251
|
Verweij SL, van der Valk FM, Stroes ESG. Novel directions in inflammation as a therapeutic target in atherosclerosis. Curr Opin Lipidol 2015; 26:580-5. [PMID: 26382552 DOI: 10.1097/mol.0000000000000233] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Atherosclerosis is a chronic disease of the arterial wall largely driven by inflammation; hence, therapeutics targeting inflammatory pathways are considered an attractive strategy in atherosclerotic cardiovascular disease (ASCVD). The purpose of this review is to describe the randomized, placebo-controlled clinical trials currently investigating the impact of anti-inflammatory strategies in ASCVD patients, to discuss novel insights and targets into the role of innate immunity in atherosclerosis and to address the promise of local drug delivery as opposed to systemic therapies in atherosclerotic disease. RECENT FINDINGS The first clinical trials using systemic anti-inflammatory drugs in ASCVD patients might be able to strengthen the case for immunomodulation once showing an improved ASCVD outcome. Several specific targets in innate immunity bear therapeutic potential, of which some have already entered the clinical arena. To prevent immunosuppression by systemic effects, drug delivery systems are increasingly being applied to locally attenuate plaque inflammation. SUMMARY Anti-inflammatory therapies seem promising for future treatment of ASCVD. In view of the risk of immunosuppression in case of long term and systemic use of anti-inflammatory drugs, there is a clinical need for highly selective and targeted therapies in patients with atherosclerosis.
Collapse
Affiliation(s)
- Simone L Verweij
- Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | |
Collapse
|
252
|
Sager HB, Heidt T, Hulsmans M, Dutta P, Courties G, Sebas M, Wojtkiewicz GR, Tricot B, Iwamoto Y, Sun Y, Weissleder R, Libby P, Swirski FK, Nahrendorf M. Targeting Interleukin-1β Reduces Leukocyte Production After Acute Myocardial Infarction. Circulation 2015; 132:1880-90. [PMID: 26358260 PMCID: PMC4651795 DOI: 10.1161/circulationaha.115.016160] [Citation(s) in RCA: 204] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Accepted: 09/02/2015] [Indexed: 02/04/2023]
Abstract
BACKGROUND Myocardial infarction (MI) is an ischemic wound that recruits millions of leukocytes. MI-associated blood leukocytosis correlates inversely with patient survival, yet the signals driving heightened leukocyte production after MI remain incompletely understood. METHODS AND RESULTS With the use of parabiosis surgery, this study shows that soluble danger signals, among them interleukin-1β, increase bone marrow hematopoietic stem cell proliferation after MI. Data obtained in bone marrow reconstitution experiments reveal that interleukin-1β enhances hematopoietic stem cell proliferation by both direct actions on hematopoietic cells and through modulation of the bone marrow's hematopoietic microenvironment. An antibody that neutralizes interleukin-1β suppresses these effects. Anti-interleukin-1β treatment dampens the post-MI increase in hematopoietic stem cell proliferation. Consequently, decreased leukocyte numbers in the blood and infarct reduce inflammation and diminish post-MI heart failure in ApoE(-/-) mice with atherosclerosis. CONCLUSIONS The presented insight into post-MI bone marrow activation identifies a mechanistic target for muting inflammation in the ischemically damaged heart.
Collapse
Affiliation(s)
- Hendrik B Sager
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Timo Heidt
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Maarten Hulsmans
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Partha Dutta
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Gabriel Courties
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Matthew Sebas
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Gregory R Wojtkiewicz
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Benoit Tricot
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Yoshiko Iwamoto
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Yuan Sun
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Ralph Weissleder
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Peter Libby
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Filip K Swirski
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.)
| | - Matthias Nahrendorf
- From Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, Boston, MA (H.B.S., T.H., M.H., P.D., G.C., M.S., G.R.W., B.T., Y.I., Y.S., R.W., F.K.S., M.N.); Department of Systems Biology, Harvard Medical School, Boston, MA (R.W.); and Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA (P.L.).
| |
Collapse
|
253
|
Turner NA. Inflammatory and fibrotic responses of cardiac fibroblasts to myocardial damage associated molecular patterns (DAMPs). J Mol Cell Cardiol 2015; 94:189-200. [PMID: 26542796 DOI: 10.1016/j.yjmcc.2015.11.002] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 10/30/2015] [Accepted: 11/01/2015] [Indexed: 02/07/2023]
Abstract
Cardiac fibroblasts (CF) are well-established as key regulators of extracellular matrix (ECM) turnover in the context of myocardial remodelling and fibrosis. Recently, this cell type has also been shown to act as a sensor of myocardial damage by detecting and responding to damage-associated molecular patterns (DAMPs) upregulated with cardiac injury. CF express a range of innate immunity pattern recognition receptors (TLRs, NLRs, IL-1R1, RAGE) that are stimulated by a host of different DAMPs that are evident in the injured or remodelling myocardium. These include intracellular molecules released by necrotic cells (heat shock proteins, high mobility group box 1 protein, S100 proteins), proinflammatory cytokines (interleukin-1α), specific ECM molecules up-regulated in response to tissue injury (fibronectin-EDA, tenascin-C) or molecules modified by a pathological environment (advanced glycation end product-modified proteins observed with diabetes). DAMP receptor activation on fibroblasts is coupled to altered cellular function including changes in proliferation, migration, myofibroblast transdifferentiation, ECM turnover and production of fibrotic and inflammatory paracrine factors, which directly impact on the heart's ability to respond to injury. This review gives an overview of the important role played by CF in responding to myocardial DAMPs and how the DAMP/CF axis could be exploited experimentally and therapeutically.
Collapse
Affiliation(s)
- Neil A Turner
- Division of Cardiovascular & Diabetes Research, and Multidisciplinary Cardiovascular Research Centre (MCRC), University of Leeds, Leeds, UK.
| |
Collapse
|
254
|
Deftereos S, Giannopoulos G, Angelidis C, Alexopoulos N, Filippatos G, Papoutsidakis N, Sianos G, Goudevenos J, Alexopoulos D, Pyrgakis V, Cleman MW, Manolis AS, Tousoulis D, Lekakis J. Anti-Inflammatory Treatment With Colchicine in Acute Myocardial Infarction: A Pilot Study. Circulation 2015; 132:1395-1403. [PMID: 26265659 DOI: 10.1161/circulationaha.115.017611] [Citation(s) in RCA: 195] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 08/05/2015] [Indexed: 12/26/2022]
Abstract
BACKGROUND Inflammatory processes have been identified as key mediators of the deleterious effects of ischemia/reperfusion in ST-segment-elevation myocardial infarction. Colchicine is a substance with potent anti-inflammatory properties, suitable for safe use in patients with cardiovascular disease. The purpose of this study was to test the hypothesis that a short course of colchicine treatment could lead to reduced infarct size. METHODS AND RESULTS Patients presenting with ST-segment-elevation myocardial infarction ≤12 hours from pain onset (treated with primary percutaneous coronary intervention) were randomly assigned to colchicine or placebo for 5 days. The primary outcome parameter was the area under the curve of creatine kinase-myocardial brain fraction concentration. A subset of patients underwent cardiac MRI with late gadolinium enhancement 6 to 9 days after the index ST-segment-elevation myocardial infarction. One hundred fifty-one patients were included (60 in the MRI substudy). The area under the creatine kinase-myocardial brain fraction curve was 3144 (interquartile range [IQR], 1754-6940) ng·h(-1)·mL(-1) in the colchicine group in comparison with 6184 (IQR, 4456-6980) ng·h(-1)·mL(-1) in controls (P<0.001). Indexed MRI-late gadolinium enhancement-defined infarct size was 18.3 (IQR, 7.6-29.9) mL/1.73 m(2) in the colchicine group versus 23.2 (18.5-33.4) mL/1.73 m(2) in controls (P=0.019). The relative infarct size (as a proportion to left ventricular myocardial volume) was 13.0 (IQR, 8.0-25.3) % and 19.8 (IQR, 13.7-29.8) %, respectively (P=0.034). CONCLUSIONS These results suggest a potential benefit of colchicine in ST-segment-elevation myocardial infarction, but further clinical trials are necessary to draw secure conclusions, especially considering the fact that the present study was not powered to assess clinical end points. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT01936285.
Collapse
Affiliation(s)
- Spyridon Deftereos
- From Department of Cardiology, Athens General Hospital "G. Gennimatas", Greece (S.D., G.G., C.A., N.P., V.P.); 2nd Department of Cardiology, University of Athens Medical School, Attikon University Hospital, Greece (S.D., G.G., G.F., J.L.); Cardiology Department, Athens Euroclinic, Greece (N.A.); AHEPA University Hospital, Thessaloniki, Greece (G.S.); 1st Department of Cardiology, University of Ioannina Medical School, Greece (J.G.); Cardiology Department, University of Patras, Greece (D.A.); Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT (M.W.C.); and 1st Department of Cardiology, University of Athens Medical School, Hippokration Hospital, Greece (A.S.M., D.T.)
| | - Georgios Giannopoulos
- From Department of Cardiology, Athens General Hospital "G. Gennimatas", Greece (S.D., G.G., C.A., N.P., V.P.); 2nd Department of Cardiology, University of Athens Medical School, Attikon University Hospital, Greece (S.D., G.G., G.F., J.L.); Cardiology Department, Athens Euroclinic, Greece (N.A.); AHEPA University Hospital, Thessaloniki, Greece (G.S.); 1st Department of Cardiology, University of Ioannina Medical School, Greece (J.G.); Cardiology Department, University of Patras, Greece (D.A.); Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT (M.W.C.); and 1st Department of Cardiology, University of Athens Medical School, Hippokration Hospital, Greece (A.S.M., D.T.).
| | - Christos Angelidis
- From Department of Cardiology, Athens General Hospital "G. Gennimatas", Greece (S.D., G.G., C.A., N.P., V.P.); 2nd Department of Cardiology, University of Athens Medical School, Attikon University Hospital, Greece (S.D., G.G., G.F., J.L.); Cardiology Department, Athens Euroclinic, Greece (N.A.); AHEPA University Hospital, Thessaloniki, Greece (G.S.); 1st Department of Cardiology, University of Ioannina Medical School, Greece (J.G.); Cardiology Department, University of Patras, Greece (D.A.); Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT (M.W.C.); and 1st Department of Cardiology, University of Athens Medical School, Hippokration Hospital, Greece (A.S.M., D.T.)
| | - Nikolaos Alexopoulos
- From Department of Cardiology, Athens General Hospital "G. Gennimatas", Greece (S.D., G.G., C.A., N.P., V.P.); 2nd Department of Cardiology, University of Athens Medical School, Attikon University Hospital, Greece (S.D., G.G., G.F., J.L.); Cardiology Department, Athens Euroclinic, Greece (N.A.); AHEPA University Hospital, Thessaloniki, Greece (G.S.); 1st Department of Cardiology, University of Ioannina Medical School, Greece (J.G.); Cardiology Department, University of Patras, Greece (D.A.); Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT (M.W.C.); and 1st Department of Cardiology, University of Athens Medical School, Hippokration Hospital, Greece (A.S.M., D.T.)
| | - Gerasimos Filippatos
- From Department of Cardiology, Athens General Hospital "G. Gennimatas", Greece (S.D., G.G., C.A., N.P., V.P.); 2nd Department of Cardiology, University of Athens Medical School, Attikon University Hospital, Greece (S.D., G.G., G.F., J.L.); Cardiology Department, Athens Euroclinic, Greece (N.A.); AHEPA University Hospital, Thessaloniki, Greece (G.S.); 1st Department of Cardiology, University of Ioannina Medical School, Greece (J.G.); Cardiology Department, University of Patras, Greece (D.A.); Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT (M.W.C.); and 1st Department of Cardiology, University of Athens Medical School, Hippokration Hospital, Greece (A.S.M., D.T.)
