1
|
Babicz RSE, Baylor N, Terlouw A, Faber DA, Fukushima K, Biondi RM, Bouley R, Brown D. Ribosomal s6 kinase is a mediator of aquaporin-2 S256 phosphorylation and membrane accumulation after EGFR inhibition with erlotinib. Am J Physiol Renal Physiol 2025; 328:F344-F359. [PMID: 39823198 DOI: 10.1152/ajprenal.00353.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025] Open
Abstract
Vasopressin (VP) activates protein kinase A (PKA), resulting in phosphorylation events and membrane accumulation of aquaporin-2 (AQP2). Epidermal growth factor receptor (EGFR) inhibition with erlotinib also induces AQP2 membrane trafficking with a phosphorylation pattern similar to VP, but without increasing PKA activity. Here, we identify the ribosomal s6 kinase (RSK) as a major mediator phosphorylating AQP2 in this novel, erlotinib-induced pathway. We found that RSK was expressed in collecting duct principal cells in rat kidneys. RSK inhibition with BI-D1870 blocked erlotinib-induced AQP2 serine 256 (S256) phosphorylation and membrane accumulation. CRISPR-generated RSK knockout (KO) cells failed to show increased S256 phosphorylation in response to erlotinib. Like PKA, RSK was able to phosphorylate AQP2 S256 in vitro. Inhibition of phosphoinositide-dependent kinase-1 (PDK1), a known activator of RSK, blocked erlotinib-induced AQP2 S256 phosphorylation and membrane accumulation. We conclude that RSK is a crucial terminal kinase phosphorylating AQP2 at S256 upon EGFR inhibition by erlotinib.NEW & NOTEWORTHY Epidermal growth factor receptor (EGFR) inhibition with erlotinib induces aquaporin-2 (AQP2) membrane accumulation with a phosphorylation pattern similar to vasopressin (VP). Here, we identify the ribosomal s6 kinase (RSK) as a major mediator phosphorylating AQP2 in this novel, erlotinib-induced pathway. In addition, we show that phosphoinositide-dependent kinase-1 (PDK1), a known activator of RSK, is implicated in this pathway: PDK1 inhibition blocks erlotinib-induced AQP2 S256 phosphorylation and membrane accumulation.
Collapse
Affiliation(s)
- Richard S E Babicz
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Noah Baylor
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Abby Terlouw
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Daphne A Faber
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Kazuhiko Fukushima
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Ricardo M Biondi
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck SocietyBuenos AiresArgentina
| | - Richard Bouley
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Dennis Brown
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
2
|
Kopaliani I, Elsaid B, Speier S, Deussen A. Immune and Metabolic Mechanisms of Endothelial Dysfunction. Int J Mol Sci 2024; 25:13337. [PMID: 39769104 PMCID: PMC11728141 DOI: 10.3390/ijms252413337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/01/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025] Open
Abstract
Endothelial dysfunction is a strong prognostic factor in predicting the development of cardiovascular diseases. Dysfunctional endothelium loses its homeostatic ability to regulate vascular tone and prevent overactivation of inflammation, leading to vascular dysfunction. These functions are critical for vascular homeostasis and arterial pressure control, the disruption of which may lead to hypertension. Hypertension itself can also cause endothelial dysfunction, as endothelial cells are susceptible to haemodynamic changes. Although it is unclear which of those factors appear first, they create a vicious circle further damaging multiple organs, including the heart and vessels. There are also sex-specific differences in homeostatic functions of the endothelium regarding vessel tone regulation, which may contribute to differences in arterial blood pressure between men and women. Even more importantly, there are sex-differences in the development of endothelial dysfunction and vessel remodelling. Hence, an understanding of the mechanisms of endothelial dysfunction and its contribution to pathological vascular remodelling during hypertension is of critical importance. This review addresses immunological and metabolic aspects in mechanisms of endothelial dysfunction and the resulting mechanisms in vascular remodelling with respect to arterial hypertension, including the potential role of sex-specific differences.
Collapse
Affiliation(s)
- Irakli Kopaliani
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
| | - Basant Elsaid
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
- Department of Physiology, Faculty of Medicine, Ain Shams University, Cairo 1181, Egypt
| | - Stephan Speier
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Andreas Deussen
- Institute of Physiology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01037 Dresden, Germany; (B.E.); (S.S.); (A.D.)
| |
Collapse
|
3
|
Elgazzaz M, Filipeanu C, Lazartigues E. Angiotensin-Converting Enzyme 2 Posttranslational Modifications and Implications for Hypertension and SARS-CoV-2: 2023 Lewis K. Dahl Memorial Lecture. Hypertension 2024; 81:1438-1449. [PMID: 38567498 PMCID: PMC11168885 DOI: 10.1161/hypertensionaha.124.22067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
ACE2 (angiotensin-converting enzyme 2), a multifunctional transmembrane protein, is well recognized as an important member of the (RAS) renin-angiotensin system with important roles in the regulation of cardiovascular function by opposing the harmful effects of Ang-II (angiotensin II) and AT1R (Ang-II type 1 receptor) activation. More recently, ACE2 was found to be the entry point for the SARS-CoV-2 virus into cells, causing COVID-19. This finding has led to an exponential rise in the number of publications focused on ACE2, albeit these studies often have opposite objectives to the preservation of ACE2 in cardiovascular regulation. However, notwithstanding accumulating data of the role of ACE2 in the generation of angiotensin-(1-7) and SARS-CoV-2 internalization, numerous other putative roles of this enzyme remain less investigated and not yet characterized. Currently, no drug modulating ACE2 function or expression is available in the clinic, and the development of new pharmacological tools should attempt targeting each step of the lifespan of the protein from synthesis to degradation. The present review expands on our presentation during the 2023 Lewis K. Dahl Memorial Lecture Sponsored by the American Heart Association Council on Hypertension. We provide a critical summary of the current knowledge of the mechanisms controlling ACE2 internalization and intracellular trafficking, the mutual regulation with GPCRs (G-protein-coupled receptors) and other proteins, and posttranslational modifications. A major focus is on ubiquitination which has become a critical step in the modulation of ACE2 cellular levels.
Collapse
Affiliation(s)
- Mona Elgazzaz
- Department of Physiology, Augusta University, Medical College of Georgia, Augusta, GA 30912, USA
- Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Catalin Filipeanu
- Department of Pharmacology, Howard University, Washington, DC 20059, USA
| | - Eric Lazartigues
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Southeast Louisiana Veterans Health Care System, New Orleans, LA 70119, USA
| |
Collapse
|
4
|
Shukla AK, Awasthi K, Usman K, Banerjee M. Role of renin-angiotensin system/angiotensin converting enzyme-2 mechanism and enhanced COVID-19 susceptibility in type 2 diabetes mellitus. World J Diabetes 2024; 15:606-622. [PMID: 38680697 PMCID: PMC11045416 DOI: 10.4239/wjd.v15.i4.606] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/22/2024] [Accepted: 02/27/2024] [Indexed: 04/11/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a disease that caused a global pandemic and is caused by infection of severe acute respiratory syndrome coronavirus 2 virus. It has affected over 768 million people worldwide, resulting in approximately 6900000 deaths. High-risk groups, identified by the Centers for Disease Control and Prevention, include individuals with conditions like type 2 diabetes mellitus (T2DM), obesity, chronic lung disease, serious heart conditions, and chronic kidney disease. Research indicates that those with T2DM face a heightened susceptibility to COVID-19 and increased mortality compared to non-diabetic individuals. Examining the renin-angiotensin system (RAS), a vital regulator of blood pressure and pulmonary stability, reveals the significance of the angiotensin-converting enzyme (ACE) and ACE2 enzymes. ACE converts angiotensin-I to the vasoconstrictor angiotensin-II, while ACE2 counters this by converting angiotensin-II to angiotensin 1-7, a vasodilator. Reduced ACE2 expression, common in diabetes, intensifies RAS activity, contributing to conditions like inflammation and fibrosis. Although ACE inhibitors and angiotensin receptor blockers can be therapeutically beneficial by increasing ACE2 levels, concerns arise regarding the potential elevation of ACE2 receptors on cell membranes, potentially facilitating COVID-19 entry. This review explored the role of the RAS/ACE2 mechanism in amplifying severe acute respiratory syndrome coronavirus 2 infection and associated complications in T2DM. Potential treatment strategies, including recombinant human ACE2 therapy, broad-spectrum antiviral drugs, and epigenetic signature detection, are discussed as promising avenues in the battle against this pandemic.
Collapse
Affiliation(s)
- Ashwin Kumar Shukla
- Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow 226007, Uttar Pradesh, India
| | - Komal Awasthi
- Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow 226007, Uttar Pradesh, India
| | - Kauser Usman
- Department of Medicine, King Georges’ Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Monisha Banerjee
- Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow 226007, Uttar Pradesh, India
- Institute of Advanced Molecular Genetics, and Infectious Diseases (IAMGID), University of Lucknow, Lucknow 226007, Uttar Pradesh, India
| |
Collapse
|
5
|
Tian Y, Fopiano KA, Buncha V, Lang L, Rudic RD, Filosa JA, Dou H, Bagi Z. Aging-induced impaired endothelial wall shear stress mechanosensing causes arterial remodeling via JAM-A/F11R shedding by ADAM17. GeroScience 2022; 44:349-369. [PMID: 34718985 PMCID: PMC8810930 DOI: 10.1007/s11357-021-00476-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 10/08/2021] [Indexed: 11/25/2022] Open
Abstract
Physiological and pathological vascular remodeling is uniquely driven by mechanical forces from blood flow in which wall shear stress (WSS) mechanosensing by the vascular endothelium plays a pivotal role. This study aimed to determine the novel role for a disintegrin and metalloproteinase 17 (ADAM17) in impaired WSS mechanosensing, which was hypothesized to contribute to aging-associated abnormal vascular remodeling. Without changes in arterial blood pressure and blood flow rate, skeletal muscle resistance arteries of aged mice (30-month-old vs. 12-week-old) exhibited impaired WSS mechanosensing and displayed inward hypertrophic arterial remodeling. These vascular changes were recapitulated by in vivo confined, AAV9-mediated overexpression of ADAM17 in the resistance arteries of young mice. An aging-related increase in ADAM17 expression reduced the endothelial junction level of its cleavage substrate, junctional adhesion molecule-A/F11 receptor (JAM-A/F11R). In cultured endothelial cells subjected to steady WSS ADAM17 activation or JAM-A/F11R knockdown inhibited WSS mechanosensing. The ADAM17-activation induced, impaired WSS mechanosensing was normalized by overexpression of ADAM17 cleavage resistant, mutated JAM-AV232Y both in cultured endothelial cells and in resistance arteries of aged mice, in vivo. These data demonstrate a novel role for ADAM17 in JAM-A/F11R cleavage-mediated impaired endothelial WSS mechanosensing and subsequently developed abnormal arterial remodeling in aging. ADAM17 could prove to be a key regulator of WSS mechanosensing, whereby it can also play a role in pathological vascular remodeling in diseases.
Collapse
Affiliation(s)
- Yanna Tian
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Katie Anne Fopiano
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Vadym Buncha
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Liwei Lang
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - R Daniel Rudic
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Jessica A Filosa
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Huijuan Dou
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
- Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Zsolt Bagi
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
6
|
Verma K, Pant M, Paliwal S, Dwivedi J, Sharma S. An Insight on Multicentric Signaling of Angiotensin II in Cardiovascular system: A Recent Update. Front Pharmacol 2021; 12:734917. [PMID: 34489714 PMCID: PMC8417791 DOI: 10.3389/fphar.2021.734917] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/09/2021] [Indexed: 12/17/2022] Open
Abstract
The multifaceted nature of the renin-angiotensin system (RAS) makes it versatile due to its involvement in pathogenesis of the cardiovascular disease. Angiotensin II (Ang II), a multifaceted member of RAS family is known to have various potential effects. The knowledge of this peptide has immensely ameliorated after meticulous research for decades. Several studies have evidenced angiotensin I receptor (AT1 R) to mediate the majority Ang II-regulated functions in the system. Functional crosstalk between AT1 R mediated signal transduction cascades and other signaling pathways has been recognized. The review will provide an up-to-date information and recent discoveries involved in Ang II receptor signal transduction and their functional significance in the cardiovascular system for potential translation in therapeutics. Moreover, the review also focuses on the role of stem cell-based therapies in the cardiovascular system.
Collapse
Affiliation(s)
- Kanika Verma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, India
| | - Malvika Pant
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, India
| | - Sarvesh Paliwal
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, India
| | - Jaya Dwivedi
- Department of Chemistry, Banasthali Vidyapith, Banasthali, India
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, India
| |
Collapse
|
7
|
Terada Y, Yayama K. Angiotensin II-Induced Vasoconstriction via Rho Kinase Activation in Pressure-Overloaded Rat Thoracic Aortas. Biomolecules 2021; 11:biom11081076. [PMID: 34439742 PMCID: PMC8391281 DOI: 10.3390/biom11081076] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/23/2021] [Accepted: 07/19/2021] [Indexed: 01/25/2023] Open
Abstract
Angiotensin II (Ang II) induces vasoconstriction through myosin light chain (MLC) kinase activation and MLC phosphatase inactivation via phosphorylation of myosin phosphatase targeting subunit 1 (MYPT1) by Rho kinase. However, the detailed mechanism underlying Rho kinase activation by Ang II is still unknown. We investigated the mechanism of Ang II-induced vasoconstriction mediated by Rho kinase in pressure-overloaded rat thoracic aortas. Pressure-overloaded rats were produced by coarctation of the suprarenal abdominal aorta in four-week-old male Wistar rats. The contractile response to Ang II was significantly enhanced in the pressure-overloaded rats. Ang II-induced vasoconstriction was attenuated by inhibitors of Rho kinase, extracellular signal-regulated kinase 1 and 2 (Erk1/2), and epidermal growth factor receptor (EGFR) in both the sham-operated and pressure-overloaded rats. The Ang II-induced vasoconstriction was attenuated by a Janus kinase 2 (JAK2) inhibitor in only the pressure-overloaded rats. The protein levels of MYPT1 and JAK2 increased only in the pressure-overloaded rat thoracic aortas. These results suggested that Ang II-induced contraction is mediated by Rho kinase activation via EGFR, Erk1/2, and JAK2 in pressure-overloaded rat thoracic aortas. Moreover, Ang II-induced contraction was enhanced in pressure-overloaded rats probably because the protein levels of MYPT1 and JAK2 increased in the thoracic aortas.
