1
|
Pietraszko P, Zorawski M, Bielecka E, Sielatycki P, Zbroch E. Mid-Regional Proadrenomedullin in COVID-19-May It Act as a Predictor of Prolonged Cardiovascular Complications? Int J Mol Sci 2023; 24:16821. [PMID: 38069140 PMCID: PMC10705931 DOI: 10.3390/ijms242316821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/06/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
The rising prevalence of cardiovascular disease (CVD) and the impact of the SARS-CoV-2 pandemic have both led to increased mortality rates, affecting public health and the global economy. Therefore, it is essential to find accessible, non-invasive prognostic markers capable of identifying patients at high risk. One encouraging avenue of exploration is the potential of mid-regional proadrenomedullin (MR-proADM) as a biomarker in various health conditions, especially in the context of CVD and COVID-19. MR-proADM presents the ability to predict mortality, heart failure, and adverse outcomes in CVD, offering promise for improved risk assessment and treatment strategies. On the other hand, an elevated MR-proADM level is associated with disease severity and cytokine storms in patients with COVID-19, making it a predictive indicator for intensive care unit admissions and mortality rates. Moreover, MR-proADM may have relevance in long COVID, aiding in the risk assessment, triage, and monitoring of individuals at increased risk of developing prolonged cardiac issues. Our review explores the potential of MR-proADM as a predictor of enduring cardiovascular complications following COVID-19 infection.
Collapse
Affiliation(s)
| | | | | | | | - Edyta Zbroch
- Department of Internal Medicine and Hypertension, Medical University of Bialystok, 15-089 Bialystok, Poland; (P.P.); (M.Z.); (E.B.); (P.S.)
| |
Collapse
|
2
|
Chang CL, Cai Z, Hsu SYT. Sustained Activation of CLR/RAMP Receptors by Gel-Forming Agonists. Int J Mol Sci 2022; 23:ijms232113408. [PMID: 36362188 PMCID: PMC9655119 DOI: 10.3390/ijms232113408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
Background: Adrenomedullin (ADM), adrenomedullin 2 (ADM2), and CGRP family peptides are important regulators of vascular vasotone and integrity, neurotransmission, and fetoplacental development. These peptides signal through CLR/RAMP1, 2, and 3 receptors, and protect against endothelial dysfunction in disease models. As such, CLR/RAMP receptor agonists are considered important therapeutic candidates for various diseases. Methods and Results: Based on the screening of a series of palmitoylated chimeric ADM/ADM2 analogs, we demonstrated a combination of lipidation and accommodating motifs at the hinge region of select peptides is important for gaining an enhanced receptor-activation activity and improved stimulatory effects on the proliferation and survival of human lymphatic endothelial cells when compared to wild-type peptides. In addition, by serendipity, we found that select palmitoylated analogs self-assemble to form liquid gels, and subcutaneous administration of an analog gel led to the sustained presence of the peptide in the circulation for >2 days. Consistently, subcutaneous injection of the analog gel significantly reduced the blood pressure in SHR rats and increased vasodilation in the hindlimbs of adult rats for days. Conclusions: Together, these data suggest gel-forming adrenomedullin analogs may represent promising candidates for the treatment of various life-threatening endothelial dysfunction-associated diseases such as treatment-resistant hypertension and preeclampsia, which are in urgent need of an effective drug.
Collapse
Affiliation(s)
- Chia Lin Chang
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital Linkou Medical Center, Chang Gung University, Kweishan, Taoyuan 20878, Taiwan
| | - Zheqing Cai
- CL Laboratory LLC, Gaithersburg, MD 20878, USA
| | - Sheau Yu Teddy Hsu
- Adepthera LLC, San Jose, CA 95138, USA
- Correspondence: ; Tel.: +1-650-799-3496
| |
Collapse
|
3
|
Kita T, Kitamura K. Translational studies of adrenomedullin and related peptides regarding cardiovascular diseases. Hypertens Res 2022; 45:389-400. [PMID: 34992239 PMCID: PMC8732970 DOI: 10.1038/s41440-021-00806-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/24/2021] [Accepted: 10/28/2021] [Indexed: 12/11/2022]
Abstract
Adrenomedullin (AM) is a vasodilative peptide with various physiological functions, including the maintenance of vascular tone and endothelial barrier function. AM levels are markedly increased during severe inflammation, such as that associated with sepsis; thus, AM is expected to be a useful clinical marker and therapeutic agent for inflammation. However, as the increase in AM levels in cardiovascular diseases (CVDs) is relatively low compared to that in infectious diseases, the value of AM as a marker of CVDs seems to be less important. Limitations pertaining to the administrative route and short half-life of AM in the bloodstream (<30 min) restrict the therapeutic applications of AM for CVDs. In early human studies, various applications of AM for CVDs were attempted, including for heart failure, myocardial infarction, pulmonary hypertension, and peripheral artery disease; however, none achieved success. We have developed AM as a therapeutic agent for inflammatory bowel disease in which the vasodilatory effect of AM is minimized. A clinical trial evaluating this AM formulation for acute cerebral infarction is ongoing. We have also developed AM derivatives that exhibit a longer half-life and less vasodilative activity. These AM derivatives can be administered by subcutaneous injection at long-term intervals. Accordingly, these derivatives will reduce the inconvenience in use compared to that for native AM and expand the possible applications of AM for treating CVDs. In this review, we present the latest translational status of AM and its derivatives.
Collapse
Affiliation(s)
- Toshihiro Kita
- Department of Projects Research, Frontier Science Research Center, University of Miyazaki, Miyazaki, Japan.
| | - Kazuo Kitamura
- Department of Projects Research, Frontier Science Research Center, University of Miyazaki, Miyazaki, Japan
| |
Collapse
|
4
|
Kita T, Kitamura K. Adrenomedullin Therapy in Moderate to Severe COVID-19. Biomedicines 2022; 10:biomedicines10030533. [PMID: 35327335 PMCID: PMC8945653 DOI: 10.3390/biomedicines10030533] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/19/2022] [Accepted: 02/21/2022] [Indexed: 12/15/2022] Open
Abstract
The 2019 coronavirus (COVID-19) pandemic is still in progress, and a significant number of patients have presented with severe illness. Recently introduced vaccines, antiviral medicines, and antibody formulations can suppress COVID-19 symptoms and decrease the number of patients exhibiting severe disease. However, complete avoidance of severe COVID-19 has not been achieved, and more importantly, there are insufficient methods to treat it. Adrenomedullin (AM) is an endogenous peptide that maintains vascular tone and endothelial barrier function. The AM plasma level is markedly increased during severe inflammatory disorders, such as sepsis, pneumonia, and COVID-19, and is associated with the severity of inflammation and its prognosis. In this study, exogenous AM administration reduced inflammation and related organ damage in rodent models. The results of this study strongly suggest that AM could be an alternative therapy in severe inflammation disorders, including COVID-19. We have previously developed an AM formulation to treat inflammatory bowel disease and are currently conducting an investigator-initiated phase 2a trial for moderate to severe COVID-19 using the same formulation. This review presents the basal AM information and the most recent translational AM/COVID-19 study.
Collapse
|
5
|
Martínez-Herrero S, Martínez A. Adrenomedullin: Not Just Another Gastrointestinal Peptide. Biomolecules 2022; 12:biom12020156. [PMID: 35204657 PMCID: PMC8961556 DOI: 10.3390/biom12020156] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/14/2022] [Accepted: 01/15/2022] [Indexed: 12/11/2022] Open
Abstract
Adrenomedullin (AM) and proadrenomedullin N-terminal 20 peptide (PAMP) are two bioactive peptides derived from the same precursor with several biological functions including vasodilation, angiogenesis, or anti-inflammation, among others. AM and PAMP are widely expressed throughout the gastrointestinal (GI) tract where they behave as GI hormones, regulating numerous physiological processes such as gastric emptying, gastric acid release, insulin secretion, bowel movements, or intestinal barrier function. Furthermore, it has been recently demonstrated that AM/PAMP have an impact on gut microbiome composition, inhibiting the growth of bacteria related with disease and increasing the number of beneficial bacteria such as Lactobacillus or Bifidobacterium. Due to their wide functions in the GI tract, AM and PAMP are involved in several digestive pathologies such as peptic ulcer, diabetes, colon cancer, or inflammatory bowel disease (IBD). AM is a key protective factor in IBD onset and development, as it regulates cytokine production in the intestinal mucosa, improves vascular and lymphatic regeneration and function and mucosal epithelial repair, and promotes a beneficial gut microbiome composition. AM and PAMP are relevant GI hormones that can be targeted to develop novel therapeutic agents for IBD, other GI disorders, or microbiome-related pathologies.
Collapse
|
6
|
Ihara M, Washida K, Yoshimoto T, Saito S. Adrenomedullin: A vasoactive agent for sporadic and hereditary vascular cognitive impairment. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2021; 2:100007. [PMID: 36324729 PMCID: PMC9616331 DOI: 10.1016/j.cccb.2021.100007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/14/2021] [Accepted: 02/26/2021] [Indexed: 04/16/2023]
Abstract
Adrenomedullin (AM) is an endogenous peptide mainly secreted from endothelial cells, which has multiple physiological actions such as anti-inflammation, vasodilation, vascular permeability regulation and angiogenesis. Blood AM levels are upregulated in a variety of pathological states including sepsis, severe COVID-19, acute ischemic stroke and vascular cognitive impairment with white matter changes, likely serving as a compensatory biological defense response against infection and ischemia. AM is currently being tested in clinical trials for ulcerative colitis, Crohn's disease, severe COVID-19 for its anti-inflammatory properties and in ischemic stroke for its additional angiogenic action. AM has been proposed as a therapeutic option for vascular cognitive impairment as its arteriogenic and angiogenic properties are thought to contribute to a slowing of cognitive decline in mice after chronic cerebral hypoperfusion. As AM promotes differentiation of oligodendrocyte precursor cells into mature oligodendrocytes under hypoxic conditions, AM could also be used in the treatment of CADASIL, where reduced oxygen delivery is thought to lead to the death of hypoxia-prone oligodendrocytes. AM therefore holds potential as an innovative therapeutic drug, which may regenerate blood vessels, while controlling inflammation in cerebrovascular diseases.