| | - Nikolaos Papoutsidakis
- From Department of Cardiology, Athens General Hospital "G. Gennimatas", Greece (S.D., G.G., C.A., N.P., V.P.); 2nd Department of Cardiology, University of Athens Medical School, Attikon University Hospital, Greece (S.D., G.G., G.F., J.L.); Cardiology Department, Athens Euroclinic, Greece (N.A.); AHEPA University Hospital, Thessaloniki, Greece (G.S.); 1st Department of Cardiology, University of Ioannina Medical School, Greece (J.G.); Cardiology Department, University of Patras, Greece (D.A.); Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT (M.W.C.); and 1st Department of Cardiology, University of Athens Medical School, Hippokration Hospital, Greece (A.S.M., D.T.)
| | - George Sianos
- From Department of Cardiology, Athens General Hospital "G. Gennimatas", Greece (S.D., G.G., C.A., N.P., V.P.); 2nd Department of Cardiology, University of Athens Medical School, Attikon University Hospital, Greece (S.D., G.G., G.F., J.L.); Cardiology Department, Athens Euroclinic, Greece (N.A.); AHEPA University Hospital, Thessaloniki, Greece (G.S.); 1st Department of Cardiology, University of Ioannina Medical School, Greece (J.G.); Cardiology Department, University of Patras, Greece (D.A.); Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT (M.W.C.); and 1st Department of Cardiology, University of Athens Medical School, Hippokration Hospital, Greece (A.S.M., D.T.)
| | - John Goudevenos
- From Department of Cardiology, Athens General Hospital "G. Gennimatas", Greece (S.D., G.G., C.A., N.P., V.P.); 2nd Department of Cardiology, University of Athens Medical School, Attikon University Hospital, Greece (S.D., G.G., G.F., J.L.); Cardiology Department, Athens Euroclinic, Greece (N.A.); AHEPA University Hospital, Thessaloniki, Greece (G.S.); 1st Department of Cardiology, University of Ioannina Medical School, Greece (J.G.); Cardiology Department, University of Patras, Greece (D.A.); Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT (M.W.C.); and 1st Department of Cardiology, University of Athens Medical School, Hippokration Hospital, Greece (A.S.M., D.T.)
| | - Dimitrios Alexopoulos
- From Department of Cardiology, Athens General Hospital "G. Gennimatas", Greece (S.D., G.G., C.A., N.P., V.P.); 2nd Department of Cardiology, University of Athens Medical School, Attikon University Hospital, Greece (S.D., G.G., G.F., J.L.); Cardiology Department, Athens Euroclinic, Greece (N.A.); AHEPA University Hospital, Thessaloniki, Greece (G.S.); 1st Department of Cardiology, University of Ioannina Medical School, Greece (J.G.); Cardiology Department, University of Patras, Greece (D.A.); Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT (M.W.C.); and 1st Department of Cardiology, University of Athens Medical School, Hippokration Hospital, Greece (A.S.M., D.T.)
| | - Vlasios Pyrgakis
- From Department of Cardiology, Athens General Hospital "G. Gennimatas", Greece (S.D., G.G., C.A., N.P., V.P.); 2nd Department of Cardiology, University of Athens Medical School, Attikon University Hospital, Greece (S.D., G.G., G.F., J.L.); Cardiology Department, Athens Euroclinic, Greece (N.A.); AHEPA University Hospital, Thessaloniki, Greece (G.S.); 1st Department of Cardiology, University of Ioannina Medical School, Greece (J.G.); Cardiology Department, University of Patras, Greece (D.A.); Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT (M.W.C.); and 1st Department of Cardiology, University of Athens Medical School, Hippokration Hospital, Greece (A.S.M., D.T.)
| | - Michael W Cleman
- From Department of Cardiology, Athens General Hospital "G. Gennimatas", Greece (S.D., G.G., C.A., N.P., V.P.); 2nd Department of Cardiology, University of Athens Medical School, Attikon University Hospital, Greece (S.D., G.G., G.F., J.L.); Cardiology Department, Athens Euroclinic, Greece (N.A.); AHEPA University Hospital, Thessaloniki, Greece (G.S.); 1st Department of Cardiology, University of Ioannina Medical School, Greece (J.G.); Cardiology Department, University of Patras, Greece (D.A.); Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT (M.W.C.); and 1st Department of Cardiology, University of Athens Medical School, Hippokration Hospital, Greece (A.S.M., D.T.)
| | - Antonis S Manolis
- From Department of Cardiology, Athens General Hospital "G. Gennimatas", Greece (S.D., G.G., C.A., N.P., V.P.); 2nd Department of Cardiology, University of Athens Medical School, Attikon University Hospital, Greece (S.D., G.G., G.F., J.L.); Cardiology Department, Athens Euroclinic, Greece (N.A.); AHEPA University Hospital, Thessaloniki, Greece (G.S.); 1st Department of Cardiology, University of Ioannina Medical School, Greece (J.G.); Cardiology Department, University of Patras, Greece (D.A.); Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT (M.W.C.); and 1st Department of Cardiology, University of Athens Medical School, Hippokration Hospital, Greece (A.S.M., D.T.)
| | - Dimitrios Tousoulis
- From Department of Cardiology, Athens General Hospital "G. Gennimatas", Greece (S.D., G.G., C.A., N.P., V.P.); 2nd Department of Cardiology, University of Athens Medical School, Attikon University Hospital, Greece (S.D., G.G., G.F., J.L.); Cardiology Department, Athens Euroclinic, Greece (N.A.); AHEPA University Hospital, Thessaloniki, Greece (G.S.); 1st Department of Cardiology, University of Ioannina Medical School, Greece (J.G.); Cardiology Department, University of Patras, Greece (D.A.); Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT (M.W.C.); and 1st Department of Cardiology, University of Athens Medical School, Hippokration Hospital, Greece (A.S.M., D.T.)
| | - John Lekakis
- From Department of Cardiology, Athens General Hospital "G. Gennimatas", Greece (S.D., G.G., C.A., N.P., V.P.); 2nd Department of Cardiology, University of Athens Medical School, Attikon University Hospital, Greece (S.D., G.G., G.F., J.L.); Cardiology Department, Athens Euroclinic, Greece (N.A.); AHEPA University Hospital, Thessaloniki, Greece (G.S.); 1st Department of Cardiology, University of Ioannina Medical School, Greece (J.G.); Cardiology Department, University of Patras, Greece (D.A.); Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT (M.W.C.); and 1st Department of Cardiology, University of Athens Medical School, Hippokration Hospital, Greece (A.S.M., D.T.)
| |
Collapse
|
255
|
Marcuzzi A, Piscianz E, Valencic E, Monasta L, Vecchi Brumatti L, Tommasini A. To Extinguish the Fire from Outside the Cell or to Shutdown the Gas Valve Inside? Novel Trends in Anti-Inflammatory Therapies. Int J Mol Sci 2015; 16:21277-21293. [PMID: 26370962 PMCID: PMC4613252 DOI: 10.3390/ijms160921277] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/19/2015] [Accepted: 08/31/2015] [Indexed: 12/26/2022] Open
Abstract
Cytokines are the most important soluble mediators of inflammation. Rare pediatric diseases provided exemplar conditions to study the anti-inflammatory efficacy of new generation therapies (biologics/biopharmaceuticals) selectively targeting single cytokines. Monoclonal antibodies and recombinant proteins have revolutionized anti-inflammatory therapies in the last two decades, allowing the specific targeting of single cytokines. They are very effective in extinguishing inflammation from outside the cell, even with the risk of an excessive and prolonged immunosuppression. Small molecules can enter the cell and shutdown the valve of inflammation by directly targeting signal proteins involved in cytokine release or in response to cytokines. They are orally-administrable drugs whose dosage can be easily adjusted to obtain the desired anti-inflammatory effect. This could make these drugs more suitable for a wide range of diseases as stroke, gout, or neurological impairment, where inflammatory activation plays a pivotal role as trigger. Autoinflammatory diseases, which have previously put anti-cytokine proteins in the limelight, can again provide a valuable model to measure the real potential of small inhibitors as anti-inflammatory agents.
Collapse
Affiliation(s)
- Annalisa Marcuzzi
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazzale Europa 1, Trieste 34128, Italy.
| | - Elisa Piscianz
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo" - , via dell'Istria, 65/1, Trieste 34137, Italy.
| | - Erica Valencic
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo" - , via dell'Istria, 65/1, Trieste 34137, Italy.
| | - Lorenzo Monasta
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo" - , via dell'Istria, 65/1, Trieste 34137, Italy.
| | - Liza Vecchi Brumatti
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo" - , via dell'Istria, 65/1, Trieste 34137, Italy.
| | - Alberto Tommasini
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo" - , via dell'Istria, 65/1, Trieste 34137, Italy.
| |
Collapse
|
256
|
Cavalli G, Dinarello CA. Treating rheumatological diseases and co-morbidities with interleukin-1 blocking therapies. Rheumatology (Oxford) 2015. [PMID: 26209330 DOI: 10.1093/rheumatology/kev269] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The inflammatory cytokines IL-1α and IL-1β orchestrate local and systemic inflammatory responses underlying a broad spectrum of diseases. Three agents for reducing IL-1 activities are currently available. Anakinra is a recombinant form of the naturally occurring IL-1 receptor antagonist. Anakinra binds to the IL-1 receptor and prevents the activity of IL-1α and IL-1β. The soluble decoy receptor rilonacept and the neutralizing mAb canakinumab block IL-1β. A mAb directed against the IL-1 receptor and a neutralizing anti-human IL-1α are in clinical trials. The availability of therapies specifically targeting IL-1 unveiled the pathological role of IL-1-mediated inflammation in a broadening list of diseases. Conditions effectively treated with agents blocking IL-1 range from classic rheumatic diseases, such as RA and gout, to autoinflammatory syndromes, such as systemic JIA and FMF. However, IL-1 antagonism is also effective against highly prevalent inflammatory diseases, namely cardiovascular diseases and type 2 diabetes, conditions that are frequently encountered as co-morbidities in patients with rheumatic diseases. Thereby, IL-1 inhibition has the potential to lift the burden of disease for patients with rheumatic conditions, but also to provide clinical benefits beyond the efficacy on osteoarticular manifestations.
Collapse
Affiliation(s)
- Giulio Cavalli
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA, Division of Internal Medicine and Clinical Immunology, IRCCS San Raffaele Scientific Institute, Milan, Italy and
| | - Charles A Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA, Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
257
|
Rider P, Carmi Y, Yossef R, Guttman O, Eini H, Azam T, Dinarello CA, Lewis EC. IL-1 Receptor Antagonist Chimeric Protein: Context-Specific and Inflammation-Restricted Activation. THE JOURNAL OF IMMUNOLOGY 2015; 195:1705-12. [PMID: 26157171 DOI: 10.4049/jimmunol.1501168] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 06/15/2015] [Indexed: 12/20/2022]
Abstract
Both IL-1α and IL-1β are highly inflammatory cytokines mediating a wide spectrum of diseases. A recombinant form of the naturally occurring IL-1R antagonist (IL-1Ra), which blocks IL-1R1, is broadly used to treat autoimmune and autoinflammatory diseases; however, blocking IL-1 increases the risk of infection. In this study, we describe the development of a novel form of recombinant IL-1Ra, termed chimeric IL-1Ra. This molecule is a fusion of the N-terminal peptide of IL-1β and IL-1Ra, resulting in inactive IL-1Ra. Because the IL-1β N-terminal peptide contains several protease sites clustered around the caspase-1 site, local proteases at sites of inflammation can cleave chimeric IL-1Ra and turn IL-1Ra active. We demonstrate that chimeric IL-1Ra reduces IL-1-mediated inflammation in vitro and in vivo. This unique approach limits IL-1 receptor blockade to sites of inflammation, while sparing a multitude of desired IL-1-related activities, including host defense against infections and IL-1-mediated repair.
