1
|
Song MH, Yoo J, Kwon DA, Chepurko E, Cho S, Fargnoli A, Hajjar RJ, Park WJ, Zangi L, Jeong D. Modified mRNA-Mediated CCN5 Gene Transfer Ameliorates Cardiac Dysfunction and Fibrosis without Adverse Structural Remodeling. Int J Mol Sci 2024; 25:6262. [PMID: 38892449 PMCID: PMC11172546 DOI: 10.3390/ijms25116262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/01/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024] Open
Abstract
Modified mRNAs (modRNAs) are an emerging delivery method for gene therapy. The success of modRNA-based COVID-19 vaccines has demonstrated that modRNA is a safe and effective therapeutic tool. Moreover, modRNA has the potential to treat various human diseases, including cardiac dysfunction. Acute myocardial infarction (MI) is a major cardiac disorder that currently lacks curative treatment options, and MI is commonly accompanied by fibrosis and impaired cardiac function. Our group previously demonstrated that the matricellular protein CCN5 inhibits cardiac fibrosis (CF) and mitigates cardiac dysfunction. However, it remains unclear whether early intervention of CF under stress conditions is beneficial or more detrimental due to potential adverse effects such as left ventricular (LV) rupture. We hypothesized that CCN5 would alleviate the adverse effects of myocardial infarction (MI) through its anti-fibrotic properties under stress conditions. To induce the rapid expression of CCN5, ModRNA-CCN5 was synthesized and administrated directly into the myocardium in a mouse MI model. To evaluate CCN5 activity, we established two independent experimental schemes: (1) preventive intervention and (2) therapeutic intervention. Functional analyses, including echocardiography and magnetic resonance imaging (MRI), along with molecular assays, demonstrated that modRNA-mediated CCN5 gene transfer significantly attenuated cardiac fibrosis and improved cardiac function in both preventive and therapeutic models, without causing left ventricular rupture or any adverse cardiac remodeling. In conclusion, early intervention in CF by ModRNA-CCN5 gene transfer is an efficient and safe therapeutic modality for treating MI-induced heart failure.
Collapse
Affiliation(s)
- Min Ho Song
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea; (M.H.S.)
| | - Jimeen Yoo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; (J.Y.); (E.C.); (A.F.)
| | - Do-A Kwon
- Department of Medicinal & Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan-si 15588, Republic of Korea; (D.-A.K.); (S.C.)
| | - Elena Chepurko
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; (J.Y.); (E.C.); (A.F.)
| | - Sunghye Cho
- Department of Medicinal & Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan-si 15588, Republic of Korea; (D.-A.K.); (S.C.)
| | - Anthony Fargnoli
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; (J.Y.); (E.C.); (A.F.)
| | - Roger J. Hajjar
- Mass General Brigham Gene and Cell Therapy Institute, Boston, MA 02139, USA;
| | - Woo Jin Park
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea; (M.H.S.)
| | - Lior Zangi
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; (J.Y.); (E.C.); (A.F.)
| | - Dongtak Jeong
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; (J.Y.); (E.C.); (A.F.)
- Department of Medicinal & Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan-si 15588, Republic of Korea; (D.-A.K.); (S.C.)
| |
Collapse
|
2
|
Gáspár R, Diószegi P, Nógrádi-Halmi D, Erdélyi-Furka B, Varga Z, Kahán Z, Csont T. The Proteoglycans Biglycan and Decorin Protect Cardiac Cells against Irradiation-Induced Cell Death by Inhibiting Apoptosis. Cells 2024; 13:883. [PMID: 38786104 PMCID: PMC11119486 DOI: 10.3390/cells13100883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/05/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024] Open
Abstract
Radiation-induced heart disease (RIHD), a common side effect of chest irradiation, is a primary cause of mortality among patients surviving thoracic cancer. Thus, the development of novel, clinically applicable cardioprotective agents which can alleviate the harmful effects of irradiation on the heart is of great importance in the field of experimental oncocardiology. Biglycan and decorin are structurally related small leucine-rich proteoglycans which have been reported to exert cardioprotective properties in certain cardiovascular pathologies. Therefore, in the present study we aimed to examine if biglycan or decorin can reduce radiation-induced damage of cardiomyocytes. A single dose of 10 Gray irradiation was applied to induce radiation-induced cell damage in H9c2 cardiomyoblasts, followed by treatment with either biglycan or decorin at various concentrations. Measurement of cell viability revealed that both proteoglycans improved the survival of cardiac cells post-irradiation. The cardiocytoprotective effect of both biglycan and decorin involved the alleviation of radiation-induced proapoptotic mechanisms by retaining the progression of apoptotic membrane blebbing and lowering the number of apoptotic cell nuclei and DNA double-strand breaks. Our findings provide evidence that these natural proteoglycans may exert protection against radiation-induced damage of cardiac cells.