Collapse
|
8
|
Kawai T, Elliott KJ, Scalia R, Eguchi S. Contribution of ADAM17 and related ADAMs in cardiovascular diseases. Cell Mol Life Sci 2021; 78:4161-4187. [PMID: 33575814 PMCID: PMC9301870 DOI: 10.1007/s00018-021-03779-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/23/2020] [Accepted: 01/27/2021] [Indexed: 02/06/2023]
Abstract
A disintegrin and metalloproteases (ADAMs) are key mediators of cell signaling by ectodomain shedding of various growth factors, cytokines, receptors and adhesion molecules at the cellular membrane. ADAMs regulate cell proliferation, cell growth, inflammation, and other regular cellular processes. ADAM17, the most extensively studied ADAM family member, is also known as tumor necrosis factor (TNF)-α converting enzyme (TACE). ADAMs-mediated shedding of cytokines such as TNF-α orchestrates immune system or inflammatory cascades and ADAMs-mediated shedding of growth factors causes cell growth or proliferation by transactivation of the growth factor receptors including epidermal growth factor receptor. Therefore, increased ADAMs-mediated shedding can induce inflammation, tissue remodeling and dysfunction associated with various cardiovascular diseases such as hypertension and atherosclerosis, and ADAMs can be a potential therapeutic target in these diseases. In this review, we focus on the role of ADAMs in cardiovascular pathophysiology and cardiovascular diseases. The main aim of this review is to stimulate new interest in this area by highlighting remarkable evidence.
Collapse
Affiliation(s)
- Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine At Temple University, Philadelphia, PA, USA
| | - Katherine J Elliott
- Cardiovascular Research Center, Lewis Katz School of Medicine At Temple University, Philadelphia, PA, USA
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine At Temple University, Philadelphia, PA, USA
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine At Temple University, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Oz M, Lorke DE, Kabbani N. A comprehensive guide to the pharmacologic regulation of angiotensin converting enzyme 2 (ACE2), the SARS-CoV-2 entry receptor. Pharmacol Ther 2021; 221:107750. [PMID: 33275999 PMCID: PMC7854082 DOI: 10.1016/j.pharmthera.2020.107750] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 02/06/2023]
Abstract
The recent emergence of coronavirus disease-2019 (COVID-19) as a global pandemic has prompted scientists to address an urgent need for defining mechanisms of disease pathology and treatment. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent for COVID-19, employs angiotensin converting enzyme 2 (ACE2) as its primary target for cell surface attachment and likely entry into the host cell. Thus, understanding factors that may regulate the expression and function of ACE2 in the healthy and diseased body is critical for clinical intervention. Over 66% of all adults in the United States are currently using a prescription drug and while earlier findings have focused on possible upregulation of ACE2 expression through the use of renin angiotensin system (RAS) inhibitors, mounting evidence suggests that various other widely administered drugs used in the treatment of hypertension, heart failure, diabetes mellitus, hyperlipidemias, coagulation disorders, and pulmonary disease may also present a varied risk for COVID-19. Specifically, we summarize mechanisms on how heparin, statins, steroids and phytochemicals, besides their established therapeutic effects, may also interfere with SARS-CoV-2 viral entry into cells. We also describe evidence on the effect of several vitamins, phytochemicals, and naturally occurring compounds on ACE2 expression and activity in various tissues and disease models. This comprehensive review aims to provide a timely compendium on the potential impact of commonly prescribed drugs and pharmacologically active compounds on COVID-19 pathology and risk through regulation of ACE2 and RAS signaling.
Collapse
Key Words
- adam17, a disintegrin and metalloprotease 17
- ace, angiotensin i converting enzyme
- ace-inh., angiotensin i converting enzyme inhibitor
- ampk, amp-activated protein kinase
- ang-ii, angiotensin ii
- arb, angiotensin ii type 1-receptor blocker
- ards, acute respiratory distress syndrome
- at1-r, angiotensin ii type 1-receptor
- βarb, β-adrenergic receptor blockers
- bk, bradykinin
- ccb, calcium channel blockers
- ch25h, cholesterol-25-hydroxylase
- copd, chronic obstructive lung disease
- cox, cyclooxygenase
- covid-19, coronavirus disease-2019
- dabk, [des-arg9]-bradykinin
- erk, extracellular signal-regulated kinase
- 25hc, 25-hydroxycholesterol
- hs, heparan sulfate
- hspg, heparan sulfate proteoglycan
- ibd, inflammatory bowel disease
- map, mitogen-activated protein
- mers, middle east respiratory syndrome
- mrb, mineralocorticoid receptor blocker
- nos, nitric oxide synthase
- nsaid, non-steroid anti-inflammatory drug
- ras, renin-angiotensin system
- sars-cov, severe acute respiratory syndrome coronavirus
- sh, spontaneously hypertensive
- s protein, spike protein
- sirt1, sirtuin 1
- t2dm, type 2 diabetes mellitus
- tcm, traditional chinese medicine
- tmprss2, transmembrane protease, serine 2
- tnf, tumor necrosis factor
- ufh, unfractionated heparin
Collapse
Affiliation(s)
- Murat Oz
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Kuwait University, Safat 13110, Kuwait.
| | - Dietrich Ernst Lorke
- Department of Anatomy and Cellular Biology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates; Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Nadine Kabbani
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
| |
Collapse
|
10
|
Russell JJ, Grisanti LA, Brown SM, Bailey CA, Bender SB, Chandrasekar B. Reversion inducing cysteine rich protein with Kazal motifs and cardiovascular diseases: The RECKlessness of adverse remodeling. Cell Signal 2021; 83:109993. [PMID: 33781845 DOI: 10.1016/j.cellsig.2021.109993] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/19/2022]
Abstract
The Reversion Inducing Cysteine Rich Protein With Kazal Motifs (RECK) is a glycosylphosphatidylinositol (GPI) anchored membrane-bound regulator of matrix metalloproteinases (MMPs). It is expressed throughout the body and plays a role in extracellular matrix (ECM) homeostasis and inflammation. In initial studies, RECK expression was found to be downregulated in various invasive cancers and associated with poor prognostic outcome. Restoring RECK, however, has been shown to reverse the metastatic phenotype. Downregulation of RECK expression is also reported in non-malignant diseases, such as periodontal disease, renal fibrosis, and myocardial fibrosis. As such, RECK induction has therapeutic potential in several chronic diseases. Mechanistically, RECK negatively regulates various matrixins involved in cell migration, proliferation, and adverse remodeling by targeting the expression and/or activation of multiple MMPs, A Disintegrin And Metalloproteinase Domain-Containing Proteins (ADAMs), and A Disintegrin And Metalloproteinase With Thrombospondin Motifs (ADAMTS). Outside of its role in remodeling, RECK has also been reported to exert anti-inflammatory effects. In cardiac diseases, for example, it has been shown to counteract several downstream effectors of Angiotensin II (Ang-II) that play a role in adverse cardiac and vascular remodeling, such as Interleukin-6 (IL-6)/IL-6 receptor (IL-6R)/glycoprotein 130 (IL-6 signal transducer) signaling and Epidermal Growth Factor Receptor (EGFR) transactivation. This review article focuses on the current understanding of the multifunctional effects of RECK and how its downregulation may contribute to adverse cardiovascular remodeling.
Collapse
Affiliation(s)
- Jacob J Russell
- Biomedical Sciences, University of Missouri, Columbia, MO, United States of America; Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States of America.
| | - Laurel A Grisanti
- Biomedical Sciences, University of Missouri, Columbia, MO, United States of America.
| | - Scott M Brown
- Biomedical Sciences, University of Missouri, Columbia, MO, United States of America; Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States of America.
| | - Chastidy A Bailey
- Biomedical Sciences, University of Missouri, Columbia, MO, United States of America; Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States of America.
| | - Shawn B Bender
- Biomedical Sciences, University of Missouri, Columbia, MO, United States of America; Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States of America; Dalton Cardiovascular Center, University of Missouri, Columbia, MO, United States of America.
| | - B Chandrasekar
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States of America; Medicine, University of Missouri School of Medicine, Columbia, MO, United States of America; Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States of America; Dalton Cardiovascular Center, University of Missouri, Columbia, MO, United States of America.
| |
Collapse
|
11
|
Iwasaki M, Saito J, Zhao H, Sakamoto A, Hirota K, Ma D. Inflammation Triggered by SARS-CoV-2 and ACE2 Augment Drives Multiple Organ Failure of Severe COVID-19: Molecular Mechanisms and Implications. Inflammation 2021; 44:13-34. [PMID: 33029758 PMCID: PMC7541099 DOI: 10.1007/s10753-020-01337-3] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/20/2020] [Accepted: 08/31/2020] [Indexed: 01/08/2023]
Abstract
The widespread occurrence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to a pandemic of coronavirus disease 2019 (COVID-19). The S spike protein of SARS-CoV-2 binds with angiotensin-converting enzyme 2 (ACE2) as a functional "receptor" and then enters into host cells to replicate and damage host cells and organs. ACE2 plays a pivotal role in the inflammation, and its downregulation may aggravate COVID-19 via the renin-angiotensin system, including by promoting pathological changes in lung injury and involving inflammatory responses. Severe patients of COVID-19 often develop acute respiratory distress syndrome and multiple organ dysfunction/failure with high mortality that may be closely related to the hyper-proinflammatory status called the "cytokine storm." Massive cytokines including interleukin-6, nuclear factor kappa B (NFκB), and tumor necrosis factor alpha (TNFα) released from SARS-CoV-2-infected macrophages and monocytes lead inflammation-derived injurious cascades causing multi-organ injury/failure. This review summarizes the current evidence and understanding of the underlying mechanisms of SARS-CoV-2, ACE2 and inflammation co-mediated multi-organ injury or failure in COVID-19 patients.
Collapse
Affiliation(s)
- Masae Iwasaki
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
- Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Junichi Saito
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
- Department of Anesthesiology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Hailin Zhao
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Atsuhiro Sakamoto
- Department of Anesthesiology and Pain Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Kazuyoshi Hirota
- Department of Anesthesiology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK.
| |
Collapse
|
12
|
Sheng L, Bayliss G, Zhuang S. Epidermal Growth Factor Receptor: A Potential Therapeutic Target for Diabetic Kidney Disease. Front Pharmacol 2021; 11:598910. [PMID: 33574751 PMCID: PMC7870700 DOI: 10.3389/fphar.2020.598910] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
Diabetic kidney disease (DKD) is a leading cause of end-stage renal disease worldwide and the major cause of renal failure among patients on hemodialysis. Numerous studies have demonstrated that transient activation of epidermal growth factor receptor (EGFR) pathway is required for promoting kidney recovery from acute injury whereas its persistent activation is involved in the progression of various chronic kidney diseases including DKD. EGFR-mediated pathogenesis of DKD is involved in hemodynamic alteration, metabolic disturbance, inflammatory response and parenchymal cellular dysfunction. Therapeutic intervention of this receptor has been available in the oncology setting. Targeting EGFR might also hold a therapeutic potential for DKD. Here we review the functional role of EGFR in the development of DKD, mechanisms involved and the perspective about use of EGFR inhibitors as a treatment for DKD.
Collapse
Affiliation(s)
- Lili Sheng
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - George Bayliss
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, United States
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, United States
| |
Collapse
|
13
|
Monteonofrio L, Florio MC, AlGhatrif M, Lakatta EG, Capogrossi MC. Aging- and gender-related modulation of RAAS: potential implications in COVID-19 disease. VASCULAR BIOLOGY 2020; 3:R1-R14. [PMID: 33537555 PMCID: PMC7849461 DOI: 10.1530/vb-20-0014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 12/11/2020] [Indexed: 12/15/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is a new infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). COVID-19 is frequently characterized by a marked inflammatory response with severe pneumonia and respiratory failure associated with multiorgan involvement. Some risk factors predispose patients to develop a more severe infection and to an increased mortality; among them, advanced age and male gender have been identified as major and independent risk factors for COVID-19 poor outcome. The renin-angiotensin-aldosterone system (RAAS) is strictly involved in COVID-19 because angiotensin converting enzyme 2 (ACE2) is the host receptor for SARS-CoV-2 and also converts pro-inflammatory angiotensin (Ang) II into anti-inflammatory Ang(1–7). In this review, we have addressed the effect of aging and gender on RAAS with emphasis on ACE2, pro-inflammatory Ang II/Ang II receptor 1 axis and anti-inflammatory Ang(1–7)/Mas receptor axis.