Collapse
Affiliation(s)
- Peleg Rider
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel;
| | - Yaron Carmi
- Department of Pathology, School of Medicine, Stanford University, Palo Alto, CA 95305; The Shraga Segal Department of Microbiology, Immunology and Genetics and The Cancer Research Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Rami Yossef
- The Shraga Segal Department of Microbiology, Immunology and Genetics and The Cancer Research Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Ofer Guttman
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Hadar Eini
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Tania Azam
- University of Colorado at Denver, Aurora, CO 80045; and
| | - Charles A Dinarello
- University of Colorado at Denver, Aurora, CO 80045; and Radboud University Medical Centre, Nijmegen 6500, the Netherlands
| | - Eli C Lewis
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| |
Collapse
|
258
|
Affiliation(s)
- Antonio Abbate
- Virginia Commonwealth University, Richmond, VA 23298, USA.
| | | | | |
Collapse
|
259
|
Abstract
Elevated levels of inflammatory mediators have been identified in patients with heart failure, including heart failure with reduced and preserved ejection fraction, as well as acute decompensated heart failure. Moreover, experimental studies have shown repeatedly that activation of inflammation in the heart provokes left ventricular remodeling and left ventricular dysfunction. Nonetheless, phase III clinical trials that have attempted to antagonize inflammatory mediators have been negative with respect to the primary end points of the trials, and in some patients, resulted in worsening heart failure or death. The following review will discuss how recent developments in the field of innate immunity have advanced our understanding of the role of inflammation in the pathogenesis of heart failure and will discuss the negative outcomes of the existing clinical trials in light of this new information.
Collapse
Affiliation(s)
- Douglas L Mann
- From the Division of Cardiology, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO.
| |
Collapse
|
260
|
Abstract
Although patients with rheumatoid arthritis (RA) are recognized to be disproportionately impacted by cardiovascular disease (CVD), effective approaches of primary and secondary CVD prevention have not been well defined in this population. Given their robust disease-modifying potential and effects on both pro-inflammatory and pro-atherogenic pathways, there has been substantial speculation that biologic treatments may serve as a means of providing highly effective RA disease control while simultaneously reducing CVD risk in this high risk group. In this review, we examine available evidence relevant to the associations of approved biologic treatments with CVD outcomes in the context of RA.
Collapse
|
261
|
Butts B, Gary RA, Dunbar SB, Butler J. The Importance of NLRP3 Inflammasome in Heart Failure. J Card Fail 2015; 21:586-93. [PMID: 25982825 DOI: 10.1016/j.cardfail.2015.04.014] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 01/08/2015] [Accepted: 04/30/2015] [Indexed: 12/11/2022]
Abstract
Patients with heart failure continue to suffer adverse health consequences despite advances in therapies over the past 2 decades. Identification of novel therapeutic targets that may attenuate disease progression is therefore needed. The inflammasome may play a central role in modulating chronic inflammation and in turn affecting heart failure progression. The inflammasome is a complex of intracellular interaction proteins that trigger maturation of proinflammatory cytokines interleukin-1β and interleukin-18 to initiate the inflammatory response. This response is amplified through production of tumor necrosis factor α and activation of inducible nitric oxide synthase. The purpose of this review is to discuss recent evidence implicating this inflammatory pathway in the pathophysiology of heart failure.
Collapse
Affiliation(s)
- Brittany Butts
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA
| | - Rebecca A Gary
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA
| | - Sandra B Dunbar
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA
| | - Javed Butler
- Cardiology Division, Stony Brook University, Stony Brook, NY.
| |
Collapse
|
262
|
Lemarié J, Boufenzer A, Popovic B, Tran N, Groubatch F, Derive M, Labroca P, Barraud D, Gibot S. Pharmacological inhibition of the triggering receptor expressed on myeloid cells-1 limits reperfusion injury in a porcine model of myocardial infarction. ESC Heart Fail 2015; 2:90-99. [PMID: 28834656 PMCID: PMC6410538 DOI: 10.1002/ehf2.12029] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 03/16/2015] [Accepted: 03/17/2015] [Indexed: 01/01/2023] Open
Abstract
Aims Limitation of ischemia/reperfusion injury is a major therapeutic target after acute myocardial infarction (AMI). Toll‐like receptors are implicated in the inflammatory response that occurs during reperfusion. The triggering receptor expressed on myeloid cells (TREM)‐1 acts as an amplifier of the immune response triggered by toll‐like receptor engagement. We hypothesized that administration of a TREM‐1 inhibitory peptide (LR12) could limit reperfusion injury in a porcine model of AMI. Methods and results AMI was induced in 15 adult minipigs by a closed‐chest coronary artery occlusion‐reperfusion technique. Animals were randomized to receive LR12 or vehicle before reperfusion (LR12 n = 7, vehicle n = 8), and were monitored during 18 h. AMI altered hemodynamics and cardiac function, as illustrated by a drop of mean arterial pressure, cardiac index, cardiac power index, ejection fraction, and real‐time pressure–volume loop‐derived parameters. TREM‐1 inhibition by LR12 significantly improved these dysfunctions (P < 0.03) and limited infarct size, as assessed by lower creatine phosphokinase and troponin I concentrations (P < 0.005). Pulmonary, renal, and hepatic impairments occurred after AMI and were attenuated by LR12 administration as assessed by a better PaO2 to FiO2 ratio, a less positive fluid balance, and lower liver enzymes levels (P < 0.05). Conclusion Inhibition of the TREM‐1 pathway by a synthetic peptide limited myocardial reperfusion injury in a clinically relevant porcine model of AMI.
Collapse
Affiliation(s)
- Jérémie Lemarié
- Inserm UMR_S1116, Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France.,Medical Intensive Care Unit, Hôpital Central, CHU Nancy, Nancy, France
| | - Amir Boufenzer
- Inserm UMR_S1116, Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France
| | - Batric Popovic
- Inserm UMR_S1116, Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France.,Department of Cardiology, Hôpital Brabois, CHU Nancy, Nancy, France
| | - Nguyen Tran
- School of Surgery, Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France
| | - Frederique Groubatch
- School of Surgery, Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France
| | | | | | - Damien Barraud
- Medical Intensive Care Unit, Hôpital Central, CHU Nancy, Nancy, France
| | - Sébastien Gibot
- Inserm UMR_S1116, Faculté de Médecine de Nancy, Université de Lorraine, Nancy, France.,Medical Intensive Care Unit, Hôpital Central, CHU Nancy, Nancy, France
| |
Collapse
|
263
|
Toldo S, Mezzaroma E, Mauro AG, Salloum F, Van Tassell BW, Abbate A. The inflammasome in myocardial injury and cardiac remodeling. Antioxid Redox Signal 2015; 22:1146-61. [PMID: 25330141 DOI: 10.1089/ars.2014.5989] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
SIGNIFICANCE An inflammatory response follows an injury of any nature, and while such a response is an attempt to promote healing, it may, itself, result in further injury. RECENT ADVANCES The inflammasome is a macromolecular structure recently recognized as a central mediator in the acute inflammatory response. The inflammasome senses the injury and it amplifies the response by leading to the release of powerful pro-inflammatory cytokines, interleukin-1β (IL-1β) and IL-18. CRITICAL ISSUES The activation of the inflammasome in the heart during ischemic and nonischemic injury represents an exaggerated response to sterile injury and promotes adverse cardiac remodeling and failure. FUTURE DIRECTIONS Pilot clinical trials have explored blockade of the inflammasome-derived IL-1β and have shown beneficial effects on cardiac function. Additional clinical studies testing this approach are warranted. Moreover, specific inflammasome inhibitors that are ready for clinical use are currently lacking.
Collapse
Affiliation(s)
- Stefano Toldo
- 1 VCU Pauley Heart Center, Virginia Commonwealth University , Richmond, Virginia
| | | | | | | | | | | |
Collapse
|
264
|
Peruzzi M, Biondi-Zoccai G, Abbate A, Giordano A, Frati G. Commentary: which comes first, the phoenix or the flame? Reflections on the role of inflammation in patients undergoing lower limb revascularization for peripheral artery disease. J Endovasc Ther 2015; 22:240-242. [PMID: 25809369 DOI: 10.1177/1526602815573217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
| | - Giuseppe Biondi-Zoccai
- Sapienza University of Rome, Latina, Italy Eleonora Lorillard Spencer Cenci Foundation, Rome, Italy
| | | | - Arturo Giordano
- Presidio Ospedaliero Pineta Grande, Castel Volturno, Italy Casa di Salute Santa Lucia, San Giuseppe Vesuviano, Italy
| | - Giacomo Frati
- Sapienza University of Rome, Latina, Italy IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
265
|
Frangogiannis NG. Interleukin-1 in cardiac injury, repair, and remodeling: pathophysiologic and translational concepts. Discoveries (Craiova) 2015; 3. [PMID: 26273700 PMCID: PMC4532433 DOI: 10.15190/d.2015.33] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
In the infarcted myocardium, necrotic cardiomyocytes release danger signals activating an intense inflammatory reaction that serves to clear the wound from dead cells and matrix debris, but may also extend injury. A growing body of evidence suggests an important role for members of the Interleukin (IL)-1 family in injury, repair and remodeling of the infarcted heart. This review manuscript discusses the pathophysiologic functions of IL-1 in the infarcted and remodeling myocardium and its potential role as a therapeutic target in patients with myocardial infarction. Dead cardiomyocytes release IL-1a that may function as a crucial alarmin triggering the post-infarction inflammatory reaction. IL-1b is markedly upregulated in the infarcted myocardium; activation of the inflammasome in both cardiomyocytes and interstitial cells results in release of bioactive IL-1b in the infarcted area. Binding of IL-1 to the type 1 receptor triggers an inflammatory cascade, inducing recruitment of pro-inflammatory leukocytes and stimulating a matrix-degrading program in fibroblasts, while delaying myofibroblast conversion. IL-1 mediates dilative remodeling following infarction and may play a role in the pathogenesis of post-infarction heart failure. As the wound is cleared from dead cells and matrix debris, endogenous inhibitory signals suppress the IL-1 response resulting in repression of inflammation and resolution of the inflammatory infiltrate. Other members of the IL-1 family (such as IL-18 and IL-33) are also implicated in regulation of the inflammatory and reparative response following myocardial infarction. IL-18 may participate in pro-inflammatory signaling, whereas IL-33 may exert cytoprotective effects. Early clinical trials suggest that IL-1 blockade may be a promising therapeutic strategy for patients with myocardial infarction.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx NY, USA
| |
Collapse
|
266
|
Affiliation(s)
- Ryan A Frieler
- From Department of Molecular and Integrative Physiology (R.A.F., R.M.M.), Department of Internal Medicine, Metabolism, Endocrinology, and Diabetes Division (R.M.M.), and Department of Pharmacology (R.M.M.), University of Michigan Medical School, Ann Arbor
| | - Richard M Mortensen
- From Department of Molecular and Integrative Physiology (R.A.F., R.M.M.), Department of Internal Medicine, Metabolism, Endocrinology, and Diabetes Division (R.M.M.), and Department of Pharmacology (R.M.M.), University of Michigan Medical School, Ann Arbor.
| |
Collapse
|
267
|
Chistiakov DA, Orekhov AN, Bobryshev YV. Contribution of neovascularization and intraplaque haemorrhage to atherosclerotic plaque progression and instability. Acta Physiol (Oxf) 2015; 213:539-53. [PMID: 25515699 DOI: 10.1111/apha.12438] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 11/13/2014] [Accepted: 12/10/2014] [Indexed: 12/14/2022]
Abstract
Atherosclerosis is a continuous pathological process that starts early in life and progresses frequently to unstable plaques. Plaque rupture leads to deleterious consequences such as acute coronary syndrome, stroke and atherothrombosis. The vulnerable lesion has several structural and functional hallmarks that distinguish it from the stable plaque. The unstable plaque has large necrotic core (over 40% plaque volume) composed of cholesterol crystals, cholesterol esters, oxidized lipids, fibrin, erythrocytes and their remnants (haeme, iron, haemoglobin), and dying macrophages. The fibrous cap is thin, depleted of smooth muscle cells and collagen, and is infiltrated with proinflammatory cells. In unstable lesion, formation of neomicrovessels is increased. These neovessels have weak integrity and leak thereby leading to recurrent haemorrhages. Haemorrhages deliver erythrocytes to the necrotic core where they degrade promoting inflammation and oxidative stress. Inflammatory cells mostly presented by monocytes/macrophages, neutrophils and mast cells extravagate from bleeding neovessels and infiltrate adventitia where they support chronic inflammation. Plaque destabilization is an evolutionary process that could start at early atherosclerotic stages and whose progression is influenced by many factors including neovascularization, intraplaque haemorrhages, formation of cholesterol crystals, inflammation, oxidative stress and intraplaque protease activity.