Collapse
Affiliation(s)
- Renáta Gáspár
- Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (R.G.); (P.D.); (D.N.-H.); (B.E.-F.)
- Interdisciplinary Centre of Excellence, University of Szeged, H-6720 Szeged, Hungary
| | - Petra Diószegi
- Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (R.G.); (P.D.); (D.N.-H.); (B.E.-F.)
- Interdisciplinary Centre of Excellence, University of Szeged, H-6720 Szeged, Hungary
| | - Dóra Nógrádi-Halmi
- Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (R.G.); (P.D.); (D.N.-H.); (B.E.-F.)
- Interdisciplinary Centre of Excellence, University of Szeged, H-6720 Szeged, Hungary
| | - Barbara Erdélyi-Furka
- Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (R.G.); (P.D.); (D.N.-H.); (B.E.-F.)
- Interdisciplinary Centre of Excellence, University of Szeged, H-6720 Szeged, Hungary
| | - Zoltán Varga
- Department of Oncotherapy, University of Szeged, H-6720 Szeged, Hungary; (Z.V.); (Z.K.)
| | - Zsuzsanna Kahán
- Department of Oncotherapy, University of Szeged, H-6720 Szeged, Hungary; (Z.V.); (Z.K.)
| | - Tamás Csont
- Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary; (R.G.); (P.D.); (D.N.-H.); (B.E.-F.)
- Interdisciplinary Centre of Excellence, University of Szeged, H-6720 Szeged, Hungary
| |
Collapse
|
3
|
Biyik I, Soysal C, Ince OUO, Durmus S, Oztas E, Keskin N, Isiklar OO, Karaagac OH, Gelisgen R, Uzun H. Prediction of Preterm Delivery Using Serum Ischemia Modified Albumin, Biglycan, and Decorin Levels in Women with Threatened Preterm Labor. REVISTA BRASILEIRA DE GINECOLOGIA E OBSTETRÍCIA 2023; 45:e754-e763. [PMID: 38141595 DOI: 10.1055/s-0043-1772593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2023] Open
Abstract
OBJECTIVE The serum ischemia modified albumin (IMA), biglycan, and decorin levels of pregnant women who were hospitalized for threatened preterm labor were measured. METHODS Fifty-one consecutive pregnant women with a single pregnancy between the 24th and 36th weeks with a diagnosis of threatened preterm labor were included in the present prospective cohort study. RESULTS As a result of multivariate logistic regression analysis for predicting preterm delivery within 24 hours, 48 hours, 7 days, 14 days, ≤ 35 gestational weeks, and ≤ 37 gestational weeks after admission, area under the curve (AUC) (95% confidence interval [CI[) values were 0.95 (0.89-1.00), 0.93 (0.86-0.99), 0.91 (0.83-0.98), 0.92 (0.85-0.99), 0.82 (0.69-0.96), and 0.89 (0.80-0.98), respectively. In the present study, IMA and biglycan levels were found to be higher and decorin levels lower in women admitted to the hospital with threatened preterm labor and who gave preterm birth within 48 hours compared with those who gave birth after 48 hours. CONCLUSION In pregnant women admitted to the hospital with threatened preterm labor, the prediction preterm delivery of the combined model created by adding IMA, decorin, and biglycan in addition to the TVS CL measurement was higher than the TVS CL measurement alone. CLINICAL TRIAL REGISTRATION The present trial was registered at ClinicalTrials.gov, number NCT04451928.
