1
|
Kehr D, Ritterhoff J, Glaser M, Jarosch L, Salazar RE, Spaich K, Varadi K, Birkenstock J, Egger M, Gao E, Koch WJ, Sauter M, Freichel M, Katus HA, Frey N, Jungmann A, Busch C, Mather PJ, Ruhparwar A, Busch M, Völkers M, Wade RC, Most P. S100A1ct: A Synthetic Peptide Derived From S100A1 Protein Improves Cardiac Performance and Survival in Preclinical Heart Failure Models. Circulation 2025; 151:548-565. [PMID: 39569500 PMCID: PMC11850016 DOI: 10.1161/circulationaha.123.066961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/15/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND The EF-hand Ca2+ sensor protein S100A1 has been identified as a molecular regulator and enhancer of cardiac performance. The ability of S100A1 to recognize and modulate the activity of targets such as SERCA2a (sarcoplasmic reticulum Ca2+ ATPase) and RyR2 (ryanodine receptor 2) in cardiomyocytes has mostly been ascribed to its hydrophobic C-terminal α-helix (residues 75-94). We hypothesized that a synthetic peptide consisting of residues 75 through 94 of S100A1 and an N-terminal solubilization tag (S100A1ct) could mimic the performance-enhancing effects of S100A1 and may be suitable as a peptide therapeutic to improve the function of diseased hearts. METHODS We applied an integrative translational research pipeline ranging from in silico computational molecular modeling and in vitro biochemical molecular assays as well as isolated rodent and human cardiomyocyte performance assessments to in vivo safety and efficacy studies in small and large animal cardiac disease models. RESULTS We characterize S100A1ct as a cell-penetrating peptide with positive inotropic and antiarrhythmic properties in normal and failing myocardium in vitro and in vivo. This activity translates into improved contractile performance and survival in preclinical heart failure models with reduced ejection fraction after S100A1ct systemic administration. S100A1ct exerts a fast and sustained dose-dependent enhancement of cardiomyocyte Ca2+ cycling and prevents β-adrenergic receptor-triggered Ca2+ imbalances by targeting SERCA2a and RyR2 activity. In line with the S100A1ct-mediated enhancement of SERCA2a activity, modeling suggests an interaction of the peptide with the transmembrane segments of the sarcoplasmic Ca2+ pump. Incorporation of a cardiomyocyte-targeting peptide tag into S100A1ct (cor-S100A1ct) further enhanced its biological and therapeutic potency in vitro and in vivo. CONCLUSIONS S100A1ct is a promising lead for the development of novel peptide-based therapeutics against heart failure with reduced ejection fraction.
Collapse
Affiliation(s)
- Dorothea Kehr
- Molecular and Translational Cardiology (D.K., J.R., K.S., K.V., J.B., M.E., A.J., M.B., P.M.), Heidelberg University Hospital (UKHD), Germany
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg, Germany (D.K., J.R., K.V., M.F., H.A.K., N.F., A.J., M.B., M.V., P.M.)
| | - Julia Ritterhoff
- Molecular and Translational Cardiology (D.K., J.R., K.S., K.V., J.B., M.E., A.J., M.B., P.M.), Heidelberg University Hospital (UKHD), Germany
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg, Germany (D.K., J.R., K.V., M.F., H.A.K., N.F., A.J., M.B., M.V., P.M.)
- Informatics for Life (I4L) consortium, Heidelberg, Germany (J.R., M.G., H.A.K., N.F., R.C.W., P.M.)
| | - Manuel Glaser
- Heidelberg Institute for Theoretical Studies (HITS), Germany (M.G., L.J., R.E.S., R.C.W.)
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany (M.G., R.C.W.)
- Informatics for Life (I4L) consortium, Heidelberg, Germany (J.R., M.G., H.A.K., N.F., R.C.W., P.M.)
| | - Lukas Jarosch
- Heidelberg Institute for Theoretical Studies (HITS), Germany (M.G., L.J., R.E.S., R.C.W.)
| | - Rafael E. Salazar
- Heidelberg Institute for Theoretical Studies (HITS), Germany (M.G., L.J., R.E.S., R.C.W.)
| | - Kristin Spaich
- Molecular and Translational Cardiology (D.K., J.R., K.S., K.V., J.B., M.E., A.J., M.B., P.M.), Heidelberg University Hospital (UKHD), Germany
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
| | - Karl Varadi
- Molecular and Translational Cardiology (D.K., J.R., K.S., K.V., J.B., M.E., A.J., M.B., P.M.), Heidelberg University Hospital (UKHD), Germany
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg, Germany (D.K., J.R., K.V., M.F., H.A.K., N.F., A.J., M.B., M.V., P.M.)
| | - Jennifer Birkenstock
- Molecular and Translational Cardiology (D.K., J.R., K.S., K.V., J.B., M.E., A.J., M.B., P.M.), Heidelberg University Hospital (UKHD), Germany
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
| | - Michael Egger
- Molecular and Translational Cardiology (D.K., J.R., K.S., K.V., J.B., M.E., A.J., M.B., P.M.), Heidelberg University Hospital (UKHD), Germany
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
| | - Erhe Gao
- Center for Translational Medicine, Temple University, Philadelphia, PA (E.G.)
| | - Walter J. Koch
- Division of Cardiovascular and Thoracic Surgery, Duke University, Durham, NC (W.J.K.)
| | - Max Sauter
- Department of Clinical Pharmacology and Pharmacoepidemiology (M.S.), Heidelberg University Hospital (UKHD), Germany
| | - Marc Freichel
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg, Germany (D.K., J.R., K.V., M.F., H.A.K., N.F., A.J., M.B., M.V., P.M.)
- Department of Pharmacology, Heidelberg Medical Faculty, Germany (M.F.)
| | - Hugo A. Katus
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg, Germany (D.K., J.R., K.V., M.F., H.A.K., N.F., A.J., M.B., M.V., P.M.)
- Informatics for Life (I4L) consortium, Heidelberg, Germany (J.R., M.G., H.A.K., N.F., R.C.W., P.M.)
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg, Germany (D.K., J.R., K.V., M.F., H.A.K., N.F., A.J., M.B., M.V., P.M.)
- Informatics for Life (I4L) consortium, Heidelberg, Germany (J.R., M.G., H.A.K., N.F., R.C.W., P.M.)
| | - Andreas Jungmann
- Molecular and Translational Cardiology (D.K., J.R., K.S., K.V., J.B., M.E., A.J., M.B., P.M.), Heidelberg University Hospital (UKHD), Germany
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg, Germany (D.K., J.R., K.V., M.F., H.A.K., N.F., A.J., M.B., M.V., P.M.)
| | - Cornelius Busch
- Department of Anesthesiology (C.B.), Heidelberg University Hospital (UKHD), Germany
| | - Paul J. Mather
- Perelman School of Medicine, University of Pennsylvania, Philadelphia (P.J.M.)
| | - Arjang Ruhparwar
- Division for Cardiothoracic-, Transplantation- and Vascular Surgery, Hannover Medical School, Hannover, Germany (A.R.)
| | - Martin Busch
- Molecular and Translational Cardiology (D.K., J.R., K.S., K.V., J.B., M.E., A.J., M.B., P.M.), Heidelberg University Hospital (UKHD), Germany
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg, Germany (D.K., J.R., K.V., M.F., H.A.K., N.F., A.J., M.B., M.V., P.M.)
| | - Mirko Völkers
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg, Germany (D.K., J.R., K.V., M.F., H.A.K., N.F., A.J., M.B., M.V., P.M.)
| | - Rebecca C. Wade
- Heidelberg Institute for Theoretical Studies (HITS), Germany (M.G., L.J., R.E.S., R.C.W.)
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany (M.G., R.C.W.)
- Interdisciplinary Center for Scientific Computing (IWR), Heidelberg, Germany (R.C.W.)
- Informatics for Life (I4L) consortium, Heidelberg, Germany (J.R., M.G., H.A.K., N.F., R.C.W., P.M.)
| | - Patrick Most
- Molecular and Translational Cardiology (D.K., J.R., K.S., K.V., J.B., M.E., A.J., M.B., P.M.), Heidelberg University Hospital (UKHD), Germany
- Department of Cardiology, Angiology and Pneumology (D.K., J.R., K.S., K.V., J.B., M.E., H.A.K., N.F., A.J., M.B., M.V., P.M.), Heidelberg University Hospital (UKHD), Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg, Germany (D.K., J.R., K.V., M.F., H.A.K., N.F., A.J., M.B., M.V., P.M.)
- Informatics for Life (I4L) consortium, Heidelberg, Germany (J.R., M.G., H.A.K., N.F., R.C.W., P.M.)
- Center for Translational Medicine, Jefferson University, Philadelphia, PA (P.M.)
| |
Collapse
|
2
|
Lee YS, Choi JR, Kim JB. Gene Therapy for Cardiovascular Disease: Clinical Perspectives. Yonsei Med J 2024; 65:557-571. [PMID: 39313446 PMCID: PMC11427124 DOI: 10.3349/ymj.2024.0127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 09/25/2024] Open
Abstract
Cardiovascular disease (CVD) stands as one of the leading causes of death in the United States, with its prevalence steadily on the rise. Traditional therapeutic approaches, such as pharmacological treatment, cardiovascular intervention, and surgery, have inherent limitations. In response to these challenges, cardiac gene therapy has emerged as a promising alternative for treating CVD patients. However, several obstacles persist, including the low efficiency of gene transduction, immune reactions to vectors or transduced cells, and the occurrence of off-target effects. While preclinical research has demonstrated significant success in various CVD model in both small and large animals, the translation of these findings to clinical applications has, for the most part, yielded disappointing results, except for some early, albeit small, trials. This review aims to provide a comprehensive summary of recent preclinical and clinical studies on gene therapy for various CVDs. Additionally, we discuss the existing limitations and challenges that hinder the widespread clinical application of cardiac gene therapy.
Collapse
Affiliation(s)
- Young Shin Lee
- Division of Cardiology, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Jung Ran Choi
- College of Medicine, Kyung Hee University, Seoul, Korea
| | - Jin-Bae Kim
- Division of Cardiology, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul, Korea
- College of Medicine, Kyung Hee University, Seoul, Korea.
| |
Collapse
|
3
|
Seitz A, Busch M, Kroemer J, Schneider A, Simon S, Jungmann A, Katus HA, Most P, Ritterhoff J. S100A1's single cysteine is an indispensable redox switch for the protection against diastolic calcium waves in cardiomyocytes. Am J Physiol Heart Circ Physiol 2024; 327:H000. [PMID: 38819384 PMCID: PMC11381028 DOI: 10.1152/ajpheart.00634.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 04/22/2024] [Accepted: 04/22/2024] [Indexed: 06/01/2024]
Abstract
The EF-hand calcium (Ca2+) sensor protein S100A1 combines inotropic with antiarrhythmic potency in cardiomyocytes (CMs). Oxidative posttranslational modification (ox-PTM) of S100A1's conserved, single-cysteine residue (C85) via reactive nitrogen species (i.e., S-nitrosylation or S-glutathionylation) has been proposed to modulate conformational flexibility of intrinsically disordered sequence fragments and to increase the molecule's affinity toward Ca2+. Considering the unknown biological functional consequence, we aimed to determine the impact of the C85 moiety of S100A1 as a potential redox switch. We first uncovered that S100A1 is endogenously glutathionylated in the adult heart in vivo. To prevent glutathionylation of S100A1, we generated S100A1 variants that were unresponsive to ox-PTMs. Overexpression of wild-type (WT) and C85-deficient S100A1 protein variants in isolated CM demonstrated equal inotropic potency, as shown by equally augmented Ca2+ transient amplitudes under basal conditions and β-adrenergic receptor (βAR) stimulation. However, in contrast, ox-PTM defective S100A1 variants failed to protect against arrhythmogenic diastolic sarcoplasmic reticulum (SR) Ca2+ waves and ryanodine receptor 2 (RyR2) hypernitrosylation during βAR stimulation. Despite diastolic performance failure, C85-deficient S100A1 protein variants exerted similar Ca2+-dependent interaction with the RyR2 than WT-S100A1. Dissecting S100A1's molecular structure-function relationship, our data indicate for the first time that the conserved C85 residue potentially acts as a redox switch that is indispensable for S100A1's antiarrhythmic but not its inotropic potency in CMs. We, therefore, propose a model where C85's ox-PTM determines S100A1's ability to beneficially control diastolic but not systolic RyR2 activity.NEW & NOTEWORTHY S100A1 is an emerging candidate for future gene-therapy treatment of human chronic heart failure. We aimed to study the significance of the conserved single-cysteine 85 (C85) residue in cardiomyocytes. We show that S100A1 is endogenously glutathionylated in the heart and demonstrate that this is dispensable to increase systolic Ca2+ transients, but indispensable for mediating S100A1's protection against sarcoplasmic reticulum (SR) Ca2+ waves, which was dependent on the ryanodine receptor 2 (RyR2) nitrosylation status.
Collapse
Affiliation(s)
- Andreas Seitz
- Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
- Department of Cardiology and Angiology, Robert-Bosch-Krankenhaus, Stuttgart, Germany
| | - Martin Busch
- Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
| | - Jasmin Kroemer
- Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
| | - Andrea Schneider
- Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
| | - Stephanie Simon
- Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
| | - Andreas Jungmann
- Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Hugo A Katus
- German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Heidelberg, Germany
- Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
| | - Patrick Most
- Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Heidelberg, Germany
- Informatics for Life consortium, Klaus Tschira Foundation, Heidelberg, Germany
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Julia Ritterhoff
- Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Heidelberg, Germany
- Informatics for Life consortium, Klaus Tschira Foundation, Heidelberg, Germany
| |
Collapse
|
4
|
Noll D, Kehr D, Most P, Ritterhoff J. S100A1: a promising therapeutic target for heart failure. Expert Opin Ther Targets 2024; 28:233-236. [PMID: 38641766 DOI: 10.1080/14728222.2024.2345746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/17/2024] [Indexed: 04/21/2024]
Affiliation(s)
- Dorothea Noll
- Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
| | - Dorothea Kehr
- Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
| | - Patrick Most
- Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
- Center for Translational Medicine, Jefferson University, Philadelphia, PA, USA
| | - Julia Ritterhoff
- Molecular and Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
| |
Collapse
|
5
|
Zhou Y, Zha Y, Yang Y, Ma T, Li H, Liang J. S100 proteins in cardiovascular diseases. Mol Med 2023; 29:68. [PMID: 37217870 DOI: 10.1186/s10020-023-00662-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 05/02/2023] [Indexed: 05/24/2023] Open
Abstract
Cardiovascular diseases have become a serious threat to human health and life worldwide and have the highest fatality rate. Therefore, the prevention and treatment of cardiovascular diseases have become a focus for public health experts. The expression of S100 proteins is cell- and tissue-specific; they are implicated in cardiovascular, neurodegenerative, and inflammatory diseases and cancer. This review article discusses the progress in the research on the role of S100 protein family members in cardiovascular diseases. Understanding the mechanisms by which these proteins exert their biological function may provide novel concepts for preventing, treating, and predicting cardiovascular diseases.