Collapse
Affiliation(s)
- Laura Monteonofrio
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Maria Cristina Florio
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Majd AlGhatrif
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA.,Longitudinal Study Section, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA.,Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Edward G Lakatta
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Maurizio C Capogrossi
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA.,Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
14
|
Factor XII blockade inhibits aortic dilatation in angiotensin II-infused apolipoprotein E-deficient mice. Clin Sci (Lond) 2020; 134:1049-1061. [PMID: 32309850 DOI: 10.1042/cs20191020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 04/05/2020] [Accepted: 04/20/2020] [Indexed: 12/23/2022]
Abstract
Abdominal aortic aneurysm (AAA) is an important cause of mortality in older adults. Chronic inflammation and excessive matrix remodelling are considered important in AAA pathogenesis. Kinins are bioactive peptides important in regulating inflammation. Stimulation of the kinin B2 receptor has been previously reported to promote AAA development and rupture in a mouse model. The endogenous B2 receptor agonist, bradykinin, is generated from the kallikrein-kinin system following activation of plasma kallikrein by Factor XII (FXII). In the current study whole-body FXII deletion, or neutralisation of activated FXII (FXIIa), inhibited expansion of the suprarenal aorta (SRA) of apolipoprotein E-deficient mice in response to angiotensin II (AngII) infusion. FXII deficiency or FXIIa neutralisation led to decreased aortic tumor necrosis factor-α-converting enzyme (TACE/a disintegrin and metalloproteinase-17 (aka tumor necrosis factor-α-converting enzyme) (ADAM-17)) activity, plasma kallikrein concentration, and epithelial growth factor receptor (EGFR) phosphorylation compared with controls. FXII deficiency or neutralisation also reduced Akt1 and Erk1/2 phosphorylation and decreased expression and levels of active matrix metalloproteinase (Mmp)-2 and Mmp-9. The findings suggest that FXII, kallikrein, ADAM-17, and EGFR are important molecular mediators by which AngII induces aneurysm in apolipoprotein E-deficient mice. This could be a novel pathway to target in the design of drugs to limit AAA progression.
Collapse
|
15
|
Palau V, Riera M, Duran X, Valdivielso JM, Betriu A, Fernández E, Pascual J, Soler MJ. Circulating ADAMs are associated with renal and cardiovascular outcomes in chronic kidney disease patients. Nephrol Dial Transplant 2020; 35:130-138. [PMID: 30102333 DOI: 10.1093/ndt/gfy240] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/11/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND A disintegrin and metalloproteinase (ADAM) 17, also known as tumour necrosis factor α-converting enzyme (TACE), is a metalloproteinase that releases the ectodomains of most growth factors, cytokines, receptors and enzymes and has been associated with the presence of chronic kidney disease (CKD) and cardiovascular (CV) disease. The role of circulating ADAMs in the progression of renal function and CV events in CKD patients is unknown. METHODS A total of 2570 subjects from an observational and multicentre study with CKD Stages 3-5, CKD Stage 5D and controls without any history of CV disease were studied. Circulating ADAM activity was assessed using a fluorometric technique. Progression of renal disease was defined as a 30% increase in serum creatinine or dialysis requirement after 24 months of follow-up. CV outcomes were assessed after 48 months of follow-up. RESULTS Patients with advanced CKD had higher ADAM activity as compared with patients with moderate CKD or controls. Male patients with progression of CKD had higher ADAM levels at baseline compared with patients with stable renal function {22.19 relative fluorescence units/μL/h [95% confidence interval (CI) 11.22-37.32] versus 12.15 (7.02-21.50)}. After multivariate adjustment, higher ADAM activity was identified as a risk factor for progression of CKD in male patients [30% increase in the creatinine odds ratio (OR) 2.72 (95% CI 1.58-4.68), P < 0.001; dialysis requirement OR 3.00 (95% CI 1.65-5.46), P < 0.001; dialysis requirement or 30% increase in the creatinine OR 3.15 (95% CI 2.06-4.81), P < 0.001]. ADAM activity was also identified as an independent risk factor for CV events [hazard ratio (HR) 1.68 (95% CI 1.20-2.36), P = 0.003]. CONCLUSIONS High ADAMs activity levels are independently associated with CKD progression in males and with CV events in CKD patients.
Collapse
Affiliation(s)
- Vanesa Palau
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain
| | - Marta Riera
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain
| | - Xavier Duran
- Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain
| | - José Manuel Valdivielso
- Unit for Detection and Treatment of Atherothrombotic Diseases, Experimental Nephrology Laboratory, Arnau de Vilanova University Hospital, Biomedical Research Institute of Lleida, Lleida, Spain
| | - Angels Betriu
- Unit for Detection and Treatment of Atherothrombotic Diseases, Experimental Nephrology Laboratory, Arnau de Vilanova University Hospital, Biomedical Research Institute of Lleida, Lleida, Spain
| | - Elvira Fernández
- Unit for Detection and Treatment of Atherothrombotic Diseases, Experimental Nephrology Laboratory, Arnau de Vilanova University Hospital, Biomedical Research Institute of Lleida, Lleida, Spain
| | - Julio Pascual
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain
| | - Maria José Soler
- Department of Nephrology, Hospital del Mar-Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain
| |
Collapse
|
16
|
de Queiroz TM, Lakkappa N, Lazartigues E. ADAM17-Mediated Shedding of Inflammatory Cytokines in Hypertension. Front Pharmacol 2020; 11:1154. [PMID: 32848763 PMCID: PMC7403492 DOI: 10.3389/fphar.2020.01154] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 07/15/2020] [Indexed: 12/12/2022] Open
Abstract
The increase of Angiontesin-II (Ang-II), one of the key peptides of the renin-angiotensin system (RAS), and its binding to the Ang-II type 1 receptor (AT1R) during hypertension is a crucial mechanism leading to AD\AM17 activation. Among the reported membrane anchored proteins cleaved by ADAM17, immunological cytokines (TNF-α, IFN-γ, TGF-β, IL-4, IL-10, IL-13, IL-6, FKN) are the major class of substrates, modulation of which triggers inflammation. The rise in ADAM17 levels has both central and peripheral implications in inflammation-mediated hypertension. This narrative review provides an overview of the role of ADAM17, with a special focus on its cellular regulation on neuronal and peripheral inflammation-mediated hypertension. Finally, it highlights the importance of ADAM17 with regards to the biology of inflammatory cytokines and their roles in hypertension.
Collapse
Affiliation(s)
- Thyago M. de Queiroz
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco - UFPE, Vitória de Santo Antão, Brazil
| | - Navya Lakkappa
- Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, United States
- Southeast Louisiana Veterans Health Care System, New Orleans, LA, United States
| | - Eric Lazartigues
- Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, United States
- Southeast Louisiana Veterans Health Care System, New Orleans, LA, United States
| |
Collapse
|
17
|
Banu N, Panikar SS, Leal LR, Leal AR. Protective role of ACE2 and its downregulation in SARS-CoV-2 infection leading to Macrophage Activation Syndrome: Therapeutic implications. Life Sci 2020; 256:117905. [PMID: 32504757 PMCID: PMC7832382 DOI: 10.1016/j.lfs.2020.117905] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/25/2020] [Accepted: 05/30/2020] [Indexed: 02/06/2023]
Abstract
In light of the outbreak of the 2019 novel coronavirus disease (COVID-19), the international scientific community has joined forces to develop effective treatment strategies. The Angiotensin-Converting Enzyme (ACE) 2, is an essential receptor for cell fusion and engulfs the SARS coronavirus infections. ACE2 plays an important physiological role, practically in all the organs and systems. Also, ACE2 exerts protective functions in various models of pathologies with acute and chronic inflammation. While ACE2 downregulation by SARS-CoV-2 spike protein leads to an overactivation of Angiotensin (Ang) II/AT1R axis and the deleterious effects of Ang II may explain the multiorgan dysfunction seen in patients. Specifically, the role of Ang II leading to the appearance of Macrophage Activation Syndrome (MAS) and the cytokine storm in COVID-19 is discussed below. In this review, we summarized the latest research progress in the strategies of treatments that mainly focus on reducing the Ang II-induced deleterious effects rather than attenuating the virus replication. Protective role of ACE2 in the organs and system Downregulation of ACE2 expression by SARS-CoV-2 leads to Ang II-induced organ damage. The appearance of MAS in COVID-19 patient Suggested treatment to diminish the deleterious effect of Ang II or appearance of MAS
Collapse
Affiliation(s)
- Nehla Banu
- Instituto de Enfermedades Crónico-Degenerativas, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Sandeep Surendra Panikar
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autonoma de México (UNAM), Apartado Postal 1-1010, Queretaro, Queretaro 76000, Mexico
| | - Lizbeth Riera Leal
- Hospital General Regional número 45, Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, Mexico
| | - Annie Riera Leal
- UC DAVIS Institute for Regenerative Cure, Department of Dermatology, University of California, 2921 Stockton Blvd, Rm 1630, 95817 Sacramento, CA, USA.
| |
Collapse
|
18
|
Peypoch O, Paüls-Vergés F, Vázquez-Santiago M, Dilme J, Romero J, Giner J, Plaza V, Escudero JR, Soria JM, Camacho M, Sabater-Lleal M. The TAGA Study: A Study of Factors Determining Aortic Diameter in Families at High Risk of Abdominal Aortic Aneurysm Reveal Two New Candidate Genes. J Clin Med 2020; 9:jcm9041242. [PMID: 32344696 PMCID: PMC7231034 DOI: 10.3390/jcm9041242] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/17/2020] [Accepted: 04/21/2020] [Indexed: 01/11/2023] Open
Abstract
A variety of disorders are known to be related with aortic geometry, among them abdominal aortic aneurysm (AAA). This work aims to present the main determinants of abdominal aortic diameter in a new cohort of families at high risk of AAA. The Triple-A Genomic Analysis (TAGA) study comprises 407 individuals related in 12 families. Each family was collected through a proband with AAA. We calculated heritability and genetic correlations between abdominal aortic diameter and clinical parameters. A genome-wide linkage scan was performed based on 4.6 million variants. A predictive model was calculated with conditional forest. Heritability of the abdominal aortic diameter was 34%. Old age, male sex, higher height, weight, creatinine levels in serum, and better lung capacity were the best predictors of aortic diameter. Linkage analyses suggested the implication of Epidermal Growth Factor Receptor (EGFR) and Betacellulin (BTC) genes with aortic diameter. This is the first study to evaluate genetic components of variation of the aortic diameter in a population of AAA high-risk individuals. These results reveal EGFR, a gene that had been previously implicated in AAA, as a determinant of aortic diameter variation in healthy genetically enriched individuals, and might indicate that a common genetic background could determine the diameter of the aorta and future risk of AAA.
Collapse
Affiliation(s)
- Olga Peypoch
- Servicios Mancomunados de Angiología, Cirugía Vascular y Endovascular, Hospitales de la Santa Creu i Sant Pau/Dos de Mayo, 08025 Barcelona, Spain; (O.P.); (J.D.); (J.R.); (J.R.E.)
- Genomics of Complex Diseases, Research Institute of Hospital de la Santa Creu i Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain; (F.P.-V.); (M.V.-S.); (J.M.S.); (M.C.)
- Universitat Autonoma de Barcelona, (IIB Sant Pau), 08025 Barcelona, Spain; (J.G.); (V.P.)
| | - Ferran Paüls-Vergés
- Genomics of Complex Diseases, Research Institute of Hospital de la Santa Creu i Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain; (F.P.-V.); (M.V.-S.); (J.M.S.); (M.C.)
| | - Miquel Vázquez-Santiago
- Genomics of Complex Diseases, Research Institute of Hospital de la Santa Creu i Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain; (F.P.-V.); (M.V.-S.); (J.M.S.); (M.C.)
- ISGlobal, Hospital Clínic-Universitat de Barcelona, 08036 Barcelona, Spain
| | - Jaime Dilme
- Servicios Mancomunados de Angiología, Cirugía Vascular y Endovascular, Hospitales de la Santa Creu i Sant Pau/Dos de Mayo, 08025 Barcelona, Spain; (O.P.); (J.D.); (J.R.); (J.R.E.)
- Genomics of Complex Diseases, Research Institute of Hospital de la Santa Creu i Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain; (F.P.-V.); (M.V.-S.); (J.M.S.); (M.C.)
- Universitat Autonoma de Barcelona, (IIB Sant Pau), 08025 Barcelona, Spain; (J.G.); (V.P.)
| | - Jose Romero
- Servicios Mancomunados de Angiología, Cirugía Vascular y Endovascular, Hospitales de la Santa Creu i Sant Pau/Dos de Mayo, 08025 Barcelona, Spain; (O.P.); (J.D.); (J.R.); (J.R.E.)
- Genomics of Complex Diseases, Research Institute of Hospital de la Santa Creu i Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain; (F.P.-V.); (M.V.-S.); (J.M.S.); (M.C.)
- Universitat Autonoma de Barcelona, (IIB Sant Pau), 08025 Barcelona, Spain; (J.G.); (V.P.)
| | - Jordi Giner
- Universitat Autonoma de Barcelona, (IIB Sant Pau), 08025 Barcelona, Spain; (J.G.); (V.P.)
- Department of Respiratory Medicine, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
| | - Vicente Plaza
- Universitat Autonoma de Barcelona, (IIB Sant Pau), 08025 Barcelona, Spain; (J.G.); (V.P.)
- Department of Respiratory Medicine, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
| | - Jose Roman Escudero
- Servicios Mancomunados de Angiología, Cirugía Vascular y Endovascular, Hospitales de la Santa Creu i Sant Pau/Dos de Mayo, 08025 Barcelona, Spain; (O.P.); (J.D.); (J.R.); (J.R.E.)
- Genomics of Complex Diseases, Research Institute of Hospital de la Santa Creu i Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain; (F.P.-V.); (M.V.-S.); (J.M.S.); (M.C.)
- Universitat Autonoma de Barcelona, (IIB Sant Pau), 08025 Barcelona, Spain; (J.G.); (V.P.)
| | - Jose Manuel Soria
- Genomics of Complex Diseases, Research Institute of Hospital de la Santa Creu i Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain; (F.P.-V.); (M.V.-S.); (J.M.S.); (M.C.)
| | - Mercedes Camacho
- Genomics of Complex Diseases, Research Institute of Hospital de la Santa Creu i Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain; (F.P.-V.); (M.V.-S.); (J.M.S.); (M.C.)