Collapse
|
7
|
Karakas M, Jarczak D, Becker M, Roedl K, Addo MM, Hein F, Bergmann A, Zimmermann J, Simon TP, Marx G, Lütgehetmann M, Nierhaus A, Kluge S. Targeting Endothelial Dysfunction in Eight Extreme-Critically Ill Patients with COVID-19 Using the Anti-Adrenomedullin Antibody Adrecizumab (HAM8101). Biomolecules 2020; 10:E1171. [PMID: 32796765 PMCID: PMC7465983 DOI: 10.3390/biom10081171] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 02/06/2023] Open
Abstract
Recently, the stabilization of the endothelium has been explicitly identified as a therapeutic goal in coronavirus disease 2019 (COVID-19). Adrecizumab (HAM8101) is a first-in-class humanized monoclonal anti-Adrenomedullin (anti-ADM) antibody, targeting the sepsis- and inflammation-based vascular and capillary leakage. Within a "treatment on a named-patient basis" approach, Adrecizumab was administered to eight extreme-critically ill COVID-19 patients with acute respiratory distress syndrome (ARDS). The patients received a single dose of Adrecizumab, which was administered between 1 and 3 days after the initiation of mechanical ventilation. The SOFA (median 12.5) and SAPS-II (median 39) scores clearly documented the population at highest risk. Moreover, six of the patients suffered from acute renal failure, of whom five needed renal replacement therapy. The length of follow-up ranged between 13 and 27 days. Following the Adrecizumab administration, one patient in the low-dose group died at day 4 due to fulminant pulmonary embolism, while four were in stable condition, and three were discharged from the intensive care unit (ICU). Within 12 days, the SOFA score, as well as the disease severity score (range 0-16, mirroring critical resources in the ICU, with higher scores indicating more severe illness), decreased in five out of the seven surviving patients (in all high-dose patients). The PaO2/FiO2 increased within 12 days, while the inflammatory parameters C-reactive protein, procalcitonin, and interleukin-6 decreased. Importantly, the mortality was lower than expected and calculated by the SOFA score. In conclusion, in this preliminary uncontrolled case series of eight shock patients with life-threatening COVID-19 and ARDS, the administration of Adrecizumab was followed by a favorable outcome. Although the non-controlled design and the small sample size preclude any definitive statement about the potential efficacy of Adrecizumab in critically ill COVID-19 patients, the results of this case series are encouraging.
Collapse
MESH Headings
- Adult
- Aged
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- COVID-19
- Coronavirus Infections/complications
- Coronavirus Infections/pathology
- Critical Illness
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/pathology
- Female
- Humans
- Male
- Middle Aged
- Pandemics
- Pneumonia, Viral/complications
- Pneumonia, Viral/pathology
- Respiratory Distress Syndrome/drug therapy
- Respiratory Distress Syndrome/etiology
- Sepsis/drug therapy
- Sepsis/etiology
Collapse
Affiliation(s)
- Mahir Karakas
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg-Kiel-Lübeck, 20251 Hamburg, Germany
- Department of Cardiology, University Heart & Vascular Center Hamburg, 20251 Hamburg, Germany;
| | - Dominik Jarczak
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (D.J.); (K.R.); (A.N.); (S.K.)
| | - Martin Becker
- Department of Cardiology, University Heart & Vascular Center Hamburg, 20251 Hamburg, Germany;
| | - Kevin Roedl
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (D.J.); (K.R.); (A.N.); (S.K.)
| | - Marylyn M. Addo
- Division of Infectious Diseases, First Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany;
- Department of Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, 20359 Hamburg, Germany
| | - Frauke Hein
- Adrenomed AG, 16761 Hennigsdorf, Germany; (F.H.); (A.B.); (J.Z.)
| | - Andreas Bergmann
- Adrenomed AG, 16761 Hennigsdorf, Germany; (F.H.); (A.B.); (J.Z.)
- SphingoTec GmbH, 16761 Hennigsdorf, Germany
- 4TEEN4 Pharmaceuticals GmbH, 16761 Hennigsdorf, Germany
| | - Jens Zimmermann
- Adrenomed AG, 16761 Hennigsdorf, Germany; (F.H.); (A.B.); (J.Z.)
| | - Tim-Philipp Simon
- Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, 52074 Aachen, Germany; (T.-P.S.); (G.M.)
| | - Gernot Marx
- Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, 52074 Aachen, Germany; (T.-P.S.); (G.M.)
| | - Marc Lütgehetmann
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany;
| | - Axel Nierhaus
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (D.J.); (K.R.); (A.N.); (S.K.)
| | - Stefan Kluge
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (D.J.); (K.R.); (A.N.); (S.K.)
| |
Collapse
|
8
|
Oatmen KE, Zile MR, Burnett JC, Spinale FG. Bioactive Signaling in Next-Generation Pharmacotherapies for Heart Failure: A Review. JAMA Cardiol 2019; 3:1232-1243. [PMID: 30484834 DOI: 10.1001/jamacardio.2018.3789] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Importance The standard pharmacotherapy for heart failure (HF), particularly HF with reduced ejection fraction (HFrEF), is primarily through the use of receptor antagonists, notably inhibition of the renin-angiotensin system by either angiotensin-converting enzyme inhibition or angiotensin II receptor blockade (ARB). However, the completed Prospective Comparison of ARNI With an ACE-Inhibitor to Determine Impact on Global Mortality and Morbidity in Heart Failure (PARADIGM-HF) trial identified that the use of a single molecule (sacubitril/valsartan), which is an ARB and the neutral endopeptidase inhibitor (NEPi) neprilysin, yielded improved clinical outcomes in HFrEF compared with angiotensin-converting enzyme inhibition alone. Observations This review examined specific bioactive signaling pathways that would be potentiated by NEPi and how these would affect key cardiovascular processes relevant to HFrEF. It also addressed potential additive/synergistic effects of ARB. A number of biological signaling pathways that may be potentiated by sacubitril/valsartan were identified, including some novel candidate molecules, which will act in a synergistic manner to favorably alter the natural history of HFrEF. Conclusions and Relevance This review identified that activation rather than inhibition of specific receptor pathways provided favorable cardiovascular effects that cannot be achieved by renin-angiotensin system inhibition alone. Thus, an entirely new avenue of translational and clinical research lies ahead in which HF pharmacotherapies will move beyond receptor antagonist strategies.
Collapse
Affiliation(s)
- Kelsie E Oatmen
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine, Columbia
| | - Michael R Zile
- Medical University of South Carolina, Charleston.,Ralph H. Johnson Department of VA Medical Center, Charleston, South Carolina
| | - John C Burnett
- Cardiorenal Research Laboratory, Mayo Clinic, Rochester, Minnesota
| | - Francis G Spinale
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine, Columbia.,William Jennings Bryan Dorn VA Medical Center, Columbia, South Carolina
| |
Collapse
|
9
|
Theuerle J, Farouque O, Vasanthakumar S, Patel SK, Burrell LM, Clark DJ, Al-Fiadh AH. Plasma endothelin-1 and adrenomedullin are associated with coronary artery function and cardiovascular outcomes in humans. Int J Cardiol 2019; 291:168-172. [PMID: 30987836 DOI: 10.1016/j.ijcard.2019.04.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/10/2019] [Accepted: 04/01/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Endothelin-1 (ET-1) is a vasoconstrictor associated with cardiovascular disease, whereas adrenomedullin (ADM) is a vasorelaxant with cardioprotective properties. We sought to determine the relationship between plasma ET-1 and ADM with coronary circulatory function and long-term major adverse cardiovascular events (MACE). METHODS Thirty-two patients undergoing coronary angiography for chest pain were recruited. Baseline plasma ET-1 and ADM levels were measured. The index of microcirculatory resistance (IMR), coronary flow mediated dilatation (cFMD) and coronary flow reserve (CFR) were measured in a non-obstructed coronary artery. Patients were assessed for MACE over a median period of 8.8 years. RESULTS Plasma ET-1 levels correlated with IMR (r = 0.57; p < 0.01) and ADM levels correlated with CFR (r = 0.50; p = 0.04) and cFMD (r = 0.62; p = 0.01). After adjustment for age, gender and cardiovascular risk factors, the association between ADM and cFMD (β = 0.79; p < 0.01) and between ET-1 and IMR (β = 5.7; p = 0.01) remained significant. IMR was higher, although not statistically significant, in patients with long-term MACE (17.9 ± 5.3 vs. 13.1 ± 6.0 units; p = 0.14). In patients free of MACE, cFMD (9.3 ± 7.6 vs. 2.8 ± 5.0%; p = 0.01) and plasma ADM levels (7.6 ± 5.3 vs. 4.0 ± 1.9 pmol/L; p = 0.07) were higher. CONCLUSIONS Plasma ET-1 and ADM were associated with measures of coronary microvascular and coronary conduit vessel function, respectively. Increased cFMD and elevated plasma ADM were associated with a cardioprotective effect.
Collapse
Affiliation(s)
- James Theuerle
- Department of Cardiology, Austin Health, Melbourne, Australia
| | - Omar Farouque
- Department of Cardiology, Austin Health, Melbourne, Australia; Department of Medicine, Austin Health, The University of Melbourne, Melbourne, Australia.
| | | | - Sheila K Patel
- Department of Medicine, Austin Health, The University of Melbourne, Melbourne, Australia
| | - Louise M Burrell
- Department of Cardiology, Austin Health, Melbourne, Australia; Department of Medicine, Austin Health, The University of Melbourne, Melbourne, Australia
| | - David J Clark
- Department of Cardiology, Austin Health, Melbourne, Australia; Department of Medicine, Austin Health, The University of Melbourne, Melbourne, Australia
| | - Ali H Al-Fiadh
- Department of Cardiology, Austin Health, Melbourne, Australia; Department of Medicine, Austin Health, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
10
|
Tsuruda T, Kato J, Kuwasako K, Kitamura K. Adrenomedullin: Continuing to explore cardioprotection. Peptides 2019; 111:47-54. [PMID: 29577955 DOI: 10.1016/j.peptides.2018.03.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/15/2018] [Accepted: 03/19/2018] [Indexed: 10/17/2022]
Abstract
Adrenomedullin (AM), a peptide isolated from an extract of human pheochromocytoma, comprises 52 amino acids with an intramolecular disulfide bond and amidation at the carboxy-terminus. AM is present in various tissues and organs in rodents and humans, including the heart. The peptide concentration increases with cardiac hypertrophy, acute myocardial infarction, and overt heart failure in the plasma and the myocardium. The principal function of AM in the cardiovascular system is the regulation of the vascular tone by vasodilation and natriuresis via cyclic adenosine monophosphate-dependent or -independent mechanism. In addition, AM may possess unique properties that inhibit aldosterone secretion, oxidative stress, apoptosis, and stimulation of angiogenesis, resulting in the protection of the structure and function of the heart. The AM receptor comprises a complex between calcitonin receptor-like receptor (CLR) and receptor activity-modifying protein (RAMP) 2 or 3, and the AM-CLR/RAMP2 system is essential for heart development during embryogenesis. Small-scale clinical trials have proven the efficacy and safety of recombinant AM peptide therapy for heart failure. Gene delivery and a modified AM peptide that prolongs the half-life of the native peptide could be an innovative method to improve the efficacy and benefit of AM in clinical settings. In this review, we focus on the pathophysiological roles of AM and its receptor system in the heart and describe the advances in AM and proAM-derived peptides as diagnostic biomarkers as well as the therapeutic application of AM and modified AM for cardioprotection.