Collapse
Affiliation(s)
- Ismail Biyik
- Department of Obstetrics and Gynecology, School of Medicine, Kutahya Health Sciences University, Kutahya, Turkey
| | - Cenk Soysal
- Department of Obstetrics and Gynecology, School of Medicine, Kutahya Health Sciences University, Kutahya, Turkey
| | - Ozlem Ulas Onur Ince
- Department of Obstetrics and Gynecology, School of Medicine, Kutahya Health Sciences University, Kutahya, Turkey
- Department of Statistics, Faculty of Arts and Sciences, Middle East Technical University, Ankara, Turkey
| | - Sinem Durmus
- Department of Medical Biochemistry, School of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Efser Oztas
- Department of Obstetrics and Gynecology, School of Medicine, Kutahya Health Sciences University, Kutahya, Turkey
| | - Nadi Keskin
- Department of Obstetrics and Gynecology, School of Medicine, Kutahya Health Sciences University, Kutahya, Turkey
| | - Ozben Ozden Isiklar
- Department of Medical Biochemistry, School of Medicine, Kutahya Health Sciences University, Kutahya, Turkey
| | - Oğuz Han Karaagac
- Department of Obstetrics and Gynecology, School of Medicine, Kutahya Health Sciences University, Kutahya, Turkey
| | - Remise Gelisgen
- Department of Medical Biochemistry, School of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Hafize Uzun
- Department of Medical Biochemistry, Faculty of Medicine, Istanbul Atlas University, Istanbul, Turkey
| |
Collapse
|
4
|
Pearce DP, Nemcek MT, Witzenburg CM. Don't go breakin' my heart: cardioprotective alterations to the mechanical and structural properties of reperfused myocardium during post-infarction inflammation. Biophys Rev 2023; 15:329-353. [PMID: 37396449 PMCID: PMC10310682 DOI: 10.1007/s12551-023-01068-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 05/21/2023] [Indexed: 07/04/2023] Open
Abstract
Myocardial infarctions (MIs) kickstart an intense inflammatory response resulting in extracellular matrix (ECM) degradation, wall thinning, and chamber dilation that leaves the heart susceptible to rupture. Reperfusion therapy is one of the most effective strategies for limiting adverse effects of MIs, but is a challenge to administer in a timely manner. Late reperfusion therapy (LRT; 3 + hours post-MI) does not limit infarct size, but does reduce incidences of post-MI rupture and improves long-term patient outcomes. Foundational studies employing LRT in the mid-twentieth century revealed beneficial reductions in infarct expansion, aneurysm formation, and left ventricle dysfunction. The mechanism by which LRT acts, however, is undefined. Structural analyses, relying largely on one-dimensional estimates of ECM composition, have found few differences in collagen content between LRT and permanently occluded animal models when using homogeneous samples from infarct cores. Uniaxial testing, on the other hand, revealed slight reductions in stiffness early in inflammation, followed soon after by an enhanced resistance to failure for cases of LRT. The use of one-dimensional estimates of ECM organization and gross mechanical function have resulted in a poor understanding of the infarct's spatially variable mechanical and structural anisotropy. To resolve these gaps in literature, future work employing full-field mechanical, structural, and cellular analyses is needed to better define the spatiotemporal post-MI alterations occurring during the inflammatory phase of healing and how they are impacted following reperfusion therapy. In turn, these studies may reveal how LRT affects the likelihood of rupture and inspire novel approaches to guide scar formation.