- Angiology, Vascular Biology and Inflammation Laboratory, Research Institute of Hospital de la Santa Creu i Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain
| | - Maria Sabater-Lleal
- Genomics of Complex Diseases, Research Institute of Hospital de la Santa Creu i Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain; (F.P.-V.); (M.V.-S.); (J.M.S.); (M.C.)
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 17176 Stockholm, Sweden
- Correspondence: ; Tel.: +93-291-9000 (ext. 8167)
| |
Collapse
|
19
|
Weskamp G, Tüshaus J, Li D, Feederle R, Maretzky T, Swendemann S, Falck-Pedersen E, McIlwain DR, Mak TW, Salmon JE, Lichtenthaler SF, Blobel CP. ADAM17 stabilizes its interacting partner inactive Rhomboid 2 (iRhom2) but not inactive Rhomboid 1 (iRhom1). J Biol Chem 2020; 295:4350-4358. [PMID: 32060096 PMCID: PMC7105298 DOI: 10.1074/jbc.ra119.011136] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 02/04/2020] [Indexed: 11/06/2022] Open
Abstract
The metalloprotease ADAM17 (a disintegrin and metalloprotease 17) is a key regulator of tumor necrosis factor α (TNFα), interleukin 6 receptor (IL-6R), and epidermal growth factor receptor (EGFR) signaling. ADAM17 maturation and function depend on the seven-membrane-spanning inactive rhomboid-like proteins 1 and 2 (iRhom1/2 or Rhbdf1/2). Most studies to date have focused on overexpressed iRhom1 and -2, so only little is known about the properties of the endogenous proteins. Here, we show that endogenous iRhom1 and -2 can be cell surface-biotinylated on mouse embryonic fibroblasts (mEFs), revealing that endogenous iRhom1 and -2 proteins are present on the cell surface and that iRhom2 also is present on the surface of lipopolysaccharide-stimulated primary bone marrow-derived macrophages. Interestingly, very little, if any, iRhom2 was detectable in mEFs or bone marrow-derived macrophages lacking ADAM17, suggesting that iRhom2 is stabilized by ADAM17. By contrast, the levels of iRhom1 were slightly increased in the absence of ADAM17 in mEFs, indicating that its stability does not depend on ADAM17. These findings support a model in which iRhom2 and ADAM17 are obligate binding partners and indicate that iRhom2 stability requires the presence of ADAM17, whereas iRhom1 is stable in the absence of ADAM17.
Collapse
Affiliation(s)
- Gisela Weskamp
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York 10021
| | - Johanna Tüshaus
- Institute for Advanced Study, Technical University Munich, 85748 Garching, Germany; Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University Munich, 81675 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Daniel Li
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York 10021
| | - Regina Feederle
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Institute for Diabetes and Obesity, Monoclonal Antibody Core Facility, Helmholtz Zentrum Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Thorsten Maretzky
- Inflammation Program and Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242
| | - Steven Swendemann
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York 10021
| | - Erik Falck-Pedersen
- Department of Biochemistry, Cellular and Molecular Biology, Weill Cornell Medicine, New York, New York 10021
| | - David R McIlwain
- Baxter Laboratory in Stem Cell Biology, Department of Microbiology and Immunology, Stanford University, Stanford, California 94305
| | - Tak W Mak
- Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, University Health Network, Toronto, Ontario M5G 2M9, Canada
| | - Jane E Salmon
- Autoimmunity and Inflammation Program, Hospital for Special Surgery, New York, New York 10021; Department of Medicine, Weill Cornell Medicine, New York, New York 10021
| | - Stefan F Lichtenthaler
- Institute for Advanced Study, Technical University Munich, 85748 Garching, Germany; Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University Munich, 81675 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Carl P Blobel
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, New York 10021; Institute for Advanced Study, Technical University Munich, 85748 Garching, Germany; Department of Medicine, Weill Cornell Medicine, New York, New York 10021; Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, New York 10021.
| |
Collapse
|
20
|
The renin-angiotensin system in the arcuate nucleus controls resting metabolic rate. Curr Opin Nephrol Hypertens 2020; 28:120-127. [PMID: 30531199 DOI: 10.1097/mnh.0000000000000477] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Obesity represents the primary challenge to improving cardiovascular health, and suppression of resting metabolic rate (RMR) is implicated in the maintenance of obesity. Increasing evidence supports a major role for the renin-angiotensin system (RAS) within the brain in the control of RMR. RECENT FINDINGS The angiotensin II (ANG) Agtr1a receptor colocalizes with the leptin receptor (Lepr) primarily within cells of the arcuate nucleus (ARC) of the hypothalamus that also express Agouti-related peptide (Agrp). This sub-population of Agtr1a receptors is required for stimulation of thermogenic sympathetic nervous activity and RMR, but not the suppression of food intake or increasing blood pressure, in response to various stimuli including high-fat diet, deoxycorticosterone acetate and salt, and leptin. Agtr1a is localized to a specific subset (SST3) of Agrp neurons within the ARC. SUMMARY The RAS within the ARC is implicated specifically in RMR control, primarily through Agtr1a localized to the SST3 subset of Agrp neurons. Ongoing research is focused on understanding the unique anatomical projections, neurotransmitter utilization, and signal transduction pathways of Agtr1a within this subset of neurons. Understanding these projections and molecular mechanisms may identify therapeutic targets for RMR and thus obesity, independent of blood pressure and appetite.
Collapse
|
21
|
Norton CE, Sheak JR, Yan S, Weise-Cross L, Jernigan NL, Walker BR, Resta TC. Augmented Pulmonary Vasoconstrictor Reactivity after Chronic Hypoxia Requires Src Kinase and Epidermal Growth Factor Receptor Signaling. Am J Respir Cell Mol Biol 2020; 62:61-73. [PMID: 31264901 PMCID: PMC6938133 DOI: 10.1165/rcmb.2018-0106oc] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 06/28/2019] [Indexed: 12/20/2022] Open
Abstract
Chronic hypoxia augments pressure- and agonist-induced pulmonary vasoconstriction through myofilament calcium sensitization. NADPH oxidases contribute to the development of pulmonary hypertension, and both epidermal growth factor receptor and Src kinases can regulate NADPH oxidase. We tested the hypothesis that Src-epidermal growth factor receptor (EGFR) signaling mediates enhanced vasoconstrictor sensitivity after chronic hypoxia through NADPH oxidase-derived superoxide generation. Protocols employed pharmacological inhibitors in isolated, pressurized rat pulmonary arteries to examine the contribution of a variety of signaling moieties to enhanced vascular tone after chronic hypoxia. Superoxide generation in pulmonary arterial smooth muscle cells was assessed using the fluorescent indicator dihydroethidium. Indices of pulmonary hypertension were measured in rats treated with the EGFR inhibitor gefitinib. Inhibition of NADPH oxidase, Rac1 (Ras-related C3 botulinum toxin substrate 1), and EGFR abolished pressure-induced pulmonary arterial tone and endothelin-1 (ET-1)-dependent calcium sensitization and vasoconstriction after chronic hypoxia. Consistently, chronic hypoxia augmented ET-1-induced superoxide production through EGFR signaling, and rats treated chronically with gefitinib displayed reduced right ventricular pressure and diminished arterial remodeling. Src kinases were also activated by ET-1 after chronic hypoxia and contributed to enhanced basal arterial tone and vasoconstriction in response to ET-1. A role for matrix metalloproteinase 2 to mediate Src-dependent EGFR activation is further supported by our findings. Our studies support a novel role for an Src kinase-EGFR-NADPH oxidase signaling axis to mediate enhanced pulmonary vascular smooth muscle Ca2+ sensitization, vasoconstriction, and pulmonary hypertension after chronic hypoxia.
Collapse
Affiliation(s)
- Charles E Norton
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Joshua R Sheak
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Simin Yan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Laura Weise-Cross
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Nikki L Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Benjimen R Walker
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Thomas C Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| |
Collapse
|
22
|
A rare loss-of-function variant of ADAM17 is associated with late-onset familial Alzheimer disease. Mol Psychiatry 2020; 25:629-639. [PMID: 29988083 PMCID: PMC7042727 DOI: 10.1038/s41380-018-0091-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 04/05/2018] [Accepted: 04/11/2018] [Indexed: 01/13/2023]
Abstract
Common variants of about 20 genes contributing to AD risk have so far been identified through genome-wide association studies (GWAS). However, there is still a large proportion of heritability that might be explained by rare but functionally important variants. One of the so far identified genes with rare AD causing variants is ADAM10. Using whole-genome sequencing we now identified a single rare nonsynonymous variant (SNV) rs142946965 [p.R215I] in ADAM17 co-segregating with an autosomal-dominant pattern of late-onset AD in one family. Subsequent genotyping and analysis of available whole-exome sequencing data of additional case/control samples from Germany, UK, and USA identified five variant carriers among AD patients only. The mutation inhibits pro-protein cleavage and the formation of the active enzyme, thus leading to loss-of-function of ADAM17 alpha-secretase. Further, we identified a strong negative correlation between ADAM17 and APP gene expression in human brain and present in vitro evidence that ADAM17 negatively controls the expression of APP. As a consequence, p.R215I mutation of ADAM17 leads to elevated Aß formation in vitro. Together our data supports a causative association of the identified ADAM17 variant in the pathogenesis of AD.
Collapse
|
23
|
Preston KJ, Rom I, Vrakas C, Landesberg G, Etwebi Z, Muraoka S, Autieri M, Eguchi S, Scalia R. Postprandial activation of leukocyte-endothelium interaction by fatty acids in the visceral adipose tissue microcirculation. FASEB J 2019; 33:11993-12007. [PMID: 31393790 DOI: 10.1096/fj.201802637rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
High-fat diet (HFD)-induced obesity is associated with accumulation of inflammatory cells predominantly in visceral adipose depots [visceral adipose tissue (VAT)] rather than in subcutaneous ones [subcutaneous adipose tissue (SAT)]. The cellular and molecular mechanisms responsible for this phenotypic difference remain poorly understood. Controversy also exists on the overall impact that adipose tissue inflammation has on metabolic health in diet-induced obesity. The endothelium of the microcirculation regulates both the transport of lipids and the trafficking of leukocytes into organ tissue. We hypothesized that the VAT and SAT microcirculations respond differently to postprandial processing of dietary fat. We also tested whether inhibition of endothelial postprandial responses to high-fat meals (HFMs) preserves metabolic health in chronic obesity. We demonstrate that administration of a single HFM or ad libitum access to a HFD for 24 h quickly induces a transient P-selectin-dependent inflammatory phenotype in the VAT but not the SAT microcirculation of lean wild-type mice. Studies in P-selectin-deficient mice confirmed a mechanistic role for P-selectin in the initiation of leukocyte trafficking, myeloperoxidase accumulation, and acute reduction in adiponectin mRNA expression by HFMs. Despite reduced VAT inflammation in response to HFMs, P-selectin-deficient mice still developed glucose intolerance and insulin resistance when chronically fed an HFD. Our data uncover a novel nutrient-sensing role of the vascular endothelium that instigates postprandial VAT inflammation. They also demonstrate that inhibition of this transient postprandial inflammatory response fails to correct metabolic dysfunction in diet-induced obesity.-Preston, K. J., Rom, I., Vrakas, C., Landesberg, G., Etwebe, Z., Muraoka, S., Autieri, M., Eguchi, S., Scalia, R. Postprandial activation of leukocyte-endothelium interaction by fatty acids in the visceral adipose tissue microcirculation.
Collapse
Affiliation(s)
| | - Inna Rom
- Cardiovascular Research Center and
| | | | | | | | | | - Michael Autieri
- Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Satoru Eguchi
- Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Rosario Scalia
- Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
24
|
Palau V, Pascual J, Soler MJ, Riera M. Role of ADAM17 in kidney disease. Am J Physiol Renal Physiol 2019; 317:F333-F342. [DOI: 10.1152/ajprenal.00625.2018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
It is known that the renin-angiotensin system plays a major role in the pathophysiology of cardiovascular disease and renal injury. Within the renin-angiotensin system, angiotensin-converting enzyme 2 (ACE2) cleaves ANG II to generate ANG(1–7) peptide, which counteracts the adverse effects of ANG II accumulation. ACE2 can undergo cleavage or shedding to release the catalytically active ectodomain into the circulation by a disintegrin and metalloprotease (ADAM)17, also known as TNF-α-converting enzyme. ADAM17 is involved in many pathological processes such as cancer, inflammatory diseases, neurological diseases, cardiovascular diseases, atherosclerosis, diabetes, and hypertension. Clinical and experimental studies have shown that ADAM17 is involved in chronic kidney disease (CKD) with a proinflammatory and profibrotic role, suggesting that it could be an important mediator of CKD progression. ADAM17 inhibition attenuates fibrosis and inflammation, suggesting that its inhibition may be a possible new valuable therapeutic tool in fibrotic kidney disease treatment. In addition, in renal disease, some experimental studies have demonstrated that ADAM17 is differently expressed in the kidney. Thus, ADAM17 is highly expressed in distal renal tubules and increased in the whole kidney in diabetic models. In this article, we will review the role of ADAM17 under physiological and pathological conditions. We will mainly focus on the importance of ADAM17 in the context of CKD.