Collapse
Affiliation(s)
- Yue Zhou
- Medical College, Yangzhou University, Yangzhou, China
| | - Yiwen Zha
- Medical College, Yangzhou University, Yangzhou, China
| | - Yongqi Yang
- Medical College, Yangzhou University, Yangzhou, China
| | - Tan Ma
- Medical College, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Hongliang Li
- Medical College, Yangzhou University, Yangzhou, China.
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China.
| | - Jingyan Liang
- Medical College, Yangzhou University, Yangzhou, China.
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China.
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China.
| |
Collapse
|
6
|
Tsoporis JN, Amatullah H, Gupta S, Izhar S, Ektesabi AM, Vaswani CM, Desjardins JF, Kabir G, Teixera Monteiro AP, Varkouhi AK, Kavantzas N, Salpeas V, Rizos I, Marshall JC, Parker TG, Leong-Poi H, Dos Santos CC. DJ-1 Deficiency Protects against Sepsis-Induced Myocardial Depression. Antioxidants (Basel) 2023; 12:antiox12030561. [PMID: 36978809 PMCID: PMC10045744 DOI: 10.3390/antiox12030561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/07/2023] [Accepted: 02/20/2023] [Indexed: 03/30/2023] Open
Abstract
Oxidative stress is considered one of the early underlying contributors of sepsis-induced myocardial depression. DJ-1, also known as PARK7, has a well-established role as an antioxidant. We have previously shown, in a clinically relevant model of polymicrobial sepsis, DJ-1 deficiency improved survival and bacterial clearance by decreasing ROS production. In the present study, we investigated the role of DJ-1 in sepsis-induced myocardial depression. Here we compared wildtype (WT) with DJ-1 deficient mice at 24 and 48 h after cecal ligation and puncture (CLP). In WT mice, DJ-1 was increased in the myocardium post-CLP. DJ-1 deficient mice, despite enhanced inflammatory and oxidative responses, had an attenuated hypertrophic phenotype, less apoptosis, improved mitochondrial function, and autophagy, that was associated with preservation of myocardial function and improved survival compared to WT mice post-CLP. Collectively, these results identify DJ-1 as a regulator of myocardial function and as such, makes it an attractive therapeutic target in the treatment of early sepsis-induced myocardial depression.
Collapse
Affiliation(s)
- James N Tsoporis
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Hajera Amatullah
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sahil Gupta
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Shehla Izhar
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Amin M Ektesabi
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Chirag M Vaswani
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jean-Francois Desjardins
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Golam Kabir
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Ana Paula Teixera Monteiro
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Amir K Varkouhi
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Nikolaos Kavantzas
- 1st Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Vasileios Salpeas
- 1st Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Ioannis Rizos
- 2nd Department of Cardiology, Attikon University Hospital, 12462 Athens, Greece
| | - John C Marshall
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Thomas G Parker
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Howard Leong-Poi
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Claudia C Dos Santos
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
7
|
Sanganalmath SK, Dubey S, Veeranki S, Narisetty K, Krishnamurthy P. The interplay of inflammation, exosomes and Ca 2+ dynamics in diabetic cardiomyopathy. Cardiovasc Diabetol 2023; 22:37. [PMID: 36804872 PMCID: PMC9942322 DOI: 10.1186/s12933-023-01755-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 01/25/2023] [Indexed: 02/22/2023] Open
Abstract
Diabetes mellitus is one of the prime risk factors for cardiovascular complications and is linked with high morbidity and mortality. Diabetic cardiomyopathy (DCM) often manifests as reduced cardiac contractility, myocardial fibrosis, diastolic dysfunction, and chronic heart failure. Inflammation, changes in calcium (Ca2+) handling and cardiomyocyte loss are often implicated in the development and progression of DCM. Although the existence of DCM was established nearly four decades ago, the exact mechanisms underlying this disease pathophysiology is constantly evolving. Furthermore, the complex pathophysiology of DCM is linked with exosomes, which has recently shown to facilitate intercellular (cell-to-cell) communication through biomolecules such as micro RNA (miRNA), proteins, enzymes, cell surface receptors, growth factors, cytokines, and lipids. Inflammatory response and Ca2+ signaling are interrelated and DCM has been known to adversely affect many of these signaling molecules either qualitatively and/or quantitatively. In this literature review, we have demonstrated that Ca2+ regulators are tightly controlled at different molecular and cellular levels during various biological processes in the heart. Inflammatory mediators, miRNA and exosomes are shown to interact with these regulators, however how these mediators are linked to Ca2+ handling during DCM pathogenesis remains elusive. Thus, further investigations are needed to understand the mechanisms to restore cardiac Ca2+ homeostasis and function, and to serve as potential therapeutic targets in the treatment of DCM.
Collapse
Affiliation(s)
- Santosh K Sanganalmath
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Nevada Las Vegas School of Medicine, Las Vegas, NV, 89102, USA.
| | - Shubham Dubey
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, University Blvd., Birmingham, AL, 35294, USA
| | - Sudhakar Veeranki
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40506, USA
| | | | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, University Blvd., Birmingham, AL, 35294, USA
| |
Collapse
|
8
|
Yan M, Yang Y, Zhou Y, Yu C, Li R, Gong W, Zheng J. Interleukin-7 aggravates myocardial ischaemia/reperfusion injury by regulating macrophage infiltration and polarization. J Cell Mol Med 2021; 25:9939-9952. [PMID: 34581005 PMCID: PMC8572772 DOI: 10.1111/jcmm.16335] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 12/24/2022] Open
Abstract
Interleukin (IL)‐7 is known to enhance the macrophages cytotoxic activity and that macrophages play a pivotal role in the development and progression of myocardial ischaemia/reperfusion (I/R) injury. However, the effects of IL‐7 on macrophages infiltration and polarization in myocardial I/R injury are currently unclear. This study aimed to evaluate the effects of the IL‐7 expression on myocardial I/R injury and their relationship with macrophages. The data showed that IL‐7 expression in mouse heart tissue increases following I/R injury and that IL‐7 knockout or anti‐IL‐7 antibody treatment significantly improve I/R injury, including reduction in myocardial infarction area, a serum troponin T level decreases and an improvement in cardiac function. On the other hand, recombinant IL‐7 (rIL‐7) supplementation induces opposite effects and the anti‐IL‐7 antibody significantly reduces the cardiomyocyte apoptosis and macrophage infiltration. rIL‐7 cannot directly cause apoptosis, but it can induce cardiomyocyte apoptosis through macrophages, in addition to increase the macrophages migration in vitro. Anti‐IL‐7 antibody affects the cytokine production in T helper (Th) 1 and Th2 cells and also promotes the macrophages differentiation to M2 macrophages. However, anti‐IL‐7 antibody does not reduce the M1 macrophage number, and it only increases the ratio of M2/M1 macrophages in mice heart tissues after I/R injury. Taking together, these data reveal that IL‐7 plays an intensifying role in myocardial I/R injury by promoting cardiomyocyte apoptosis through the regulation of macrophage infiltration and polarization.
Collapse
Affiliation(s)
- Mengwen Yan
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Yaliu Yang
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Ying Zhou
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Changan Yu
- Central Laboratory of Cardiovascular Disease, China-Japan Friendship Hospital, Beijing, China
| | - Rui Li
- Department of Health Care, China-Japan Freindship Hospital, Ministry of Health, Beijing, China
| | - Wei Gong
- Emergency and Critical Care Center, Beijing Anzhen Hospital Capital Medical University, Beijing, China.,Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Jingang Zheng
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China.,Department of Cardiology, China-Japan Friendship School of Clinical Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.,Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| |
Collapse
|
9
|
Glaser M, Bruce NJ, Han SB, Wade RC. Simulation of the Positive Inotropic Peptide S100A1ct in Aqueous Environment by Gaussian Accelerated Molecular Dynamics. J Phys Chem B 2021; 125:4654-4666. [PMID: 33944558 DOI: 10.1021/acs.jpcb.1c00902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The S100A1ct peptide, consisting of the C-terminal 20 residues of the S100A1 protein fused to an N-terminal 6-residue hydrophilic tag, has been found to exert a positive inotropic effect, resulting in improved contractile performance of failing cardiac and skeletal muscle without arrhythmic side-effects. The S100A1ct peptide thus has high potential for the treatment of acute heart failure. As a step toward understanding its molecular mechanism of action, and to provide a basis for peptidomimetic design to optimize its properties, we here describe de novo structure predictions and molecular dynamics simulations to characterize the conformational landscape of S100A1ct in aqueous environment. In S100A1, the C-terminal 20 residues form an α-helix, but de novo peptide structure predictions indicate that other conformations are also possible. Conventional molecular dynamics simulations in implicit and explicit solvent corroborated this finding. To ensure adequate sampling, we performed simulations of a tagged 10-residue segment of S100A1ct, and we carried out Gaussian accelerated molecular dynamics simulations of the peptides. These simulations showed that although the helical conformation of S100A1ct was the most energetically stable, the peptide can adopt a range of kinked conformations, suggesting that its activity may be related to its ability to act as a conformational switch.
Collapse
Affiliation(s)
- Manuel Glaser
- Heidelberg Institute for Theoretical Studies (HITS), Schloss-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany.,Informatics for Life, Heidelberg, Germany
| | - Neil J Bruce
- Heidelberg Institute for Theoretical Studies (HITS), Schloss-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany.,Informatics for Life, Heidelberg, Germany
| | - Sungho Bosco Han
- Heidelberg Institute for Theoretical Studies (HITS), Schloss-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Rebecca C Wade
- Heidelberg Institute for Theoretical Studies (HITS), Schloss-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany.,Informatics for Life, Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany.,Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Alliance and Interdisciplinary Center for Scientific Computing (IWR), 69120 Heidelberg, Germany
| |
Collapse
|
10
|
Li X, Wang X, Sun T, Ping Y, Dai Y, Liu Z, Wang Y, Wang D, Xia X, Shan H, Zhang W, Tao Z. S100A1 is a sensitive and specific cardiac biomarker for early diagnosis and prognostic assessment of acute myocardial infarction measured by chemiluminescent immunoassay. Clin Chim Acta 2021; 516:71-76. [PMID: 33476588 DOI: 10.1016/j.cca.2021.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 12/20/2020] [Accepted: 01/12/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND A member of the S100 family of Ca2+-binding proteins, S100A1 is highly expressed in cardiac muscle tissue. Although this protein is considered an indicator of acute myocardial infarction (AMI), high-throughput and sensitive detection methods are still urgently needed. We constructed a rapid and sensitive method for detecting S100A1 and to investigate the clinical utility of S100A1 as a biomarker for the early diagnosis of AMI and subsequent prognostic assessments. We developed an automated chemiluminescent immunoassay to detect S100A1. We then analyzed the performance of the newly developed assay including evaluation of the analytical sensitivity, analytical selectivity, linear range, accuracy and repeatability. METHODS We recruited 87 patients with AMI or angina pectoris to explore the value of this marker for the early diagnosis and prognostic assessment. RESULTS The chemiluminescent-immune-based S100A1 assay had functional analytical sensitivity with a detection limit of 0.13 ng/ml, and a wide linear range of 0.13-31.66 ng/ml. It also exhibited good repeatability with intra-assay and inter-assay findings of <5% and <15%, respectively. Plasma S100A1 was found to have a higher diagnostic sensitivity than conventional cardiac biomarkers (creatine kinase-MB and cardiac troponin T). The survival analysis showed that a higher concentration of plasma S100A1 (>1.02 ng/ml) was notably associated with the poor prognosis of AMI patients after first PCI. CONCLUSIONS Measurement of circulating S100A1 concentrations with our newly developed chemiluminescent-immune-based assay shows potential for use in the clinic. This assay could enable early identification and prognostic assessment of AMI.
Collapse
Affiliation(s)
- Xiaoying Li
- The First People's Hospital of Hangzhou Lin'an District, China
| | - Xuchu Wang
- Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, China
| | - Tao Sun
- Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, China
| | - Ying Ping
- Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, China
| | - Yibei Dai
- Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, China
| | - Zhenping Liu
- Yuhang Branch of the Second Affiliated Hospital of Zhejiang University, China
| | - Yiyun Wang
- Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, China
| | - Danhua Wang
- Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, China
| | - Xiaofen Xia
- The First People's Hospital of Hangzhou Lin'an District, China
| | | | - Weiqun Zhang
- The First People's Hospital of Hangzhou Lin'an District, China
| | - Zhihua Tao
- Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, China.
| |
Collapse
|
11
|
Abstract
Supplemental Digital Content is Available in the Text. Ischemia and anoxia-induced mitochondrial impairment may be a key factor leading to heart injury during myocardial infarction (MI). Calpain 1 and 2 are involved in the MI-induced mitochondria injury. G protein-coupled receptor 35 (GPR35) could be triggered by hypoxia. Whether or not GPR35 regulates calpain 1/2 in the pathogenesis of MI is still unclear. In this study, we determined that MI increases GPR35 expression in myocardial tissue. Suppression of GPR35 protects heart from MI injury in mice through reduction of reactive oxygen species activity and mitochondria-dependent apoptosis. Further studies show that GPR35 regulates calpain 1/2. Suppression of GPR35 reduces the expression and activity of calpain 1/2, and alleviates calpain 1/2-associated mitochondrial injury to preserve cardiac function. Based on these data, we conclude that a functional inhibition of GPR35 downregulates calpain 1/2 and contributes to maintenance of cardiac function under pathologic conditions with mitochondrial disorder. In conclusion, our study showed that the identified regulation by GPR35 of calpain 1/2 has important implications for the pathogenesis of MI. Targeting the action of GPR35 and calpain 1/2 in mitochondria presents a potential therapeutic intervention for MI.