Collapse
Affiliation(s)
- Daniel P. Pearce
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706 USA
| | - Mark T. Nemcek
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706 USA
| | - Colleen M. Witzenburg
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706 USA
| |
Collapse
|
5
|
Chen J, Chapski DJ, Jong J, Awada J, Wang Y, Slamon DJ, Vondriska TM, Packard RRS. Integrative transcriptomics and cell systems analyses reveal protective pathways controlled by Igfbp-3 in anthracycline-induced cardiotoxicity. FASEB J 2023; 37:e22977. [PMID: 37219486 PMCID: PMC10286824 DOI: 10.1096/fj.202201885rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 04/24/2023] [Accepted: 05/03/2023] [Indexed: 05/24/2023]
Abstract
Anthracyclines such as doxorubicin (Dox) are effective chemotherapeutic agents; however, their use is hampered by subsequent cardiotoxicity risk. Our understanding of cardiomyocyte protective pathways activated following anthracycline-induced cardiotoxicity (AIC) remains incomplete. Insulin-like growth factor binding protein (IGFBP) 3 (Igfbp-3), the most abundant IGFBP family member in the circulation, is associated with effects on the metabolism, proliferation, and survival of various cells. Whereas Igfbp-3 is induced by Dox in the heart, its role in AIC is ill-defined. We investigated molecular mechanisms as well as systems-level transcriptomic consequences of manipulating Igfbp-3 in AIC using neonatal rat ventricular myocytes and human-induced pluripotent stem cell-derived cardiomyocytes. Our findings reveal that Dox induces the nuclear enrichment of Igfbp-3 in cardiomyocytes. Furthermore, Igfbp-3 reduces DNA damage, impedes topoisomerase IIβ expression (Top2β) which forms Top2β-Dox-DNA cleavage complex leading to DNA double-strand breaks (DSB), alleviates detyrosinated microtubule accumulation-a hallmark of increased cardiomyocyte stiffness and heart failure-and favorably affects contractility following Dox treatment. These results indicate that Igfbp-3 is induced by cardiomyocytes in an effort to mitigate AIC.
Collapse
Affiliation(s)
- Junjie Chen
- Molecular, Cellular, and Integrative Physiology Program,
College of Letters and Science, and David Geffen School of Medicine, University of
California, Los Angeles, CA
| | - Douglas J. Chapski
- Department of Anesthesiology & Perioperative Medicine,
David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Jeremy Jong
- Division of Cardiology, Department of Medicine, David
Geffen School of Medicine, University of California, Los Angeles, CA
| | - Jerome Awada
- Division of Cardiology, Department of Medicine, David
Geffen School of Medicine, University of California, Los Angeles, CA
| | - Yijie Wang
- Division of Cardiology, Department of Medicine, David
Geffen School of Medicine, University of California, Los Angeles, CA
| | - Dennis J. Slamon
- Division of Hematology & Oncology, Department of
Medicine, David Geffen School of Medicine, University of California, Los Angeles,
CA
- Jonsson Comprehensive Cancer Center, University of
California, Los Angeles, CA
| | - Thomas M. Vondriska
- Molecular, Cellular, and Integrative Physiology Program,
College of Letters and Science, and David Geffen School of Medicine, University of
California, Los Angeles, CA
- Department of Anesthesiology & Perioperative Medicine,
David Geffen School of Medicine, University of California, Los Angeles, CA
- Division of Cardiology, Department of Medicine, David
Geffen School of Medicine, University of California, Los Angeles, CA
- Department of Physiology, David Geffen School of Medicine,
University of California, Los Angeles, CA
- Molecular Biology Institute, University of California, Los
Angeles, CA
| | - René R. Sevag Packard
- Molecular, Cellular, and Integrative Physiology Program,
College of Letters and Science, and David Geffen School of Medicine, University of
California, Los Angeles, CA
- Division of Cardiology, Department of Medicine, David
Geffen School of Medicine, University of California, Los Angeles, CA
- Jonsson Comprehensive Cancer Center, University of
California, Los Angeles, CA
- Department of Physiology, David Geffen School of Medicine,
University of California, Los Angeles, CA
- Molecular Biology Institute, University of California, Los
Angeles, CA
- Ronald Reagan UCLA Medical Center, Los Angeles, CA
- Veterans Affairs West Los Angeles Medical Center, Los
Angeles, CA
- California NanoSystems Institute, University of
California, Los Angeles, CA
| |
Collapse
|
6
|
Portokallidou K, Dovrolis N, Ragia G, Atzemian N, Kolios G, Manolopoulos VG. Multi-omics integration to identify the genetic expression and protein signature of dilated and ischemic cardiomyopathy. Front Cardiovasc Med 2023; 10:1115623. [PMID: 36860278 PMCID: PMC9968758 DOI: 10.3389/fcvm.2023.1115623] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 01/30/2023] [Indexed: 02/16/2023] Open