Collapse
Affiliation(s)
- Toshihiro Tsuruda
- Department of Internal Medicine, Circulatory and Body Fluid Regulation, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan.
| | - Johji Kato
- Frontier Science Research Center, Faculty of Medicine, University of Miyazaki, Japan
| | - Kenji Kuwasako
- Frontier Science Research Center, Faculty of Medicine, University of Miyazaki, Japan
| | - Kazuo Kitamura
- Department of Internal Medicine, Circulatory and Body Fluid Regulation, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| |
Collapse
|
11
|
Fraty M, Velho G, Gand E, Fumeron F, Ragot S, Sosner P, Mohammedi K, Gellen B, Saulnier PJ, Halimi JM, Montaigne D, Ducrocq G, Rehman M, Marre M, Roussel R, Hadjadj S. Prognostic value of plasma MR-proADM vs NT-proBNP for heart failure in people with type 2 diabetes: the SURDIAGENE prospective study. Diabetologia 2018; 61:2643-2653. [PMID: 30232509 DOI: 10.1007/s00125-018-4727-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 08/03/2018] [Indexed: 11/29/2022]
Abstract
AIMS/HYPOTHESIS N-terminal pro-B-type natriuretic peptide (NT-proBNP) is the gold standard prognostic biomarker for diagnosis and occurrence of heart failure. Here, we compared its prognostic value for the occurrence of congestive heart failure with that of plasma mid-region pro-adrenomedullin (MR-proADM), a surrogate for adrenomedullin, a vasoactive peptide with vasodilator and natriuretic properties, in people with type 2 diabetes. METHODS Plasma MR-proADM concentration was measured in baseline samples of a hospital-based cohort of consecutively recruited participants with type 2 diabetes. Our primary endpoint was heart failure requiring hospitalisation. RESULTS We included 1438 participants (age 65 ± 11 years; 604 women and 834 men). Hospitalisation for heart failure occurred during follow-up (median 64 months) in 206 participants; the incidence rate of heart failure was 2.5 (95% CI 2.2, 2.9) per 100 person-years. Plasma concentrations of MR-proADM and NT-proBNP were significantly associated with heart failure in a Cox multivariable analysis model when adjusted for age, diabetes duration, history of coronary heart disease, proteinuria and baseline eGFR (adjHR [95%CI] 1.83 [1.51, 2.21] and 2.20 [1.86, 2.61], respectively, per 1 SD log10 increment, both p < 0.001). MR-proADM contributed significant supplementary information to the prognosis of heart failure when we considered the clinical risk factors (integrated discrimination improvement [IDI, mean ± SEM] 0.021 ± 0.007, p = 0.001) (Table 3). Inclusion of NT-proBNP in the multivariable model including MR-proADM contributed significant complementary information on prediction of heart failure (IDI [mean ± SEM] 0.028 ± 0.008, p < 0.001). By contrast, MR-proADM did not contribute supplementary information on prediction of heart failure in a model including NT-proBNP (IDI [mean ± SEM] 0.003 ± 0.003, p = 0.27), with similar results for heart failure with reduced ejection fraction and preserved ejection fraction. CONCLUSIONS/INTERPRETATION MR-proADM is a prognostic biomarker for heart failure in people with type 2 diabetes but gives no significant complementary information on prediction of heart failure compared with NT-proBNP.
Collapse
Affiliation(s)
- Mathilde Fraty
- Centre d'Investigation Clinique, CHU de Poitiers, Poitiers, France.
- Service d'Endocrinologie-Diabétologie, CHU de Poitiers, 2 rue de la Milétrie, 86021, Poitiers, France.
| | - Gilberto Velho
- Centre de Recherche des Cordeliers, Inserm UMR_S 1138, Paris, France
| | - Elise Gand
- Pole DUNE (Digestif, Urologie, Néphrologie, Endocrinologie), CHU de Poitiers, Poitiers, France
| | - Fréderic Fumeron
- Centre de Recherche des Cordeliers, Inserm UMR_S 1138, Paris, France
- UFR de Médecine, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Stéphanie Ragot
- Centre d'Investigation Clinique, CHU de Poitiers, Poitiers, France
- UFR de Médecine et Pharmacie, Université de Poitiers, Poitiers, France
- CIC 1402, Inserm Poitiers, Poitiers, France
| | - Philippe Sosner
- Centre médico-sportif MON STADE, Paris, France
- Centre de Diagnostic et de Thérapeutique, AP-HP Hôpital Universitaire Hôtel-Dieu, Paris, France
- Laboratoire MOVE (EA 6314), Faculté des Sciences du Sport, Université de Poitiers, Poitiers, France
| | - Kamel Mohammedi
- Service Endocrinologie, Diabétologie, Nutrition, Hôpital Haut Lévêque, CHU de Bordeaux, Pessac, France
| | - Barnabas Gellen
- Service de Cardiologie, Polyclinique de Poitiers, Poitiers, France
| | - Pierre-Jean Saulnier
- Centre d'Investigation Clinique, CHU de Poitiers, Poitiers, France
- UFR de Médecine et Pharmacie, Université de Poitiers, Poitiers, France
- CIC 1402, Inserm Poitiers, Poitiers, France
| | - Jean-Michel Halimi
- Service Néphrologie, Dialyse et Transplantation, CHU de Tours, Tours, France
- Inserm CIC0202, Tours, France
| | - David Montaigne
- Clinique de Physiologie et Département d'Échocardiographie, CHRU Lille, Lille, France
- Inserm U1011, EGID, Institut Pasteur de Lille, Université de Lille, Lille, France
| | - Grégory Ducrocq
- UFR de Médecine, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- Service de Cardiologie, AP-HP, Hôpital Bichat Claude Bernard, Paris, France
| | - Michaela Rehman
- Centre d'Investigation Clinique, CHU de Poitiers, Poitiers, France
- Service de Cardiologie, CHU de Poitiers, Poitiers, France
| | - Michel Marre
- Centre de Recherche des Cordeliers, Inserm UMR_S 1138, Paris, France
- UFR de Médecine, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- Service de Diabétologie, Endocrinologie et Nutrition, DHU FIRE, AP-HP, Hôpital Bichat Claude Bernard, Paris, France
| | - Ronan Roussel
- Centre de Recherche des Cordeliers, Inserm UMR_S 1138, Paris, France
- UFR de Médecine, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- Service de Diabétologie, Endocrinologie et Nutrition, DHU FIRE, AP-HP, Hôpital Bichat Claude Bernard, Paris, France
| | - Samy Hadjadj
- Centre d'Investigation Clinique, CHU de Poitiers, Poitiers, France.
- Service d'Endocrinologie-Diabétologie, CHU de Poitiers, 2 rue de la Milétrie, 86021, Poitiers, France.
- UFR de Médecine et Pharmacie, Université de Poitiers, Poitiers, France.
- CIC 1402, Inserm Poitiers, Poitiers, France.
| | | |
Collapse
|
12
|
Natural and synthetic peptides in the cardiovascular diseases: An update on diagnostic and therapeutic potentials. Arch Biochem Biophys 2018; 662:15-32. [PMID: 30481494 DOI: 10.1016/j.abb.2018.11.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/31/2018] [Accepted: 11/21/2018] [Indexed: 02/07/2023]
Abstract
Several peptides play an important role in physiological and pathological conditions into the cardiovascular system. In addition to well-known vasoactive agents such as angiotensin II, endothelin, serotonin or natriuretic peptides, the vasoconstrictor Urotensin-II (Uro-II) and the vasodilators Urocortins (UCNs) and Adrenomedullin (AM) have been implicated in the control of vascular tone and blood pressure as well as in cardiovascular disease states including congestive heart failure, atherosclerosis, coronary artery disease, and pulmonary and systemic hypertension. Therefore these peptides, together with their receptors, become important therapeutic targets in cardiovascular diseases (CVDs). Circulating levels of these agents in the blood are markedly modified in patients with specific CVDs compared with those in healthy patients, becoming also potential biomarkers for these pathologies. This review will provide an overview of current knowledge about the physiological roles of Uro-II, UCN and AM in the cardiovascular system and their implications in cardiovascular diseases. It will further focus on the structural modifications carried out on original peptide sequences in the search of analogues with improved physiochemical properties as well as in the delivery methods. Finally, we have overviewed the possible application of these peptides and/or their precursors as biomarkers of CVDs.
Collapse
|
13
|
Precursor proadrenomedullin influences cardiomyocyte survival and local inflammation related to myocardial infarction. Proc Natl Acad Sci U S A 2018; 115:E8727-E8736. [PMID: 30166452 DOI: 10.1073/pnas.1721635115] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Increased adrenomedullin (ADM) levels are associated with various cardiac diseases such as myocardial infarction (MI). ADM is cleaved off from the full-length precursor protein proadrenomedullin (ProADM) during its posttranslational processing. To date, no biological effect of ProADM is reported, while ADM infusion leads to antiapoptotic effects and improved cardiac function. Using an MI mouse model, we found an induction of ProADM gene as well as protein expression during the early phase of MI. This was accompanied by apoptosis and increasing inflammation, which substantially influence the post-MI remodeling processes. Simulating ischemia in vitro, we demonstrate that ProADM expression was increased in cardiomyocytes and cardiac fibroblasts. Subsequently, we treated ischemic cardiomyocytes with either ProADM or ADM and found that both proteins increased survival. This effect was diminishable by blocking the ADM1 receptor. To investigate whether ProADM and ADM play a role in the regulation of cardiac inflammation, we analyzed chemokine expression after treatment of cells with both proteins. While ProADM induced an expression of proinflammatory cytokines, thus promoting inflammation, ADM reduced chemokine expression. On leukocytes, both proteins repressed chemokine expression, revealing antiinflammatory effects. However, ProADM but not ADM dampened concurrent activation of leukocytes. Our data show that the full-length precursor ProADM is biologically active by reducing apoptosis to a similar extent as ADM. We further assume that ProADM induces local inflammation in affected cardiac tissue but attenuates exaggerated inflammation, whereas ADM has low impact. Our data suggest that both proteins are beneficial during MI by influencing apoptosis and inflammation.