Collapse
Affiliation(s)
- D. A. Chistiakov
- Department of Medical Nanobiotechnology; Pirogov Russian State Medical University; Moscow Russia
- The Mount Sinai Community Clinical Oncology Program; Mount Sinai Comprehensive Cancer Center; Mount Sinai Medical Center; Miami Beach FL USA
- Research Center for Children's Health; Moscow Russia
| | - A. N. Orekhov
- Laboratory of Angiopathology; Institute of General Pathology and Pathophysiology; Russian Academy of Sciences; Moscow Russia
- Skolkovo Innovative Center; Institute for Atherosclerosis Research; Moscow Russia
| | - Y. V. Bobryshev
- Laboratory of Angiopathology; Institute of General Pathology and Pathophysiology; Russian Academy of Sciences; Moscow Russia
- Faculty of Medicine and St Vincent's Centre for Applied Medical Research; University of New South Wales; Sydney NSW Australia
- School of Medicine; University of Western Sydney; Campbelltown NSW Australia
| |
Collapse
|
268
|
Role of genetic alterations in the NLRP3 and CARD8 genes in health and disease. Mediators Inflamm 2015; 2015:846782. [PMID: 25788762 PMCID: PMC4348606 DOI: 10.1155/2015/846782] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 01/05/2015] [Indexed: 12/30/2022] Open
Abstract
The complexity of a common inflammatory disease is influenced by multiple genetic and environmental factors contributing to the susceptibility of disease. Studies have reported that these exogenous and endogenous components may perturb the balance of innate immune response by activating the NLRP3 inflammasome. The multimeric NLRP3 complex results in the caspase-1 activation and the release of potent inflammatory cytokines, like IL-1β. Several studies have been performed on the association of the genetic alterations in genes encoding NLRP3 and CARD8 with the complex diseases with inflammatory background, like inflammatory bowel disease, cardiovascular diseases, rheumatoid arthritis, and type 1 diabetes. The aim of the present review is therefore to summarize the literature regarding genetic alterations in these genes and their association with health and disease.
Collapse
|
269
|
Seropian IM, Sonnino C, Van Tassell BW, Biasucci LM, Abbate A. Inflammatory markers in ST-elevation acute myocardial infarction. EUROPEAN HEART JOURNAL-ACUTE CARDIOVASCULAR CARE 2015; 5:382-95. [PMID: 25681486 DOI: 10.1177/2048872615568965] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 01/02/2015] [Indexed: 01/05/2023]
Abstract
After acute myocardial infarction, ventricular remodeling is characterized by changes at the molecular, structural, geometrical and functional level that determine progression to heart failure. Inflammation plays a key role in wound healing and scar formation, affecting ventricular remodeling. Several, rather different, components of the inflammatory response were studied as biomarkers in ST-elevation acute myocardial infarction. Widely available and inexpensive tests, such as leukocyte count at admission, as well as more sophisticated immunoassays provide powerful predictors of adverse outcome in patients with ST-elevation acute myocardial infarction. We review the value of inflammatory markers in ST-elevation acute myocardial infarction and their association with ventricular remodeling, heart failure and sudden death. In conclusion, the use of these biomarkers may identify subjects at greater risk of adverse events and perhaps provide an insight into the mechanisms of disease progression.
Collapse
Affiliation(s)
- Ignacio M Seropian
- Interventional Cardiology Department, Hospital Italiano de Buenos Aires, Argentina
| | - Chiara Sonnino
- VCU Pauley Heart Center, Virginia Commonwealth University, USA Victoria Johnson Research Laboratory, Virginia Commonwealth University, USA Department of Cardiovascular Medicine, Catholic University, Italy
| | - Benjamin W Van Tassell
- VCU Pauley Heart Center, Virginia Commonwealth University, USA Victoria Johnson Research Laboratory, Virginia Commonwealth University, USA School of Pharmacy, Virginia Commonwealth University, USA
| | - Luigi M Biasucci
- Department of Cardiovascular Medicine, Catholic University, Italy
| | - Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, USA Victoria Johnson Research Laboratory, Virginia Commonwealth University, USA
| |
Collapse
|
270
|
Morton AC, Rothman AMK, Greenwood JP, Gunn J, Chase A, Clarke B, Hall AS, Fox K, Foley C, Banya W, Wang D, Flather MD, Crossman DC. The effect of interleukin-1 receptor antagonist therapy on markers of inflammation in non-ST elevation acute coronary syndromes: the MRC-ILA Heart Study. Eur Heart J 2015; 36:377-84. [PMID: 25079365 PMCID: PMC4320321 DOI: 10.1093/eurheartj/ehu272] [Citation(s) in RCA: 260] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 04/21/2014] [Accepted: 06/12/2014] [Indexed: 12/15/2022] Open
Abstract
AIMS Acute coronary syndromes (ACSs) are driven by inflammation within coronary plaque. Interleukin-1 (IL-1) has an established role in atherogenesis and the vessel-response to injury. ACS patients have raised serum markers of inflammation. We hypothesized that if IL-1 is a driving influence of inflammation in non-ST elevation ACS (NSTE-ACS), IL-1 inhibition would reduce the inflammatory response at the time of ACS. METHODS AND RESULTS A phase II, double-blinded, randomized, placebo-controlled, study recruited 182 patients with NSTE-ACS, presenting <48 h from onset of chest pain. Treatment was 1:1 allocation to daily, subcutaneous IL-1receptor antagonist (IL-1ra) or placebo for 14 days. Baseline characteristics were well matched. Treatment compliance was 85% at 7 days. The primary endpoint (area-under-the-curve for C-reactive protein over the first 7 days) was: IL-1ra group, 21.98 mg day/L (95%CI 16.31-29.64); placebo group, 43.5 mg day/L (31.15-60.75) (geometric mean ratio = 0.51 mg/L; 95%CI 0.32-0.79; P = 0.0028). In the IL-1ra group, 14-day achieved high-sensitive C-reactive protein (P < 0.0001) and IL-6 levels (P = 0.02) were lower than Day 1. Sixteen days after discontinuation of treatment (Day 30) high-sensitive C-reactive protein levels had risen again in the IL-1ra group [IL-1ra; 3.50 mg/L (2.65-4.62): placebo; 2.21 mg/L (1.67-2.92), P = 0.022]. MACE at Day 30 and 3 months was similar but at 1 year there was a significant excess of events in the IL-1ra group. CONCLUSION IL-1 drives C-reactive protein elevation at the time of NSTE-ACS. Following 14 days IL-1ra treatment inflammatory markers were reduced. These results show the importance of IL-1 as a target in ACS, but also indicate the need for additional studies with anti-IL-1 therapy in ACS to assess duration and safety. CLINICAL TRIAL REGISTRATION EUCTR 2006-001767-31-GB: www.clinicaltrialsregister.eu/ctr-search/trial/2006-001767-31/GB.
Collapse
Affiliation(s)
- Allison C Morton
- Department of Cardiology, Northern General Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Alexander M K Rothman
- Department of Cardiology, Northern General Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK Department of Cardiovascular Science, University of Sheffield, Sheffield, UK
| | - John P Greenwood
- Academic Unit of Cardiovascular Medicine, Yorkshire Heart Centre, Leeds, UK
| | - Julian Gunn
- Department of Cardiology, Northern General Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK Department of Cardiovascular Science, University of Sheffield, Sheffield, UK
| | - Alex Chase
- Abertawe Bro Morgannwg University NHS Trust, Morriston Hospital, Swansea, UK
| | - Bernard Clarke
- Institute of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Alistair S Hall
- Academic Unit of Cardiovascular Medicine, Yorkshire Heart Centre, Leeds, UK
| | - Keith Fox
- Royal Infirmary of Edinburgh and University of Edinburgh, Edinburgh, UK
| | - Claire Foley
- Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, London, UK
| | - Winston Banya
- Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, London, UK
| | - Duolao Wang
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Marcus D Flather
- Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, London, UK Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
| | | |
Collapse
|
271
|
Abbate A, Dinarello CA. Anti-inflammatory therapies in acute coronary syndromes: is IL-1 blockade a solution? Eur Heart J 2015; 36:337-9. [PMID: 25205529 DOI: 10.1093/eurheartj/ehu369] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2024] Open
Affiliation(s)
- Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, 1200 E. Broad street, Box 980281, Richmond, VA, 23298, USA
| | | |
Collapse
|
272
|
Ulland TK, Ferguson PJ, Sutterwala FS. Evasion of inflammasome activation by microbial pathogens. J Clin Invest 2015; 125:469-77. [PMID: 25642707 DOI: 10.1172/jci75254] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Activation of the inflammasome occurs in response to infection with a wide array of pathogenic microbes. The inflammasome serves as a platform to activate caspase-1, which results in the subsequent processing and secretion of the proinflammatory cytokines IL-1β and IL-18 and the initiation of an inflammatory cell death pathway termed pyroptosis. Effective inflammasome activation is essential in controlling pathogen replication as well as initiating adaptive immune responses against the offending pathogens. However, a number of pathogens have developed strategies to evade inflammasome activation. In this Review, we discuss these pathogen evasion strategies as well as the potential infectious complications of therapeutic blockade of IL-1 pathways.
Collapse
|
273
|
Abbate A, Kontos MC, Abouzaki NA, Melchior RD, Thomas C, Van Tassell BW, Oddi C, Carbone S, Trankle CR, Roberts CS, Mueller GH, Gambill ML, Christopher S, Markley R, Vetrovec GW, Dinarello CA, Biondi-Zoccai G. Comparative safety of interleukin-1 blockade with anakinra in patients with ST-segment elevation acute myocardial infarction (from the VCU-ART and VCU-ART2 pilot studies). Am J Cardiol 2015; 115:288-292. [PMID: 25482680 DOI: 10.1016/j.amjcard.2014.11.003] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Revised: 11/03/2014] [Accepted: 11/03/2014] [Indexed: 02/08/2023]
Abstract
Two pilot studies of interleukin-1 (IL-1) blockade in ST-segment elevation myocardial infarction (STEMI) showed blunted acute inflammatory response and overall favorable outcomes at 3 months follow-up. We hereby present a patient-level pooled analysis with extended follow-up of 40 patients with clinically stable STEMI randomized to anakinra, a recombinant IL-1 receptor antagonist, 100 mg/day for 14 days or placebo in a double-blinded fashion. End points included death, cardiac death, recurrent acute myocardial infarction (AMI), stroke, unstable angina, and symptomatic heart failure. Median follow-up was 28 (interquartile range 3 to 38) months. Sixteen patients (40%) had a total of 22 adverse cardiovascular events: 1 cardiac death, 4 recurrent AMI, 5 episodes of unstable angina pectoris requiring hospitalization and/or urgent revascularization, and 11 new diagnoses of heart failure. Treatment with anakinra was associated with a hazard ratio of 1.08 (95% confidence interval 0.31 to 3.74, p = 0.90) for the combined end point of death, recurrent AMI, unstable angina pectoris, or stroke and a hazard ratio of 0.16 (95% confidence interval 0.03 to 0.76, p = 0.008) for death or heart failure. In conclusion, IL-1 blockade with anakinra for 2 weeks appears, therefore, to have a neutral effect on recurrent ischemic events, whereas it may prevent new-onset heart failure long term after STEMI.