Abstract
Introduction Heart failure (HF) is a complex clinical syndrome leading to high morbidity. In this study, we aimed to identify the gene expression and protein signature of HF main causes, namely dilated cardiomyopathy (DCM) and ischemic cardiomyopathy (ICM). Methods Omics data were accessed through GEO repository for transcriptomic and PRIDE repository for proteomic datasets. Sets of differentially expressed genes and proteins comprising DCM (DiSig) and ICM (IsSig) signatures were analyzed by a multilayered bioinformatics approach. Enrichment analysis via the Gene Ontology was performed through the Metascape platform to explore biological pathways. Protein-protein interaction networks were analyzed via STRING db and Network Analyst. Results Intersection of transcriptomic and proteomic analysis showed 10 differentially expressed genes/proteins in DiSig (AEBP1, CA3, HBA2, HBB, HSPA2, MYH6, SERPINA3, SOD3, THBS4, UCHL1) and 15 differentially expressed genes/proteins in IsSig (AEBP1, APOA1, BGN, CA3, CFH, COL14A1, HBA2, HBB, HSPA2, LTBP2, LUM, MFAP4, SOD3, THBS4, UCHL1). Common and distinct biological pathways between DiSig and IsSig were retrieved, allowing for their molecular characterization. Extracellular matrix organization, cellular response to stress and transforming growth factor-beta were common between two subphenotypes. Muscle tissue development was dysregulated solely in DiSig, while immune cells activation and migration in IsSig. Discussion Our bioinformatics approach sheds light on the molecular background of HF etiopathology showing molecular similarities as well as distinct expression differences between DCM and ICM. DiSig and IsSig encompass an array of "cross-validated" genes at both transcriptomic and proteomic level, which can serve as novel pharmacological targets and possible diagnostic biomarkers.
Collapse
Affiliation(s)
- Konstantina Portokallidou
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Alexandroupolis, Greece,Individualised Medicine and Pharmacological Research Solutions Center, Alexandroupolis, Greece
| | - Nikolas Dovrolis
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Alexandroupolis, Greece,Individualised Medicine and Pharmacological Research Solutions Center, Alexandroupolis, Greece,Nikolas Dovrolis,
| | - Georgia Ragia
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Alexandroupolis, Greece,Individualised Medicine and Pharmacological Research Solutions Center, Alexandroupolis, Greece
| | - Natalia Atzemian
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Alexandroupolis, Greece,Individualised Medicine and Pharmacological Research Solutions Center, Alexandroupolis, Greece
| | - George Kolios
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Alexandroupolis, Greece,Individualised Medicine and Pharmacological Research Solutions Center, Alexandroupolis, Greece
| | - Vangelis G. Manolopoulos
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Alexandroupolis, Greece,Individualised Medicine and Pharmacological Research Solutions Center, Alexandroupolis, Greece,Clinical Pharmacology Unit, Academic General Hospital of Alexandroupolis, Alexandroupolis, Greece,*Correspondence: Vangelis G. Manolopoulos,
| |
Collapse
|
7
|
Scuruchi M, Mannino F, Imbesi C, Pallio G, Vermiglio G, Bagnato G, Minutoli L, Bitto A, Squadrito F, Irrera N. Biglycan Involvement in Heart Fibrosis: Modulation of Adenosine 2A Receptor Improves Damage in Immortalized Cardiac Fibroblasts. Int J Mol Sci 2023; 24:ijms24021784. [PMID: 36675295 PMCID: PMC9866951 DOI: 10.3390/ijms24021784] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/29/2022] [Accepted: 01/13/2023] [Indexed: 01/17/2023] Open
Abstract
Cardiac fibrosis is a common pathological feature of different cardiovascular diseases, characterized by the aberrant deposition of extracellular matrix (ECM) proteins in the cardiac interstitium, myofibroblast differentiation and increased fibrillar collagen deposition stimulated by transforming growth factor (TGF)-β activation. Biglycan (BGN), a small leucine-rich proteoglycan (SLRPG) integrated within the ECM, plays a key role in matrix assembly and the phenotypic control of cardiac fibroblasts. Moreover, BGN is critically involved in pathological cardiac remodeling through TGF-β binding, thus causing myofibroblast differentiation and proliferation. Adenosine receptors (ARs), and in particular A2AR, may play a key role in stimulating fibrotic damage through collagen production/deposition, as a consequence of cyclic AMP (cAMP) and AKT activation. For this reason, A2AR modulation could be a useful tool to manage cardiac fibrosis in order to reduce fibrotic scar