Collapse
|
12
|
Soltani L, Kheirouri S, Enamzadeh E. Elevated serum levels of S100A1 and zinc α2-glycoprotein in patients with heart failure. Nutr Metab Cardiovasc Dis 2021; 31:162-168. [PMID: 33257194 DOI: 10.1016/j.numecd.2020.07.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 06/27/2020] [Accepted: 07/20/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND AIMS Heart failure (HF) is a growing concern worldwide. S100A1 and zinc α2-glycoprotein (ZAG) play an important role in heart function. We examined serum levels of S100A1 and ZAG in HF patients and their association with anthropometric indices and body composition. METHODS AND RESULTS Sixty-four patients with HF, mean age 56.2, 48 male and 16 females, with ejection fraction <30-35%, were recruited from Shahid Madani Heart Hospital in Tabriz, Iran, from April to October 2019. Two groups, cachexia (n = 32) and non-cachexia (n = 32), which were divided based on weight loss of at least 7.5% in the last six months, were compared with the control group (n = 26). S100A1 and ZAG serum levels were determined by ELISA. Serum median (min-max) levels of S100A1 and ZAG were significantly greater in HF patients [326 (184.8-635.2) and 150.4 (61.5-520.7)] than healthy controls [265.4 (43.6-658.8) and 119.8 (16.7-533)], both p = 0.001. S100A1 Serum levels in cachexia group was significantly higher than non-cachexia group [331 (245.6-469.6) vs. 318 (184.8-635.2), p = 0.03]. A strong positive association was observed between S100A1 and ZAG serum levels in patients (r = 0.70, p < 0.0001). Serum levels of these two proteins negatively and significantly associated with BMI (r = -0.25, p = 0.044 and r = -0.28, p = 0.024, respectively) and arm circumference (r = -0.26, p = 0.037 and r = -0.25, p = 0.047, respectively). CONCLUSION The results indicate that S100A1 and ZAG are likely to contribute to the pathogenesis of HF disease and weight loss, as well as the strong association between S100A1 and ZAG possibly indicating a similar mechanism of action for these two proteins.
Collapse
Affiliation(s)
- Leila Soltani
- Students Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sorayya Kheirouri
- Department of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Elgar Enamzadeh
- Cardiovascular Research Center, Madani Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
13
|
Immunohistochemistry in the Detection of Early Myocardial Infarction: Systematic Review and Analysis of Limitations Because of Autolysis and Putrefaction. Appl Immunohistochem Mol Morphol 2020; 28:95-102. [PMID: 32044877 DOI: 10.1097/pai.0000000000000688] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The postmortem diagnosis of acute myocardial infarction is one of the main problems in forensic practice, especially in cases in which death occurs soon after (from minutes to a few hours) the onset of the ischemic damage. Several authors have highlighted the possibility to overcome the limits of conventional histology in this diagnosis by utilizing immunohistochemistry. In the present research, we examined over 30 scientific studies and picked out over 20 main immunohistochemical antigens analyzed with a view to enabling the rapid diagnosis of early myocardial infarction. The aim of our review was to examine and summarize all the principal markers studied to date and also to consider their limitations, including protein alteration because of cadaveric autolysis and putrefaction.
Collapse
|
14
|
Njegic A, Wilson C, Cartwright EJ. Targeting Ca 2 + Handling Proteins for the Treatment of Heart Failure and Arrhythmias. Front Physiol 2020; 11:1068. [PMID: 33013458 PMCID: PMC7498719 DOI: 10.3389/fphys.2020.01068] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/04/2020] [Indexed: 12/18/2022] Open
Abstract
Diseases of the heart, such as heart failure and cardiac arrhythmias, are a growing socio-economic burden. Calcium (Ca2+) dysregulation is key hallmark of the failing myocardium and has long been touted as a potential therapeutic target in the treatment of a variety of cardiovascular diseases (CVD). In the heart, Ca2+ is essential for maintaining normal cardiac function through the generation of the cardiac action potential and its involvement in excitation contraction coupling. As such, the proteins which regulate Ca2+ cycling and signaling play a vital role in maintaining Ca2+ homeostasis. Changes to the expression levels and function of Ca2+-channels, pumps and associated intracellular handling proteins contribute to altered Ca2+ homeostasis in CVD. The remodeling of Ca2+-handling proteins therefore results in impaired Ca2+ cycling, Ca2+ leak from the sarcoplasmic reticulum and reduced Ca2+ clearance, all of which contributes to increased intracellular Ca2+. Currently, approved treatments for targeting Ca2+ handling dysfunction in CVD are focused on Ca2+ channel blockers. However, whilst Ca2+ channel blockers have been successful in the treatment of some arrhythmic disorders, they are not universally prescribed to heart failure patients owing to their ability to depress cardiac function. Despite the progress in CVD treatments, there remains a clear need for novel therapeutic approaches which are able to reverse pathophysiology associated with heart failure and arrhythmias. Given that heart failure and cardiac arrhythmias are closely associated with altered Ca2+ homeostasis, this review will address the molecular changes to proteins associated with both Ca2+-handling and -signaling; their potential as novel therapeutic targets will be discussed in the context of pre-clinical and, where available, clinical data.
Collapse
Affiliation(s)
- Alexandra Njegic
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, United Kingdom.,Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Claire Wilson
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, United Kingdom.,Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Elizabeth J Cartwright
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
15
|
Zhou X, Xia N, Lv B, Tang T, Nie S, Zhang M, Jiao J, Liu J, Xu C, Hou G, Yang X, Hu Y, Liao Y, Cheng X. Interleukin 35 ameliorates myocardial ischemia‐reperfusion injury by activating the gp130‐STAT3 axis. FASEB J 2020; 34:3224-3238. [PMID: 31917470 DOI: 10.1096/fj.201901718rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 12/17/2019] [Accepted: 12/20/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Xingdi Zhou
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Biological Targeted Therapy of Education Ministry and Hubei Province Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Ni Xia
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Biological Targeted Therapy of Education Ministry and Hubei Province Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Bingjie Lv
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Biological Targeted Therapy of Education Ministry and Hubei Province Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Tingting Tang
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Biological Targeted Therapy of Education Ministry and Hubei Province Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Shaofang Nie
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Biological Targeted Therapy of Education Ministry and Hubei Province Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Min Zhang
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Biological Targeted Therapy of Education Ministry and Hubei Province Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Jiao Jiao
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Biological Targeted Therapy of Education Ministry and Hubei Province Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Jianfeng Liu
- Sino‐France Laboratory of cellular signaling, Key Laboratory of Molecular Biophysics of Ministry of Education College of Life Science and Technology and Collaborative Innovation Center for Genetics and Development Huazhong University of Science and Technology Wuhan Hubei China
| | - Chanjuan Xu
- Sino‐France Laboratory of cellular signaling, Key Laboratory of Molecular Biophysics of Ministry of Education College of Life Science and Technology and Collaborative Innovation Center for Genetics and Development Huazhong University of Science and Technology Wuhan Hubei China
| | - Guofei Hou
- Sino‐France Laboratory of cellular signaling, Key Laboratory of Molecular Biophysics of Ministry of Education College of Life Science and Technology and Collaborative Innovation Center for Genetics and Development Huazhong University of Science and Technology Wuhan Hubei China
| | - Xiangping Yang
- School of Basic Medicine Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Yu Hu
- Key Laboratory of Biological Targeted Therapy of Education Ministry and Hubei Province Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Institute of Hematology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Yuhua Liao
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Biological Targeted Therapy of Education Ministry and Hubei Province Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Xiang Cheng
- Department of Cardiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Biological Targeted Therapy of Education Ministry and Hubei Province Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| |
Collapse
|
16
|
Sreejit G, Flynn MC, Patil M, Krishnamurthy P, Murphy AJ, Nagareddy PR. S100 family proteins in inflammation and beyond. Adv Clin Chem 2020; 98:173-231. [PMID: 32564786 DOI: 10.1016/bs.acc.2020.02.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The S100 family proteins possess a variety of intracellular and extracellular functions. They interact with multiple receptors and signal transducers to regulate pathways that govern inflammation, cell differentiation, proliferation, energy metabolism, apoptosis, calcium homeostasis, cell cytoskeleton and microbial resistance. S100 proteins are also emerging as novel diagnostic markers for identifying and monitoring various diseases. Strategies aimed at targeting S100-mediated signaling pathways hold a great potential in developing novel therapeutics for multiple diseases. In this chapter, we aim to summarize the current knowledge about the role of S100 family proteins in health and disease with a major focus on their role in inflammatory conditions.
Collapse
Affiliation(s)
| | - Michelle C Flynn
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Mallikarjun Patil
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Andrew J Murphy
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Department of Immunology, Monash University, Melbourne, VIC, Australia
| | | |
Collapse
|
17
|
Riuzzi F, Chiappalupi S, Arcuri C, Giambanco I, Sorci G, Donato R. S100 proteins in obesity: liaisons dangereuses. Cell Mol Life Sci 2020; 77:129-147. [PMID: 31363816 PMCID: PMC11104817 DOI: 10.1007/s00018-019-03257-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/19/2019] [Accepted: 07/24/2019] [Indexed: 02/07/2023]
Abstract
Obesity is an endemic pathophysiological condition and a comorbidity associated with hypercholesterolemia, hypertension, cardiovascular disease, type 2 diabetes mellitus, and cancer. The adipose tissue of obese subjects shows hypertrophic adipocytes, adipocyte hyperplasia, and chronic low-grade inflammation. S100 proteins are Ca2+-binding proteins exclusively expressed in vertebrates in a cell-specific manner. They have been implicated in the regulation of a variety of functions acting as intracellular Ca2+ sensors transducing the Ca2+ signal and extracellular factors affecting cellular activity via ligation of a battery of membrane receptors. Certain S100 proteins, namely S100A4, the S100A8/S100A9 heterodimer and S100B, have been implicated in the pathophysiology of obesity-promoting macrophage-based inflammation via toll-like receptor 4 and/or receptor for advanced glycation end-products ligation. Also, serum levels of S100A4, S100A8/S100A9, S100A12, and S100B correlate with insulin resistance/type 2 diabetes, metabolic risk score, and fat cell size. Yet, secreted S100B appears to exert neurotrophic effects on sympathetic fibers in brown adipose tissue contributing to the larger sympathetic innervation of this latter relative to white adipose tissue. In the present review we first briefly introduce S100 proteins and then critically examine their role(s) in adipose tissue and obesity.
Collapse
Affiliation(s)
- Francesca Riuzzi
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
- Interuniversity Institute of Myology (IIM), University of Perugia, 06132, Perugia, Italy
| | - Sara Chiappalupi
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
- Interuniversity Institute of Myology (IIM), University of Perugia, 06132, Perugia, Italy
| | - Cataldo Arcuri
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
| | - Ileana Giambanco
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
| | - Guglielmo Sorci
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
- Interuniversity Institute of Myology (IIM), University of Perugia, 06132, Perugia, Italy
- Centro Universitario di Ricerca sulla Genomica Funzionale, University of Perugia, 06132, Perugia, Italy
| | - Rosario Donato
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy.
| |
Collapse
|
18
|
Zhao J, Xu T, Zhou Y, Zhou Y, Xia Y, Li D. B-type natriuretic peptide and its role in altering Ca 2+-regulatory proteins in heart failure-mechanistic insights. Heart Fail Rev 2019; 25:861-871. [PMID: 31820203 DOI: 10.1007/s10741-019-09883-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Heart failure (HF) is a worldwide disease with high levels of morbidity and mortality. The pathogenesis of HF is complicated and involves imbalances in hormone and electrolyte. B-type natriuretic peptide (BNP) has served as a biomarker of HF severity, and in recent years, it has been used to treat the disease, thanks to its cardio-protective effects, such as diuresis, natriuresis, and vasodilatation. In stage C/D HF, symptoms are severe despite elevated BNP. Disturbances in Ca2+ homeostasis are often a dominating feature of the disease, causing Ca2+-regulatory protein dysfunction, including reduced expression and activity of sarcoplasmic reticulum Ca2+-ATPase2a (SERCA2a), impaired ryanodine receptors (RYRs) function, intensive Na+-Ca2+ exchanger (NCX), and downregulation of S100A1. The relationship between natriuretic peptides (NPs) and Ca2+-regulatory proteins has been widely studied and represents important mechanisms in the etiology of HF. In this review, we present evidence that BNP may regulate Ca2+-regulatory proteins, in particular, suppressing SERCA2a and S100A1 expression. However, relationships between BNP and other Ca2+-regulatory proteins remain vague.