Collapse
Affiliation(s)
- Vanesa Palau
- Department of Nephrology, Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Julio Pascual
- Department of Nephrology, Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Maria José Soler
- Department of Nephrology, Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Marta Riera
- Department of Nephrology, Hospital del Mar Medical Research Institute, Barcelona, Spain
| |
Collapse
|
25
|
Dougherty U, Mustafi R, Haider HI, Khalil A, Souris JS, Joseph L, Hart J, Konda VJ, Zhang W, Pekow J, Li YC, Bissonnette M. Losartan and Vitamin D Inhibit Colonic Tumor Development in a Conditional Apc-Deleted Mouse Model of Sporadic Colon Cancer. Cancer Prev Res (Phila) 2019; 12:433-448. [PMID: 31088824 DOI: 10.1158/1940-6207.capr-18-0380] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 04/02/2019] [Accepted: 05/06/2019] [Indexed: 12/20/2022]
Abstract
Colorectal cancer is a leading cause of cancer deaths. The renin-angiotensin system (RAS) is upregulated in colorectal cancer, and epidemiologic studies suggest RAS inhibitors reduce cancer risk. Because vitamin D (VD) receptor negatively regulates renin, we examined anticancer efficacy of VD and losartan (L), an angiotensin receptor blocker. Control Apc+/LoxP mice and tumor-forming Apc+/LoxP Cdx2P-Cre mice were randomized to unsupplemented Western diet (UN), or diets supplemented with VD, L, or VD+L, the latter to assess additive or synergistic effects. At 6 months, mice were killed. Plasma Ca2+, 25(OH)D3, 1α, 25(OH)2D3, renin, and angiotensin II (Ang II) were quantified. Colonic transcripts were assessed by qPCR and proteins by immunostaining and blotting. Cancer incidence and tumor burden were significantly lower in Cre+ VD and Cre+ L, but not in the Cre+ VD+L group. In Apc+/LoxP mice, VD increased plasma 1,25(OH)2D3 and colonic VDR. In Apc+/LoxP-Cdx2P-Cre mice, plasma renin and Ang II, and colonic tumor AT1, AT2, and Cyp27B1 were increased and VDR downregulated. L increased, whereas VD decreased plasma renin and Ang II in Cre+ mice. VD or L inhibited tumor development, while exerting differential effects on plasma VD metabolites and RAS components. We speculate that AT1 is critical for tumor development, whereas RAS suppression plays a key role in VD chemoprevention. When combined with L, VD no longer increases active VD and colonic VDR in Cre- mice nor suppresses renin and Ang II in Cre+ mice, likely contributing to lack of chemopreventive efficacy of the combination.
Collapse
Affiliation(s)
| | - Reba Mustafi
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Haider I Haider
- Department of Medicine, University of Chicago, Chicago, Illinois
| | | | - Jeffrey S Souris
- Department of Radiology, University of Chicago, Chicago, Illinois
| | - Loren Joseph
- Department of Pathology, Beth Israel, Harvard Medical School, Boston, Massachusetts
| | - John Hart
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Vani J Konda
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Wei Zhang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Joel Pekow
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Yan Chun Li
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Marc Bissonnette
- Department of Medicine, University of Chicago, Chicago, Illinois.
| |
Collapse
|
26
|
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: 714] [Impact Index Per Article: 102.0] [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
|
27
|
Evidence That ADAM17 Mediates the Protective Action of CGRP against Angiotensin II-Induced Inflammation in Vascular Smooth Muscle Cells. Mediators Inflamm 2018; 2018:2109352. [PMID: 30046277 PMCID: PMC6038660 DOI: 10.1155/2018/2109352] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/30/2018] [Accepted: 04/15/2018] [Indexed: 12/14/2022] Open
Abstract
Calcitonin gene-related peptide (CGRP) has a potent protective action on the cardiovascular system; however, little is known about the role of CGRP in angiotensin II- (Ang II-) induced inflammation of vascular smooth muscle cells (VSMCs). This study is aimed at determining the anti-inflammatory effect of CGRP in Ang II-treated VSMCs and whether a disintegrin and metalloproteinase 17 (ADAM17) modulates this protective action. Small interference RNA (siRNA) and inhibitors of CGRP, epidermal growth factor receptor (EGFR), and extracellular signal-regulated kinase 1/2 (ERK1/2) were adopted to investigate their effect on Ang II-induced inflammation in VSMCs. Here, we found that CGRP could inhibit inflammation and decrease ADAM17 expression and activation of EGFR and ERK1/2 in VSMCs stimulated with Ang II. Results of siRNA demonstrated that ADAM17 siRNA attenuated Ang II-induced inflammation and up-regulation of activities of EGFR and ERK1/2 in VSMCs. Furthermore, the EGFR-ERK1/2 pathway promoted Ang II-induced VSMC inflammation. In summary, these findings identify the anti-inflammatory effect of CGRP in VSMCs stimulated by Ang II and suggest that ADAM17 is involved in the protective effect of CGRP against Ang II-induced inflammation via the EGFR-ERK1/2 pathway in VSMCs.
Collapse
|
28
|
Ogola B, Zhang Y, Iyer L, Thekkumkara T. 2-Methoxyestradiol causes matrix metalloproteinase 9-mediated transactivation of epidermal growth factor receptor and angiotensin type 1 receptor downregulation in rat aortic smooth muscle cells. Am J Physiol Cell Physiol 2018; 314:C554-C568. [DOI: 10.1152/ajpcell.00152.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Studies have demonstrated the therapeutic potential of estrogen metabolite 2-methoxyestradiol (2ME2) in several cardiovascular disorders, including hypertension. However, the exact mechanism(s) remains unknown. In this study, primary rat aortic smooth muscle cells (RASMCs) were exposed to 2ME2, and angiotensin type 1 receptor (AT1R) expression, function, and associated signaling pathways were evaluated. In RASMCs, 2ME2 downregulated AT1R expression in a concentration- and time-dependent manner, which was correlated with reduced mRNA expression. The 2ME2 effect was through G protein-coupled receptor 30 (GPR30) that inhibits second messenger cAMP. Moreover, 2ME2 exposure phosphorylated ERK1/2 that was sensitive to MEK inhibitor PD98059. Selective epidermal growth factor receptor (EGFR) inhibitor AG1478 blocked 2ME2-induced EGFR transactivation and attenuated subsequent phosphorylation of ERK1/2 preventing AT1R downregulation. The transactivation was dependent on 2ME2-induced release of matrix metalloproteinase 9 (MMP9) and epidermal growth factor demonstrated by ELISA. Furthermore, transfection with small interfering (si) RNA targeting MMP9 impeded ERK1/2 activation and AT1R downregulation in response to 2ME2 and G1 stimulation. Interestingly, under similar conditions, stimulation of GPR30 with the selective agonist G1 elicited similar signaling pathways and downregulated the AT1R expression that was reversed by GPR30 antagonist G15. Furthermore, 2ME2 and G1 inhibited angiotensin II (ANG II) induced Ca2+ release, a response consistent with AT1R downregulation. Collectively, our study demonstrates for the first time that 2ME2 binding to GPR30 induces MMP9 specific transactivation of EGFR that mediates ERK1/2-dependent downregulation of AT1R in RASMCs. The study provides critical insights into the newly discovered role and signaling pathways of 2ME2 in the regulation of AT1R in vascular cells and its potential to be developed as a therapeutic agent that ameliorates hypertension.
Collapse
Affiliation(s)
- Benard Ogola
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Amarillo, Texas
| | - Yong Zhang
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Amarillo, Texas
| | - Laxmi Iyer
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Amarillo, Texas
| | - Thomas Thekkumkara
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Amarillo, Texas
| |
Collapse
|
29
|
Eguchi S, Kawai T, Scalia R, Rizzo V. Understanding Angiotensin II Type 1 Receptor Signaling in Vascular Pathophysiology. Hypertension 2018; 71:804-810. [PMID: 29581215 DOI: 10.1161/hypertensionaha.118.10266] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Satoru Eguchi
- From the Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA.
| | - Tatsuo Kawai
- From the Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Rosario Scalia
- From the Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Victor Rizzo
- From the Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| |
Collapse
|
30
|
Schreier B, Hünerberg M, Mildenberger S, Rabe S, Bethmann D, Wickenhauser C, Gekle M. Deletion of the EGF receptor in vascular smooth muscle cells prevents chronic angiotensin II-induced arterial wall stiffening and media thickening. Acta Physiol (Oxf) 2018; 222. [PMID: 29152859 DOI: 10.1111/apha.12996] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 11/09/2017] [Accepted: 11/10/2017] [Indexed: 01/13/2023]
Abstract
AIM In vivo vascular smooth muscle cell (VSMC) EGF receptor (EGFR) contributes to acute angiotensin II (AII) effects on vascular tone and blood pressure. The ubiquitously expressed EGFR has been implicated in vascular remodelling preceding end-organ damage by pharmacological inhibition, and AII signalling in cultured vascular cells is partly EGFR-dependent. However, the role of VSMC-EGFR in vivo during AII-induced pathophysiological processes is not known. METHODS This study assesses the in vivo relevance of VSMC-EGFR during chronic AII challenge without further stressors, using a mouse model with inducible, VSMC-specific EGFR knock out (VSMC-EGFR-KO). In these mice functional and structural vascular, renal and cardiac effects or biomarkers were investigated in vivo and ex vivo. RESULTS Vascular smooth muscle cell-EGFR-KO prevented AII-induced media hypertrophy of mesenteric arteries, renal arterioles and the aorta, VSMC ERK1/2-phosphorylation as well as the impairment of vascular compliance. Furthermore, induction of vascular fibrosis, creatinineamia, renal interstitial fibrosis as well as the increase in fractional water excretion was prevented. AII-induced increase in systolic blood pressure was mitigated. By contrast, endothelial dysfunction, induction of vascular inflammatory marker mRNA and albuminuria were not inhibited. Cardiac and cardiomyocyte hypertrophy were also not prevented by VSMC-EGFR-KO. CONCLUSION Vascular smooth muscle cell-EGFRs are relevant for pathological AII action in vivo. Our data show in vivo and ex vivo the necessity of VSMC-EGFR for AII-induced structural and functional vascular remodelling, not including endothelial dysfunction. Hereby, VSMC-EGFR gains importance for complete AII-induced renal end-organ damage succeeding vascular remodelling.
Collapse
Affiliation(s)
- B. Schreier
- Julius-Bernstein-Institute of Physiology; Martin Luther University Halle-Wittenberg; Halle Germany
| | - M. Hünerberg
- Julius-Bernstein-Institute of Physiology; Martin Luther University Halle-Wittenberg; Halle Germany
| | - S. Mildenberger
- Julius-Bernstein-Institute of Physiology; Martin Luther University Halle-Wittenberg; Halle Germany
| | - S. Rabe
- Julius-Bernstein-Institute of Physiology; Martin Luther University Halle-Wittenberg; Halle Germany
| | - D. Bethmann
- Institute of Pathology; Martin Luther University Halle-Wittenberg; Halle Germany
| | - C. Wickenhauser
- Institute of Pathology; Martin Luther University Halle-Wittenberg; Halle Germany
| | - M. Gekle
- Julius-Bernstein-Institute of Physiology; Martin Luther University Halle-Wittenberg; Halle Germany
| |
Collapse
|
31
|
Sperm-egg fusion disorder in a Chinese male patient was associated with a rare ADAM20 variant. Oncotarget 2017; 9:2086-2091. [PMID: 29416755 PMCID: PMC5788623 DOI: 10.18632/oncotarget.23331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 12/13/2017] [Indexed: 11/27/2022] Open
Abstract
We report here a 28-year-old male with infertility. No abnormality was found in his semen examination. The couple achieved a successful pregnancy under the help of intracytoplasmic sperm injection during which we found that sperm could enter the zona pellucida, but could not fuse with the egg within the short insemination period. We then performed whole-exome sequencing technology on this patient and found a rare variant (c.641A>C:p.D214A) in ADAM20, which encoded a disintegrin and metalloprotease 20 protein. To further verify the pathogenicity of this variant, we analyzed ADAM20 protein expression in spermatozoa by immunostaining analysis, which showed a mis-localization of ADAM20 in the patient's spermatozoa. Therefore, we concluded that mutation in ADAM20 may be associated with sperm-egg fusion disorder in this patient.
Collapse
|
32
|
Lin N, Ji Z, Huang C. Smad7 alleviates glomerular mesangial cell proliferation via the ROS-NF-κB pathway. Exp Cell Res 2017; 361:210-216. [PMID: 28988741 DOI: 10.1016/j.yexcr.2017.10.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 09/11/2017] [Accepted: 10/03/2017] [Indexed: 12/01/2022]
Abstract
OBJECTIVE The aim of this study was to demonstrate that altered gene expression of Smad7regulated NF-κB expression and ROS production on Ang II (Angiotensin II)-induced rat glomerular mesangial cell (GMC) proliferation. METHODS pAdTrack-CMV-Smad7 was transduced into rat GMC by adeno-transduction using an ADV (adenovirus)-mediated vector in vivo. Diphenylene iodonium chloride (DPI) pre-treated GMC, and blocked ROS generation as determined by DCFH-DA method. Altered expressions of IκBα and p65 were monitored by Western blot analysis and immunofluorescence. GMC proliferation was tested by the Cell Counting Kit-8 assay. Apoptosis of GMC was detected by flow cytometric analysis. RESULTS Over-expression of Smad7 dampened the ability of Ang II to promote ROS synthesis and inhibited the ability of Ang II to decrease functional expression of IκBα. Moreover, Smad7 increased nuclear IκBα expression. Smad7 did not significantly influence the capacity of Ang II to increase protein expression of NF-κB p65. However, immunofluorescence analysis showed that Smad7 reduced nuclear NF-κB p65 level. Further, over-expression of Smad7 promoted GMC apoptosis by inhibiting NF-κB activation, which alleviated the Ang II-promoted proliferation of GMC. CONCLUSIONS Smad7 influenced NF-κB expression by regulating ROS generation, and induced GMC apoptosis to counter the Ang II-promoted proliferation.