Collapse
|
14
|
Falkentoft AC, Rørth R, Iversen K, Høfsten DE, Kelbæk H, Holmvang L, Frydland M, Schoos MM, Helqvist S, Axelsson A, Clemmensen P, Jørgensen E, Saunamäki K, Tilsted HH, Pedersen F, Torp-Pedersen C, Kofoed KF, Goetze JP, Engstrøm T, Køber L. MR-proADM as a Prognostic Marker in Patients With ST-Segment-Elevation Myocardial Infarction-DANAMI-3 (a Danish Study of Optimal Acute Treatment of Patients With STEMI) Substudy. J Am Heart Assoc 2018; 7:JAHA.117.008123. [PMID: 29776961 PMCID: PMC6015359 DOI: 10.1161/jaha.117.008123] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Midregional proadrenomedullin (MR‐proADM) has demonstrated prognostic potential after myocardial infarction (MI). Yet, the prognostic value of MR‐proADM at admission has not been examined in patients with ST‐segment–elevation MI (STEMI). Methods and Results The aim of this substudy, DANAMI‐3 (The Danish Study of Optimal Acute Treatment of Patients with ST‐segment–elevation myocardial infarction), was to examine the associations of admission concentrations of MR‐proADM with short‐ and long‐term mortality and hospital admission for heart failure in patients with ST‐segment–elevation myocardial infarction. Outcomes were assessed using Cox proportional hazard models and area under the curve using receiver operating characteristics. In total, 1122 patients were included. The median concentration of MR‐proADM was 0.64 nmol/L (25th–75th percentiles, 0.53–0.79). Within 30 days 23 patients (2.0%) died and during a 3‐year follow‐up 80 (7.1%) died and 38 (3.4%) were admitted for heart failure. A doubling of MR‐proADM was, in adjusted models, associated with an increased risk of 30‐day mortality (hazard ratio, 2.67; 95% confidence interval, 1.01–7.11; P=0.049), long‐term mortality (hazard ratio, 3.23; 95% confidence interval, 1.97–5.29; P<0.0001), and heart failure (hazard ratio, 2.71; 95% confidence interval, 1.32–5.58; P=0.007). For 30‐day and 3‐year mortality, the area under the curve for MR‐proADM was 0.77 and 0.78, respectively. For 3‐year mortality, area under the curve (0.84) of the adjusted model marginally changed (0.85; P=0.02) after addition of MR‐proADM. Conclusions Elevation of admission MR‐proADM was associated with long‐term mortality and heart failure, whereas the association with short‐term mortality was borderline significant. MR‐proADM may be a marker of prognosis after ST‐segment–elevation myocardial infarction but does not seem to add substantial prognostic information to established clinical models. Clinical Trial Registration URL: http://www.ClinicalTrials.gov/. Unique identifiers: NCT01435408 and NCT01960933.
Collapse
Affiliation(s)
| | - Rasmus Rørth
- Department of Cardiology, Rigshospitalet University of Copenhagen, Denmark
| | - Kasper Iversen
- Department of Cardiology, Herlev Hospital University of Copenhagen, Herlev, Denmark
| | - Dan E Høfsten
- Department of Cardiology, Rigshospitalet University of Copenhagen, Denmark
| | - Henning Kelbæk
- Department of Cardiology, Rigshospitalet University of Copenhagen, Denmark
| | - Lene Holmvang
- Department of Cardiology, Rigshospitalet University of Copenhagen, Denmark
| | - Martin Frydland
- Department of Cardiology, Rigshospitalet University of Copenhagen, Denmark
| | - Mikkel M Schoos
- Department of Cardiology, Rigshospitalet University of Copenhagen, Denmark
| | - Steffen Helqvist
- Department of Cardiology, Rigshospitalet University of Copenhagen, Denmark
| | - Anna Axelsson
- Department of Cardiology, Rigshospitalet University of Copenhagen, Denmark
| | - Peter Clemmensen
- Division of Cardiology, Department of Medicine, Nykøbing Falster Hospital, Nykøbing Falster, Denmark.,Department of General and Interventional Cardiology, University Heart Center Hamburg-Eppendorf, Hamburg, Germany
| | - Erik Jørgensen
- Department of Cardiology, Rigshospitalet University of Copenhagen, Denmark
| | - Kari Saunamäki
- Department of Cardiology, Rigshospitalet University of Copenhagen, Denmark
| | | | - Frants Pedersen
- Department of Cardiology, Rigshospitalet University of Copenhagen, Denmark
| | | | - Klaus F Kofoed
- Department of Cardiology, Rigshospitalet University of Copenhagen, Denmark
| | - Jens P Goetze
- Department of Clinical Biochemistry, Rigshospitalet University of Copenhagen, Denmark
| | - Thomas Engstrøm
- Department of Cardiology, Rigshospitalet University of Copenhagen, Denmark
| | - Lars Køber
- Department of Cardiology, Rigshospitalet University of Copenhagen, Denmark
| |
Collapse
|
15
|
Vairamani K, Wang HS, Medvedovic M, Lorenz JN, Shull GE. RNA SEQ Analysis Indicates that the AE3 Cl -/HCO 3- Exchanger Contributes to Active Transport-Mediated CO 2 Disposal in Heart. Sci Rep 2017; 7:7264. [PMID: 28779178 PMCID: PMC5544674 DOI: 10.1038/s41598-017-07585-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/29/2017] [Indexed: 02/06/2023] Open
Abstract
Loss of the AE3 Cl−/HCO3− exchanger (Slc4a3) in mice causes an impaired cardiac force-frequency response and heart failure under some conditions but the mechanisms are not known. To better understand the functions of AE3, we performed RNA Seq analysis of AE3-null and wild-type mouse hearts and evaluated the data with respect to three hypotheses (CO2 disposal, facilitation of Na+-loading, and recovery from an alkaline load) that have been proposed for its physiological functions. Gene Ontology and PubMatrix analyses of differentially expressed genes revealed a hypoxia response and changes in vasodilation and angiogenesis genes that strongly support the CO2 disposal hypothesis. Differential expression of energy metabolism genes, which indicated increased glucose utilization and decreased fatty acid utilization, were consistent with adaptive responses to perturbations of O2/CO2 balance in AE3-null myocytes. Given that the myocardium is an obligate aerobic tissue and consumes large amounts of O2, the data suggest that loss of AE3, which has the potential to extrude CO2 in the form of HCO3−, impairs O2/CO2 balance in cardiac myocytes. These results support a model in which the AE3 Cl−/HCO3− exchanger, coupled with parallel Cl− and H+-extrusion mechanisms and extracellular carbonic anhydrase, is responsible for active transport-mediated disposal of CO2.
Collapse
Affiliation(s)
- Kanimozhi Vairamani
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267, USA
| | - Hong-Sheng Wang
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267, USA
| | - Mario Medvedovic
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267, USA
| | - John N Lorenz
- Department of Cellular and Molecular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267, USA
| | - Gary E Shull
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267, USA.
| |
Collapse
|
16
|
Supeł K, Kacprzak M, Zielińska M. The prognostic value of MR-proadrenomedullin in patients with acute coronary syndrome complicated by cardiogenic shock. Biomarkers 2016; 22:296-303. [DOI: 10.1080/1354750x.2016.1252962] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Karolina Supeł
- Department of Interventional Cardiology and Electrocardiology, Intensive Cardiac Therapy Clinic, Medical University of Lodz, Poland
| | - Michał Kacprzak
- Department of Interventional Cardiology and Electrocardiology, Intensive Cardiac Therapy Clinic, Medical University of Lodz, Poland
| | - Marzenna Zielińska
- Department of Interventional Cardiology and Electrocardiology, Intensive Cardiac Therapy Clinic, Medical University of Lodz, Poland
| |
Collapse
|
17
|
Plasma mid-regional pro-adrenomedullin levels are inversely associated with anxiety but unrelated to depression: Results from the observational DIAST-CHF study in patients with cardiovascular risk factors. Psychoneuroendocrinology 2015; 62:227-32. [PMID: 26342564 DOI: 10.1016/j.psyneuen.2015.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 07/22/2015] [Accepted: 08/05/2015] [Indexed: 11/21/2022]
Abstract
OBJECTIVES It has been postulated that patients with heart failure have a high risk of ventricular arrhythmias and sudden cardiac death resulting from anxiety-induced autonomic arousal. In the prospective and multicenter DIAST-CHF (Diagnostic Trial on Prevalence and Clinical Course of Diastolic Dysfunction and Heart Failure) study, we therefore, tested the hypothesis that adrenomedullin (ADM), a well-established predictor for cardiovascular outcome, is associated with self-rated anxiety symptoms in patients at risk of suffering from or actually with overt heart failure. PARTICIPANTS AND MEASURES Study participants with risk factors for diastolic dysfunction were requested to complete the Hospital Anxiety and Depression Scale (HADS), and plasma mid-regional pro-adrenomedullin (MR-proADM) concentrations were measured. RESULTS In bivariate analysis, we found significantly lower plasma MR-proADM levels in patients with elevated HADS-anxiety scores above the clinically relevant cut-off level of ≥11 (n=118, 536pmol/l, interquartile range [IQR] 449-626) as compared to non-anxious study participants (n=1,292, 573pmol/l, IQR 486-702, p=0.001). A set of multivariate models adjusted for potential confounders confirmed the negative association between self-rated anxiety symptoms and plasma MR-proADM. In similar models, no significant association was detected between HADS-depression scores and MR-proADM. CONCLUSIONS The inverse relationship between plasma MR-proADM and anxiety observed in patients with cardiovascular risk factors supports a previous experimental study using a mutant mouse line with a brain-specific loss of ADM expression which displayed hyperactive and over-anxious behavior. Further experimental and clinical studies are warranted to test the hypothesis that also in humans ADM acts as a neuromodulator with anxiolytic properties.