Collapse
Affiliation(s)
- Jiaqi Zhao
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Tongda Xu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Yao Zhou
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - You Zhou
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China
| | - Yong Xia
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China.
| | - Dongye Li
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China. .,Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People's Republic of China.
| |
Collapse
|
19
|
Fan L, Liu B, Guo R, Luo J, Li H, Li Z, Xu W. Elevated plasma S100A1 level is a risk factor for ST-segment elevation myocardial infarction and associated with post-infarction cardiac function. Int J Med Sci 2019; 16:1171-1179. [PMID: 31523180 PMCID: PMC6743283 DOI: 10.7150/ijms.35037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 07/17/2019] [Indexed: 01/20/2023] Open
Abstract
AIM To investigate the association between plasma S100A1 level and ST-segment elevation myocardial infarction (STEMI) and potential significance of S100A1 in post-infarction cardiac function. METHODS We examined the plasma S100A1 level in 207 STEMI patients (STEMI group) and 217 clinically healthy subjects for routine physical examination without a history of coronary artery disease (Control group). Baseline characteristics and concentrations of relevant biomarkers were compared. The relationship between S100A1 and other plasma biomarkers was detected using correlation analysis. The predictive role of S100A1 on occurrence of STEMI was then assessed using multivariate ordinal regression model analysis after adjusting for other covariates. RESULTS The plasma S100A1 level was found to be significantly higher (P<0.001) in STEMI group (3197.7±1576.0 pg/mL) than in Control (1423.5±1315.5 pg/mL) group. Furthermore, the correlation analysis demonstrated plasma S100A1 level was significantly associated correlated with hypersensitive cardiac troponin T (hs-cTnT) (r = 0.32; P < 0.001), creatine kinase MB (CK-MB) (r = 0.42, P < 0.001), left ventricular eject fraction (LVEF) (r = -0.12, P = 0.01), N-terminal prohormone of brain natriuretic peptide (NT-proBNP) (r = 0.61; P < 0.001) and hypersensitive C reactive protein (hs-CRP) (r = 0.38; P < 0.001). Moreover, the enrolled subjects who with a S100A1 concentration ≤ 1965.9 pg/mL presented significantly better cardiac function than the rest population. Multivariate Logistic regression analysis revealed that S100A1 was an independent predictor for STEMI patients (OR: 0.671, 95% CI 0.500-0.891, P<0.001). In addition, higher S100A1 concentration (> 1965.9 pg/mL) significantly increased the risk of STEMI as compared with the lower level (OR: 6.925; 95% CI: 4.15-11.375; P<0.001). CONCLUSION These results indicated that the elevated plasma S100A1 level is an important predictor of STEMI in combination with several biomarkers and also potentially reflects the cardiac function following the acute coronary ischemia.
Collapse
Affiliation(s)
- Linlin Fan
- Institute of Biomedical Sciences, Department of Cardiology, Shanghai Institute of Cardiovascular Disease, Fudan University, Shanghai, 200032, China
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Baoxin Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Rong Guo
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Jiachen Luo
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Hongqiang Li
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Zhiqiang Li
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Weigang Xu
- Community Health Service Center of Pengpu New Estate, Jing'an District, Shanghai, 200435, China
| |
Collapse
|
20
|
Yu Z, Zhang Y, Tang Z, Song J, Gao X, Sun T, Liu Y, Yang J, Wang T, Liu J. Intracavernosal Adeno-Associated Virus-Mediated S100A1 Gene Transfer Enhances Erectile Function in Diabetic Rats by Promoting Cavernous Angiogenesis via VEGF-A/VEGFR2 Signaling. J Sex Med 2019; 16:1344-1354. [PMID: 31378707 DOI: 10.1016/j.jsxm.2019.06.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/26/2019] [Accepted: 06/17/2019] [Indexed: 10/26/2022]
Abstract
INTRODUCTION Novel therapeutic targets for diabetes-induced erectile dysfunction (DED) are urgently needed. Previous studies have proved that S100A1, a small Ca2+-binding protein, is a pluripotent regulator of cardiovascular pathophysiology. Its absence is associated with endothelial dysfunction, the central event linking cardiovascular changes in diabetes. However, the role of S100A1 in DED remains unknown. AIM To explore the effect and underlying mechanisms of S100A1 in restoring erectile function in type I diabetic rat model. METHODS Diabetes was induced by intraperitoneal injection of streptozotocin and then screened by apomorphine (APO) to confirm erectile dysfunction. Rats that met the criteria of penile erection were marked as APO-positive; otherwise, the result was APO-negative. In experiment 1, S100A1 gene expression alterations in the corpus cavernosum in moderate and established stages of DED were analyzed. In experiment 2, S100A1 and control GFP gene were delivered into the corpus cavernosum in APO-negative rats by adeno-associated virus (AAV) serotype 9. Erectile function was assessed at 4 weeks after gene therapy. MAIN OUTCOME MEASURES Erectile response, histologic and molecular alterations. RESULTS S100A1 protein was localized to the area surrounding the cavernosal sinusoids in the penis, and it was gradually downregulated synchronized with the progression of DED. Compared with an injection of AAV-GFP, a single injection of AAV-S100A1 significantly restored erectile function in diabetic rats. S100A1 overexpression significantly upregulated the expression of endogenous VEGF-A, promoted VEGFR2 internalization, and subsequently triggered the protein kinase B-endothelial nitric oxide synthase pathway in diabetic erectile tissues. Marked increases in nitric oxide and endothelial content were noted in AAV-S100A1-treated diabetic rats. CLINICAL IMPLICATIONS Local S100A1 overexpression may be an alternative therapy for DED and should be further investigated by future clinical studies. STRENGTH & LIMITATIONS This is the first study demonstrating the angiogenic role of S100A1 in DED, but does not preclude the contribution of the effects of S100A1 in other tissues such as the neuronal tissue on the functional effects observed in erectile responses. CONCLUSION The decreased expression of S100A1 during hyperglycemia might be important in the development of erectile dysfunction. S100A1 may play a potential role in restoring erectile function in rats with DED through modulating cavernous angiogenesis. Yu Z, Zhang Y, Tang Z, et al. Intracavernosal Adeno-Associated Virus-Mediated S100A1 Gene Transfer Enhances Erectile Function in Diabetic Rats by Promoting Cavernous Angiogenesis via VEGF-A/VEGFR2 Signaling. J Sex Med 2019;16:1344-1354.
Collapse
Affiliation(s)
- Zhe Yu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yan Zhang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhe Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jingyu Song
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xintao Gao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Taotao Sun
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yang Liu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jun Yang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Tao Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jihong Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
21
|
Heizmann CW. S100 proteins: Diagnostic and prognostic biomarkers in laboratory medicine. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:1197-1206. [DOI: 10.1016/j.bbamcr.2018.10.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 10/12/2018] [Indexed: 01/04/2023]
|
22
|
Heizmann CW. Ca 2+-Binding Proteins of the EF-Hand Superfamily: Diagnostic and Prognostic Biomarkers and Novel Therapeutic Targets. Methods Mol Biol 2019; 1929:157-186. [PMID: 30710273 DOI: 10.1007/978-1-4939-9030-6_11] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A multitude of Ca2+-sensor proteins containing the specific Ca2+-binding motif (helix-loop-helix, called EF-hand) are of major clinical relevance in a many human diseases. Measurements of troponin, the first intracellular Ca-sensor protein to be discovered, is nowadays the "gold standard" in the diagnosis of patients with acute coronary syndrome (ACS). Mutations have been identified in calmodulin and linked to inherited ventricular tachycardia and in patients affected by severe cardiac arrhythmias. Parvalbumin, when introduced into the diseased heart by gene therapy to increase contraction and relaxation speed, is considered to be a novel therapeutic strategy to combat heart failure. S100 proteins, the largest subgroup with the EF-hand protein family, are closely associated with cardiovascular diseases, various types of cancer, inflammation, and autoimmune pathologies. The intention of this review is to summarize the clinical importance of this protein family and their use as biomarkers and potential drug targets, which could help to improve the diagnosis of human diseases and identification of more selective therapeutic interventions.
Collapse
Affiliation(s)
- Claus W Heizmann
- Department of Pediatrics, Division of Clinical Chemistry and Biochemistry, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
23
|
Teissier T, Boulanger É. The receptor for advanced glycation end-products (RAGE) is an important pattern recognition receptor (PRR) for inflammaging. Biogerontology 2019; 20:279-301. [PMID: 30968282 DOI: 10.1007/s10522-019-09808-3] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/02/2019] [Indexed: 12/20/2022]
Abstract
The receptor for advanced glycation end-products (RAGE) was initially characterized and named for its ability to bind to advanced glycation end-products (AGEs) that form upon the irreversible and non-enzymatic interaction between nucleophiles, such as lysine, and carbonyl compounds, such as reducing sugars. The concentrations of AGEs are known to increase in conditions such as diabetes, as well as during ageing. However, it is now widely accepted that RAGE binds with numerous ligands, many of which can be defined as pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs). The interaction between RAGE and its ligands mainly results in a pro-inflammatory response, and can lead to stress events often favouring mitochondrial dysfunction or cellular senescence. Thus, RAGE should be considered as a pattern recognition receptor (PRR), similar to those that regulate innate immunity. Innate immunity itself plays a central role in inflammaging, the chronic low-grade and sterile inflammation that increases with age and is a potentially important contributory factor in ageing. Consequently, and in addition to the age-related accumulation of PAMPs and DAMPs and increases in pro-inflammatory cytokines from senescent cells and damaged cells, PRRs are therefore important in inflammaging. We suggest here that, through its interconnection with immunity, senescence, mitochondrial dysfunction and inflammasome activation, RAGE is a key contributor to inflammaging and that the pro-longevity effects seen upon blocking RAGE, or upon its deletion, are thus the result of reduced inflammaging.
Collapse
Affiliation(s)
- Thibault Teissier
- Univ. Lille, Inserm, CHU Lille, U995 - LIRIC - Lille Inflammation Research International Center, 59000, Lille, France.
| | - Éric Boulanger
- Univ. Lille, Inserm, CHU Lille, U995 - LIRIC - Lille Inflammation Research International Center, 59000, Lille, France.,Department of Geriatrics and Ageing Biology, School of Medicine, Lille University, Lille, France.,Department of Geriatrics, Lille University Hospital, Lille, France
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW The current knowledge of pathophysiological and molecular mechanisms responsible for the genesis and development of heart failure (HF) is absolutely vast. Nonetheless, the hiatus between experimental findings and therapeutic options remains too deep, while the available pharmacological treatments are mostly seasoned and display limited efficacy. The necessity to identify new, non-pharmacological strategies to target molecular alterations led investigators, already many years ago, to propose gene therapy for HF. Here, we will review some of the strategies proposed over the past years to target major pathogenic mechanisms/factors responsible for severe cardiac injury developing into HF and will provide arguments in favor of the necessity to keep alive research on this topic. RECENT FINDINGS After decades of preclinical research and phases of enthusiasm and disappointment, clinical trials were finally launched in recent years. The first one to reach phase II and testing gene delivery of sarcoendoplasmic reticulum calcium ATPase did not yield encouraging results; however, other trials are ongoing, more efficient viral vectors are being developed, and promising new potential targets have been identified. For instance, recent research is focused on gene repair, in vivo, to treat heritable forms of HF, while strong experimental evidence indicates that specific microRNAs can be delivered to post-ischemic hearts to induce regeneration, a result that was previously thought possible only by using stem cell therapy. Gene therapy for HF is aging, but exciting perspectives are still very open.
Collapse
Affiliation(s)
- Khatia Gabisonia
- Institute of Life Sciences, Fondazione Toscana Gabriele Monasterio, Scuola Superiore Sant'Anna, Piazza Martiri della Liberta` 33, 56127, Pisa, Italy
| | - Fabio A Recchia
- Institute of Life Sciences, Fondazione Toscana Gabriele Monasterio, Scuola Superiore Sant'Anna, Piazza Martiri della Liberta` 33, 56127, Pisa, Italy.
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
25
|
Medkova A, Srovnal J, Potomkova J, Volejnikova J, Mihal V. Multifarious diagnostic possibilities of the S100 protein family: predominantly in pediatrics and neonatology. World J Pediatr 2018; 14:315-321. [PMID: 29858979 DOI: 10.1007/s12519-018-0163-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 05/11/2018] [Indexed: 01/05/2023]
Abstract
BACKGROUND Numerous articles related to S100 proteins have been recently published. This review aims to introduce this large protein family and its importance in the diagnostics of many pathological conditions in children and adults. DATA SOURCES Based on original publications found in database systems, we summarize the current knowledge about the S100 protein group and highlight the most important proteins with focus on pediatric use. RESULTS The S100 family is composed of Ca2+ and Zn2+ binding proteins, which are present only in vertebrates. Some of these proteins can be used as diagnostic markers in cardiology (S100A1, S100A12), oncology (S100A2, S100A5, S100A6, S100A14, S100A16, S100P, S100B), neurology (S100B), rheumatology (S100A8/A9, S100A4, S100A6, and S100A12), nephrology and infections (S100A8, S100A9, S100A8/A9, S100A12). The most useful S100 proteins in pediatrics are S100A8, S100A9, heterodimers S100A8/A9, S100B and S100A12. CONCLUSIONS The S100 family members are promising biomarkers and provide numerous possibilities for implementation into clinical practice to optimize the differential diagnostic process.
Collapse
Affiliation(s)
- Anna Medkova
- Department of Pediatrics, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, I. P. Pavlova 6, 779 00, Olomouc, Czech Republic.
| | - Josef Srovnal
- Department of Pediatrics, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, I. P. Pavlova 6, 779 00, Olomouc, Czech Republic
- Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University Olomouc, Hněvotínská, 1333/5, 779 00, Olomouc, Czech Republic
| | - Jarmila Potomkova
- Department of Pediatrics, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, I. P. Pavlova 6, 779 00, Olomouc, Czech Republic
- Department of Science and Research, University Hospital Olomouc, I. P. Pavlova 6, 779 00, Olomouc, Czech Republic
| | - Jana Volejnikova
- Department of Pediatrics, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, I. P. Pavlova 6, 779 00, Olomouc, Czech Republic
- Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University Olomouc, Hněvotínská, 1333/5, 779 00, Olomouc, Czech Republic
| | - Vladimir Mihal
- Department of Pediatrics, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, I. P. Pavlova 6, 779 00, Olomouc, Czech Republic
- Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University Olomouc, Hněvotínská, 1333/5, 779 00, Olomouc, Czech Republic
| |
Collapse
|
26
|
Hernández‐Ochoa EO, Melville Z, Vanegas C, Varney KM, Wilder PT, Melzer W, Weber DJ, Schneider MF. Loss of S100A1 expression leads to Ca 2+ release potentiation in mutant mice with disrupted CaM and S100A1 binding to CaMBD2 of RyR1. Physiol Rep 2018; 6:e13822. [PMID: 30101473 PMCID: PMC6087734 DOI: 10.14814/phy2.13822] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 11/24/2022] Open
Abstract
Calmodulin (CaM) and S100A1 fine-tune skeletal muscle Ca2+ release via opposite modulation of the ryanodine receptor type 1 (RyR1). Binding to and modulation of RyR1 by CaM and S100A1 occurs predominantly at the region ranging from amino acid residue 3614-3640 of RyR1 (here referred to as CaMBD2). Using synthetic peptides, it has been shown that CaM binds to two additional regions within the RyR1, specifically residues 1975-1999 and 4295-4325 (CaMBD1 and CaMBD3, respectively). Because S100A1 typically binds to similar motifs as CaM, we hypothesized that S100A1 could also bind to CaMBD1 and CaMBD3. Our goals were: (1) to establish whether S100A1 binds to synthetic peptides containing CaMBD1 and CaMBD3 using isothermal calorimetry (ITC), and (2) to identify whether S100A1 and CaM modulate RyR1 Ca2+ release activation via sites other than CaMBD2 in RyR1 in its native cellular context. We developed the mouse model (RyR1D-S100A1KO), which expresses point mutation RyR1-L3625D (RyR1D) that disrupts the modulation of RyR1 by CaM and S100A1 at CaMBD2 and also lacks S100A1 (S100A1KO). ITC assays revealed that S100A1 binds with different affinities to CaMBD1 and CaMBD3. Using high-speed Ca2+ imaging and a model for Ca2+ binding and transport, we show that the RyR1D-S100A1KO muscle fibers exhibit a modest but significant increase in myoplasmic Ca2+ transients and enhanced Ca2+ release flux following field stimulation when compared to fibers from RyR1D mice, which were used as controls to eliminate any effect of binding at CaMBD2, but with preserved S100A1 expression. Our results suggest that S100A1, similar to CaM, binds to CaMBD1 and CaMBD3 within the RyR1, but that CaMBD2 appears to be the primary site of RyR1 regulation by CaM and S100A1.