Collapse
Affiliation(s)
- Antonio Abbate
- VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, Virginia.
| | - Michael Christopher Kontos
- VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, Virginia
| | - Nayef Antar Abouzaki
- VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, Virginia
| | - Ryan David Melchior
- VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, Virginia
| | - Christopher Thomas
- VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, Virginia
| | - Benjamin Wallace Van Tassell
- VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, Virginia
| | - Claudia Oddi
- VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, Virginia
| | - Salvatore Carbone
- VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, Virginia
| | - Cory Ross Trankle
- VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, Virginia
| | - Charlotte Susan Roberts
- VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, Virginia
| | - George Herman Mueller
- VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, Virginia
| | - Michael Lucas Gambill
- VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, Virginia
| | - Sanah Christopher
- VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, Virginia
| | - Roshanak Markley
- VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, Virginia
| | - George Wayne Vetrovec
- VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, Virginia
| | | | - Giuseppe Biondi-Zoccai
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University, Latina, Italy
| |
Collapse
|
274
|
van Hout G, Jansen of Lorkeers S, Wever K, Sena E, van Solinge W, Doevendans P, Pasterkamp G, Chamuleau S, Hoefer I. Anti-inflammatory compounds to reduce infarct size in large-animal models of myocardial infarction: A meta-analysis. ACTA ACUST UNITED AC 2015. [DOI: 10.1002/ebm2.4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- G.P.J. van Hout
- Experimental Cardiology Laboratory; University Medical Center Utrecht; Utrecht the Netherlands
| | | | - K.E. Wever
- Systematic Review Centre for Laboratory animal Experimentation; Radboud University Nijmegen Medical Center; Nijmegen the Netherlands
| | - E.S. Sena
- Centre for Clinical Brain Sciences; University of Edinburgh; Edinburgh UK
| | - W.W. van Solinge
- Department of Clinical Chemistry and Haematology; University Medical Center Utrecht; Utrecht the Netherlands
| | - P.A. Doevendans
- Experimental Cardiology Laboratory; University Medical Center Utrecht; Utrecht the Netherlands
- Department of Cardiology; University Medical Center Utrecht; Utrecht the Netherlands
| | - G. Pasterkamp
- Experimental Cardiology Laboratory; University Medical Center Utrecht; Utrecht the Netherlands
| | - S.A.J. Chamuleau
- Department of Cardiology; University Medical Center Utrecht; Utrecht the Netherlands
| | - I.E. Hoefer
- Experimental Cardiology Laboratory; University Medical Center Utrecht; Utrecht the Netherlands
| |
Collapse
|
275
|
Huynh K, Van Tassell B, Chow SL. Predicting therapeutic response in patients with heart failure: the story of C-reactive protein. Expert Rev Cardiovasc Ther 2015; 13:153-61. [PMID: 25578159 DOI: 10.1586/14779072.2015.1000307] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Heart failure continues to be a major public health burden in the USA. With markedly high rates of morbidity and mortality upon diagnosis, effective treatment and prognosis are critical in the management of chronic heart failure. Growing evidence now supports the hypothesis that inflammation plays a key role in the progression and worsening of heart failure. Of the various inflammatory mediators identified, C-reactive protein, an acute phase inflammatory marker, has been associated with poor prognosis in patients with heart failure. Several interventional studies have been investigated to explore C-reactive protein modulation and potential treatment options and health outcomes; however, further studies are warranted before C-reactive protein-targeted therapy may be recommended in the management of heart failure.
Collapse
Affiliation(s)
- Kitty Huynh
- Western University of Health Sciences, Pharmacy Practice, 309 E. Second Street, Pomona, 91766, USA
| | | | | |
Collapse
|
276
|
Abstract
Both experimental and clinical evidence accumulated over the last couple of decades has linked inflammatory activation to the initiation and progression of chronic heart failure (HF). Circulating levels of inflammatory mediators are associated with cardiac function and inform risk prediction in patients, but the effect of anti-inflammatory therapy in HF remains uncertain. Interleukin (IL)-6 type cytokines are central to the inflammatory response, and convey their signals through the ubiquitously expressed glycoprotein (gp) 130 receptor subunit. IL-6-type/gp130 signaling therefore represents an inflammatory nexus, with inherent potential for disease modification. This review focuses on the current knowledge of IL-6/gp130 signaling in relation to HF, with a particular emphasis on the role of soluble gp130 (sgp130), a signaling pathway modulator. Biological aspects of sgp130 and IL-6 signaling are discussed, as are potential novel therapeutic approaches to modulate this central inflammatory signaling pathway.
Collapse
|
277
|
Abbate A, Van Tassell BW, Christopher S, Abouzaki NA, Sonnino C, Oddi C, Carbone S, Melchior RD, Gambill ML, Roberts CS, Kontos MC, Peberdy MA, Toldo S, Vetrovec GW, Biondi-Zoccai G, Dinarello CA. Effects of Prolastin C (Plasma-Derived Alpha-1 Antitrypsin) on the acute inflammatory response in patients with ST-segment elevation myocardial infarction (from the VCU-alpha 1-RT pilot study). Am J Cardiol 2015; 115:8-12. [PMID: 25456867 DOI: 10.1016/j.amjcard.2014.09.043] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 09/21/2014] [Accepted: 09/21/2014] [Indexed: 02/08/2023]
Abstract
Alpha-1 antitrypsin (AAT) has broad anti-inflammatory and immunomodulating properties in addition to inhibiting serine proteases. Administration of human plasma-derived AAT is protective in models of acute myocardial infarction in mice. The objective of this study was to determine the safety and tolerability of human plasma-derived AAT and its effects on the acute inflammatory response in non-AAT deficient patients with ST-segment elevation myocardial infarction (STEMI). Ten patients with acute STEMI were enrolled in an open-label, single-arm treatment study of AAT at 60 mg/kg infused intravenously within 12 hours of admission and following standard of care treatment. C-reactive protein (CRP) and plasma AAT levels were determined at admission, 72 hours, and 14 days, and patients were followed clinically for 12 weeks for the occurrence of new onset heart failure, recurrent myocardial infarction, or death. Twenty patients with STEMI enrolled in previous randomized trials with identical inclusion and/or exclusion criteria, but who received placebo, served as historical controls. Prolastin C was well tolerated and there were no in-hospital adverse events. Compared with historical controls, the area under the curve of CRP levels was significantly lower 14 days after admission in the Prolastin C group (75.9 [31.4 to 147.8] vs 205.6 [78.8 to 410.9] mg/l, p = 0.048), primarily due to a significant blunting of the increase occurring between admission and 72 hours (delta CRP +1.7 [0.2 to 9.4] vs +21.1 [3.1 to 38.0] mg/l, p = 0.007). Plasma AAT levels increased from admission (149 [116 to 189]) to 203 ([185 to 225] mg/dl) to 72 hours (p = 0.005). In conclusion, a single administration of Prolastin C in patients with STEMI is well tolerated and is associated with a blunted acute inflammatory response.
Collapse
Affiliation(s)
- Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia.
| | | | - Sanah Christopher
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| | | | - Chiara Sonnino
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia; Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, Virginia
| | - Claudia Oddi
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia; Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, Virginia
| | - Salvatore Carbone
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia; Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, Virginia
| | - Ryan David Melchior
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia; Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, Virginia
| | - Michael Lucas Gambill
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia; Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, Virginia
| | | | | | - Mary Ann Peberdy
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia; Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, Virginia
| | - Stefano Toldo
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| | | | | | | |
Collapse
|
278
|
Vallejo S, Palacios E, Romacho T, Villalobos L, Peiró C, Sánchez-Ferrer CF. The interleukin-1 receptor antagonist anakinra improves endothelial dysfunction in streptozotocin-induced diabetic rats. Cardiovasc Diabetol 2014; 13:158. [PMID: 25518980 PMCID: PMC4276125 DOI: 10.1186/s12933-014-0158-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 11/21/2014] [Indexed: 01/06/2023] Open
Abstract
Background Endothelial dysfunction is a crucial early phenomenon in vascular diseases linked to diabetes mellitus and associated to enhanced oxidative stress. There is increasing evidence about the role for pro-inflammatory cytokines, like interleukin-1β (IL-1β), in developing diabetic vasculopathy. We aimed to determine the possible involvement of this cytokine in the development of diabetic endothelial dysfunction, analysing whether anakinra, an antagonist of IL-1 receptors, could reduce this endothelial alteration by interfering with pro-oxidant and pro-inflammatory pathways into the vascular wall. Results In control and two weeks evolution streptozotocin-induced diabetic rats, either untreated or receiving anakinra, vascular reactivity and NADPH oxidase activity were measured, respectively, in isolated rings and homogenates from mesenteric microvessels, while nuclear factor (NF)-κB activation was determined in aortas. Plasma levels of IL-1β and tumor necrosis factor (TNF)-α were measured by ELISA. In isolated mesenteric microvessels from control rats, two hours incubation with IL-1β (1 to 10 ng/mL) produced a concentration-dependent impairment of endothelium-dependent relaxations, which were mediated by enhanced NADPH oxidase activity via IL-1 receptors. In diabetic rats treated with anakinra (100 or 160 mg/Kg/day for 3 or 7 days before sacrifice) a partial improvement of diabetic endothelial dysfunction occurred, together with a reduction of vascular NADPH oxidase and NF-κB activation. Endothelial dysfunction in diabetic animals was also associated to higher activities of the pro-inflammatory enzymes cyclooxygenase (COX) and the inducible isoform of nitric oxide synthase (iNOS), which were markedly reduced after anakinra treatment. Circulating IL-1β and TNF-α levels did not change in diabetic rats, but they were lowered by anakinra treatment. Conclusions In this short-term model of type 1 diabetes, endothelial dysfunction is associated to an IL-1 receptor-mediated activation of vascular NADPH oxidase and NF-κB, as well as to vascular inflammation. Moreover, endothelial dysfunction, vascular oxidative stress and inflammation were reduced after anakinra treatment. Whether this mechanism can be extrapolated to a chronic situation or whether it may apply to diabetic patients remain to be established. However, it may provide new insights to further investigate the therapeutic use of IL-1 receptor antagonists to obtain vascular benefits in patients with diabetes mellitus and/or atherosclerosis.
Collapse
Affiliation(s)
- Susana Vallejo
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Calle Arzobispo Morcillo 4, 29029, Madrid, Spain.
| | - Erika Palacios
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Calle Arzobispo Morcillo 4, 29029, Madrid, Spain. .,Present address: Departamento de Ciencias de la Salud, Edificio CN208, Oficina O, Universidad de las Américas, Puebla, México.
| | - Tania Romacho
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Calle Arzobispo Morcillo 4, 29029, Madrid, Spain. .,Present address: Paul Langerhans-Group, Integrative Physiology, German Diabetes Center, Auf'm Hennekamp 65, 40225, Düsseldorf, Germany.
| | - Laura Villalobos
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Calle Arzobispo Morcillo 4, 29029, Madrid, Spain.
| | - Concepción Peiró
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Calle Arzobispo Morcillo 4, 29029, Madrid, Spain.
| | - Carlos F Sánchez-Ferrer
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Calle Arzobispo Morcillo 4, 29029, Madrid, Spain.
| |
Collapse
|
279
|
Dinarello CA. An expanding role for interleukin-1 blockade from gout to cancer. Mol Med 2014; 20 Suppl 1:S43-58. [PMID: 25549233 PMCID: PMC4374514 DOI: 10.2119/molmed.2014.00232] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 11/17/2014] [Indexed: 01/08/2023] Open
Abstract
There is an expanding role for interleukin (IL)-1 in diseases from gout to cancer. More than any other cytokine family, the IL-1 family is closely linked to innate inflammatory and immune responses. This linkage is because the cytoplasmic segment of all members of the IL-1 family of receptors contains a domain, which is highly homologous to the cytoplasmic domains of all toll-like receptors (TLRs). This domain, termed "toll IL-1 receptor (TIR) domain," signals as does the IL-1 receptors; therefore, inflammation due to the TLR and the IL-1 families is nearly the same. Fundamental responses such as the induction of cyclo-oxygenase type 2, increased surface expression of cellular adhesion molecules and increased gene expression of a broad number of inflammatory molecules characterizes IL-1 signal transduction as it does for TLR agonists. IL-1β is the most studied member of the IL-1 family because of its role in mediating autoinflammatory disease. However, a role for IL-1α in disease is being validated because of the availability of a neutralizing monoclonal antibody to human IL-1α. There are presently three approved therapies for blocking IL-1 activity. Anakinra is a recombinant form of the naturally occurring IL-1 receptor antagonist, which binds to the IL-1 receptor and prevents the binding of IL-1β as well as IL-1α. Rilonacept is a soluble decoy receptor that neutralizes primarily IL-1β but also IL-1α. Canakinumab is a human monoclonal antibody that neutralizes only IL-1β. Thus, a causal or significant contributing role can be established for IL-1β and IL-1α in human disease.