Collapse
|
18
|
Stenberg TA, Kildal AB, How OJ, Myrmel T. Adrenomedullin-epinephrine cotreatment enhances cardiac output and left ventricular function by energetically neutral mechanisms. Am J Physiol Heart Circ Physiol 2012; 302:H1584-90. [PMID: 22307666 DOI: 10.1152/ajpheart.00887.2011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Adrenomedullin (AM) used therapeutically reduces mortality in the acute phase of experimental myocardial infarction. However, AM is potentially deleterious in acute heart failure as it is vasodilative and inotropically neutral. AM and epinephrine (EPI) are cosecreted from chromaffin cells, indicating a physiological interaction. We assessed the hemodynamic and energetic profile of AM-EPI cotreatment, exploring whether drug interaction improves cardiac function. Left ventricular (LV) mechanoenergetics were evaluated in 14 open-chest pigs using pressure-volume analysis and the pressure-volume area-myocardial O(2) consumption (PVA-MVo(2)) framework. AM (15 ng·kg(-1)·min(-1), n = 8) or saline (controls, n = 6) was infused for 120 min. Subsequently, a concurrent infusion of EPI (50 ng·kg(-1)·min(-1)) was added in both groups (AM-EPI vs. EPI). AM increased cardiac output (CO) and coronary blood flow by 20 ± 10% and 39 ± 14% (means ± SD, P < 0.05 vs. baseline), whereas controls were unaffected. AM-EPI increased CO and coronary blood flow by 55 ± 17% and 75 ± 16% (P < 0.05, AM-EPI interaction) compared with 13 ± 12% (P < 0.05 vs. baseline) and 18 ± 31% (P = not significant) with EPI. LV systolic capacitance decreased by -37 ± 22% and peak positive derivative of LV pressure (dP/dt(max)) increased by 32 ± 7% with AM-EPI (P < 0.05, AM-EPI interaction), whereas no significant effects were observed with EPI. Mean arterial pressure was maintained by AM-EPI and tended to decrease with EPI (+2 ± 13% vs. -11 ± 10%, P = not significant). PVA-MVo(2) relationships were unaffected by all treatments. In conclusion, AM-EPI cotreatment has an inodilator profile with CO and LV function augmented beyond individual drug effects and is not associated with relative increases in energetic cost. This can possibly take the inodilator treatment strategy beyond hemodynamic goals and exploit the cardioprotective effects of AM in acute heart failure.
Collapse
Affiliation(s)
- Thor Allan Stenberg
- Surgical Research Laboratory, Department of Clinical Medicine, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway.
| | | | | | | |
Collapse
|
19
|
The first clinical pilot study of intravenous adrenomedullin administration in patients with acute myocardial infarction. J Cardiovasc Pharmacol 2011; 56:413-9. [PMID: 20930593 DOI: 10.1097/fjc.0b013e3181f15b45] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Adrenomedullin (AM) is a 52-amino-acid vasodilator peptide that was originally isolated from human pheochromocytoma. In the previous experimental study with rat ischemia/reperfusion model, AM reduced infarct size and inhibited myocyte apoptosis. AM also suppressed the production of oxygen-free radicals. The present study was designed to evaluate the feasibility of intravenous administration of AM in patients with acute myocardial infarction. We studied 10 patients with first acute myocardial infarction [male to female ratio: 9 to 1, age: 65 ± 9 (mean ± SD) years, peak creatine phosphokinase level: 4215 ± 1933 (SD) U/L], who were hospitalized within 12 hours of symptom onset. Proceeding reperfusion therapy, AM infusion was initiated and continued at concentration of 0.0125-0.025 μg·kg·min for 12 hours. Follow-up coronary angiography and left ventriculography were performed at 3 months. Cardiac magnetic resonance was examined at 1 month and 3 months after AM therapy. During infusion of AM, hemodynamics kept stable except 2 patients. Wall motion index in the infarct area at 3 months was significantly improved compared with that at baseline, and infarct size evaluated by cardiac magnetic resonance was significantly decreased at 3 months. In conclusion, intravenous administration of AM, which possesses a variety of potential cardiovascular protective actions, can be adjunctive to percutaneous coronary intervention.
Collapse
|
20
|
Dhillon OS, Khan SQ, Narayan HK, Ng KH, Struck J, Quinn PA, Morgenthaler NG, Squire IB, Davies JE, Bergmann A, Ng LL. Prognostic Value of Mid-Regional Pro-Adrenomedullin Levels Taken on Admission and Discharge in Non–ST-Elevation Myocardial Infarction. J Am Coll Cardiol 2010; 56:125-33. [DOI: 10.1016/j.jacc.2010.01.060] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2009] [Revised: 01/07/2010] [Accepted: 01/18/2010] [Indexed: 11/28/2022]
|
21
|
The role of nitric oxide and potassium channels in the effect of adrenomedullin in human internal thoracic arteries. ACTA ACUST UNITED AC 2010; 161:92-6. [DOI: 10.1016/j.regpep.2009.12.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Revised: 11/24/2009] [Accepted: 12/30/2009] [Indexed: 01/11/2023]
|
22
|
Nishikimi T, Karasawa T, Inaba C, Ishimura K, Tadokoro K, Koshikawa S, Yoshihara F, Nagaya N, Sakio H, Kangawa K, Matsuoka H. Effects of long-term intravenous administration of adrenomedullin (AM) plus hANP therapy in acute decompensated heart failure: a pilot study. Circ J 2009; 73:892-8. [PMID: 19346663 DOI: 10.1253/circj.cj-08-0487] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND It was reported previously that 30 min administration of adrenomedullin (AM) improves hemodynamics in chronic stable heart failure patients. The present study was designed to examine whether long-term AM + human atrial natriuretic peptide (hANP) administration can be used as a therapeutic drug in patients with acute decompensated heart failure (ADHF) in clinical setting. METHODS AND RESULTS Seven acute heart failure patients (74 +/- 5 years) with dyspnea and pulmonary congestion were studied. AM (0.02 microg x kg(-1) x min(-1)) + hANP (0.05 microg x kg(-1) x min(-1)) was infused for 12 h and then hANP (0.05 microg x kg(-1) x min(-1)) was infused for 12 h. Hemodynamic, renal, hormonal and oxidative stress responses were evaluated. AM + hANP significantly reduced mean arterial pressure, pulmonary arterial pressure and systemic and pulmonary vascular resistance without changing heart rate, and increased cardiac output for most time-points compared with those at baseline. In addition, AM + hANP reduced aldosterone, brain natriuretic peptide and free-radical metabolites compared with those at baseline (all P<0.05). AM + hANP increased urine volume and U(Na)V compared with baseline data. CONCLUSIONS In this small, pilot trial, AM + hANP therapy had beneficial hemodynamic and hormonal effects in ADHF. Intravenous infusion of AM with hANP could be used as a therapeutic drug in ADHF. These data are preliminary and require confirmation in a larger clinical study.
Collapse
Affiliation(s)
- Toshio Nishikimi
- Department of Hypertension and Cardiorenal Medicine, Dokkyo Medical University, Mibu, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Rademaker MT, Charles CJ, Nicholls MG, Richards AM. Hemodynamic, hormonal, and renal actions of adrenomedullin 2 in experimental heart failure. Circ Heart Fail 2008; 1:134-42. [PMID: 19808282 DOI: 10.1161/circheartfailure.107.755504] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Adrenomedullin 2 (AM2) is a novel member of the calcitonin gene-related peptide family that is thought to play a regulatory role in circulatory homeostasis under normal physiological conditions. The effects of AM2 in heart failure have not been investigated previously. METHODS AND RESULTS Two incremental doses of human AM2 (10 and 100 ng[kg.min] for 90 minutes each) were given by intravenous infusion to 8 sheep with pacing-induced heart failure. Compared with time-matched control infusions, AM2 produced dose-dependent increases in left ventricular dP/dt(max) (control 1168+/-138 mm Hg/s versus AM2 high-dose 1402+/-130 mm Hg/s; P<0.01) and cardiac output (2.09+/-0.66 L/min versus 3.81+/-0.30 L/min; P<0.001) and reductions in calculated total peripheral resistance (40+/-6 mm Hg(L.min) versus 21+/-4 mm Hg(L.min); P<0.001), mean arterial pressure (74.4+/-2.4 mm Hg versus 66.2+/-2.5 mm Hg; P<0.001), and left atrial pressure (23.3+/-1.0 mm Hg versus 18.8+/-1.3 mm Hg; P<0.001). AM2 administration also induced significant elevations in plasma cAMP (P<0.01) in association with rises in atrial (P<0.05) and brain (P<0.01) natriuretic peptides and plasma renin activity (P<0.01). Despite the increase in renin activity, plasma aldosterone levels were not significantly altered, whereas the aldosterone/plasma renin activity ratio was reduced (P=0.08). Plasma vasopressin, endothelin-1, and catecholamines levels were also unchanged by AM2. Renal effects of AM2 included increased excretion of sodium (P<0.05), cAMP (P<0.01), and creatinine (P<0.05), with augmented creatinine clearance (P<0.05), and a trend for urine output to rise (P=0.068). CONCLUSIONS These results indicate that AM2 administration has favorable effects on cardiovascular, endocrine, and renal indexes in heart failure and identify the peptide as a potential therapeutic target in this disease.