Collapse
Affiliation(s)
- Erick O. Hernández‐Ochoa
- Department of Biochemistry and Molecular BiologyUniversity of Maryland School of MedicineBaltimoreMaryland
| | - Zephan Melville
- Department of Biochemistry and Molecular BiologyUniversity of Maryland School of MedicineBaltimoreMaryland
| | - Camilo Vanegas
- Department of Biochemistry and Molecular BiologyUniversity of Maryland School of MedicineBaltimoreMaryland
| | - Kristen M. Varney
- Department of Biochemistry and Molecular BiologyUniversity of Maryland School of MedicineBaltimoreMaryland
- Center for Biomolecular Therapeutics (CBT)University of Maryland School of MedicineMaryland
| | - Paul T. Wilder
- Department of Biochemistry and Molecular BiologyUniversity of Maryland School of MedicineBaltimoreMaryland
- Center for Biomolecular Therapeutics (CBT)University of Maryland School of MedicineMaryland
| | - Werner Melzer
- Institute of Applied PhysiologyUlm UniversityUlmGermany
| | - David J. Weber
- Department of Biochemistry and Molecular BiologyUniversity of Maryland School of MedicineBaltimoreMaryland
- Center for Biomolecular Therapeutics (CBT)University of Maryland School of MedicineMaryland
| | - Martin F. Schneider
- Department of Biochemistry and Molecular BiologyUniversity of Maryland School of MedicineBaltimoreMaryland
| |
Collapse
|
27
|
Liu S, Xia Y, Liu X, Wang Y, Chen Z, Xie J, Qian J, Shen H, Yang P. In-depth proteomic profiling of left ventricular tissues in human end-stage dilated cardiomyopathy. Oncotarget 2018; 8:48321-48332. [PMID: 28427148 PMCID: PMC5564650 DOI: 10.18632/oncotarget.15689] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/12/2017] [Indexed: 01/30/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is caused by reduced left ventricular (LV) myocardial function, which is one of the most common causes of heart failure (HF). We performed iTRAQ-coupled 2D-LC-MS/MS to profile the cardiac proteome of LV tissues from healthy controls and patients with end-stage DCM. We identified 4263 proteins, of which 125 were differentially expressed in DCM tissues compared to LV controls. The majority of these were membrane proteins related to cellular junctions and neuronal metabolism. In addition, these proteins were involved in membrane organization, mitochondrial organization, translation, protein transport, and cell death process. Four key proteins involved in the cell death process were also detected by western blotting, indicated that cell death was activated in DCM tissues. Furthermore, S100A1 and eEF2 were enriched in the “cellular assembly and organization” and “cell cycle” networks, respectively. We verified decreases in these two proteins in end-stage DCM LV samples through multiple reaction monitoring (MRM). These observations demonstrate that our understanding of the mechanisms underlying DCM can be deepened through comparison of the proteomes of normal LV tissues with that from end-stage DCM in humans.
Collapse
Affiliation(s)
- Shanshan Liu
- Institutes of Biomedical Sciences of Shanghai Medical School and Minhang Hospital, Fudan University, Shanghai, China.,Department of Systems Biology for Medicine and School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yan Xia
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaohui Liu
- Department of chemistry, Fudan University, Shanghai, China
| | - Yi Wang
- Institutes of Biomedical Sciences of Shanghai Medical School and Minhang Hospital, Fudan University, Shanghai, China.,Department of Systems Biology for Medicine and School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhangwei Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Juanjuan Xie
- Institutes of Biomedical Sciences of Shanghai Medical School and Minhang Hospital, Fudan University, Shanghai, China.,Department of Systems Biology for Medicine and School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Juying Qian
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huali Shen
- Institutes of Biomedical Sciences of Shanghai Medical School and Minhang Hospital, Fudan University, Shanghai, China.,Department of Systems Biology for Medicine and School of Basic Medical Sciences, Fudan University, Shanghai, China.,Department of chemistry, Fudan University, Shanghai, China
| | - Pengyuan Yang
- Institutes of Biomedical Sciences of Shanghai Medical School and Minhang Hospital, Fudan University, Shanghai, China.,Department of Systems Biology for Medicine and School of Basic Medical Sciences, Fudan University, Shanghai, China.,Department of chemistry, Fudan University, Shanghai, China
| |
Collapse
|
28
|
Imbalzano E, Mandraffino G, Casciaro M, Quartuccio S, Saitta A, Gangemi S. Pathophysiological mechanism and therapeutic role of S100 proteins in cardiac failure: a systematic review. Heart Fail Rev 2018; 21:463-73. [PMID: 26833319 DOI: 10.1007/s10741-016-9529-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
S100 proteins are a family of highly acidic calcium-binding proteins involved in calcium handling in many tissues and organs. Some of these proteins are highly expressed in cardiac tissue, and an impairment of some specific S100 proteins has been related to heart failure. To check this hypothesis, we decided to review the literature since 2008 until May 2015. According to the studies collected, recovering S100A1 levels may enhance contractile/relaxing performance in heart failure, reverse negative force-frequency relationship, improve contractile reserve, reverse diastolic dysfunction and protect against pro-arrhythmic reductions of sarcoplasmic reticulum calcium. The safety profile of gene therapy was also confirmed. Increased S100B protein levels were related to a worse outcome in chronic heart failure. S100A8/A9 complex plasma levels, as well as other inflammatory biomarkers, were significantly higher in chronic heart failure patients. S100A2 seems to increase both contractile and relaxation performance in animal cardiomyocytes. Otherwise, S100A6 cardiac expression seems to have no effects on contractility. S100A4 KO mice showed reduced cardiac interstitial fibrosis. Data collected encourage a potential prospective application in human. These proteins could be exploited as biomarkers in stadiation and prognosis of chronic heart failure, as well as therapeutic target to rescue failing heart. Registration details The study protocol has been registered in PROSPERO ( http://www.crd.york.ac.uk/PROSPERO/ ) under registration number CRD42015027932.
Collapse
Affiliation(s)
- Egidio Imbalzano
- Department of Clinical and Experimental Medicine, Policlinic University of Messina, Via Consolare Valeria n.1, 98125, Messina, Italy.
| | - Giuseppe Mandraffino
- Department of Clinical and Experimental Medicine, Policlinic University of Messina, Via Consolare Valeria n.1, 98125, Messina, Italy
| | - Marco Casciaro
- School and Division of Allergy and Clinical Immunology, University of Messina, Messina, Italy
| | - Sebastiano Quartuccio
- Department of Clinical and Experimental Medicine, Policlinic University of Messina, Via Consolare Valeria n.1, 98125, Messina, Italy
| | - Antonino Saitta
- Department of Clinical and Experimental Medicine, Policlinic University of Messina, Via Consolare Valeria n.1, 98125, Messina, Italy
| | - Sebastiano Gangemi
- School and Division of Allergy and Clinical Immunology, University of Messina, Messina, Italy.,Institute of Applied Sciences and Intelligent Systems (ISASI) - Messina Unit, Messina, Italy
| |
Collapse
|
29
|
Abstract
The occlusion of a coronary artery by a thrombus generated on a ruptured atherosclerotic plaque has been pursued in the last decades as a determining event for the clinical outcome after myocardial infarction (MI). Yet, MI causes a cell death wave front, which triggers an inflammatory response to clear cellular debris, and which in excess can double the myocardial lesion and influence the clinical prognosis in the short and long term. Accordingly, proper, timely regulated inflammatory response has now been considered a second pivotal player in cardiac recovery after MI justifying the search for pharmacological strategies to modulate inflammatory effectors. This chapter reviews the key events and the main effectors of inflammation after myocardial ischemic insult, as well as the contribution of this phenomenon to the progression of atherosclerosis.
Collapse
Affiliation(s)
- Joaquim B Oliveira
- Laboratory of Atherosclerosis and Vascular Biology, State University of Campinas, Campinas, Brazil
| | - Alexandre A S M Soares
- Laboratory of Atherosclerosis and Vascular Biology, State University of Campinas, Campinas, Brazil
| | - Andrei C Sposito
- Laboratory of Atherosclerosis and Vascular Biology, State University of Campinas, Campinas, Brazil.
| |
Collapse
|
30
|
Jungi S, Fu X, Segiser A, Busch M, Most P, Fiedler M, Carrel T, Tevaearai Stahel H, Longnus SL, Most H. Enhanced Cardiac S100A1 Expression Improves Recovery from Global Ischemia-Reperfusion Injury. J Cardiovasc Transl Res 2018; 11:236-245. [PMID: 29392537 DOI: 10.1007/s12265-018-9788-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 01/08/2018] [Indexed: 12/14/2022]
Abstract
Gene-targeted therapy with the inotropic Ca2 + -sensor protein S100A1 rescues contractile function in post-ischemic heart failure and is being developed towards clinical trials. Its proven beneficial effect on cardiac metabolism and mitochondrial function suggests a cardioprotective effect of S100A1 in myocardial ischemia-reperfusion injury (IRI). Fivefold cardiomyocyte-specific S100A1 overexpressing, isolated rat hearts perfused in working mode were subjected to 28 min ischemia (37 °C) followed by 60 min reperfusion. S100A1 overexpressing hearts showed superior hemodynamic recover: Left ventricular pressure recovered to 57 ± 7.3% of baseline compared to 51 ± 4.6% in control (p = 0.025), this effect mirrored in LV work and dP/dt(max). Troponin T and lactate dehydrogenase was decreased in the S100A1 group, as well as FoxO pro-apoptotic transcription factor, indicating less tissue necrosis, whereas phosphocreatine content was higher after reperfusion. This is the first report of a cardioprotective effect of S100A1 overexpression in a global IRI model.
Collapse
Affiliation(s)
- S Jungi
- Department of Cardiovascular Surgery, Inselspital University Hospital, University of Bern, 3010, Bern, Switzerland
| | - X Fu
- Department of Cardiovascular Surgery, Inselspital University Hospital, University of Bern, 3010, Bern, Switzerland
| | - A Segiser
- Department of Cardiovascular Surgery, Inselspital University Hospital, University of Bern, 3010, Bern, Switzerland
| | - M Busch
- Section for Molecular and Translational Cardiology, Department of Cardiology, Pneumology and Angiology, Karl-Ruprechts University of Heidelberg, Heidelberg, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - P Most
- Section for Molecular and Translational Cardiology, Department of Cardiology, Pneumology and Angiology, Karl-Ruprechts University of Heidelberg, Heidelberg, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - M Fiedler
- Center for Laboratory Medicine, Inselspital University Hospital, University of Bern, Bern, Switzerland
| | - T Carrel
- Department of Cardiovascular Surgery, Inselspital University Hospital, University of Bern, 3010, Bern, Switzerland
| | - H Tevaearai Stahel
- Department of Cardiovascular Surgery, Inselspital University Hospital, University of Bern, 3010, Bern, Switzerland
| | - S L Longnus
- Department of Cardiovascular Surgery, Inselspital University Hospital, University of Bern, 3010, Bern, Switzerland
| | - Henriette Most
- Department of Cardiovascular Surgery, Inselspital University Hospital, University of Bern, 3010, Bern, Switzerland.
| |
Collapse
|
31
|
Gouveia A, Bajwa E, Klegeris A. Extracellular cytochrome c as an intercellular signaling molecule regulating microglial functions. Biochim Biophys Acta Gen Subj 2017; 1861:2274-2281. [DOI: 10.1016/j.bbagen.2017.06.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 06/05/2017] [Accepted: 06/22/2017] [Indexed: 01/13/2023]
|
32
|
Ebrahimi E, Kheirouri S, Alizadeh M. Down-regulation of S100A1 protein in patients with metabolic syndrome and its association with zinc-α2-glycoprotein. Scott Med J 2017; 62:88-95. [PMID: 28825380 DOI: 10.1177/0036933017727433] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Objectives It has been proposed that zinc-α2-glycoprotein and S100A1 are possibly linked to the development of lipogenesis and obesity. We aimed to measure serum levels of S100A1 and zinc-α2-glycoprotein in patients with metabolic syndrome and investigate any associations of these two novel peptides with each other or components of metabolic syndrome. Methods Forty-four patients with metabolic syndrome and the equivalent number of healthy controls participated in this study. The participants' body mass index, waist circumference, systolic and diastolic blood pressure were measured. Serum levels of low- and high-density lipoprotein cholesterol, total cholesterol, triglyceride, fasting blood sugar, insulin, zinc-α2-glycoprotein and S100A1 protein were determined. Results Higher levels of anthropometric and lipid indices, metabolic factors and also SBP and DBP were observed in the metabolic syndrome group. Serum S100A1 levels were significantly lower in the metabolic syndrome group than the control group ( P = 0.008). There was a strong positive correlation between serum zinc-α2-glycoprotein and S100A1 levels ( r = 0.80, P < 0.0001). Serum levels of both S100A1 ( P = 0.03) and zinc-α2-glycoprotein ( P = 0.02) were potentially higher in subjects with hypertension than those with normal blood pressure, though these were found as part of multiple testing. Conclusion The results indicate that changes in the circulating level of S100A1 protein occur in metabolic syndrome patients. The strong correlation between serum zinc-α2-glycoprotein and S100A1 might suggest that production or release of these two proteins could be related mechanistically.