Collapse
Affiliation(s)
- Charles Anthony Dinarello
- Department of Medicine, Division of Infectious Diseases, University of Colorado Denver, Aurora, Colorado, United States of America; and Department of Medicine, Radboud University, Nijmegen, the Netherlands
| |
Collapse
|
280
|
A novel pharmacologic inhibitor of the NLRP3 inflammasome limits myocardial injury after ischemia-reperfusion in the mouse. J Cardiovasc Pharmacol 2014; 63:316-322. [PMID: 24336017 DOI: 10.1097/fjc.0000000000000053] [Citation(s) in RCA: 218] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND The formation of the NLRP3 inflammasome in the heart during acute myocardial infarction amplifies the inflammatory response and mediates further damage. Glyburide has NLRP3 inhibitory activity in vitro but requires very high doses in vivo, associated with hypoglycemia. The aim of this study was to measure the effects on the NLRP3 inflammasome of 16673-34-0, an intermediate substrate free of the cyclohexylurea moiety, involved in insulin release. METHODS AND RESULTS We synthesized 16673-34-0 (5-chloro-2-methoxy-N-[2-(4-sulfamoylphenyl)ethyl]benzamide) that displayed no effect on glucose metabolism. HL-1 cardiomyocytes were treated with lipopolysaccharide and ATP to induce the formation of the NLRP3 inflammasome, measured as increased caspase-1 activity and cell death, and 16673-34-0 prevented such effects. 16673-34-0 was well tolerated with no effects on the glucose levels in vivo. Treatment with 16673-34-0 in a model of acute myocardial infarction because of ischemia and reperfusion significantly inhibited the activity of inflammasome (caspase-1) in the heart by 90% (P < 0.01) and reduced infarct size, measured at pathology (by >40%, P < 0.01) and with troponin I levels (by >70%, P < 0.01). CONCLUSIONS The small molecule 16673-34-0, an intermediate substrate in the glyburide synthesis free of the cyclohexylurea moiety, inhibits the formation of the NLRP3 inflammasome in cardiomyocytes and limits the infarct size after myocardial ischemia-reperfusion in the mouse, without affecting glucose metabolism.
Collapse
|
281
|
Sonnino C, Christopher S, Oddi C, Toldo S, Falcao RA, Melchior RD, Mueller GH, Abouzaki NA, Varma A, Gambill ML, Van Tassell BW, Dinarello CA, Abbate A. Leukocyte activity in patients with ST-segment elevation acute myocardial infarction treated with anakinra. Mol Med 2014; 20:486-9. [PMID: 25121719 PMCID: PMC4277550 DOI: 10.2119/molmed.2014.00121] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 08/07/2014] [Indexed: 12/13/2022] Open
Abstract
Anakinra, the recombinant form of the human interleukin (IL)-1 receptor antagonist, blunts the acute systemic inflammatory response in patients with ST-segment elevation myocardial infarction (STEMI), by determining a fall in peripheral blood leukocyte and plasma C-reactive protein levels. The aim of the present study was to determine the effects of anakinra on the activity of leukocytes measured ex vivo. Blood was collected 72 h after admission in 17 patients enrolled in the Virginia Commonwealth University-Anakirna Remodeling Trial (2) (VCU-ART2) and randomly treated with anakinra (N=7) or placebo (N=10). Whole blood was cultured at 37°C for 24 h to measure spontaneous production of IL-6 or stimulated with Escherichia coli lipopolysaccharide (LPS) for toll-like receptor (TLR)-4 or heat-killed Staphylococcus epidermidis (SE) for TLR-2 activation. The cultures of anakinra-treated patients produced significantly less IL-6 spontaneously (71 pg/mL [27-114]) compared with placebo-treated patients (290 pg/mL [211-617], p=0.005). LPS- or SE-induced IL-6 production, on the other hand, was not statistically different between anakinra-versus placebo-treated patients (344 pg/mL [94-560] versus 370 pg/mL [306-991], p=0.32 for LPS, and 484 pg/mL [77-612] versus 615 pg/mL [413-871], p=0.31 for SE, respectively). IL-1 blockade with anakinra in STEMI patients results in reduced spontaneous leukocyte activity ex vivo without impairing the responsiveness to bacterial stimuli.
Collapse
Affiliation(s)
- Chiara Sonnino
- Virginia Commonwealth University, VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Richmond, Virginia, United States of America
| | - Sanah Christopher
- Virginia Commonwealth University, VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Richmond, Virginia, United States of America
| | - Claudia Oddi
- Virginia Commonwealth University, VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Richmond, Virginia, United States of America
| | - Stefano Toldo
- Virginia Commonwealth University, VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Richmond, Virginia, United States of America
| | - Raquel Appa Falcao
- Virginia Commonwealth University, VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Richmond, Virginia, United States of America
| | - Ryan D Melchior
- Virginia Commonwealth University, VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Richmond, Virginia, United States of America
| | - George H Mueller
- Virginia Commonwealth University, VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Richmond, Virginia, United States of America
| | - Nayef A Abouzaki
- Virginia Commonwealth University, VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Richmond, Virginia, United States of America
| | - Amit Varma
- Virginia Commonwealth University, VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Richmond, Virginia, United States of America
| | - Michael L Gambill
- Virginia Commonwealth University, VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Richmond, Virginia, United States of America
| | - Benjamin W Van Tassell
- Virginia Commonwealth University, VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Richmond, Virginia, United States of America
| | - Charles A Dinarello
- Department of Medicine, University of Colorado, Aurora, Colorado, United States of America
| | - Antonio Abbate
- Virginia Commonwealth University, VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Richmond, Virginia, United States of America
| |
Collapse
|
282
|
The immune system and the remodeling infarcted heart: cell biological insights and therapeutic opportunities. J Cardiovasc Pharmacol 2014; 63:185-95. [PMID: 24072174 DOI: 10.1097/fjc.0000000000000003] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Extensive necrosis of ischemic cardiomyocytes in the infarcted myocardium activates the innate immune response triggering an intense inflammatory reaction. Release of danger signals from dying cells and damaged matrix activates the complement cascade and stimulates Toll-like receptor/interleukin-1 signaling, resulting in the activation of the nuclear factor-κB system and induction of chemokines, cytokines, and adhesion molecules. Subsequent infiltration of the infarct with neutrophils and mononuclear cells serves to clear the wound from dead cells and matrix debris, while stimulating reparative pathways. In addition to its role in repair of the infarcted heart and formation of a scar, the immune system is also involved in adverse remodeling of the infarcted ventricle. Overactive immune responses and defects in suppression, containment, and resolution of the postinfarction inflammatory reaction accentuate dilative remodeling in experimental models and may be associated with chamber dilation, systolic dysfunction, and heart failure in patients surviving a myocardial infarction. Interventions targeting the inflammatory response to attenuate adverse remodeling may hold promise in patients with myocardial infarction that exhibit accentuated, prolonged, or dysregulated immune responses to the acute injury.
Collapse
|
283
|
|
284
|
Yoshida T, Friehs I, Mummidi S, del Nido PJ, Addulnour-Nakhoul S, Delafontaine P, Valente AJ, Chandrasekar B. Pressure overload induces IL-18 and IL-18R expression, but markedly suppresses IL-18BP expression in a rabbit model. IL-18 potentiates TNF-α-induced cardiomyocyte death. J Mol Cell Cardiol 2014; 75:141-51. [PMID: 25108227 PMCID: PMC4157969 DOI: 10.1016/j.yjmcc.2014.07.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 07/15/2014] [Accepted: 07/16/2014] [Indexed: 10/24/2022]
Abstract
Recurrent or sustained inflammation plays a causal role in the development and progression of left ventricular hypertrophy (LVH) and its transition to failure. Interleukin (IL)-18 is a potent pro-hypertrophic inflammatory cytokine. We report that induction of pressure overload in the rabbit, by constriction of the descending thoracic aorta induces compensatory hypertrophy at 4weeks (mass/volume ratio: 1.7±0.11) and ventricular dilatation indicative of heart failure at 6weeks (mass/volume ratio: 0.7±0.04). In concordance with this, fractional shortening was preserved at 4weeks, but markedly attenuated at 6weeks. We cloned rabbit IL-18, IL-18Rα, IL-18Rβ, and IL-18 binding protein (IL-18BP) cDNA, and show that pressure overload, while enhancing IL-18 and IL-18R expression in hypertrophied and failing hearts, markedly attenuated the level of expression of the endogenous IL-18 antagonist IL-18BP. Cyclical mechanical stretch (10% cyclic equibiaxial stretch, 1Hz) induced hypertrophy of primary rabbit cardiomyocytes in vitro and enhanced ANP, IL-18, and IL-18Rα expression. Further, treatment with rhIL-18 induced its own expression and that of IL-18Rα via AP-1 activation, and induced cardiomyocyte hypertrophy in part via PI3K/Akt/GATA4 signaling. In contrast, IL-18 potentiated TNF-α-induced cardiomyocyte death, and by itself induced cardiac endothelial cell death. These results demonstrate that pressure overload is associated with enhanced IL-18 and its receptor expression in hypertrophied and failingrabbit hearts. Since IL-18BP expression is markedly inhibited, our results indicate a positive amplification in IL-18 proinflammatory signaling during pressure overload, and suggest IL-18 as a potential therapeutic target in pathological hypertrophy and cardiac failure.
Collapse
Affiliation(s)
- Tadashi Yoshida
- Heart and Vascular Institute, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Ingeborg Friehs
- Department of Cardiac Surgery, Children's Hospital Boston, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Srinivas Mummidi
- South Texas Veterans Health Care System and Department of Medicine, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Pedro J del Nido
- Department of Cardiac Surgery, Children's Hospital Boston, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Solange Addulnour-Nakhoul
- Department of Medicine-Gastroenterology, Tulane University School of Medicine, New Orleans, LA 70112, USA; Research Service, Southeast Louisiana Veterans Health Care System, New Orleans, LA 70161, USA
| | - Patrice Delafontaine
- Heart and Vascular Institute, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Anthony J Valente
- Department of Cardiac Surgery, Children's Hospital Boston, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Bysani Chandrasekar
- Heart and Vascular Institute, Tulane University School of Medicine, New Orleans, LA 70112, USA; Research Service, Southeast Louisiana Veterans Health Care System, New Orleans, LA 70161, USA.
| |
Collapse
|
285
|
Abstract
Monogenic autoinflammatory syndromes present with excessive systemic inflammation including fever, rashes, arthritis, and organ-specific inflammation and are caused by defects in single genes encoding proteins that regulate innate inflammatory pathways. Pathogenic variants in two interleukin-1 (IL-1)-regulating genes, NLRP3 and IL1RN, cause two severe and early-onset autoinflammatory syndromes, CAPS (cryopyrin associated periodic syndromes) and DIRA (deficiency of IL-1 receptor antagonist). The discovery of the mutations that cause CAPS and DIRA led to clinical and basic research that uncovered the key role of IL-1 in an extended spectrum of immune dysregulatory conditions. NLRP3 encodes cryopyrin, an intracellular "molecular sensor" that forms a multimolecular platform, the NLRP3 inflammasome, which links "danger recognition" to the activation of the proinflammatory cytokine IL-1β. The success and safety profile of drugs targeting IL -1 in the treatment of CAPS and DIRA have encouraged their wider use in other autoinflammatory syndromes including the classic hereditary periodic fever syndromes (familial Mediterranean fever, TNF receptor-associated periodic syndrome, and hyperimmunoglobulinemia D with periodic fever syndrome) and additional immune dysregulatory conditions that are not genetically well defined, including Still's, Behcet's, and Schnitzler diseases. The fact that the accumulation of metabolic substrates such as monosodium urate, ceramide, cholesterol, and glucose can trigger the NLRP3 inflammasome connects metabolic stress to IL-1β-mediated inflammation and provides a rationale for therapeutically targeting IL-1 in prevalent diseases such as gout, diabetes mellitus, and coronary artery disease.