Collapse
Affiliation(s)
- Miriam T Rademaker
- Christchurch Cardioendocrine Research Group, University of Otago, Christchurch, New Zealand.
| | | | | | | |
Collapse
|
24
|
Bisping E, Tenderich G, Barckhausen P, Stumme B, Bruns S, von Lewinski D, Pieske B. Atrial myocardium is the predominant inotropic target of adrenomedullin in the human heart. Am J Physiol Heart Circ Physiol 2007; 293:H3001-7. [PMID: 17766467 DOI: 10.1152/ajpheart.01276.2006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Adrenomedullin (ADM) is an endogenous peptide with favorable hemodynamic effects in vivo. In this study, we characterized the direct functional effects of ADM in isolated preparations from human atria and ventricles. In electrically stimulated human nonfailing right atrial trabeculae, ADM (0.0001–1 μmol/l) increased force of contraction in a concentration-dependent manner, with a maximal increase by 35 ± 8% (at 1 μmol/l; P < 0.05). The positive inotropic effect was accompanied by a disproportionate increase in calcium transients assessed by aequorin light emission [by 76 ± 20%; force/light ratio (ΔF/ΔL) 0.58 ± 0.15]. In contrast, elevation of extracellular calcium (from 2.5 to 3.2 mmol/l) proportionally increased force and aequorin light emission (ΔF/ΔL 1.0 ± 0.1; P < 0.05 vs. ADM). Consistent with a cAMP-dependent mechanism, ADM (1 μmol/l) increased atrial cAMP levels by 90 ± 12%, and its inotropic effects could be blocked by the protein kinase A (PKA) inhibitor H-89. ADM also exerted positive inotropic effects in failing atrial myocardium and in nonfailing and failing ventricular myocardium. The inotropic response was significantly weaker in ventricular vs. atrial myocardium and in failing vs. nonfailing myocardium. In conclusion, ADM exerts Ca2+-dependent positive inotropic effects in human atrial and less-pronounced effects in ventricular myocardium. The inotropic effects are related to increased cAMP levels and stimulation of PKA. In heart failure, the responsiveness to ADM is reduced in atria and ventricles.
Collapse
Affiliation(s)
- Egbert Bisping
- Department of Cardiology, University of Göttingen, Göttingen, Germany
| | | | | | | | | | | | | |
Collapse
|
25
|
Ertmer C, Van Aken H, Westphal M. Adrenomedullin in the Treatment of Cardiovascular Dysfunction and Sepsis. Intensive Care Med 2007. [DOI: 10.1007/978-0-387-49518-7_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
26
|
|
27
|
Westphal M, Sander J, Van Aken H, Ertmer C, Stubbe HD, Booke M. [Role of adrenomedullin in the pathogenesis and treatment of cardiovascular dysfunctions and sepsis]. Anaesthesist 2006; 55:171-8. [PMID: 15997387 DOI: 10.1007/s00101-005-0888-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Adrenomedullin (AM) is an endogenous vasodilatory peptide hormone, which plays a key role in the regulation and preservation of cardiovascular and pulmonary functions. Clinical and experimental studies have demonstrated that AM represents an alternative therapeutic option in the treatment of pulmonary hypertension. In addition, AM proved to be useful in the treatment of cardiovascular dysfunctions, such as arterial hypertension and congestive heart failure following myocardial infarction. Recent research has also shown that AM plays a pivotal role in the development of sepsis-associated hemodynamic and microcirculatory disorders. Experimental studies also suggest that infusion of exogenous AM might be a rational approach to prevent and treat hypodynamic septic shock. The objectives of this review article are to characterize the regulative properties of AM and to discuss clinical and experimental studies which allow to judge the role of AM in the setting of cardiovascular dysfunction and sepsis.
Collapse
Affiliation(s)
- M Westphal
- Klinik und Poliklinik für Anästhesiologie und operative Intensivmedizin, Universitätsklinikum, Münster.
| | | | | | | | | | | |
Collapse
|
28
|
Yanagawa B, Nagaya N. Adrenomedullin: molecular mechanisms and its role in cardiac disease. Amino Acids 2006; 32:157-64. [PMID: 16583314 DOI: 10.1007/s00726-005-0279-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2005] [Accepted: 12/03/2005] [Indexed: 10/24/2022]
Abstract
Adrenomedullin (AM) is a potent, long-lasting vasoactive peptide originally isolated from human pheochromocytoma. Since its discovery, serum and tissue AM expression have been shown to be increased in experimental models and in patients with cardiac hypertrophy, myocardial infarction and end-stage heart failure with several beneficial effects. Considerable evidence exists for a wide range of autocrine, paracrine and endocrine mechanisms for AM which include vasodilatory, anti-apoptotic, angiogenic, anti-fibrotic, natriuretic, diuretic and positive inotropic. Thus, through regulation of body fluid or direct cardiac mechanisms, AM has additive and beneficial effects in the context of heart disease. Notable molecular mechanisms of AM include cyclic adenosine monophosphate, guanosine-3',5'-monophosphate, PI3K/Akt and MAPK-ERK-mediated cascades. Given the endogenous and multifunctional nature of AM, we consider this molecule to have great potential in the treatment of cardiovascular diseases. In agreement, early experimental and preliminary clinical studies suggest that AM is a new and promising therapy for cardiovascular diseases.
Collapse
Affiliation(s)
- B Yanagawa
- Department of Regenerative Medicine and Tissue Engineering, National Cardiovascular Center Research Institute, Osaka, Japan
| | | |
Collapse
|
29
|
García MA, Martín-Santamaría S, de Pascual-Teresa B, Ramos A, Julián M, Martínez A. Adrenomedullin: a new and promising target for drug discovery. Expert Opin Ther Targets 2006; 10:303-17. [PMID: 16548778 DOI: 10.1517/14728222.10.2.303] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Adrenomedullin (AM) is a 52 amino acid peptide that plays a critical role in several diseases such as hypertension, cancer, diabetes, cardiovascular and renal disorders, among others. Interestingly, AM behaves as a protective agent against some pathologies, yet is a stimulating factor for other disorders. Thus, AM can be considered as a new and promising target for the design of non-peptidic modulators that could be useful for the treatment of those pathologies, by regulating AM levels or the activity of AM. A full decade on from its discovery, much more is known about AM molecular biology and pharmacology, but this knowledge still needs to be applied to the development of clinically useful drugs.
Collapse
Affiliation(s)
- Mario A García
- Universidad San Pablo CEU, Departamento de Química, Facultad de Farmacia, Urbanización Montepríncipe, 28668 Boadilla del Monte, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
30
|
Ueda K, Teragawa H, Kimura M, Matsuda K, Higashi Y, Yamagata T, Oshima T, Yoshizumi M, Chayama K. Adrenomedullin causes coronary vasodilation in humans: effects of inhibition of nitric oxide synthesis. J Cardiovasc Pharmacol 2006; 46:534-9. [PMID: 16160609 DOI: 10.1097/01.fjc.0000179156.51985.db] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Experimental studies have shown that adrenomedullin (AM) causes vasodilation, in part, mediated by endothelium-derived nitric oxide (NO). However, it remains to be clarified how NO is involved in AM-induced coronary vasoreactivity in humans. We examined whether NO contributes to the vasodilatory effects of adrenomedullin on human coronary arteries. In 10 patients with angiographically normal coronary arteries, adrenomedullin (low dose: 1 ng/kg/min; high dose: 10 ng/kg/min) was infused into the left coronary ostium before and after an infusion of N-monomethyl-L-arginine (L-NMMA, 40 micromol/min for 5 min), an NO synthase inhibitor. Coronary diameter and coronary blood flow (CBF) were evaluated by quantitative angiography and Doppler flow velocity measurements. Changes in these parameters in response to adrenomedullin were expressed as percent changes from baseline values. Adrenomedullin at a high dose dilated coronary arteries (3.7+/-0.5%, P<0.001). Adrenomedullin increased the coronary blood flow at both doses (low: 55.7+/-13.9%, P<0.01; high: 48.8+/-9.8%, P<0.001). After the infusion of L-NMMA, adrenomedullin-induced coronary vasodilation and increase in coronary blood flow were attenuated. These findings suggest that adrenomedullin dilates human coronary arteries through an increase in NO production, at least in part.
Collapse
Affiliation(s)
- Kentaro Ueda
- Department of Medicine and Molecular Science, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Yasu T, Nishikimi T, Kobayashi N, Ikeda N, Ueba H, Nakamura T, Funayama H, Kubo N, Kawakami M, Matsuoka H, Kangawa K, Saito M. Up-regulated synthesis of mature-type adrenomedullin in coronary circulation immediately after reperfusion in patients with anterior acute myocardial infarction. ACTA ACUST UNITED AC 2005; 129:161-6. [PMID: 15927712 DOI: 10.1016/j.regpep.2005.02.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2004] [Accepted: 02/04/2005] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Levels of adrenomedullin (AM), a potent vasodilatory peptide, have been shown to increase in the early stage of acute myocardial infarction (AMI). The purpose of this study was to determine whether coronary sinus-aortic step-up of mature forms of AM is accelerated in patients with AMI after reperfusion. METHODS The subjects were 29 consecutive patients with a first episode of anterior AMI and 10 normal controls. All patients with AMI underwent balloon reperfusion therapy within 24 h after symptom onset. Plasma levels of two molecular forms of AM (an active, mature form [AM-m] and an intermediate, inactive glycine-extended form [AM-Gly]) in the aorta and coronary sinus (CS) were measured by specific immunoradiometric assay after reperfusion. RESULTS Plasma levels of AM-m and AM-Gly in the aorta and CS were higher in AMI patients than in controls. CS-aortic step-up of AM-m, which is an index of myocardial production of AM-m, was significantly greater in AMI patients than in controls (1.7 +/- 1.4 vs. 0.4 +/- 0.3 pmol/L, P < 0.01). However, there was no significant difference in CS-aortic step-up of AM-Gly (P = 0.30). AMI patients with left ventricular dysfunction (n = 10) had a significantly higher CS-aortic AM-m step-up than AMI patients without left ventricular dysfunction (n = 19). AM-m in the aorta and CS negatively correlated with the left ventricular ejection fraction (r = -0.50, r = -0.48, P < 0.01). CONCLUSIONS Myocardial synthesis of AM-m is accelerated in patients with reperfused AMI, especially in patients with critical left ventricular dysfunction. Increased myocardial synthesis of active AM may protect against cardiac dysfunction, myocardial remodeling, or both after the onset of AMI.
Collapse
Affiliation(s)
- Takanori Yasu
- Department of Integrated Medicine I, Omiya Medical Center, Jichi Medical School, Saitama, Saitama, 330-8503 Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Hamid SA, Baxter GF. Adrenomedullin: regulator of systemic and cardiac homeostasis in acute myocardial infarction. Pharmacol Ther 2005; 105:95-112. [PMID: 15670621 DOI: 10.1016/j.pharmthera.2004.08.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
During and following acute myocardial infarction, a variety of endogenous mediators are elevated, one of which is adrenomedullin (AM). AM is a multifunctional peptide that has been identified as having a putative beneficial role following an ischemic insult at both systemic and local levels. Classically described as a potent vasodilator, natriuretic, and diuretic agent, experimental infarct models also demonstrate AM to exhibit antiproliferative and antiapoptotic functions in the myocardium, counterregulating the effects of mediators such as angiotensin-II and endothelin-1. Less well documented are the angiogenic and inflammatory modulating potentials of AM, which may also contribute toward reducing adverse ventricular remodeling. The review examines clinical and experimental studies, looking at the effects of AM and cellular mechanisms that could be involved in mediating cardioprotective effects and ultimately optimizing left ventricular remodeling. Finally, the possibility of enhancing endogenous actions of AM by pharmacological intervention is considered.