Collapse
Affiliation(s)
- Nana Lin
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zequan Ji
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Cuiwen Huang
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
33
|
Kawai T, Takayanagi T, Forrester SJ, Preston KJ, Obama T, Tsuji T, Kobayashi T, Boyer MJ, Cooper HA, Kwok HF, Hashimoto T, Scalia R, Rizzo V, Eguchi S. Vascular ADAM17 (a Disintegrin and Metalloproteinase Domain 17) Is Required for Angiotensin II/β-Aminopropionitrile-Induced Abdominal Aortic Aneurysm. Hypertension 2017; 70:959-963. [PMID: 28947615 DOI: 10.1161/hypertensionaha.117.09822] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 06/22/2017] [Accepted: 08/31/2017] [Indexed: 01/05/2023]
Abstract
Angiotensin II (AngII)-activated epidermal growth factor receptor has been implicated in abdominal aortic aneurysm (AAA) development. In vascular smooth muscle cells (VSMCs), AngII activates epidermal growth factor receptor via a metalloproteinase, ADAM17 (a disintegrin and metalloproteinase domain 17). We hypothesized that AngII-dependent AAA development would be prevented in mice lacking ADAM17 in VSMCs. To test this concept, control and VSMC ADAM17-deficient mice were cotreated with AngII and a lysyl oxidase inhibitor, β-aminopropionitrile, to induce AAA. We found that 52.4% of control mice did not survive because of aortic rupture. All other surviving control mice developed AAA and demonstrated enhanced expression of ADAM17 in the AAA lesions. In contrast, all AngII and β-aminopropionitrile-treated VSMC ADAM17-deficient mice survived and showed reduction in external/internal diameters (51%/28%, respectively). VSMC ADAM17 deficiency was associated with lack of epidermal growth factor receptor activation, interleukin-6 induction, endoplasmic reticulum/oxidative stress, and matrix deposition in the abdominal aorta of treated mice. However, both VSMC ADAM17-deficient and control mice treated with AngII and β-aminopropionitrile developed comparable levels of hypertension. Treatment of C57Bl/6 mice with an ADAM17 inhibitory antibody but not with control IgG also prevented AAA development. In conclusion, VSMC ADAM17 silencing or systemic ADAM17 inhibition seems to protect mice from AAA formation. The mechanism seems to involve suppression of epidermal growth factor receptor activation.
Collapse
Affiliation(s)
- Tatsuo Kawai
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Takehiko Takayanagi
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Steven J Forrester
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Kyle J Preston
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Takashi Obama
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Toshiyuki Tsuji
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Tomonori Kobayashi
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Michael J Boyer
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Hannah A Cooper
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Hang Fai Kwok
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Tomoki Hashimoto
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Rosario Scalia
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.)
| | - Victor Rizzo
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.).
| | - Satoru Eguchi
- From the Cardiovascular Research Center, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (T. Kawai, T. Takayanagi, S.J.F., K.J.P., T.O., T. Tsuji, T. Kobayashi, M.J.B., H.A.C., R.S., V.R., S.E.); Faculty of Health Sciences, Macau Special Administrative Region, University of Macau, Taipa (H.F.K.); and Department of Anesthesia and Perioperative Care, University of California, San Francisco (T.H.).
| |
Collapse
|
34
|
Kim S, Yang L, Kim S, Lee RG, Graham MJ, Berliner JA, Lusis AJ, Cai L, Temel RE, Rateri DL, Lee S. Targeting hepatic heparin-binding EGF-like growth factor (HB-EGF) induces anti-hyperlipidemia leading to reduction of angiotensin II-induced aneurysm development. PLoS One 2017; 12:e0182566. [PMID: 28792970 PMCID: PMC5549937 DOI: 10.1371/journal.pone.0182566] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 07/20/2017] [Indexed: 01/02/2023] Open
Abstract
Objective The upregulated expression of heparin binding EGF-like growth factor (HB-EGF) in the vessel and circulation is associated with risk of cardiovascular disease. In this study, we tested the effects of HB-EGF targeting using HB-EGF-specific antisense oligonucleotide (ASO) on the development of aortic aneurysm in a mouse aneurysm model. Approach and results Low-density lipoprotein receptor (LDLR) deficient mice (male, 16 weeks of age) were injected with control and HB-EGF ASOs for 10 weeks. To induce aneurysm, the mice were fed a high fat diet (22% fat, 0.2% cholesterol; w/w) at 5 week point of ASO administration and infused with angiotensin II (AngII, 1,000ng/kg/min) for the last 4 weeks of ASO administration. We confirmed that the HB-EGF ASO administration significantly downregulated HB-EGF expression in multiple tissues including the liver. Importantly, the HB-EGF ASO administration significantly suppressed development of aortic aneurysms including thoracic and abdominal types. Interestingly, the HB-EGF ASO administration induced a remarkable anti-hyperlipidemic effect by suppressing very low density lipoprotein (VLDL) level in the blood. Mechanistically, the HB-EGF targeting suppressed hepatic VLDL secretion rate without changing heparin-releasable plasma triglyceride (TG) hydrolytic activity or fecal neutral cholesterol excretion rate. Conclusion This result suggested that the HB-EGF targeting induced protection against aneurysm development through anti-hyperlipidemic effects. Suppression of hepatic VLDL production process appears to be a key mechanism for the anti-hyperlipidemic effects by the HB-EGF targeting.
Collapse
Affiliation(s)
- Seonwook Kim
- Saha Cardiovascular Research Center at the University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Lihua Yang
- Saha Cardiovascular Research Center at the University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Seongu Kim
- Saha Cardiovascular Research Center at the University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Richard G. Lee
- Cardiovascular Antisense Drug Discovery Group at the Ionis Pharmaceuticals, Inc., Carlsbad, California, United States of America
| | - Mark J. Graham
- Cardiovascular Antisense Drug Discovery Group at the Ionis Pharmaceuticals, Inc., Carlsbad, California, United States of America
| | - Judith A. Berliner
- Department of Medicine-Cardiology, University of California-Los Angeles School of Medicine, Los Angeles, California, United States of America
| | - Aldons J. Lusis
- Department of Medicine-Cardiology, University of California-Los Angeles School of Medicine, Los Angeles, California, United States of America
| | - Lei Cai
- Saha Cardiovascular Research Center at the University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Ryan E. Temel
- Saha Cardiovascular Research Center at the University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- Department of Pharmacology & Nutritional Sciences at the University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Debra L. Rateri
- Saha Cardiovascular Research Center at the University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Sangderk Lee
- Saha Cardiovascular Research Center at the University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- Department of Pharmacology & Nutritional Sciences at the University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
35
|
AT1 receptor signaling pathways in the cardiovascular system. Pharmacol Res 2017; 125:4-13. [PMID: 28527699 DOI: 10.1016/j.phrs.2017.05.008] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 05/10/2017] [Accepted: 05/11/2017] [Indexed: 01/14/2023]
Abstract
The importance of the renin angiotensin aldosterone system in cardiovascular physiology and pathophysiology has been well described whereas the detailed molecular mechanisms remain elusive. The angiotensin II type 1 receptor (AT1 receptor) is one of the key players in the renin angiotensin aldosterone system. The AT1 receptor promotes various intracellular signaling pathways resulting in hypertension, endothelial dysfunction, vascular remodeling and end organ damage. Accumulating evidence shows the complex picture of AT1 receptor-mediated signaling; AT1 receptor-mediated heterotrimeric G protein-dependent signaling, transactivation of growth factor receptors, NADPH oxidase and ROS signaling, G protein-independent signaling, including the β-arrestin signals and interaction with several AT1 receptor interacting proteins. In addition, there is functional cross-talk between the AT1 receptor signaling pathway and other signaling pathways. In this review, we will summarize an up to date overview of essential AT1 receptor signaling events and their functional significances in the cardiovascular system.
Collapse
|
36
|
Xu J, Sriramula S, Xia H, Moreno-Walton L, Culicchia F, Domenig O, Poglitsch M, Lazartigues E. Clinical Relevance and Role of Neuronal AT 1 Receptors in ADAM17-Mediated ACE2 Shedding in Neurogenic Hypertension. Circ Res 2017; 121:43-55. [PMID: 28512108 DOI: 10.1161/circresaha.116.310509] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 05/11/2017] [Accepted: 05/16/2017] [Indexed: 12/16/2022]
Abstract
RATIONALE Neurogenic hypertension is characterized by an increase in sympathetic activity and often resistance to drug treatments. We previously reported that it is also associated with a reduction of angiotensin-converting enzyme type 2 (ACE2) and an increase in a disintegrin and metalloprotease 17 (ADAM17) activity in experimental hypertension. In addition, while multiple cells within the central nervous system have been involved in the development of neurogenic hypertension, the contribution of ADAM17 has not been investigated. OBJECTIVE To assess the clinical relevance of this ADAM17-mediated ACE2 shedding in hypertensive patients and further identify the cell types and signaling pathways involved in this process. METHODS AND RESULTS Using a mass spectrometry-based assay, we identified ACE2 as the main enzyme converting angiotensin II into angiotensin-(1-7) in human cerebrospinal fluid. We also observed an increase in ACE2 activity in the cerebrospinal fluid of hypertensive patients, which was correlated with systolic blood pressure. Moreover, the increased level of tumor necrosis factor-α in those cerebrospinal fluid samples confirmed that ADAM17 was upregulated in the brain of hypertensive patients. To further assess the interaction between brain renin-angiotensin system and ADAM17, we generated mice lacking angiotensin II type 1 receptors specifically on neurons. Our data reveal that despite expression on astrocytes and other cells types in the brain, ADAM17 upregulation during deoxycorticosterone acetate-salt hypertension occurs selectively on neurons, and neuronal angiotensin II type 1 receptors are indispensable to this process. Mechanistically, reactive oxygen species and extracellular signal-regulated kinase were found to mediate ADAM17 activation. CONCLUSIONS Our data demonstrate that angiotensin II type 1 receptors promote ADAM17-mediated ACE2 shedding in the brain of hypertensive patients, leading to a loss in compensatory activity during neurogenic hypertension.
Collapse
Affiliation(s)
- Jiaxi Xu
- From the Department of Pharmacology and Experimental Therapeutics (J.X., S.S., H.X., E.L.), Cardiovascular Center of Excellence (J.X., S.S., H.X., E.L.), Neurosciences Center of Excellence (E.L.), Department of Emergency Medicine (L.M.-W.), and Department of Neurological Surgery (F.C.), Louisiana State University Health Sciences Center, New Orleans, LA; and Attoquant Diagnostics GmbH, Vienna, Austria (O.D., M.P.)
| | - Srinivas Sriramula
- From the Department of Pharmacology and Experimental Therapeutics (J.X., S.S., H.X., E.L.), Cardiovascular Center of Excellence (J.X., S.S., H.X., E.L.), Neurosciences Center of Excellence (E.L.), Department of Emergency Medicine (L.M.-W.), and Department of Neurological Surgery (F.C.), Louisiana State University Health Sciences Center, New Orleans, LA; and Attoquant Diagnostics GmbH, Vienna, Austria (O.D., M.P.)
| | - Huijing Xia
- From the Department of Pharmacology and Experimental Therapeutics (J.X., S.S., H.X., E.L.), Cardiovascular Center of Excellence (J.X., S.S., H.X., E.L.), Neurosciences Center of Excellence (E.L.), Department of Emergency Medicine (L.M.-W.), and Department of Neurological Surgery (F.C.), Louisiana State University Health Sciences Center, New Orleans, LA; and Attoquant Diagnostics GmbH, Vienna, Austria (O.D., M.P.)
| | - Lisa Moreno-Walton
- From the Department of Pharmacology and Experimental Therapeutics (J.X., S.S., H.X., E.L.), Cardiovascular Center of Excellence (J.X., S.S., H.X., E.L.), Neurosciences Center of Excellence (E.L.), Department of Emergency Medicine (L.M.-W.), and Department of Neurological Surgery (F.C.), Louisiana State University Health Sciences Center, New Orleans, LA; and Attoquant Diagnostics GmbH, Vienna, Austria (O.D., M.P.)
| | - Frank Culicchia
- From the Department of Pharmacology and Experimental Therapeutics (J.X., S.S., H.X., E.L.), Cardiovascular Center of Excellence (J.X., S.S., H.X., E.L.), Neurosciences Center of Excellence (E.L.), Department of Emergency Medicine (L.M.-W.), and Department of Neurological Surgery (F.C.), Louisiana State University Health Sciences Center, New Orleans, LA; and Attoquant Diagnostics GmbH, Vienna, Austria (O.D., M.P.)
| | - Oliver Domenig
- From the Department of Pharmacology and Experimental Therapeutics (J.X., S.S., H.X., E.L.), Cardiovascular Center of Excellence (J.X., S.S., H.X., E.L.), Neurosciences Center of Excellence (E.L.), Department of Emergency Medicine (L.M.-W.), and Department of Neurological Surgery (F.C.), Louisiana State University Health Sciences Center, New Orleans, LA; and Attoquant Diagnostics GmbH, Vienna, Austria (O.D., M.P.)
| | - Marko Poglitsch
- From the Department of Pharmacology and Experimental Therapeutics (J.X., S.S., H.X., E.L.), Cardiovascular Center of Excellence (J.X., S.S., H.X., E.L.), Neurosciences Center of Excellence (E.L.), Department of Emergency Medicine (L.M.-W.), and Department of Neurological Surgery (F.C.), Louisiana State University Health Sciences Center, New Orleans, LA; and Attoquant Diagnostics GmbH, Vienna, Austria (O.D., M.P.)