Collapse
Affiliation(s)
- Elham Ebrahimi
- 1 Student, Department of Nutrition, Tabriz University of Medical Sciences, Iran
| | - Sorayya Kheirouri
- 2 Associate Professor, Department of Nutrition, Tabriz University of Medical Sciences, Iran
| | - Mohammad Alizadeh
- 2 Associate Professor, Department of Nutrition, Tabriz University of Medical Sciences, Iran
| |
Collapse
|
33
|
Bera A, Sen D. Promise of adeno-associated virus as a gene therapy vector for cardiovascular diseases. Heart Fail Rev 2017; 22:795-823. [DOI: 10.1007/s10741-017-9622-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
34
|
Knezevic T, Myers VD, Su F, Wang J, Song J, Zhang XQ, Gao E, Gao G, Muniswamy M, Gupta MK, Gordon J, Weiner KN, Rabinowitz J, Ramsey FV, Tilley DG, Khalili K, Cheung JY, Feldman AM. Adeno-associated Virus Serotype 9 - Driven Expression of BAG3 Improves Left Ventricular Function in Murine Hearts with Left Ventricular Dysfunction Secondary to a Myocardial Infarction. ACTA ACUST UNITED AC 2016; 1:647-656. [PMID: 28164169 PMCID: PMC5289821 DOI: 10.1016/j.jacbts.2016.08.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BAG3 is a highly conserved protein having pleiotropic effects that is expressed at high levels in the heart, skeletal muscles, and many cancers. BAG3 levels are reduced in many forms of LV dysfunction including mice after ligation of the left coronary artery. Retro-orbital injection of mice with an adeno-associated virus coupled to murine BAG3 under the control of a CMV promoter (rAAV9-BAG3) increased myocardial levels of BAG3 by 7 days post-injection. Retro-orbital injection of rAAV9-BAG3 in mice post-myocardial infarction improved LV function, whereas rAAV9-BAG3 had no effect on LV function in the absence of an MI. BAG3 may prove to be a new therapeutic target in the treatment of heart failure.
Mutations in Bcl-2–associated athanogene 3 (BAG3) were associated with skeletal muscle dysfunction and dilated cardiomyopathy. Retro-orbital injection of an adeno-associated virus serotype 9 expressing BAG3 (rAAV9-BAG3) significantly (p < 0.0001) improved left ventricular ejection fraction, fractional shortening, and stroke volume 9 days post-injection in mice with cardiac dysfunction secondary to a myocardial infarction. Furthermore, myocytes isolated from mice 3 weeks after injection showed improved cell shortening, enhanced systolic [Ca2+]i and increased [Ca2+]i transient amplitudes, and increased maximal L-type Ca2+ current amplitude. These results suggest that BAG3 gene therapy may provide a novel therapeutic option for the treatment of heart failure.
Collapse
Affiliation(s)
- Tijana Knezevic
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennslyvnaia; Department of Neuroscience, the Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Valerie D Myers
- Department of Medicine, the Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Feifei Su
- Department of Medicine, the Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania; Department of Cardiology, TangDu Hospital, Fourth Military Medical University, Xi'an, China
| | - JuFang Wang
- Center for Translational Medicine, the Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Jianliang Song
- Center for Translational Medicine, the Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Xue-Qian Zhang
- Center for Translational Medicine, the Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Erhe Gao
- Center for Translational Medicine, the Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Guofeng Gao
- Department of Medicine, the Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Madesh Muniswamy
- Center for Translational Medicine, the Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Manish K Gupta
- Department of Neuroscience, the Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Jennifer Gordon
- Department of Neuroscience, the Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Kristen N Weiner
- Department of Medicine, the Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Joseph Rabinowitz
- Center for Translational Medicine, the Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Frederick V Ramsey
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennslyvnaia
| | - Douglas G Tilley
- Center for Translational Medicine, the Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Kamel Khalili
- Department of Neuroscience, the Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Joseph Y Cheung
- Department of Medicine, the Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania; Center for Translational Medicine, the Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Arthur M Feldman
- Department of Medicine, the Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
35
|
Immunohistochemical detection of early myocardial infarction: a systematic review. Int J Legal Med 2016; 131:411-421. [PMID: 27885432 DOI: 10.1007/s00414-016-1494-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 11/08/2016] [Indexed: 12/11/2022]
Abstract
The postmortem diagnosis of early myocardial infarction is a challenge for forensic pathologists because the routine histology is neither specific. Many authors have suggested the use of the immunohistochemistry to fill the gaps in the histological diagnosis of early myocardial infarction. This review aims to analyse advances of immunohistochemical detection of early cardiac damage due to ischaemia. To this purpose, we reviewed experimental studies that investigated immunohistochemical markers and their estimated timing of expression. The review was performed according to specific inclusion and exclusion criteria, and a total of 23 studies assessing the immunohistochemical markers for the diagnosis and timing of early myocardial infarction were identified. The literature review highlights that the analysed markers are complement components, others being inflammatory mediators, cardiac cell proteins, plasma proteins, stress or hypoxia-induced factors and proteins associated with heart failure. All studies demonstrate the effectiveness of the tested markers in the early detection of myocardial infarction in both animal and human samples.
Collapse
|
36
|
Guo Y, Cui L, Jiang S, Wang D, Jiang S, Xie C, Jia Y. S100A1 transgenic treatment of acute heart failure causes proteomic changes in rats. Mol Med Rep 2016; 14:1538-52. [PMID: 27357314 PMCID: PMC4940056 DOI: 10.3892/mmr.2016.5440] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 03/23/2016] [Indexed: 12/11/2022] Open
Abstract
S100 Ca2+-binding protein A1 (S100A1) is an important regulator of myocardial contractility. The aim of the present study was to identify the underlying mechanisms of S100A1 activity via profiling the protein expression in rats administered with an S100A1 adenovirus (Ad-S100A1-EGFP) following acute myocardial infarction (AMI). LTQ OrbiTrap mass spectrometry was used to profile the protein expression in the Ad-S100A1-EGFP and control groups post-AMI. Using Protein Analysis Through Evolutionary Relationships (PANTHER) analysis, 134 energy metabolism-associated proteins, which comprised 20 carbohydrate metabolism-associated and 27 lipid metabolism associated proteins, were identified as differentially expressed in the Ad-S100A1-EGFP hearts compared with controls. The majority of the differentially expressed proteins identified were important enzymes involved in energy metabolism. The present study identified 12 Ca2+-binding proteins and 22 cytoskeletal proteins. The majority of the proteins expressed in the Ad-S100A1-EGFP group were upregulated compared with the control group. These results were further validated using western blot analysis. Following AMI, Ca2+ is crucial for the recovery of myocardial function in S100A1 transgenic rats as indicated by the upregulation of proteins associated with energy metabolism and Ca2+-binding. Thus, the current study ascertained that energy production and contractile ability were enhanced after AMI in the ventricular myocardium of the Ad-S100A1-EGFP group.
Collapse
Affiliation(s)
- Yichen Guo
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Lianqun Cui
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Shiliang Jiang
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Dongmei Wang
- Department of Radiology, Shandong Jiao Tong Hospital, Jinan, Shandong 250063, P.R. China
| | - Shu Jiang
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Chen Xie
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Yanping Jia
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
37
|
Rebbeck RT, Nitu FR, Rohde D, Most P, Bers DM, Thomas DD, Cornea RL. S100A1 Protein Does Not Compete with Calmodulin for Ryanodine Receptor Binding but Structurally Alters the Ryanodine Receptor·Calmodulin Complex. J Biol Chem 2016; 291:15896-907. [PMID: 27226555 DOI: 10.1074/jbc.m115.713107] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Indexed: 11/06/2022] Open
Abstract
S100A1 has been suggested as a therapeutic agent to enhance myocyte Ca(2+) cycling in heart failure, but its molecular mode of action is poorly understood. Using FRET, we tested the hypothesis that S100A1 directly competes with calmodulin (CaM) for binding to intact, functional ryanodine receptors type I (RyR1) and II (RyR2) from skeletal and cardiac muscle, respectively. Our FRET readout provides an index of acceptor-labeled CaM binding near donor-labeled FKBP (FK506-binding protein 12.6) on the cytoplasmic domain of RyR in isolated sarcoplasmic reticulum vesicles. S100A1 (0.01-400 μm) partially inhibited FRET (i.e. CaM binding), with Ki > 10 μm, for both RyR1 and RyR2. The high [S100A1] required for partial effects on FRET indicates a lack of competition by S100A1 on CaM/RyR binding under normal physiological conditions. High-resolution analysis of time-resolved FRET detects two structural states of RyR-bound CaM, which respond to [Ca(2+)] and are isoform-specific. The distribution of these structural states was perturbed only by high micromolar [S100A1], which promoted a shift of bound CaM to a lower FRET orientation (without altering the amount of CaM bound to RyR). Thus, high micromolar S100A1 does alter the CaM/RyR interaction, without involving competition. Nevertheless, submicromolar S100A1 can alter RyR function, an effect that is influenced by both [Ca(2+)] and [CaM]. We conclude that CaM and S100A1 can concurrently bind to and functionally modulate RyR1 and RyR2, but this does not involve direct competition at the RyR CaM binding site.
Collapse
Affiliation(s)
- Robyn T Rebbeck
- From the Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis Minnesota 55455
| | - Florentin R Nitu
- From the Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis Minnesota 55455
| | - David Rohde
- the Center for Molecular and Translational Cardiology, Department of Internal Medicine III, University of Heidelberg, INF 410, 69120, Heidelberg, Germany, and
| | - Patrick Most
- the Center for Molecular and Translational Cardiology, Department of Internal Medicine III, University of Heidelberg, INF 410, 69120, Heidelberg, Germany, and
| | - Donald M Bers
- the Department of Pharmacology, University of California, Davis, California 95616
| | - David D Thomas
- From the Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis Minnesota 55455
| | - Razvan L Cornea
- From the Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis Minnesota 55455,
| |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW The use of adeno-associated virus (AAV) as an efficient, cardiotropic, and safe vector, coupled with the identification of key molecular targets, has placed gene-based therapies within reach of cardiovascular diseases. The purpose of this review is to provide a focused update on the current advances related to AAV-mediated gene therapy in cardiovascular diseases, and particularly in heart failure (HF), wherein gene therapy has recently made important progress. RECENT FINDINGS Multiple successful preclinical studies suggest a potential utility of AAV gene therapy for arrhythmias and biological heart pacing, as well as RNA overexpression. Moreover, AAV-mediated overexpression of several molecular targets involved in HF has demonstrated promising results in clinically relevant large animal models. In humans, a safe and successful completion of a phase 2 clinical trial targeting the sarcoplasmic reticulum calcium ATPase pump with AAV has been reported. Serial studies are ongoing to further prove the efficacy of AAV-mediated sarcoplasmic reticulum calcium ATPase pump gene transfer in human HF. SUMMARY Significant progress in clinical translation of AAV-mediated cardiac gene therapy has been achieved in recent years. This will prompt further clinical trials, and positive results could open a new era for cardiac gene therapy.
Collapse
|
39
|
Eryilmaz U, Demirci B, Aksun S, Boyacioglu M, Akgullu C, Ilgenli TF, Yalinkilinc HS, Bilgen M. S100A1 as a Potential Diagnostic Biomarker for Assessing Cardiotoxicity and Implications for the Chemotherapy of Certain Cancers. PLoS One 2015; 10:e0145418. [PMID: 26682543 PMCID: PMC4687715 DOI: 10.1371/journal.pone.0145418] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 12/03/2015] [Indexed: 01/02/2023] Open
Abstract
This study examined the value of blood marker S100A1 in detecting cardiotoxicity induced by chemotherapy agents; trastuzumab and lapatinib, in normal rat heart. The rats were divided into three groups: control (n = 8, no treatment), T (n = 8, one time ip treatment with 10 mg/kg trastuzumab) and L (n = 8, oral treatment with 100 mg/kg/day lapatinib for 7 days). The activities of oxidative stress parameters Malondialdehyde (MDA), Superoxide dismutase (SOD), Catalase (CAT) and Glutathione (GSH) were measured from the extracted cardiac tissues. The levels of troponinI and S100A1 expressions were measured from blood samples. All biomarkers responded to the treatments as they exhibited alterations from their normative values, validating the chemically induced cardiotoxicity. S100A1 expression attenuated significantly (75%), which made the sensitive detection of cardiotoxicity feasible. Assessment of cardiotoxicity with S100A1 may be a valuable alternative in clinical oncology of cancers in some organs such as breast and prostate, as they do not overexpress it to compete against.
Collapse
Affiliation(s)
- Ufuk Eryilmaz
- Department of Cardiology, Medical Faculty, Adnan Menderes University, Aydin, Turkey
- * E-mail:
| | - Buket Demirci
- Department of Medical Pharmacology, Medical Faculty, Adnan Menderes University, Aydin, Turkey
| | - Saliha Aksun
- Department of Medical Biochemistry, Medical Faculty, Katip Celebi University, Izmir, Turkey
| | - Murat Boyacioglu
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Adnan Menderes University, Aydin, Turkey
| | - Cagdas Akgullu
- Department of Cardiology, Medical Faculty, Adnan Menderes University, Aydin, Turkey
| | | | - Hande Sultan Yalinkilinc
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Adnan Menderes University, Aydin, Turkey
| | - Mehmet Bilgen
- Department of Biophysics, Medical Faculty, Adnan Menderes University, Aydin, Turkey
| |
Collapse
|
40
|
Adipokines and their receptors: potential new targets in cardiovascular diseases. Future Med Chem 2015; 7:139-57. [PMID: 25686003 DOI: 10.4155/fmc.14.147] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue is an 'endocrine organ' that influences diverse physiological and pathological processes via adipokines secretion. Strong evidences suggest that epicardial and perivascular adipose tissue can directly regulate heart and vessels' structure and function. Indeed, in obesity there is a shift toward the secretion of adipokines that promote a pro-inflammatory status and contribute to obesity cardiomyopathy. The prospect of modulating adipokines and/or their receptors represents an attractive perspective to the treatment of cardiovascular diseases. In this paper, we described the most important actions of certain adipokines and their receptors that are capable of influencing cardiovascular physiology as well as their possible use as therapeutic targets.
