11/2022] Open
Abstract
Activated immune cell infiltration into organs contributes to the development and maintenance of hypertension. Studies targeting specific immune cell populations or reducing their inflammatory signalling have demonstrated a reduction in BP. Lymphatic vessels play a key role in immune cell trafficking and in resolving inflammation, but little is known about their role in hypertension. Studies from our laboratory and others suggest that inflammation-associated or induction of lymphangiogenesis is organ protective and anti-hypertensive. This review provides the basis for hypertension as a disease of chronic inflammation in various tissues and highlights how renal lymphangiogenesis is a novel regulator of kidney health and BP. LINKED ARTICLES: This article is part of a themed section on Immune Targets in Hypertension. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.12/issuetoc.
Collapse
Affiliation(s)
| | | | - Joseph M Rutkowski
- Department of Medical Physiology, Texas A&M College of Medicine, College Station, TX, USA
| | - Brett M Mitchell
- Department of Medical Physiology, Texas A&M College of Medicine, College Station, TX, USA
| |
Collapse
|
249
|
Alexander MR, Norlander AE, Elijovich F, Atreya RV, Gaye A, Gnecco JS, Laffer CL, Galindo CL, Madhur MS. Human monocyte transcriptional profiling identifies IL-18 receptor accessory protein and lactoferrin as novel immune targets in hypertension. Br J Pharmacol 2018; 176:2015-2027. [PMID: 29774543 DOI: 10.1111/bph.14364] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 03/30/2018] [Accepted: 04/30/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Monocytes play a critical role in hypertension. The purpose of our study was to use an unbiased approach to determine whether hypertensive individuals on conventional therapy exhibit an altered monocyte gene expression profile and to perform validation studies of selected genes to identify novel therapeutic targets for hypertension. EXPERIMENTAL APPROACH Next generation RNA sequencing identified differentially expressed genes in a small discovery cohort of normotensive and hypertensive individuals. Several of these genes were further investigated for association with hypertension in multiple validation cohorts using qRT-PCR, regression analysis, phenome-wide association study and case-control analysis of a missense polymorphism. KEY RESULTS We identified 60 genes that were significantly differentially expressed in hypertensive monocytes, many of which are related to IL-1β. Uni- and multivariate regression analyses of the expression of these genes with mean arterial pressure (MAP) revealed four genes that significantly correlated with MAP in normotensive and/or hypertensive individuals. Of these, lactoferrin (LTF), peptidoglycan recognition protein 1 and IL-18 receptor accessory protein (IL18RAP) remained significantly elevated in peripheral monocytes of hypertensive individuals in a separate validation cohort. Interestingly, IL18RAP expression associated with MAP in a cohort of African Americans. Furthermore, homozygosity for a missense single nucleotide polymorphism in LTF that decreases antimicrobial function and increases protein levels (rs1126478) was over-represented in patients with hypertension relative to controls (odds ratio 1.16). CONCLUSIONS AND IMPLICATIONS These data demonstrate that monocytes exhibit enhanced pro-inflammatory gene expression in hypertensive individuals and identify IL18RAP and LTF as potential novel mediators of human hypertension. LINKED ARTICLES This article is part of a themed section on Immune Targets in Hypertension. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.12/issuetoc.
Collapse
Affiliation(s)
- Matthew R Alexander
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Allison E Norlander
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Fernando Elijovich
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ravi V Atreya
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Amadou Gaye
- Metabolic, Cardiovascular and Inflammatory Disease Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Juan S Gnecco
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN, USA
| | - Cheryl L Laffer
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cristi L Galindo
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Meena S Madhur
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.,Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
250
|
Münzel T, Daiber A, Steven S, Tran LP, Ullmann E, Kossmann S, Schmidt FP, Oelze M, Xia N, Li H, Pinto A, Wild P, Pies K, Schmidt ER, Rapp S, Kröller-Schön S. Effects of noise on vascular function, oxidative stress, and inflammation: mechanistic insight from studies in mice. Eur Heart J 2018; 38:2838-2849. [PMID: 28329261 DOI: 10.1093/eurheartj/ehx081] [Citation(s) in RCA: 182] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 02/06/2017] [Indexed: 02/07/2023] Open
Abstract
Aims Epidemiological studies indicate that traffic noise increases the incidence of coronary artery disease, hypertension and stroke. The underlying mechanisms remain largely unknown. Field studies with nighttime noise exposure demonstrate that aircraft noise leads to vascular dysfunction, which is markedly improved by vitamin C, suggesting a key role of oxidative stress in causing this phenomenon. Methods and results We developed a novel animal model to study the vascular consequences of aircraft noise exposure. Peak sound levels of 85 and mean sound level of 72 dBA applied by loudspeakers for 4 days caused an increase in systolic blood pressure, plasma noradrenaline and angiotensin II levels and induced endothelial dysfunction. Noise increased eNOS expression but reduced vascular NO levels because of eNOS uncoupling. Noise increased circulating levels of nitrotyrosine, interleukine-6 and vascular expression of the NADPH oxidase subunit Nox2, nitrotyrosine-positive proteins and of endothelin-1. FACS analysis demonstrated an increase in infiltrated natural killer-cells and neutrophils into the vasculature. Equal mean sound pressure levels of white noise for 4 days did not induce these changes. Comparative Illumina sequencing of transcriptomes of aortic tissues from aircraft noise-treated animals displayed significant changes of genes in part responsible for the regulation of vascular function, vascular remodelling, and cell death. Conclusion We established a novel and unique aircraft noise stress model with increased blood pressure and vascular dysfunction associated with oxidative stress. This animal model enables future studies of molecular mechanisms, mitigation strategies, and pharmacological interventions to protect from noise-induced vascular damage.
Collapse
Affiliation(s)
- Thomas Münzel
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main
| | - Andreas Daiber
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main
| | - Sebastian Steven
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Lan P Tran
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Elisabeth Ullmann
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Sabine Kossmann
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Frank P Schmidt
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Matthias Oelze
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Ning Xia
- Department of Pharmacology, University Medical Center, Obere Zahlbacher , Mainz, Germany
| | - Huige Li
- Department of Pharmacology, University Medical Center, Obere Zahlbacher , Mainz, Germany
| | - Antonio Pinto
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Philipp Wild
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main.,Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Kai Pies
- Engineering Office for Noise Protection, Mainz, Germany
| | - Erwin R Schmidt
- Institute for Molecular Genetics, Johannes Gutenberg University, Mainz, Germany
| | - Steffen Rapp
- Institute for Molecular Genetics, Johannes Gutenberg University, Mainz, Germany
| | - Swenja Kröller-Schön
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| |
Collapse
|