Collapse
Affiliation(s)
- Adriana A Jesus
- Translational Autoinflammatory Disease Section, National Institute of Arthritis, Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health, Bethesda, Maryland 20982;
| | | |
Collapse
|
286
|
Wagner KB, Felix SB, Riad A. Innate immune receptors in heart failure: Side effect or potential therapeutic target? World J Cardiol 2014; 6:791-801. [PMID: 25228958 PMCID: PMC4163708 DOI: 10.4330/wjc.v6.i8.791] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 04/18/2014] [Accepted: 06/11/2014] [Indexed: 02/06/2023] Open
Abstract
Heart failure (HF) is a leading cause of mortality and morbidity in western countries and occasions major expenses for public health systems. Although optimal medical treatment is widely available according to current guidelines, the prognosis of patients with HF is still poor. Despite the etiology of the disease, increased systemic or cardiac activation of the innate immune system is well documented in several types of HF. In some cases there is evidence of an association between innate immune activation and clinical outcome of patients with this disease. However, the few large trials conducted with the use of anti-inflammatory medication in HF have not revealed its benefits. Thus, greater understanding of the relationship between alteration in the immune system and development and progression of HF is urgently necessary: prior to designing therapeutic interventions that target pathological inflammatory processes in preventing harmful cardiac effects of immune modulatory therapy. In this regard, relatively recently discovered receptors of the innate immune system, i.e., namely toll-like receptors (TLRs) and nod-like receptors (NLRs)-are the focus of intense cardiovascular research. These receptors are main up-stream regulators of cytokine activation. This review will focus on current knowledge of the role of TLRs and NLRs, as well as on downstream cytokine activation, and will discuss potential therapeutic implications.
Collapse
|
287
|
Cogle CR, Wise E, Meacham AM, Zierold C, Traverse JH, Henry TD, Perin EC, Willerson JT, Ellis SG, Carlson M, Zhao DXM, Bolli R, Cooke JP, Anwaruddin S, Bhatnagar A, da Graca Cabreira-Hansen M, Grant MB, Lai D, Moyé L, Ebert RF, Olson RE, Sayre SL, Schulman IH, Bosse RC, Scott EW, Simari RD, Pepine CJ, Taylor DA. Detailed analysis of bone marrow from patients with ischemic heart disease and left ventricular dysfunction: BM CD34, CD11b, and clonogenic capacity as biomarkers for clinical outcomes. Circ Res 2014; 115:867-74. [PMID: 25136078 DOI: 10.1161/circresaha.115.304353] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
RATIONALE Bone marrow (BM) cell therapy for ischemic heart disease (IHD) has shown mixed results. Before the full potency of BM cell therapy can be realized, it is essential to understand the BM niche after acute myocardial infarction (AMI). OBJECTIVE To study the BM composition in patients with IHD and severe left ventricular (LV) dysfunction. METHODS AND RESULTS BM from 280 patients with IHD and LV dysfunction were analyzed for cell subsets by flow cytometry and colony assays. BM CD34(+) cell percentage was decreased 7 days after AMI (mean of 1.9% versus 2.3%-2.7% in other cohorts; P<0.05). BM-derived endothelial colonies were significantly decreased (P<0.05). Increased BM CD11b(+) cells associated with worse LV ejection fraction (LVEF) after AMI (P<0.05). Increased BM CD34(+) percentage associated with greater improvement in LVEF (+9.9% versus +2.3%; P=0.03, for patients with AMI and +6.6% versus -0.02%; P=0.021 for patients with chronic IHD). In addition, decreased BM CD34(+) percentage in patients with chronic IHD correlated with decrement in LVEF (-2.9% versus +0.7%; P=0.0355). CONCLUSIONS In this study, we show a heterogeneous mixture of BM cell subsets, decreased endothelial colony capacity, a CD34+ cell nadir 7 days after AMI, a negative correlation between CD11b percentage and postinfarct LVEF, and positive correlation of CD34 percentage with change in LVEF after cell therapy. These results serve as a possible basis for the small clinical improvement seen in autologous BM cell therapy trials and support selection of potent cell subsets and reversal of comorbid BM impairment. CLINICAL TRIAL REGISTRATIONS URL http://www.clinicaltrials.gov. Unique identifiers: NCT00684021, NCT00684060, and NCT00824005.
Collapse
Affiliation(s)
- Christopher R Cogle
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - Elizabeth Wise
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - Amy M Meacham
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - Claudia Zierold
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - Jay H Traverse
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - Timothy D Henry
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - Emerson C Perin
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - James T Willerson
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - Stephen G Ellis
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - Marjorie Carlson
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - David X M Zhao
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - Roberto Bolli
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - John P Cooke
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - Saif Anwaruddin
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - Aruni Bhatnagar
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - Maria da Graca Cabreira-Hansen
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - Maria B Grant
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - Dejian Lai
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - Lem Moyé
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.).
| | - Ray F Ebert
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - Rachel E Olson
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - Shelly L Sayre
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - Ivonne H Schulman
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - Raphael C Bosse
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - Edward W Scott
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - Robert D Simari
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - Carl J Pepine
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | - Doris A Taylor
- From the University of Florida College of Medicine, Gainesville (C.R.C., E.W., A.M.M., M.B.G., R.C.B., E.W.S., C.J.P.); University of Minnesota School of Medicine, Minneapolis (C.Z., M.C.); Minneapolis Heart Institute Foundation at Abbott, MN (J.H.T., T.D.H., R.E.O.); Texas Heart Institute, Houston (E.C.P., J.T.W., M.d.G.C.-H., D.A.T.); Cleveland Clinic Foundation, OH (S.G.E.); Wake Forest Baptist Health, Winston-Salem, NC (D.X.M.Z.); University of Louisville, School of Medicine, KY (R.B., A.B.); Houston Methodist Research Institute, TX (J.P.C.); University of Pennsylvania School of Medicine, Philadelphia (S.A.); University of Texas School of Public Health, Houston (D.L., L.M., S.L.S.); National Heart, Lung and Blood Institute, Bethesda, MD (R.F.E.); University of Miami School of Medicine, FL (I.H.S.), and Mayo Clinic College of Medicine, Rochester, MN (R.D.S.)
| | | |
Collapse
|
288
|
van der Meer JWM, Simon A, Dinarello CA. Comment on "Power of rare diseases: found in translation". Sci Transl Med 2014; 6:219le1. [PMID: 24431110 DOI: 10.1126/scitranslmed.3007911] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The recent Perspective entitled "Power of rare diseases: Found in translation" undervalues the contributions of academic research in first-in-human studies.
Collapse
Affiliation(s)
- Jos W M van der Meer
- Department of Medicine, Radboud University Medical Center, 6500HB Nijmegen, the Netherlands
| | | | | |
Collapse
|
289
|
Interleukin-1β Blockade Improves Left Ventricular Systolic/Diastolic Function and Restores Contractility Reserve in Severe Ischemic Cardiomyopathy in the Mouse. J Cardiovasc Pharmacol 2014; 64:1-6. [DOI: 10.1097/fjc.0000000000000106] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
290
|
|
291
|
Serhal M, Longenecker CT. Preventing Heart Failure in Inflammatory and Immune Disorders. CURRENT CARDIOVASCULAR RISK REPORTS 2014; 8. [PMID: 26316924 DOI: 10.1007/s12170-014-0392-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Patients with chronic inflammatory diseases are at increased risk for heart failure due to ischemic heart disease and other causes including heart failure with preserved ejection fraction. Using rheumatoid arthritis and treated HIV infection as two prototypical examples, we review the epidemiology and potential therapies to prevent heart failure in these populations. Particular focus is given to anti-inflammatory therapies including statins and biologic disease modifying drugs. There is also limited evidence for lifestyle changes and blockade of the renin-angiotensin-aldosterone system. We conclude by proposing how a strategy for heart failure prevention, such as the model tested in the Screening To Prevent Heart Failure (STOP-HF) trial, may be adapted to chronic inflammatory disease.
Collapse
Affiliation(s)
- Maya Serhal
- University Hospitals Case Medical Center, Cleveland, OH, USA
| | - Chris T Longenecker
- University Hospitals Case Medical Center, Cleveland, OH, USA ; Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
292
|
O'Brien LC, Mezzaroma E, Van Tassell BW, Marchetti C, Carbone S, Abbate A, Toldo S. Interleukin-18 as a therapeutic target in acute myocardial infarction and heart failure. Mol Med 2014; 20:221-9. [PMID: 24804827 DOI: 10.2119/molmed.2014.00034] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 04/28/2014] [Indexed: 12/13/2022] Open
Abstract
Interleukin 18 (IL-18) is a proinflammatory cytokine in the IL-1 family that has been implicated in a number of disease states. In animal models of acute myocardial infarction (AMI), pressure overload, and LPS-induced dysfunction, IL-18 regulates cardiomyocyte hypertrophy and induces cardiac contractile dysfunction and extracellular matrix remodeling. In patients, high IL-18 levels correlate with increased risk of developing cardiovascular disease (CVD) and with a worse prognosis in patients with established CVD. Two strategies have been used to counter the effects of IL-18:IL-18 binding protein (IL-18BP), a naturally occurring protein, and a neutralizing IL-18 antibody. Recombinant human IL-18BP (r-hIL-18BP) has been investigated in animal studies and in phase I/II clinical trials for psoriasis and rheumatoid arthritis. A phase II clinical trial using a humanized monoclonal IL-18 antibody for type 2 diabetes is ongoing. Here we review the literature regarding the role of IL-18 in AMI and heart failure and the evidence and challenges of using IL-18BP and blocking IL-18 antibodies as a therapeutic strategy in patients with heart disease.