Collapse
Affiliation(s)
- Shabaz A Hamid
- Department of Basic Sciences, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK.
| | | |
Collapse
|
33
|
Katayama T, Nakashima H, Takagi C, Honda Y, Suzuki S, Yano K. Predictors of mortality in patients with acute myocardial infarction and cardiogenic shock. Circ J 2005; 69:83-8. [PMID: 15635209 DOI: 10.1253/circj.69.83] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Although cardiogenic shock (CS) is the leading cause of death for acute myocardial infarction (AMI) patients, reliable predictive factors in the acute stage, such as cardiovascular peptides, have not yet been identified. METHODS AND RESULTS In 42 consecutive AMI patients with CS on admission, successfully treated by primary percutaneous coronary intervention (PCI) within 12 h of onset, related factors including brain natriuretic peptide (BNP), atrial natriuretic peptide (ANP), renin, aldosterone, catecholamines, and adrenomedullin, were investigated 24 h from onset, as well as the 1-year mortality rates. During the 12-month follow-up period, 15 patients died from cardiovascular causes (group D). There were no significant differences in patient characteristics, angiographic findings, and left ventricular systolic function between group D subjects and the survivors (group S: n=27). Multivariate analysis identified high levels of adrenomedullin as an independent predictor of 1-year mortality (risk ratio: 6.42, 95% confidence interval, 1.49-43.31, p<0.05). CONCLUSIONS The acute-phase plasma concentration of adrenomedullin may be a reliable predictor of mortality in patients with AMI complicated by CS and successfully treated by direct PCI, as may be BNP concentration, peak-creatine kinase value, and ventricular fibrillation.
Collapse
Affiliation(s)
- Toshiro Katayama
- Department of Cardiovascular Medicine, Course of Medical and Dental Sciences, Graduate School of Biomedical Sciences, Nagasaki University School of Medicine, Nagasaki, Japan.
| | | | | | | | | | | |
Collapse
|
34
|
Katayama T, Nakashima H, Honda Y, Suzuki S, Yano K. Relationship between adrenomedullin and left-ventricular systolic function and mortality in acute myocardial infarction. Angiology 2005; 56:35-42. [PMID: 15678254 DOI: 10.1177/000331970505600105] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The purpose of this study was to investigate the relationship between plasma adrenomedullin concentration levels and left-ventricular systolic function in patients with acute myocardial infarction (AMI), and to assess whether these findings can be used to predict clinical outcomes, including mortality. One hundred twenty-four consecutive first AMI attack subjects were successfully reperfused with primary percutaneous coronary intervention therapy. Plasma adrenomedullin concentrations were evaluated at 24 hours from onset. Left ventriculograms of all patients taken in the acute (soon after reperfusion therapy) and subacute (21 +/-9 days after onset) phases were used to evaluate left-ventricular ejection fraction (LVEF), and the difference in LVEF (delta-LVEF) between the two stages calculated. There were significantly more patients with cardiogenic shock in the H-Adm group (above the median value of plasma adrenomedullin concentrations > or =3.5 Fmol/mL) than in the L-Adm (< 3.5 Fmol/mL) group (p<0.0001). There was significantly higher mortality in the H-Adm group (p<0.01). Multivariate analysis identified plasma adrenomedullin concentrations alone as an independent predictor of mortality (p<0.05). There were no significant differences in acute-stage LVEF between the groups. LVEF in the subacute stage was, however, significantly lower in the H-Adm group than in the L-Adm group (52 +/-12% vs 59 +/-11%, p<0.05). Also, delta-LVEF was significantly lower in the H-Adm group than in the L-Adm group (1.9 +/-9.7% vs 6.3 +/-10.3%, p<0.01). Plasma adrenomedullin concentrations in the early phase of AMI correlate closely with the severity of heart failure, and may offer important prognostic information about the risk of mortality. Our data suggest that plasma adrenomedullin concentrations may be an independent predictor of the deterioration of left-ventricular systolic function.
Collapse
Affiliation(s)
- Toshiro Katayama
- Department of Cardiology, Nagasaki Citizens Hospital, Nagasaki, Japan.
| | | | | | | | | |
Collapse
|
35
|
Bunton DC, Petrie MC, Hillier C, Johnston F, McMurray JJV. The clinical relevance of adrenomedullin: a promising profile? Pharmacol Ther 2005; 103:179-201. [PMID: 15464589 DOI: 10.1016/j.pharmthera.2004.07.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Adrenomedullin (AM) is a peptide that possesses potentially beneficial properties. Since the initial discovery of the peptide by Kitamura et al. in 1993, the literature has been awash with reports describing its novel mechanisms of action and huge potential as a therapeutic target. Strong evidence now exists that AM is able to act as an autocrine, paracrine, or endocrine mediator in a number of biologically significant functions, including the endothelial regulation of blood pressure, protection against organ damage in sepsis or hypoxia, and the control of blood volume through the regulation of thirst. Its early promise as a potential mediator/modulator of disease was not, however, entirely as a result of the discovery of physiological functions but due more to the observation of increasing levels measured in plasma in direct correlation with disease progression. In health, AM circulates at low picomolar concentrations in plasma in 2 forms, a mature 52-amino acid peptide and an immature 53-amino acid peptide. Plasma levels of AM have now been shown to be increased in a number of pathological states, including congestive heart failure, sepsis, essential hypertension, acute myocardial infarction, and renal impairment. These earliest associations have been further supplemented with evidence of a role for AM in other pathologies including, most intriguingly, cancer. In this review, we offer a timely review of our current knowledge on AM and give a detailed account of the putative role of AM in those clinical areas in which the best therapeutic opportunities might exist.
Collapse
Affiliation(s)
- David C Bunton
- Vascular Assessment Unit, Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Cowcaddens Road, Glasgow G4 0BA, UK.
| | | | | | | | | |
Collapse
|
36
|
Katayama T, Nakashima H, Furudono S, Honda Y, Suzuki S, Yano K. Evaluation of neurohumoral activation (adrenomedullin, BNP, catecholamines, etc.) in patients with acute myocardial infarction. Intern Med 2004; 43:1015-22. [PMID: 15609694 DOI: 10.2169/internalmedicine.43.1015] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE The object of our study was to identify the most useful predictor of patient prognosis in acute myocardial infarction (AMI), from 7 acute-phase cardiovascular peptides which take part in neurohumoral activation [brain natriuretic peptide (BNP), atrial natriuretic peptide (ANP), renin, aldosterone, adrenomedullin, epinephrine and norepinephrine]. METHODS In 141 consecutive AMI patients, 24 hours from onset, we evaluated plasma concentration levels of the 7 types of cardiovascular peptides and the relationships between the values of these peptides and short-term clinical prognosis, including mortality. RESULTS Plasma levels of all cardiovascular peptides were significantly higher in patients who suffered mortality than in surviving patients (BNP: 1,267+/-997 pg/ml vs. 293+/-327 pg/ml, p<0.0001; ANP: 164+/-186 pg/ml vs. 64+/-76 pg/ml, p<0.001; adrenomedullin: 13.61+/-3.29 Fmol/l vs. 3.45+/-1.52 Fmol/I, p<0.0001; renin: 8.79+/-7.15 ng/ml/h vs. 4.34+/-5.10 ng/ml/h, p<0.01; aldosterone: 249+/-210 pg/ml vs. 68+/-74 pg/ml, p<0.0001; epinephrine: 3,191+/-8,360 pg/ml vs. 68+/-74 pg/ml, p<0.0001; norepinephrine: 21.8+/-46.2 ng/ml vs. 0.9+/-0.8, ng/ml p<0.0001). Multivariate analysis identified only high levels of adrenomedullin as an independent related factor of cardiogenic shock (risk ratio: 5.84, 95% C.I.: 1.80-18.95, p=0.003), and as an independent predictor of short-term mortality (risk ratio: 16.16, 95% C.I.: 1.38-189.71, p=0.03). CONCLUSIONS Acute-phase neurohumoral activation, involving renin, aldosterone, epinephrine, norepinephrine, BNP, ANP, and adrenomedullin may be closely related to poor patient outcomes, including mortality. Our results suggest that acute-phase plasma adrenomedullin concentrations may be the most useful predictor of patient prognosis in the setting of AMI, out of the 7 types of cardiovascular peptides involved in neurohumoral activation.
Collapse
Affiliation(s)
- Toshiro Katayama
- Department of Cardiovascular Medicine and Course of Medical and Dental Science, Graduate School of Biomedical Science, Nagasaki University, Nagasaki
| | | | | | | | | | | |
Collapse
|
37
|
Abstract
The vasodilator hydralazine, used clinically in cardiovascular therapy, relaxes arterial smooth muscle by inhibiting accumulation of intracellular free Ca2+ via an unidentified primary target. Collagen prolyl hydroxylase is a known target of hydralazine. We therefore investigated whether inhibition of other members of this enzyme family, namely the hypoxia-inducible factor (HIF)-regulating O2-dependent prolyl hydroxylase domain (PHD) enzymes, could represent a novel mechanism of action. Hydralazine induced rapid and transient expression of HIF-1alpha and downstream targets of HIF (endothelin-1, adrenomedullin, haem oxygenase 1, and vascular endothelial growth factor [VEGF]) in endothelial and smooth muscle cells and induced endothelial cell-specific proliferation. Hydralazine dose-dependently inhibited PHD activity and induced nonhydroxylated HIF-1alpha, evidence for HIF stabilization specifically by inhibition of PHD enzyme activity. In vivo, hydralazine induced HIF-1alpha and VEGF protein in tissue extracts and elevated plasma VEGF levels. In sponge angiogenesis assays, hydralazine increased stromal cell infiltration and blood vessel density versus control animals. Thus, hydralazine activates the HIF pathway through inhibition of PHD activity and initiates a pro-angiogenic phenotype. This represents a novel mechanism of action for hydralazine and presents HIF as a potential target for treatment of ischemic disease.