Collapse
|
41
|
Fargnoli AS, Katz MG, Williams RD, Kendle AP, Steuerwald N, Bridges CR. Liquid jet delivery method featuring S100A1 gene therapy in the rodent model following acute myocardial infarction. Gene Ther 2015; 23:151-7. [PMID: 26461176 PMCID: PMC4742412 DOI: 10.1038/gt.2015.100] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/12/2015] [Accepted: 09/17/2015] [Indexed: 01/16/2023]
Abstract
The S100A1 gene is a promising target enhancing contractility and survival post myocardial infarction (MI). Achieving sufficient gene delivery within safety limits is a major translational problem. This proof of concept study evaluates viral-mediated S100A1 overexpression featuring a novel liquid jet delivery (LJ) method. 24 rats after successful MI were divided into 3 groups (n=8 ea.): saline control (SA), ssAAV9.S100A1 (SS) delivery, and scAAV9.S100A1 (SC) delivery (both 1.2×1011 viral particles). For each post MI rat, the LJ device fired three separate 100 μL injections into the myocardium. Following 10 weeks, all rats were evaluated with echocardiography, quantitative polymerase chain reaction (qPCR), and overall S100A1 and CD38 immune protein. At 10 weeks all groups demonstrated a functional decline from baseline, but the S100A1 therapy groups displayed preserved LV function with significantly higher ejection fraction %; SS group [60±3] and SC group [57±4] versus saline [46±3], p<0.05. Heart qPCR testing showed robust S100A1 in the SS [10,147±3993] and SC [35,155±5808] copies per 100 ng DNA, while off target liver detection was lower in both SS [40±40], SC [34,841±3164] respectively. Cardiac S100A1 protein expression was [4.3±0.2] and [6.1±0.3] fold higher than controls in the SS and SC groups respectively, p<0.05.
Collapse
Affiliation(s)
- A S Fargnoli
- Department of Thoracic and Cardiac Surgery, Sanger Heart and Vascular Institute, Carolinas HealthCare System, Charlotte, NC, USA
| | - M G Katz
- Department of Thoracic and Cardiac Surgery, Sanger Heart and Vascular Institute, Carolinas HealthCare System, Charlotte, NC, USA
| | - R D Williams
- Department of Thoracic and Cardiac Surgery, Sanger Heart and Vascular Institute, Carolinas HealthCare System, Charlotte, NC, USA
| | - A P Kendle
- Department of Thoracic and Cardiac Surgery, Sanger Heart and Vascular Institute, Carolinas HealthCare System, Charlotte, NC, USA
| | - N Steuerwald
- Molecular Biology Core, Department of Therapeutic Research and Development, Cannon Research Center, Carolinas HealthCare System, Charlotte, NC, USA
| | - C R Bridges
- Department of Thoracic and Cardiac Surgery, Sanger Heart and Vascular Institute, Carolinas HealthCare System, Charlotte, NC, USA
| |
Collapse
|
42
|
van Hout GPJ, Arslan F, Pasterkamp G, Hoefer IE. Targeting danger-associated molecular patterns after myocardial infarction. Expert Opin Ther Targets 2015; 20:223-39. [DOI: 10.1517/14728222.2016.1088005] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
43
|
Rincon MY, VandenDriessche T, Chuah MK. Gene therapy for cardiovascular disease: advances in vector development, targeting, and delivery for clinical translation. Cardiovasc Res 2015; 108:4-20. [PMID: 26239654 PMCID: PMC4571836 DOI: 10.1093/cvr/cvv205] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 07/22/2015] [Indexed: 01/06/2023] Open
Abstract
Gene therapy is a promising modality for the treatment of inherited and acquired cardiovascular diseases. The identification of the molecular pathways involved in the pathophysiology of heart failure and other associated cardiac diseases led to encouraging preclinical gene therapy studies in small and large animal models. However, the initial clinical results yielded only modest or no improvement in clinical endpoints. The presence of neutralizing antibodies and cellular immune responses directed against the viral vector and/or the gene-modified cells, the insufficient gene expression levels, and the limited gene transduction efficiencies accounted for the overall limited clinical improvements. Nevertheless, further improvements of the gene delivery technology and a better understanding of the underlying biology fostered renewed interest in gene therapy for heart failure. In particular, improved vectors based on emerging cardiotropic serotypes of the adeno-associated viral vector (AAV) are particularly well suited to coax expression of therapeutic genes in the heart. This led to new clinical trials based on the delivery of the sarcoplasmic reticulum Ca2+-ATPase protein (SERCA2a). Though the first clinical results were encouraging, a recent Phase IIb trial did not confirm the beneficial clinical outcomes that were initially reported. New approaches based on S100A1 and adenylate cyclase 6 are also being considered for clinical applications. Emerging paradigms based on the use of miRNA regulation or CRISPR/Cas9-based genome engineering open new therapeutic perspectives for treating cardiovascular diseases by gene therapy. Nevertheless, the continuous improvement of cardiac gene delivery is needed to allow the use of safer and more effective vector doses, ultimately bringing gene therapy for heart failure one step closer to reality.
Collapse
Affiliation(s)
- Melvin Y Rincon
- Department of Gene Therapy and Regenerative Medicine, Free University of Brussels (VUB), Building D, room D306, Laarbeeklaan 103, Brussels, Belgium Centro de Investigaciones, Fundacion Cardiovascular de Colombia, Floridablanca, Colombia
| | - Thierry VandenDriessche
- Department of Gene Therapy and Regenerative Medicine, Free University of Brussels (VUB), Building D, room D306, Laarbeeklaan 103, Brussels, Belgium Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Marinee K Chuah
- Department of Gene Therapy and Regenerative Medicine, Free University of Brussels (VUB), Building D, room D306, Laarbeeklaan 103, Brussels, Belgium Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| |
Collapse
|
44
|
Maxwell JT, Somasuntharam I, Gray WD, Shen M, Singer JM, Wang B, Saafir T, Crawford BH, Jiang R, Murthy N, Davis ME, Wagner MB. Bioactive nanoparticles improve calcium handling in failing cardiac myocytes. Nanomedicine (Lond) 2015. [PMID: 26223412 DOI: 10.2217/nnm.15.126] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIMS To evaluate the ability of N-acetylglucosamine (GlcNAc) decorated nanoparticles and their cargo to modulate calcium handling in failing cardiac myocytes (CMs). MATERIALS & METHODS Primary CMs isolated from normal and failing hearts were treated with GlcNAc nanoparticles in order to assess the ability of the nanoparticles and their cargo to correct dysfunctional calcium handling in failing myocytes. RESULTS & CONCLUSION GlcNAc particles reduced aberrant calcium release in failing CMs and restored sarcomere function. Additionally, encapsulation of a small calcium-modulating protein, S100A1, in GlcNAc nanoparticles also showed improved calcium regulation. Thus, the development of our bioactive nanoparticle allows for a 'two-hit' treatment, by which the cargo and also the nanoparticle itself can modulate intracellular protein activity.
Collapse
Affiliation(s)
- Joshua T Maxwell
- Wallace H Coulter Department of Biomedical Engineering, Emory University School of Medicine, 1648 Pierce Dr NE, Atlanta, GA 30307, USA.,Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, 1648 Pierce Dr NE, Atlanta, GA 30307, USA.,Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA, USA
| | - Inthirai Somasuntharam
- Wallace H Coulter Department of Biomedical Engineering, Emory University School of Medicine, 1648 Pierce Dr NE, Atlanta, GA 30307, USA
| | - Warren D Gray
- Wallace H Coulter Department of Biomedical Engineering, Emory University School of Medicine, 1648 Pierce Dr NE, Atlanta, GA 30307, USA
| | - Ming Shen
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, 1648 Pierce Dr NE, Atlanta, GA 30307, USA.,Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA, USA
| | - Jason M Singer
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, 1648 Pierce Dr NE, Atlanta, GA 30307, USA.,Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA, USA
| | - Bo Wang
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, 1648 Pierce Dr NE, Atlanta, GA 30307, USA.,Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA, USA
| | - Talib Saafir
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, 1648 Pierce Dr NE, Atlanta, GA 30307, USA.,Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA, USA
| | - Brian H Crawford
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, 1648 Pierce Dr NE, Atlanta, GA 30307, USA.,Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA, USA
| | - Rong Jiang
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, 1648 Pierce Dr NE, Atlanta, GA 30307, USA.,Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA, USA
| | - Niren Murthy
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, USA
| | - Michael E Davis
- Wallace H Coulter Department of Biomedical Engineering, Emory University School of Medicine, 1648 Pierce Dr NE, Atlanta, GA 30307, USA.,Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, 1648 Pierce Dr NE, Atlanta, GA 30307, USA.,Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA, USA
| | - Mary B Wagner
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, 1648 Pierce Dr NE, Atlanta, GA 30307, USA.,Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA, USA
| |
Collapse
|
45
|
S100A1 DNA-based Inotropic Therapy Protects Against Proarrhythmogenic Ryanodine Receptor 2 Dysfunction. Mol Ther 2015; 23:1320-1330. [PMID: 26005840 DOI: 10.1038/mt.2015.93] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 04/28/2015] [Indexed: 12/21/2022] Open
Abstract
Restoring expression levels of the EF-hand calcium (Ca(2+)) sensor protein S100A1 has emerged as a key factor in reconstituting normal Ca(2+) handling in failing myocardium. Improved sarcoplasmic reticulum (SR) function with enhanced Ca(2+) resequestration appears critical for S100A1's cyclic adenosine monophosphate-independent inotropic effects but raises concerns about potential diastolic SR Ca(2+) leakage that might trigger fatal arrhythmias. This study shows for the first time a diminished interaction between S100A1 and ryanodine receptors (RyR2s) in experimental HF. Restoring this link in failing cardiomyocytes, engineered heart tissue and mouse hearts, respectively, by means of adenoviral and adeno-associated viral S100A1 cDNA delivery normalizes diastolic RyR2 function and protects against Ca(2+)- and β-adrenergic receptor-triggered proarrhythmogenic SR Ca(2+) leakage in vitro and in vivo. S100A1 inhibits diastolic SR Ca(2+) leakage despite aberrant RyR2 phosphorylation via protein kinase A and calmodulin-dependent kinase II and stoichiometry with accessory modulators such as calmodulin, FKBP12.6 or sorcin. Our findings demonstrate that S100A1 is a regulator of diastolic RyR2 activity and beneficially modulates diastolic RyR2 dysfunction. S100A1 interaction with the RyR2 is sufficient to protect against basal and catecholamine-triggered arrhythmic SR Ca(2+) leak in HF, combining antiarrhythmic potency with chronic inotropic actions.
Collapse
|
46
|
Rohde D, Busch M, Volkert A, Ritterhoff J, Katus HA, Peppel K, Most P. Cardiomyocytes, endothelial cells and cardiac fibroblasts: S100A1's triple action in cardiovascular pathophysiology. Future Cardiol 2015; 11:309-21. [PMID: 26021637 PMCID: PMC11544369 DOI: 10.2217/fca.15.18] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Over the past decade, basic and translational research delivered comprehensive evidence for the relevance of the Ca(2+)-binding protein S100A1 in cardiovascular diseases. Aberrant expression levels of S100A1 surfaced as molecular key defects, driving the pathogenesis of chronic heart failure, arterial and pulmonary hypertension, peripheral artery disease and disturbed myocardial infarction healing. Loss of intracellular S100A1 renders entire Ca(2+)-controlled networks dysfunctional, thereby leading to cardiomyocyte failure and endothelial dysfunction. Lack of S100A1 release in ischemic myocardium compromises cardiac fibroblast function, entailing impaired damage healing. This review focuses on molecular pathways and signaling cascades regulated by S100A1 in cardiomyocytes, endothelial cells and cardiac fibroblasts in order to provide an overview of our current mechanistic understanding of S100A1's action in cardiovascular pathophysiology.
Collapse
Affiliation(s)
- David Rohde
- Molecular & Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg University, INF 410, 69120 Heidelberg, Germany
| | - Martin Busch
- Molecular & Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg University, INF 410, 69120 Heidelberg, Germany
| | - Anne Volkert
- Molecular & Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg University, INF 410, 69120 Heidelberg, Germany
| | - Julia Ritterhoff
- Molecular & Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg University, INF 410, 69120 Heidelberg, Germany
| | - Hugo A. Katus
- Molecular & Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg University, INF 410, 69120 Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Heidelberg University Hospital, Heidelberg University, INF 410, 69120 Heidelberg, Germany
| | - Karsten Peppel
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
- uniQure GmbH, INF 410, 69120 Heidelberg, Germany
| | - Patrick Most
- Molecular & Translational Cardiology, Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg University, INF 410, 69120 Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Heidelberg University Hospital, Heidelberg University, INF 410, 69120 Heidelberg, Germany
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
- uniQure GmbH, INF 410, 69120 Heidelberg, Germany
| |
Collapse
|
47
|
Liu Z, Ye P, Wang S, Wu J, Sun Y, Zhang A, Ren L, Cheng C, Huang X, Wang K, Deng P, Wu C, Yue Z, Xia J. MicroRNA-150 Protects the Heart From Injury by Inhibiting Monocyte Accumulation in a Mouse Model of Acute Myocardial Infarction. ACTA ACUST UNITED AC 2015; 8:11-20. [PMID: 25466411 DOI: 10.1161/circgenetics.114.000598] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Background—
MicroRNAs (miRs) and inflammatory monocytes participate in many cardiac pathophysiological processes including acute myocardial infarction (AMI). Recently, we observed that miR-150 is downregulated in injured mouse plasma after AMI as well as in human infarcted monocytes. However, the precise functional role of miR-150 in response to AMI remains unknown.