Collapse
Affiliation(s)
- Laura C O'Brien
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Eleonora Mezzaroma
- Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, Virginia, United States of America Virginia Commonwealth University Pauley Heart Center, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America Pharmacotherapy and Outcome Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Benjamin W Van Tassell
- Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, Virginia, United States of America Virginia Commonwealth University Pauley Heart Center, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America Pharmacotherapy and Outcome Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Carlo Marchetti
- Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, Virginia, United States of America Virginia Commonwealth University Pauley Heart Center, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Salvatore Carbone
- Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, Virginia, United States of America Virginia Commonwealth University Pauley Heart Center, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Antonio Abbate
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia, United States of America Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, Virginia, United States of America Virginia Commonwealth University Pauley Heart Center, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Stefano Toldo
- Victoria Johnson Research Laboratories, Virginia Commonwealth University, Richmond, Virginia, United States of America Virginia Commonwealth University Pauley Heart Center, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| |
Collapse
|
293
|
The emerging role of biotechnological drugs in the treatment of gout. BIOMED RESEARCH INTERNATIONAL 2014; 2014:264859. [PMID: 24839602 PMCID: PMC4009249 DOI: 10.1155/2014/264859] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 03/16/2014] [Indexed: 12/21/2022]
Abstract
One of the most important therapeutic advances obtained in the field of rheumatology is the availability of the so-called bio(techno)logical drugs, which have deeply changed treatment perspectives in diseases such as rheumatoid arthritis and ankylosing spondylitis. According to the steadily increasing attention on gout, due to well-established prognostic and epidemiology implications, in the last 5 years, the same change of perspective has been observed also for this disease. In fact, several bio(techno)logical agents have been investigated both for the management of the articular gout symptoms, targeting mainly interleukin-1β, as well as urate-lowering therapies such as recombinant uricases. Among the IL-1β inhibitors, the majority of studies involve drugs such as anakinra, canakinumab, and rilonacept, but other compounds are under development. Moreover, other potential targets have been suggested, as, for example, the TNF alpha and IL-6, even if data obtained are less robust than those of IL-1β inhibitors. Regarding urate-lowering therapies, the recombinant uricases pegloticase and rasburicase clearly showed their effectiveness in gout patients. Also in this case, new compounds are under development. The aim of this review is to focus on the various aspects of different bio(techno)logical drugs in gouty patients.
Collapse
|
294
|
Anti-Inflammatory Treatment With Colchicine in Stable Chronic Heart Failure. JACC-HEART FAILURE 2014; 2:131-7. [DOI: 10.1016/j.jchf.2013.11.006] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 11/25/2013] [Accepted: 11/28/2013] [Indexed: 11/24/2022]
|
295
|
Toldo S, Mezzaroma E, O'Brien L, Marchetti C, Seropian IM, Voelkel NF, Van Tassell BW, Dinarello CA, Abbate A. Interleukin-18 mediates interleukin-1-induced cardiac dysfunction. Am J Physiol Heart Circ Physiol 2014; 306:H1025-31. [PMID: 24531812 PMCID: PMC3962640 DOI: 10.1152/ajpheart.00795.2013] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 02/12/2014] [Indexed: 12/15/2022]
Abstract
Patients with heart failure (HF) have enhanced systemic IL-1 activity, and, in the experimental mouse model, IL-1 induces left ventricular (LV) systolic dysfunction. Whether the effects of IL-1 are direct or mediated by an inducible cytokine, such as IL-18, is unknown. Recombinant human IL-18-binding protein (IL-18BP) or an IL-18-blocking antibody (IL-18AB) was used to neutralize endogenous IL-18 after challenge with the plasma of patients with HF or with recombinant murine IL-1β in adult male mice. Plasma levels of IL-18 and IL-6 (a key mediator of IL-1-induced systemic effects) and LV fractional shortening were measured in mice sedated with pentobarbital sodium (30-50 mg/kg). Mice with genetic deletion of IL-18 or IL-18 receptors were compared with matching wild-type mice. A group of mice received murine IL-18 to evaluate the effects on LV fractional shortening. Plasma from HF patients and IL-1β induced LV systolic dysfunction that was prevented by pretreatment with IL-18AB or IL-18BP. IL-1β failed to induce LV systolic dysfunction in mice with genetic deletion of IL-18 signaling. IL-1β induced a significant increase in plasma IL-18 and IL-6 levels. Genetic or pharmacological inhibition of IL-18 signaling failed to block the induction of IL-6 by IL-1β. In conclusion, IL-1 induces a release of active IL-18 in the mouse that mediates the LV systolic dysfunction but not the induction of IL-6. IL-18 blockade may therefore represent a novel and more targeted therapeutic approach to treat HF.
Collapse
Affiliation(s)
- Stefano Toldo
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| | | | | | | | | | | | | | | | | |
Collapse
|
296
|
Abstract
Myocardial infarction triggers an intense inflammatory response that is essential for cardiac repair, but which is also implicated in the pathogenesis of postinfarction remodelling and heart failure. Signals in the infarcted myocardium activate toll-like receptor signalling, while complement activation and generation of reactive oxygen species induce cytokine and chemokine upregulation. Leukocytes recruited to the infarcted area, remove dead cells and matrix debris by phagocytosis, while preparing the area for scar formation. Timely repression of the inflammatory response is critical for effective healing, and is followed by activation of myofibroblasts that secrete matrix proteins in the infarcted area. Members of the transforming growth factor β family are critically involved in suppression of inflammation and activation of a profibrotic programme. Translation of these concepts to the clinic requires an understanding of the pathophysiological complexity and heterogeneity of postinfarction remodelling in patients with myocardial infarction. Individuals with an overactive and prolonged postinfarction inflammatory response might exhibit left ventricular dilatation and systolic dysfunction and might benefit from targeted anti-IL-1 or anti-chemokine therapies, whereas patients with an exaggerated fibrogenic reaction can develop heart failure with preserved ejection fraction and might require inhibition of the Smad3 (mothers against decapentaplegic homolog 3) cascade. Biomarker-based approaches are needed to identify patients with distinct pathophysiologic responses and to rationally implement inflammation-modulating strategies.
Collapse
|
297
|
Li X, Deroide N, Mallat Z. The role of the inflammasome in cardiovascular diseases. J Mol Med (Berl) 2014; 92:307-19. [PMID: 24638861 DOI: 10.1007/s00109-014-1144-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 02/26/2014] [Accepted: 03/04/2014] [Indexed: 12/17/2022]
Abstract
Inflammasome is a very important signaling platform sensing a variety of triggers of the innate immune system. Inflammasome promotes the production of important pro-inflammatory cytokines such as IL-1β and IL-18. Tight control of inflammasome activity is, therefore, essential and occurs at multiple levels. The activation of inflammasome pathways is linked to the pathogenesis of various prevalent disorders including cardiovascular disease such as atherosclerosis, ischemic injury, cardiomyopathy, and Kawasaki disease. The study of the inflammasome in the cardiovascular system has led to the identification of important triggers and endogenous modulators, and to the exploration of new treatment strategies based on the inhibition of inflammasome activation or its end products, i.e., IL-1β and IL-18. In summary, the discovery of the inflammasome has greatly advanced our understanding of how the innate immune system interferes with cardiovascular disease development and progression, and targeting inflammasome provides new avenues for the treatment and management of cardiovascular diseases.
Collapse
Affiliation(s)
- Xuan Li
- Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 2QQ, UK
| | | | | |
Collapse
|
298
|
Falcao RA, Christopher S, Oddi C, Reznikov L, Grizzard JD, Abouzaki NA, Varma A, Van Tassell BW, Dinarello CA, Abbate A. Interleukin-10 in patients with ST-segment elevation myocardial infarction. Int J Cardiol 2014; 172:e6-8. [DOI: 10.1016/j.ijcard.2013.12.126] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 12/20/2013] [Indexed: 11/26/2022]
|
299
|
Seropian IM, Toldo S, Van Tassell BW, Abbate A. Anti-inflammatory strategies for ventricular remodeling following ST-segment elevation acute myocardial infarction. J Am Coll Cardiol 2014; 63:1593-603. [PMID: 24530674 DOI: 10.1016/j.jacc.2014.01.014] [Citation(s) in RCA: 225] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 12/28/2013] [Accepted: 01/08/2014] [Indexed: 12/21/2022]
Abstract
Acute myocardial infarction (AMI) leads to molecular, structural, geometric, and functional changes in the heart in a process known as ventricular remodeling. An intense organized inflammatory response is triggered after myocardial ischemia and necrosis and involves all components of the innate immunity, affecting both cardiomyocytes and noncardiomyocyte cells. Inflammation is triggered by tissue injury; it mediates wound healing and scar formation and affects ventricular remodeling. Many therapeutic attempts aimed at reducing inflammation in AMI during the past 3 decades presented issues of impaired healing or increased risk of cardiac rupture or failed to show any additional benefit in addition to standard therapies. More recent strategies aimed at selectively blocking one of the key factors upstream rather than globally suppressing the response downstream have shown some promising results in pilot trials. We herein review the pathophysiological mechanisms of inflammation and ventricular remodeling after AMI and the results of clinical trials with anti-inflammatory strategies.
Collapse
Affiliation(s)
| | - Stefano Toldo
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia; Victoria Johnson Research Laboratory, Virginia Commonwealth University, Richmond, Virginia
| | - Benjamin W Van Tassell
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia; Victoria Johnson Research Laboratory, Virginia Commonwealth University, Richmond, Virginia; School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia; Victoria Johnson Research Laboratory, Virginia Commonwealth University, Richmond, Virginia.
| |
Collapse
|
300
|
Van Tassell BW, Arena R, Biondi-Zoccai G, Canada JM, Oddi C, Abouzaki NA, Jahangiri A, Falcao RA, Kontos MC, Shah KB, Voelkel NF, Dinarello CA, Abbate A. Effects of interleukin-1 blockade with anakinra on aerobic exercise capacity in patients with heart failure and preserved ejection fraction (from the D-HART pilot study). Am J Cardiol 2014; 113:321-327. [PMID: 24262762 PMCID: PMC4899612 DOI: 10.1016/j.amjcard.2013.08.047] [Citation(s) in RCA: 212] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2013] [Revised: 08/20/2013] [Accepted: 08/20/2013] [Indexed: 02/08/2023]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a clinical syndrome of exercise intolerance due to impaired myocardial relaxation and/or increased stiffness. Patients with HFpEF often show signs of chronic systemic inflammation, and experimental studies have shown that interleukin-1 (IL-1), a key proinflammatory cytokine, impairs myocardial relaxation. The aim of the present study was to determine the effects of IL-1 blockade with anakinra on aerobic exercise capacity in patients with HFpEF and plasma C-reactive protein (CRP) >2 mg/L (reflecting increased IL-1 activity). A total of 12 patients were enrolled in a double-blind, randomized, placebo-controlled, crossover trial and assigned 1:1 to receive 1 of the 2 treatments (anakinra 100 mg or placebo) for 14 days and an additional 14 days of the alternate treatment (placebo or anakinra). The cardiopulmonary exercise test was performed at baseline, after the first 14 days, and after the second 14 days of treatment. The placebo-corrected interval change in peak oxygen consumption was chosen as the primary end point. All 12 patients enrolled in the present study and receiving treatment completed both phases and experienced no major adverse events. Anakinra led to a statistically significant improvement in peak oxygen consumption (+1.2 ml/kg/min, p = 0.009) and a significant reduction in plasma CRP levels (-74%, p = 0.006). The reduction in CRP levels correlated with the improvement in peak oxygen consumption (R = -0.60, p = 0.002). Three patients (25%) had mild and self-limiting injection site reactions. In conclusion, IL-1 blockade with anakinra for 14 days significantly reduced the systemic inflammatory response and improved the aerobic exercise capacity of patients with HFpEF and elevated plasma CRP levels.
Collapse
Affiliation(s)
- Benjamin Wallace Van Tassell
- Virginia Commonwealth University, VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Richmond, VA, USA
| | | | | | - Justin McNair Canada
- Virginia Commonwealth University, VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Richmond, VA, USA
| | - Claudia Oddi
- Virginia Commonwealth University, VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Richmond, VA, USA
| | - Nayef Antar Abouzaki
- Virginia Commonwealth University, VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Richmond, VA, USA
| | - Arehzo Jahangiri
- Virginia Commonwealth University, VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Richmond, VA, USA
| | - Raquel Appa Falcao
- Virginia Commonwealth University, VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Richmond, VA, USA
| | - Michael Christopher Kontos
- Virginia Commonwealth University, VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Richmond, VA, USA
| | - Keyur Bharat Shah
- Virginia Commonwealth University, VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Richmond, VA, USA
| | - Norbert Felix Voelkel
- Virginia Commonwealth University, VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Richmond, VA, USA
| | | | - Antonio Abbate
- Virginia Commonwealth University, VCU Pauley Heart Center and Victoria Johnson Research Laboratories, Richmond, VA, USA
| |
Collapse
|