Collapse
MESH Headings
- Adrenomedullin
- Angiogenesis Inducing Agents/pharmacology
- Animals
- Breast Neoplasms/pathology
- Carcinoma/pathology
- Carcinoma, Renal Cell/pathology
- Cell Hypoxia
- Cell Line, Tumor/drug effects
- Cell Line, Tumor/metabolism
- Cells, Cultured/drug effects
- Cells, Cultured/metabolism
- DNA-Binding Proteins/biosynthesis
- DNA-Binding Proteins/genetics
- Dose-Response Relationship, Drug
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Endothelin-1/biosynthesis
- Endothelin-1/genetics
- Enzyme Inhibitors/pharmacology
- Gene Expression Regulation/drug effects
- Heme Oxygenase (Decyclizing)/biosynthesis
- Heme Oxygenase (Decyclizing)/genetics
- Heme Oxygenase-1
- Humans
- Hydralazine/pharmacology
- Hypoxia-Inducible Factor 1
- Hypoxia-Inducible Factor 1, alpha Subunit
- Implants, Experimental
- Kidney Neoplasms/pathology
- Membrane Proteins
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Nude
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Neovascularization, Physiologic/drug effects
- Nuclear Proteins/biosynthesis
- Nuclear Proteins/genetics
- Peptides/genetics
- Peptides/metabolism
- Procollagen-Proline Dioxygenase/antagonists & inhibitors
- Procollagen-Proline Dioxygenase/physiology
- Transcription Factors/biosynthesis
- Transcription Factors/genetics
- Vascular Endothelial Growth Factor A/biosynthesis
- Vascular Endothelial Growth Factor A/genetics
- Vasodilator Agents/pharmacology
Collapse
Affiliation(s)
- Helen J Knowles
- Cancer Research UK Molecular Oncology Laboratory, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK
| | | | | | | |
Collapse
|
38
|
Okumura H, Nagaya N, Itoh T, Okano I, Hino J, Mori K, Tsukamoto Y, Ishibashi-Ueda H, Miwa S, Tambara K, Toyokuni S, Yutani C, Kangawa K. Adrenomedullin Infusion Attenuates Myocardial Ischemia/Reperfusion Injury Through the Phosphatidylinositol 3-Kinase/Akt-Dependent Pathway. Circulation 2004; 109:242-8. [PMID: 14691041 DOI: 10.1161/01.cir.0000109214.30211.7c] [Citation(s) in RCA: 115] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Background—
Infusion of adrenomedullin (AM) has beneficial hemodynamic effects in patients with heart failure. However, the effect of AM on myocardial ischemia/reperfusion remains unknown.
Methods and Results—
Male Sprague-Dawley rats were exposed to a 30-minute period of ischemia induced by ligation of the left coronary artery. They were randomized to receive AM, AM plus wortmannin (a phosphatidylinositol 3-kinase [PI3K] inhibitor), or saline for 60 minutes after coronary ligation. Hemodynamics and infarct size were examined 24 hours after reperfusion. Myocardial apoptosis was also examined 6 hours after reperfusion. The effect of AM on Akt phosphorylation in cardiac tissues was examined by Western blotting. Intravenous administration of AM significantly reduced myocardial infarct size (28±4% to 16±1%,
P
<0.01), left ventricular end-diastolic pressure (19±2 to 8±2 mm Hg,
P
<0.05), and myocardial apoptotic death (19±2% to 9±4%,
P
<0.05). Western blot analysis showed that AM infusion accelerated Akt phosphorylation in cardiac tissues and that pretreatment with wortmannin significantly attenuated AM-induced Akt phosphorylation. Moreover, pretreatment with wortmannin abolished the beneficial effects of AM: a reduction of infarct size, a decrease in left ventricular end-diastolic pressure, and inhibition of myocardial apoptosis after ischemia/reperfusion.
Conclusions—
Short-term infusion of AM significantly attenuated myocardial ischemia/reperfusion injury. These cardioprotective effects are attributed mainly to antiapoptotic effects of AM via a PI3K/Akt-dependent pathway.
Collapse
Affiliation(s)
- Hiroyuki Okumura
- Department of Biochemistry, National Cardiovascular Center Research Institute, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Matteo RD, May CN. Direct coronary vasodilator action of adrenomedullin is mediated by nitric oxide. Br J Pharmacol 2003; 140:1414-20. [PMID: 14623767 PMCID: PMC1574158 DOI: 10.1038/sj.bjp.0705572] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Increased circulating levels of adrenomedullin (ADM) cause peripheral vasodilatation and hypotension, accompanied by cardiac actions including tachycardia and increases in cardiac contractility, cardiac output, coronary conductance (CC) and coronary blood flow (CBF). It is unclear to what extent these cardiac effects are direct actions of ADM or secondary to the hypotension and altered cardiac loading. The direct cardiac actions of ADM were examined in conscious sheep previously implanted with aortic and coronary flow probes, and an indwelling left coronary artery cannula. Responses to infusion of ADM (0.5 microg kg(-1) h(-1) for 1 h) into the left coronary artery or jugular vein were compared (n=6). The effect of blockade of nitric oxide (NO) synthase with intracoronary (i.c.) N(omega)-nitro-l-arginine (l-NNA; 1.5 mg kg(-1) h(-1), infused for 2 h before and during ADM infusion, was assessed to determine whether the responses to ADM were mediated by NO (n=5). I.c. ADM caused large and sustained increases in CC (0.35+/-0.07-0.55+/-0.13 ml min(-1) mmHg-1, P<0.05) and CBF (28+/-6-42+/-9 ml min(-1), P<0.05), but had no effect on arterial pressure or indices of cardiac contractility (first differential of the upstroke of systole and peak aortic flow rate). Intravenous infusion of ADM had no effects. I.c. l-NNA, at a dose that abolished the coronary vasodilator action of acetylcholine, blocked ADM-induced coronary vasodilatation. In conclusion, ADM had a direct coronary vasodilator action that was mediated by release of endogenous NO and resulted in increased CBF. There was no evidence for a direct inotropic action of ADM.
Collapse
Affiliation(s)
- R De Matteo
- Howard Florey Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | - C N May
- Howard Florey Institute, University of Melbourne, Parkville, Victoria 3010, Australia
- Author for correspondence:
| |
Collapse
|
40
|
Nishikimi T, Yoshihara F, Mori Y, Kangawa K, Matsuoka H. Cardioprotective effect of adrenomedullin in heart failure. Hypertens Res 2003; 26 Suppl:S121-7. [PMID: 12630822 DOI: 10.1291/hypres.26.s121] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Many neurohumoral factors participate in the pathophysiology of heart failure, and adrenomedullin (AM) may be involved in their derangement. This work reviews the accumulating evidence in support of a compensatory role of AM in heart failure, and describes the possible mechanisms of this role. It has been established that plasma AM levels are increased in patients with heart failure in proportion to the severity of the disease. Furthermore, recent studies suggest that plasma AM level is an independent prognostic indicator of heart failure. Thus, AM may be not only a biochemical marker for evaluating the severity of heart failure, but also a prognostic indicator of this syndrome. In patients with heart failure, AM production is increased not only in the plasma, but also in the heart. AM secretion from the failing human heart is also increased, but this increase is small and responds slowly to the stimulus. This phenomenon may be explained by the fact that AM is secreted via a constitutive pathway and that AM is an autocrine and/or a paracrine factor in the heart. An experiment using cultured myocytes suggested that cytokines and mechanical stress are important stimuli for AM production in the heart. Regarding the action of AM in the heart, recent studies have suggested that AM exerts an inotropic action both in vitro and in vivo. AM also attenuates cardiac hypertrophy in myocytes and inhibits proliferation and collagen production in cardiac fibroblasts. These results suggest that AM may be an antifibrotic, antihypertrophic, and positive inotropic factor in the failing and hypertrophied heart. Because AM has many cardiorenal actions, AM administration may be useful for the treatment of heart failure. Indeed, acute administration of AM has been shown to improve the hemodynamics, renal function, and hormonal parameters in patients with heart failure. Moreover, recent studies have shown that AM gene therapy or long-term AM infusion significantly improved cardiac hypertrophy and fibrosis, and prolonged the survival time in an animal model of hypertension and heart failure. In conclusion, these findings suggest that AM plays a compensatory role in the pathophysiology of heart failure and that administration of AM may be a new and promising approach for the treatment of patients with this syndrome.
Collapse
Affiliation(s)
- Toshio Nishikimi
- Department of Hypertension and Cardiorenal Medicine, Dokkyo University School of Medicine, 880 Kitakobayashi, Mibu-machi, Shimotsuga-gun, Tochigi 321-0293, Japan.
| | | | | | | | | |
Collapse
|
41
|
Kis B, Abrahám CS, Deli MA, Kobayashi H, Niwa M, Yamashita H, Busija DW, Ueta Y. Adrenomedullin, an autocrine mediator of blood-brain barrier function. Hypertens Res 2003; 26 Suppl:S61-70. [PMID: 12630813 DOI: 10.1291/hypres.26.s61] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Since the discovery that adrenomedullin gene expression is 20- to 40-fold higher in endothelial cells than even in the adrenal medulla, this peptide has been regarded as an important secretory product of the vascular endothelium, together with nitric oxide, eicosanoids, endothelin-1, and other vasoactive metabolites. Cerebral endothelial cells secrete an exceptionally large amount of adrenomedullin, and the adrenomedullin concentration is about 50% higher in the cerebral circulation than in the peripheral vasculature. The adrenomedullin production of cerebral endothelial cells is induced by astrocyte-derived factors. Adrenomedullin causes vasodilation in the cerebral circulation, may participate in the maintenance of the resting cerebral blood flow, and may be protective against ischemic brain injury. Recent data from our laboratory indicate that adrenomedullin, as an endothelium-derived autocrine/paracrine hormone, plays an important role in the regulation of specific blood-brain barrier properties. Adrenomedullin is suggested to be one of the physiological links between astrocyte-derived factors, cyclic adenosine 3'5'-monophosphate (cAMP), and the induction and maintenance of the blood-brain barrier. Moreover, the role of adrenomedullin in the differentiation and proliferation of endothelial cells and in angiogenesis suggests a more complex function for adrenomedullin in the cerebral circulation and in the development of the blood-brain barrier.
Collapse
Affiliation(s)
- Béla Kis
- Department of Physiology and Pharmacology, Wake Forest University, Medical Center Boulevard, Winston-Salem, NC 27157, USA.
| | | | | | | | | | | | | | | |
Collapse
|