| | - Eric Lazartigues
- From the Department of Pharmacology and Experimental Therapeutics (J.X., S.S., H.X., E.L.), Cardiovascular Center of Excellence (J.X., S.S., H.X., E.L.), Neurosciences Center of Excellence (E.L.), Department of Emergency Medicine (L.M.-W.), and Department of Neurological Surgery (F.C.), Louisiana State University Health Sciences Center, New Orleans, LA; and Attoquant Diagnostics GmbH, Vienna, Austria (O.D., M.P.).
| |
Collapse
|
37
|
Shen M, Morton J, Davidge ST, Kassiri Z. Loss of smooth muscle cell disintegrin and metalloproteinase 17 transiently suppresses angiotensin II-induced hypertension and end-organ damage. J Mol Cell Cardiol 2017; 103:11-21. [DOI: 10.1016/j.yjmcc.2016.12.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 11/08/2016] [Accepted: 12/02/2016] [Indexed: 11/27/2022]
|
38
|
In Vitro Analysis of Hypertensive Signal Transduction: Kinase Activation, Kinase Manipulation, and Physiologic Outputs. Hypertension 2017; 1527:201-211. [DOI: 10.1007/978-1-4939-6625-7_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
39
|
Chodavarapu H, Chhabra KH, Xia H, Shenoy V, Yue X, Lazartigues E. High-fat diet-induced glucose dysregulation is independent of changes in islet ACE2 in mice. Am J Physiol Regul Integr Comp Physiol 2016; 311:R1223-R1233. [PMID: 27806985 DOI: 10.1152/ajpregu.00362.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 10/12/2016] [Accepted: 10/28/2016] [Indexed: 12/14/2022]
Abstract
While restoration of ACE2 activity in the pancreas leads to improvement of glycemia in experimental models of Type 2 diabetes, global deficiency in ACE2 disrupts β-cell function and impairs glucose tolerance in mice, demonstrating the physiological role of ACE2 in glucose homeostasis. Although the contribution of pancreatic ACE2 to glucose regulation has been demonstrated in genetic models of diabetes and in models with overexpression of the renin-angiotensin system (RAS), it is unclear whether islet ACE2 is involved in glycemic control in common models of human Type 2 diabetes. To determine whether diet-induced diabetes deregulates glucose homeostasis via reduction of ACE2 in the pancreatic islets, wild-type (WT) and ACE2 knockout (KO) male mice were fed a high-fat diet (HFD) for 16 wk. ACE2 KO mice were more susceptible than WT mice to HFD-mediated glycemic dysregulation. Islet ACE2 activity and expression of various genes, including ANG II type 1a receptor (mAT1aR) were then assessed. Surprisingly, we observed no change in islet ACE2 activity and expression despite local RAS overactivity, indicated by an upregulation of mAT1aR expression. Despite a predominant expression in islet α-cells, further investigation highlighted a minor role for ACE2 on glucagon expression. Further, pancreatic ACE2 gene therapy improved glycemia in HFD-fed WT mice, leading to enhanced glucose-stimulated insulin secretion, reduced pancreatic ANG II levels, fibrosis, and ADAM17 activity. Altogether, our study demonstrates that HFD feeding increases RAS activity and mediates glycemic dysregulation likely through loss of ACE2 present outside the islets but independently of changes in islet ACE2.
Collapse
Affiliation(s)
- Harshita Chodavarapu
- Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Kavaljit H Chhabra
- Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana.,Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Huijing Xia
- Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Vinayak Shenoy
- Department of Pharmacology, California Health Sciences University, Clovis, California; and
| | - Xinping Yue
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Eric Lazartigues
- Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana;
| |
Collapse
|
40
|
Xu J, Mukerjee S, Silva-Alves CRA, Carvalho-Galvão A, Cruz JC, Balarini CM, Braga VA, Lazartigues E, França-Silva MS. A Disintegrin and Metalloprotease 17 in the Cardiovascular and Central Nervous Systems. Front Physiol 2016; 7:469. [PMID: 27803674 PMCID: PMC5067531 DOI: 10.3389/fphys.2016.00469] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 09/30/2016] [Indexed: 01/19/2023] Open
Abstract
ADAM17 is a metalloprotease and disintegrin that lodges in the plasmatic membrane of several cell types and is able to cleave a wide variety of cell surface proteins. It is somatically expressed in mammalian organisms and its proteolytic action influences several physiological and pathological processes. This review focuses on the structure of ADAM17, its signaling in the cardiovascular system and its participation in certain disorders involving the heart, blood vessels, and neural regulation of autonomic and cardiovascular modulation.
Collapse
Affiliation(s)
- Jiaxi Xu
- Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - Snigdha Mukerjee
- Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | | | | | - Josiane C Cruz
- Centro de Biotecnologia, Universidade Federal da Paraíba João Pessoa, Brazil
| | - Camille M Balarini
- Centro de Ciências da Saúde, Universidade Federal da Paraíba João Pessoa, Brazil
| | - Valdir A Braga
- Centro de Biotecnologia, Universidade Federal da Paraíba João Pessoa, Brazil
| | - Eric Lazartigues
- Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | | |
Collapse
|
41
|
Loria AS. Can we fight chronic kidney disease by targeting endothelial HB-EGF? Am J Physiol Renal Physiol 2016; 311:F406-8. [PMID: 27335378 DOI: 10.1152/ajprenal.00345.2016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 06/14/2016] [Indexed: 11/22/2022] Open
Affiliation(s)
- Analia S Loria
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
42
|
Takayanagi T, Forrester SJ, Kawai T, Obama T, Tsuji T, Elliott KJ, Nuti E, Rossello A, Kwok HF, Scalia R, Rizzo V, Eguchi S. Vascular ADAM17 as a Novel Therapeutic Target in Mediating Cardiovascular Hypertrophy and Perivascular Fibrosis Induced by Angiotensin II. Hypertension 2016; 68:949-955. [PMID: 27480833 DOI: 10.1161/hypertensionaha.116.07620] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 06/28/2016] [Indexed: 12/13/2022]
Abstract
Angiotensin II (AngII) has been strongly implicated in hypertension and its complications. Evidence suggests the mechanisms by which AngII elevates blood pressure and enhances cardiovascular remodeling and damage may be distinct. However, the signal transduction cascade by which AngII specifically initiates cardiovascular remodeling, such as hypertrophy and fibrosis, remains insufficiently understood. In vascular smooth muscle cells, a metalloproteinase ADAM17 mediates epidermal growth factor receptor transactivation, which may be responsible for cardiovascular remodeling but not hypertension induced by AngII. Thus, the objective of this study was to test the hypothesis that activation of vascular ADAM17 is indispensable for vascular remodeling but not for hypertension induced by AngII. Vascular ADAM17-deficient mice and control mice were infused with AngII for 2 weeks. Control mice infused with AngII showed cardiac hypertrophy, vascular medial hypertrophy, and perivascular fibrosis. These phenotypes were prevented in vascular ADAM17-deficient mice independent of blood pressure alteration. AngII infusion enhanced ADAM17 expression, epidermal growth factor receptor activation, and endoplasmic reticulum stress in the vasculature, which were diminished in ADAM17-deficient mice. Treatment with a human cross-reactive ADAM17 inhibitory antibody also prevented cardiovascular remodeling and endoplasmic reticulum stress but not hypertension in C57Bl/6 mice infused with AngII. In vitro data further supported these findings. In conclusion, vascular ADAM17 mediates AngII-induced cardiovascular remodeling via epidermal growth factor receptor activation independent of blood pressure regulation. ADAM17 seems to be a unique therapeutic target for the prevention of hypertensive complications.
Collapse
Affiliation(s)
- Takehiko Takayanagi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia PA (T.T., S.J.F., T.K., T.O., T.T., Y.F., K.J.E., R.S., V.R., S.E.), Department of Pharmacy, University of Pisa, Pisa, Italy (E.N., A.R.), and Faculty of Health Sciences, University of Macau, Macau, China (HF.K.)
| | - Steven J Forrester
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia PA (T.T., S.J.F., T.K., T.O., T.T., Y.F., K.J.E., R.S., V.R., S.E.), Department of Pharmacy, University of Pisa, Pisa, Italy (E.N., A.R.), and Faculty of Health Sciences, University of Macau, Macau, China (HF.K.)
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia PA (T.T., S.J.F., T.K., T.O., T.T., Y.F., K.J.E., R.S., V.R., S.E.), Department of Pharmacy, University of Pisa, Pisa, Italy (E.N., A.R.), and Faculty of Health Sciences, University of Macau, Macau, China (HF.K.)
| | - Takashi Obama
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia PA (T.T., S.J.F., T.K., T.O., T.T., Y.F., K.J.E., R.S., V.R., S.E.), Department of Pharmacy, University of Pisa, Pisa, Italy (E.N., A.R.), and Faculty of Health Sciences, University of Macau, Macau, China (HF.K.)
| | - Toshiyuki Tsuji
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia PA (T.T., S.J.F., T.K., T.O., T.T., Y.F., K.J.E., R.S., V.R., S.E.), Department of Pharmacy, University of Pisa, Pisa, Italy (E.N., A.R.), and Faculty of Health Sciences, University of Macau, Macau, China (HF.K.)
| | - Katherine J Elliott
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia PA (T.T., S.J.F., T.K., T.O., T.T., Y.F., K.J.E., R.S., V.R., S.E.), Department of Pharmacy, University of Pisa, Pisa, Italy (E.N., A.R.), and Faculty of Health Sciences, University of Macau, Macau, China (HF.K.)
| | - Elisa Nuti
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia PA (T.T., S.J.F., T.K., T.O., T.T., Y.F., K.J.E., R.S., V.R., S.E.), Department of Pharmacy, University of Pisa, Pisa, Italy (E.N., A.R.), and Faculty of Health Sciences, University of Macau, Macau, China (HF.K.)
| | - Armando Rossello
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia PA (T.T., S.J.F., T.K., T.O., T.T., Y.F., K.J.E., R.S., V.R., S.E.), Department of Pharmacy, University of Pisa, Pisa, Italy (E.N., A.R.), and Faculty of Health Sciences, University of Macau, Macau, China (HF.K.)
| | - Hang Fai Kwok
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia PA (T.T., S.J.F., T.K., T.O., T.T., Y.F., K.J.E., R.S., V.R., S.E.), Department of Pharmacy, University of Pisa, Pisa, Italy (E.N., A.R.), and Faculty of Health Sciences, University of Macau, Macau, China (HF.K.)
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia PA (T.T., S.J.F., T.K., T.O., T.T., Y.F., K.J.E., R.S., V.R., S.E.), Department of Pharmacy, University of Pisa, Pisa, Italy (E.N., A.R.), and Faculty of Health Sciences, University of Macau, Macau, China (HF.K.)
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia PA (T.T., S.J.F., T.K., T.O., T.T., Y.F., K.J.E., R.S., V.R., S.E.), Department of Pharmacy, University of Pisa, Pisa, Italy (E.N., A.R.), and Faculty of Health Sciences, University of Macau, Macau, China (HF.K.)
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia PA (T.T., S.J.F., T.K., T.O., T.T., Y.F., K.J.E., R.S., V.R., S.E.), Department of Pharmacy, University of Pisa, Pisa, Italy (E.N., A.R.), and Faculty of Health Sciences, University of Macau, Macau, China (HF.K.)
| |
Collapse
|
43
|
Chen J, Zeng F, Forrester SJ, Eguchi S, Zhang MZ, Harris RC. Expression and Function of the Epidermal Growth Factor Receptor in Physiology and Disease. Physiol Rev 2016; 96:1025-1069. [DOI: 10.1152/physrev.00030.2015] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The epidermal growth factor receptor (EGFR) is the prototypical member of a family of membrane-associated intrinsic tyrosine kinase receptors, the ErbB family. EGFR is activated by multiple ligands, including EGF, transforming growth factor (TGF)-α, HB-EGF, betacellulin, amphiregulin, epiregulin, and epigen. EGFR is expressed in multiple organs and plays important roles in proliferation, survival, and differentiation in both development and normal physiology, as well as in pathophysiological conditions. In addition, EGFR transactivation underlies some important biologic consequences in response to many G protein-coupled receptor (GPCR) agonists. Aberrant EGFR activation is a significant factor in development and progression of multiple cancers, which has led to development of mechanism-based therapies with specific receptor antibodies and tyrosine kinase inhibitors. This review highlights the current knowledge about mechanisms and roles of EGFR in physiology and disease.