Methods and Results—
In a mouse model of AMI and in human subjects with AMI, we found that miR-150 expression was reduced in monocytes. In vitro studies showed that ectopic expression of miR-150 markedly reduced monocyte migration and proinflammatory cytokine production, whereas blockade of miR-150 had opposing effects. In vivo studies showed that overexpression of miR-150 in mice resulted in improved cardiac function, reduced myocardial infarction size, inhibition of apoptosis, and reduced inflammatory Ly-6C
high
monocyte invasion levels after AMI. Wild-type mice transplanted with miR-150 null (−/−) bone marrow cells could reverse this protective effect. Mechanistic studies demonstrated that miR-150 inhibited the expression of chemokine receptor 4 (CXCR4), thereby promoting monocyte migration.
Conclusions—
Our findings indicate that miR-150 acts as a critical regulator of monocyte cell migration and production of proinflammatory cytokines, leading to cardioprotective effects against AMI-induced injury. Thus, miR-150 may be a suitable target for therapeutic intervention in the setting of ischemic heart disease.
Collapse
Affiliation(s)
- Zheng Liu
- From the Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China (Z.L.); Department of Cardiovascular Surgery (Z.L., J.W., Y.S., A.Z., L.R., C.C., X.H., K.W., P.D., C.W., Z.Y., J.X.) and Department of Thoracic Surgery (S.W.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Department of Cardiovascular Medicine (P.Y.) and Department of Cardiovascular Surgery (J.X.), Central Hospital of Wuhan, Wuhan, China
| | - Ping Ye
- From the Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China (Z.L.); Department of Cardiovascular Surgery (Z.L., J.W., Y.S., A.Z., L.R., C.C., X.H., K.W., P.D., C.W., Z.Y., J.X.) and Department of Thoracic Surgery (S.W.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Department of Cardiovascular Medicine (P.Y.) and Department of Cardiovascular Surgery (J.X.), Central Hospital of Wuhan, Wuhan, China
| | - Sihua Wang
- From the Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China (Z.L.); Department of Cardiovascular Surgery (Z.L., J.W., Y.S., A.Z., L.R., C.C., X.H., K.W., P.D., C.W., Z.Y., J.X.) and Department of Thoracic Surgery (S.W.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Department of Cardiovascular Medicine (P.Y.) and Department of Cardiovascular Surgery (J.X.), Central Hospital of Wuhan, Wuhan, China
| | - Jie Wu
- From the Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China (Z.L.); Department of Cardiovascular Surgery (Z.L., J.W., Y.S., A.Z., L.R., C.C., X.H., K.W., P.D., C.W., Z.Y., J.X.) and Department of Thoracic Surgery (S.W.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Department of Cardiovascular Medicine (P.Y.) and Department of Cardiovascular Surgery (J.X.), Central Hospital of Wuhan, Wuhan, China
| | - Yuan Sun
- From the Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China (Z.L.); Department of Cardiovascular Surgery (Z.L., J.W., Y.S., A.Z., L.R., C.C., X.H., K.W., P.D., C.W., Z.Y., J.X.) and Department of Thoracic Surgery (S.W.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Department of Cardiovascular Medicine (P.Y.) and Department of Cardiovascular Surgery (J.X.), Central Hospital of Wuhan, Wuhan, China
| | - Anchen Zhang
- From the Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China (Z.L.); Department of Cardiovascular Surgery (Z.L., J.W., Y.S., A.Z., L.R., C.C., X.H., K.W., P.D., C.W., Z.Y., J.X.) and Department of Thoracic Surgery (S.W.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Department of Cardiovascular Medicine (P.Y.) and Department of Cardiovascular Surgery (J.X.), Central Hospital of Wuhan, Wuhan, China
| | - Linyun Ren
- From the Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China (Z.L.); Department of Cardiovascular Surgery (Z.L., J.W., Y.S., A.Z., L.R., C.C., X.H., K.W., P.D., C.W., Z.Y., J.X.) and Department of Thoracic Surgery (S.W.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Department of Cardiovascular Medicine (P.Y.) and Department of Cardiovascular Surgery (J.X.), Central Hospital of Wuhan, Wuhan, China
| | - Chao Cheng
- From the Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China (Z.L.); Department of Cardiovascular Surgery (Z.L., J.W., Y.S., A.Z., L.R., C.C., X.H., K.W., P.D., C.W., Z.Y., J.X.) and Department of Thoracic Surgery (S.W.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Department of Cardiovascular Medicine (P.Y.) and Department of Cardiovascular Surgery (J.X.), Central Hospital of Wuhan, Wuhan, China
| | - Xiaofan Huang
- From the Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China (Z.L.); Department of Cardiovascular Surgery (Z.L., J.W., Y.S., A.Z., L.R., C.C., X.H., K.W., P.D., C.W., Z.Y., J.X.) and Department of Thoracic Surgery (S.W.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Department of Cardiovascular Medicine (P.Y.) and Department of Cardiovascular Surgery (J.X.), Central Hospital of Wuhan, Wuhan, China
| | - Ke Wang
- From the Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China (Z.L.); Department of Cardiovascular Surgery (Z.L., J.W., Y.S., A.Z., L.R., C.C., X.H., K.W., P.D., C.W., Z.Y., J.X.) and Department of Thoracic Surgery (S.W.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Department of Cardiovascular Medicine (P.Y.) and Department of Cardiovascular Surgery (J.X.), Central Hospital of Wuhan, Wuhan, China
| | - Peng Deng
- From the Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China (Z.L.); Department of Cardiovascular Surgery (Z.L., J.W., Y.S., A.Z., L.R., C.C., X.H., K.W., P.D., C.W., Z.Y., J.X.) and Department of Thoracic Surgery (S.W.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Department of Cardiovascular Medicine (P.Y.) and Department of Cardiovascular Surgery (J.X.), Central Hospital of Wuhan, Wuhan, China
| | - Chuangyan Wu
- From the Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China (Z.L.); Department of Cardiovascular Surgery (Z.L., J.W., Y.S., A.Z., L.R., C.C., X.H., K.W., P.D., C.W., Z.Y., J.X.) and Department of Thoracic Surgery (S.W.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Department of Cardiovascular Medicine (P.Y.) and Department of Cardiovascular Surgery (J.X.), Central Hospital of Wuhan, Wuhan, China
| | - Zhang Yue
- From the Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China (Z.L.); Department of Cardiovascular Surgery (Z.L., J.W., Y.S., A.Z., L.R., C.C., X.H., K.W., P.D., C.W., Z.Y., J.X.) and Department of Thoracic Surgery (S.W.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Department of Cardiovascular Medicine (P.Y.) and Department of Cardiovascular Surgery (J.X.), Central Hospital of Wuhan, Wuhan, China
| | - Jiahong Xia
- From the Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China (Z.L.); Department of Cardiovascular Surgery (Z.L., J.W., Y.S., A.Z., L.R., C.C., X.H., K.W., P.D., C.W., Z.Y., J.X.) and Department of Thoracic Surgery (S.W.), Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and Department of Cardiovascular Medicine (P.Y.) and Department of Cardiovascular Surgery (J.X.), Central Hospital of Wuhan, Wuhan, China
| |
Collapse
|
48
|
Teichert-Kuliszewska K, Tsoporis JN, Desjardins JF, Yin J, Wang L, Kuebler WM, Parker TG. Absence of the calcium-binding protein, S100A1, confers pulmonary hypertension in mice associated with endothelial dysfunction and apoptosis. Cardiovasc Res 2014; 105:8-19. [PMID: 25395393 DOI: 10.1093/cvr/cvu241] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AIMS S100A1, a 10-kDa, Ca(2+)-binding protein, is expressed in endothelial cells (ECs) and binds eNOS. Its absence is associated with impaired production of nitric oxide (NO) and mild systemic hypertension. As endothelial dysfunction contributes to clinical and experimental pulmonary hypertension (PH), we investigated the impact of deleting S100A1 in mice, on pulmonary haemodynamics, endothelial function, NO production, associated signalling pathways, and apoptosis. METHODS AND RESULTS Compared with wild-type (WT), S100A1-knock-out mice (KO) exhibited increased right ventricular (RV) weight/body weight ratio and elevated RV pressure in the absence of altered left ventricular filling pressures, accompanied by increase in wall thickness of muscularized pulmonary arteries and a reduction in microvascular perfusion. In isolated lung preparations, KO revealed reduced basal NO, blunted dose-responsiveness to acetylcholine, and augmented basal and angiotensin (AII)-induced pulmonary vascular resistance (R₀) compared with WT. Pre-treatment of KO lungs with S100A1 attenuated the AII-induced increase in pulmonary arterial pressure and R₀. S100A1-induced phosphorylation of eNOS, Akt, and ERK1/2 is attenuated in pulmonary EC of KO compared with WT. Basal and TNF-α-induced EC apoptosis is greater in KO vs. WT, and cell survival is enhanced by S100A1 treatment. CONCLUSION Our data demonstrate that the absence of S100A1 results in PH by disruption of its normal capacity to (i) enhance pulmonary EC function by induction of eNOS activity and NO levels via Akt/ERK1/2 pathways and (ii) promote EC survival. The ability of exogenously administered S100A1 to rescue this phenotype makes it an attractive therapeutic target in the treatment of PH.
Collapse
Affiliation(s)
| | | | | | - Jun Yin
- Department of Surgery, Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Toronto, Canada
| | - Liming Wang
- Department of Surgery, Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Toronto, Canada
| | - Wolfgang M Kuebler
- Department of Surgery, Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Toronto, Canada
| | | |
Collapse
|
49
|
Tilley DG, Zhu W, Myers VD, Barr LA, Gao E, Li X, Song J, Carter RL, Makarewich CA, Yu D, Troupes CD, Grisanti LA, Coleman RC, Koch WJ, Houser SR, Cheung JY, Feldman AM. β-adrenergic receptor-mediated cardiac contractility is inhibited via vasopressin type 1A-receptor-dependent signaling. Circulation 2014; 130:1800-11. [PMID: 25205804 DOI: 10.1161/circulationaha.114.010434] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Enhanced arginine vasopressin levels are associated with increased mortality during end-stage human heart failure, and cardiac arginine vasopressin type 1A receptor (V1AR) expression becomes increased. Additionally, mice with cardiac-restricted V1AR overexpression develop cardiomyopathy and decreased β-adrenergic receptor (βAR) responsiveness. This led us to hypothesize that V1AR signaling regulates βAR responsiveness and in doing so contributes to development of heart failure. METHODS AND RESULTS Transaortic constriction resulted in decreased cardiac function and βAR density and increased cardiac V1AR expression, effects reversed by a V1AR-selective antagonist. Molecularly, V1AR stimulation led to decreased βAR ligand affinity, as well as βAR-induced Ca(2+) mobilization and cAMP generation in isolated adult cardiomyocytes, effects recapitulated via ex vivo Langendorff analysis. V1AR-mediated regulation of βAR responsiveness was demonstrated to occur in a previously unrecognized Gq protein-independent/G protein receptor kinase-dependent manner. CONCLUSIONS This newly discovered relationship between cardiac V1AR and βAR may be informative for the treatment of patients with acute decompensated heart failure and elevated arginine vasopressin.
Collapse
Affiliation(s)
- Douglas G Tilley
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.).
| | - Weizhong Zhu
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Valerie D Myers
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Larry A Barr
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Erhe Gao
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Xue Li
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Jianliang Song
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Rhonda L Carter
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Catherine A Makarewich
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Daohai Yu
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Constantine D Troupes
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Laurel A Grisanti
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Ryan C Coleman
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Walter J Koch
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Steven R Houser
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Joseph Y Cheung
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| | - Arthur M Feldman
- From the Center for Translational Medicine (D.G.T., E.G., J.S, R.L.C., L.A.G., W.J.K., J.Y.C.), Department of Pharmacology (D.G.T., W.J.K.), Cardiovascular Research Center (W.Z., V.D.M., L.A.B., C.A.M., C.D.T., R.C.C., S.R.H.), Department of Physiology (L.A.B., C.A.M., S.R.H., A.M.F.), Department of Clinical Sciences (D.Y.), and Department of Medicine (J.Y.C., A.M.F.), Temple University School of Medicine, Philadelphia, PA; and the Division of Cardiology, Fourth Military Medical University, Xian, People's Republic of China (X.L.)
| |
Collapse
|
50
|
Duarte-Costa S, Castro-Ferreira R, Neves JS, Leite-Moreira AF. S100A1: a major player in cardiovascular performance. Physiol Res 2014; 63:669-81. [PMID: 25157660 DOI: 10.33549/physiolres.932712] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Calcium cycling is a major determinant of cardiac function. S100A1 is the most abundant member of the calcium-binding S100 protein family in myocardial tissue. S100A1 interacts with a variety of calcium regulatory proteins such as SERCA2a, ryanodine receptors, L-type calcium channels and Na(+)/Ca(2+) exchangers, thus enhancing calcium cycling. Aside from this major function, S100A1 has an important role in energy balance, myofilament sliding, myofilament calcium sensibility, titin-actin interaction, apoptosis and cardiac remodeling. Apart from its properties regarding cardiomyocytes, S100A1 is also important in vessel relaxation and angiogenesis. S100A1 potentiates cardiac function thus increasing the cardiomyocytes' functional reserve; this is an important feature in heart failure. In fact, S100A1 seems to normalize cardiac function after myocardial infarction. Also, S100A1 is essential in the acute response to adrenergic stimulation. Gene therapy experiments show promising results, although further studies are still needed to reach clinical practice. In this review, we aim to describe the molecular basis and regulatory function of S100A1, exploring its interactions with a myriad of target proteins. We also explore its functional effects on systolic and diastolic function as well as its acute actions. Finally, we discuss S100A1 gene therapy and its progression so far.
Collapse
Affiliation(s)
- S Duarte-Costa
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal.
| | | | | | | |
Collapse
|