Collapse
Affiliation(s)
- Jianchun Chen
- Departments of Medicine, Cancer Biology, and Molecular Physiology and Biophysics, Vanderbilt University School of Medicine and Nashville Veterans Affairs Hospital, Nashville, Tennessee; and Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Fenghua Zeng
- Departments of Medicine, Cancer Biology, and Molecular Physiology and Biophysics, Vanderbilt University School of Medicine and Nashville Veterans Affairs Hospital, Nashville, Tennessee; and Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Steven J. Forrester
- Departments of Medicine, Cancer Biology, and Molecular Physiology and Biophysics, Vanderbilt University School of Medicine and Nashville Veterans Affairs Hospital, Nashville, Tennessee; and Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Satoru Eguchi
- Departments of Medicine, Cancer Biology, and Molecular Physiology and Biophysics, Vanderbilt University School of Medicine and Nashville Veterans Affairs Hospital, Nashville, Tennessee; and Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Ming-Zhi Zhang
- Departments of Medicine, Cancer Biology, and Molecular Physiology and Biophysics, Vanderbilt University School of Medicine and Nashville Veterans Affairs Hospital, Nashville, Tennessee; and Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Raymond C. Harris
- Departments of Medicine, Cancer Biology, and Molecular Physiology and Biophysics, Vanderbilt University School of Medicine and Nashville Veterans Affairs Hospital, Nashville, Tennessee; and Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
44
|
Wang M, Tang YB, Ma MM, Chen JH, Hu CP, Zhao SP, Peng DQ, Zhou JG, Guan YY, Zhang Z. TRPC3 channel confers cerebrovascular remodelling during hypertension via transactivation of EGF receptor signalling. Cardiovasc Res 2015; 109:34-43. [DOI: 10.1093/cvr/cvv246] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 10/11/2015] [Indexed: 01/07/2023] Open
|
45
|
Forrester SJ, Kawai T, O'Brien S, Thomas W, Harris RC, Eguchi S. Epidermal Growth Factor Receptor Transactivation: Mechanisms, Pathophysiology, and Potential Therapies in the Cardiovascular System. Annu Rev Pharmacol Toxicol 2015; 56:627-53. [PMID: 26566153 DOI: 10.1146/annurev-pharmtox-070115-095427] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Epidermal growth factor receptor (EGFR) activation impacts the physiology and pathophysiology of the cardiovascular system, and inhibition of EGFR activity is emerging as a potential therapeutic strategy to treat diseases including hypertension, cardiac hypertrophy, renal fibrosis, and abdominal aortic aneurysm. The capacity of G protein-coupled receptor (GPCR) agonists, such as angiotensin II (AngII), to promote EGFR signaling is called transactivation and is well described, yet delineating the molecular processes and functional relevance of this crosstalk has been challenging. Moreover, these critical findings are dispersed among many different fields. The aim of our review is to highlight recent advancements in defining the signaling cascades and downstream consequences of EGFR transactivation in the cardiovascular renal system. We also focus on studies that link EGFR transactivation to animal models of the disease, and we discuss potential therapeutic applications.
Collapse
Affiliation(s)
- Steven J Forrester
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140;
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140;
| | - Shannon O'Brien
- The School of Biomedical Sciences, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Walter Thomas
- The School of Biomedical Sciences, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Raymond C Harris
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140;
| |
Collapse
|
46
|
Karnik SS, Unal H, Kemp JR, Tirupula KC, Eguchi S, Vanderheyden PML, Thomas WG. International Union of Basic and Clinical Pharmacology. XCIX. Angiotensin Receptors: Interpreters of Pathophysiological Angiotensinergic Stimuli [corrected]. Pharmacol Rev 2015; 67:754-819. [PMID: 26315714 PMCID: PMC4630565 DOI: 10.1124/pr.114.010454] [Citation(s) in RCA: 225] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The renin angiotensin system (RAS) produced hormone peptides regulate many vital body functions. Dysfunctional signaling by receptors for RAS peptides leads to pathologic states. Nearly half of humanity today would likely benefit from modern drugs targeting these receptors. The receptors for RAS peptides consist of three G-protein-coupled receptors—the angiotensin II type 1 receptor (AT1 receptor), the angiotensin II type 2 receptor (AT2 receptor), the MAS receptor—and a type II trans-membrane zinc protein—the candidate angiotensin IV receptor (AngIV binding site). The prorenin receptor is a relatively new contender for consideration, but is not included here because the role of prorenin receptor as an independent endocrine mediator is presently unclear. The full spectrum of biologic characteristics of these receptors is still evolving, but there is evidence establishing unique roles of each receptor in cardiovascular, hemodynamic, neurologic, renal, and endothelial functions, as well as in cell proliferation, survival, matrix-cell interaction, and inflammation. Therapeutic agents targeted to these receptors are either in active use in clinical intervention of major common diseases or under evaluation for repurposing in many other disorders. Broad-spectrum influence these receptors produce in complex pathophysiological context in our body highlights their role as precise interpreters of distinctive angiotensinergic peptide cues. This review article summarizes findings published in the last 15 years on the structure, pharmacology, signaling, physiology, and disease states related to angiotensin receptors. We also discuss the challenges the pharmacologist presently faces in formally accepting newer members as established angiotensin receptors and emphasize necessary future developments.
Collapse
Affiliation(s)
- Sadashiva S Karnik
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Hamiyet Unal
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Jacqueline R Kemp
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Kalyan C Tirupula
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Satoru Eguchi
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Patrick M L Vanderheyden
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Walter G Thomas
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| |
Collapse
|
47
|
de Queiroz TM, Xia H, Filipeanu CM, Braga VA, Lazartigues E. α-Lipoic acid reduces neurogenic hypertension by blunting oxidative stress-mediated increase in ADAM17. Am J Physiol Heart Circ Physiol 2015; 309:H926-34. [PMID: 26254330 PMCID: PMC4591409 DOI: 10.1152/ajpheart.00259.2015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 07/30/2015] [Indexed: 02/07/2023]
Abstract
We previously reported that type 2 angiotensin-converting enzyme (ACE2) compensatory activity is impaired by the disintegrin and metalloprotease 17 (ADAM17), and lack of ACE2 is associated with oxidative stress in neurogenic hypertension. To investigate the relationship between ADAM17 and oxidative stress, Neuro2A cells were treated with ANG II (100 nM) 24 h after vehicle or α-lipoic acid (LA, 500 μM). ADAM17 expression was increased by ANG II (120.5 ± 9.1 vs. 100.2 ± 0.8%, P < 0.05) and decreased after LA (69.0 ± 0.3 vs. 120.5 ± 9.1%, P < 0.05). In another set of experiments, LA reduced ADAM17 (92.9 ± 5.3 vs. 100.0 ± 11.2%, P < 0.05) following its overexpression. Moreover, ADAM17 activity was reduced by LA in ADAM17-overexpressing cells [109.5 ± 19.8 vs. 158.0 ± 20.0 fluorescence units (FU)·min(-1)·μg protein(-1), P < 0.05], in which ADAM17 overexpression increased oxidative stress (114.1 ± 2.5 vs. 101.0 ± 1.0%, P < 0.05). Conversely, LA-treated cells attenuated ADAM17 overexpression-induced oxidative stress (76.0 ± 9.1 vs. 114.1 ± 2.5%, P < 0.05). In deoxycorticosterone acetate (DOCA)-salt hypertensive mice, a model in which ADAM17 expression and activity are increased, hypertension was blunted by pretreatment with LA (119.0 ± 2.4 vs. 131.4 ± 2.2 mmHg, P < 0.05). In addition, LA improved dysautonomia and baroreflex sensitivity. Furthermore, LA blunted the increase in NADPH oxidase subunit expression, as well as the increase in ADAM17 and decrease in ACE2 activity in the hypothalamus of DOCA-salt hypertensive mice. Taken together, these data suggest that LA might preserve ACE2 compensatory activity by breaking the feedforward cycle between ADAM17 and oxidative stress, resulting in a reduction of neurogenic hypertension.
Collapse
Affiliation(s)
- Thyago M de Queiroz
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana; Department of Biotechnology, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Huijing Xia
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana; Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana; and
| | - Catalin M Filipeanu
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana; Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana; and
| | - Valdir A Braga
- Department of Biotechnology, Federal University of Paraíba, João Pessoa, PB, Brazil
| | - Eric Lazartigues
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, Louisiana; Neurosciences Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana; Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana; and
| |
Collapse
|
48
|
Dong DD, Zhou H, Li G. ADAM15 targets MMP9 activity to promote lung cancer cell invasion. Oncol Rep 2015; 34:2451-60. [PMID: 26323669 DOI: 10.3892/or.2015.4203] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 07/20/2015] [Indexed: 11/05/2022] Open
Abstract
ADAM15 is a membrane-associated proteinase belonging to a disintegrin and metalloproteinase (ADAM) family. Recent studies suggested that ADAM15 is overexpressed in several types of cancer and is involved in metastatic tumor progression. However, the function of ADAM15 in non-small cell lung cancer (NSCLC) is currently unknown. In the present study, we found that high expression of ADAM15 was associated with decreased overall survival (OS) and disease-free survival (DFS) in NSCLC patients. Furthermore, shRNA-mediated knockdown of ADAM15 attenuated cell migration and invasion. Mechanistic study demonstrated that ADAM15 upregulated MMP9 expression in lung cancer cells via activation of the MEK-ERK pathway. Moreover, ADAM15 proteolytically cleaved and activated pro-MMP9 in vitro and interacted with MMP9 in vivo. Overexpression of ADAM15 in A549 cells promoted cell invasion, while knocking down MMP9 attenuated cell invasive ability. Therefore, our data not only support a pro-metastatic role of ADAM15 in lung cancer progression, but also reveal a novel mechanism of ADAM15 in promoting cancer cell invasion through directly targeting MMP9 activation.
Collapse
Affiliation(s)
- Dan-Dan Dong
- Department of Pathology, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Hui Zhou
- Department of Thoracic Medicine, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, P.R. China
| | - Gao Li
- Department of Thoracic Surgery, Hainan General Hospital, Haikou, Hainan 570311, P.R. China
| |
Collapse
|
49
|
Paricalcitol Inhibits Aldosterone-Induced Proinflammatory Factors by Modulating Epidermal Growth Factor Receptor Pathway in Cultured Tubular Epithelial Cells. BIOMED RESEARCH INTERNATIONAL 2015; 2015:783538. [PMID: 26064952 PMCID: PMC4438184 DOI: 10.1155/2015/783538] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 01/11/2015] [Indexed: 11/17/2022]
Abstract
Chronic kidney disease is characterized by Vitamin D deficiency and activation of the renin-angiotensin-aldosterone system. Increasing data show that vitamin D receptor agonists (VDRAs) exert beneficial effects in renal disease and possess anti-inflammatory properties, but the underlying mechanism remains unknown. Emerging evidence suggests that "a disintegrin and metalloproteinase" (ADAM)/epidermal growth factor receptor (EGFR) signalling axis contributes to renal damage. Aldosterone induces EGFR transactivation regulating several processes including cell proliferation and fibrosis. However, data on tubular epithelial cells is scarce. We have found that, in cultured tubular epithelial cells, aldosterone induced EGFR transactivation via TGF-α/ADAM17. Blockade of the TGF-α/ADAM17/EGFR pathway inhibited aldosterone-induced proinflammatory gene upregulation. Moreover, among the potential downstream mechanisms, we found that TGF-α/ADAM17/EGFR inhibition blocked ERK and STAT-1 activation in response to aldosterone. Next, we investigated the involvement of TGF-α/ADAM17/EGFR axis in VDRA anti-inflammatory effects. Preincubation with the VDRA paricalcitol inhibited aldosterone-induced EGFR transactivation, TGF-α/ADAM-17 gene upregulation, and downstream mechanisms, including proinflammatory factors overexpression. In conclusion, our data suggest that the anti-inflammatory actions of paricalcitol in tubular cells could depend on the inhibition of TGF-α/ADAM17/EGFR pathway in response to aldosterone, showing an important mechanism of VDRAs action.
Collapse
|
50
|
Takayanagi T, Kawai T, Forrester SJ, Obama T, Tsuji T, Fukuda Y, Elliott KJ, Tilley DG, Davisson RL, Park JY, Eguchi S. Role of epidermal growth factor receptor and endoplasmic reticulum stress in vascular remodeling induced by angiotensin II. Hypertension 2015; 65:1349-55. [PMID: 25916723 DOI: 10.1161/hypertensionaha.115.05344] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/01/2015] [Indexed: 12/18/2022]
Abstract
The mechanisms by which angiotensin II (AngII) elevates blood pressure and enhances end-organ damage seem to be distinct. However, the signal transduction cascade by which AngII specifically mediates vascular remodeling such as medial hypertrophy and perivascular fibrosis remains incomplete. We have previously shown that AngII-induced epidermal growth factor receptor (EGFR) transactivation is mediated by disintegrin and metalloproteinase domain 17 (ADAM17), and that this signaling is required for vascular smooth muscle cell hypertrophy but not for contractile signaling in response to AngII. Recent studies have implicated endoplasmic reticulum (ER) stress in hypertension. Interestingly, EGFR is capable of inducing ER stress. The aim of this study was to test the hypothesis that activation of EGFR and ER stress are critical components required for vascular remodeling but not hypertension induced by AngII. Mice were infused with AngII for 2 weeks with or without treatment of EGFR inhibitor, erlotinib, or ER chaperone, 4-phenylbutyrate. AngII infusion induced vascular medial hypertrophy in the heart, kidney and aorta, and perivascular fibrosis in heart and kidney, cardiac hypertrophy, and hypertension. Treatment with erlotinib as well as 4-phenylbutyrate attenuated vascular remodeling and cardiac hypertrophy but not hypertension. In addition, AngII infusion enhanced ADAM17 expression, EGFR activation, and ER/oxidative stress in the vasculature, which were diminished in both erlotinib-treated and 4-phenylbutyrate-treated mice. ADAM17 induction and EGFR activation by AngII in vascular cells were also prevented by inhibition of EGFR or ER stress. In conclusion, AngII induces vascular remodeling by EGFR activation and ER stress via a signaling mechanism involving ADAM17 induction independent of hypertension.
Collapse
Affiliation(s)
- Takehiko Takayanagi
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Tatsuo Kawai
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Steven J Forrester
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Takashi Obama
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Toshiyuki Tsuji
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Yamato Fukuda
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Katherine J Elliott
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Douglas G Tilley
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Robin L Davisson
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Joon-Young Park
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.)
| | - Satoru Eguchi
- From the Department of Physiology, Cardiovascular Research Center (T. Takayanagi, T.K., S.J.F., T.O., T. Tsuji, Y.F., K.J.E., J.-Y.P., S.E.) and Department of Pharmacology, Center for Translational Medicine (D.G.T.), Temple University School of Medicine, Philadelphia, PA; Department of Kinesiology, Temple University College of Public Health, Philadelphia, PA (S.J.F., J.-Y.P.); and Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY (R.L.D.).
| |
Collapse
|