deposition in heart tissue. Therefore, the aim of the present study was to investigate the possible crosstalk between A2AR and BGN modulation in an in vitro model of TGF-β-induced fibrosis. Immortalized human cardiac fibroblasts (IM-HCF) were stimulated with TGF-β at the concentration of 10 ng/mL for 24 h to induce a fibrotic phenotype. After applying the TGF-β stimulus, cells were treated with two different A2AR antagonists, Istradefylline and ZM241385, for an additional 24 h, at the concentration of 10 µM and 1 µM, respectively. Both A2AR antagonists were able to regulate the oxidative stress induced by TGF-β through intracellular reactive oxygen species (ROS) reduction in IM-HCFs. Moreover, collagen1a1, MMPs 3/9, BGN, caspase-1 and IL-1β gene expression was markedly decreased following A2AR antagonist treatment in TGF-β-challenged human fibroblasts. The results obtained for collagen1a1, SMAD3, α-SMA and BGN were also confirmed when protein expression was evaluated; phospho-Akt protein levels were also reduced following Istradefylline and ZM241385 use, thus suggesting that collagen production involves AKT recruited by the A2AR. These results suggest that A2AR modulation might be an effective therapeutic option to reduce the fibrotic processes involved in heart pathological remodeling.
Collapse
Affiliation(s)
- Michele Scuruchi
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy
| | - Federica Mannino
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy
| | - Chiara Imbesi
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy
| | - Giovanni Pallio
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy
| | - Giovanna Vermiglio
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Via C. Valeria, 98125 Messina, Italy
| | - Gianluca Bagnato
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy
| | - Letteria Minutoli
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy
| | - Francesco Squadrito
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy
- Correspondence:
| | - Natasha Irrera
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125 Messina, Italy
| |
Collapse
|
8
|
Azimzadeh O, Moertl S, Ramadan R, Baselet B, Laiakis EC, Sebastian S, Beaton D, Hartikainen JM, Kaiser JC, Beheshti A, Salomaa S, Chauhan V, Hamada N. Application of radiation omics in the development of adverse outcome pathway networks: an example of radiation-induced cardiovascular disease. Int J Radiat Biol 2022; 98:1722-1751. [PMID: 35976069 DOI: 10.1080/09553002.2022.2110325] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Epidemiological studies have indicated that exposure of the heart to doses of ionizing radiation as low as 0.5 Gy increases the risk of cardiac morbidity and mortality with a latency period of decades. The damaging effects of radiation to myocardial and endothelial structures and functions have been confirmed radiobiologically at high dose, but much less is known at low dose. Integration of radiation biology and epidemiology data is a recommended approach to improve the radiation risk assessment process. The adverse outcome pathway (AOP) framework offers a comprehensive tool to compile and translate mechanistic information into pathological endpoints which may be relevant for risk assessment at the different levels of a biological system. Omics technologies enable the generation of large volumes of biological data at various levels of complexity, from molecular pathways to functional organisms. Given the quality and quantity of available data across levels of biology, omics data can be attractive sources of information for use within the AOP framework. It is anticipated that radiation omics studies could improve our understanding of the molecular mechanisms behind the adverse effects of radiation on the cardiovascular system. In this review, we explored the available omics studies on radiation-induced cardiovascular disease (CVD) and their applicability to the proposed AOP for CVD. RESULTS The results of 80 omics studies published on radiation-induced CVD over the past 20 years have been discussed in the context of the AOP of CVD proposed by Chauhan et al. Most of the available omics data on radiation-induced CVD are from proteomics, transcriptomics, and metabolomics, whereas few datasets were available from epigenomics and multi-omics. The omics data presented here show great promise in providing information for several key events of the proposed AOP of CVD, particularly oxidative stress, alterations of energy metabolism, extracellular matrix and vascular remodeling. CONCLUSIONS The omics data presented here shows promise to inform the various levels of the proposed AOP of CVD. However, the data highlight the urgent need of designing omics studies to address the knowledge gap concerning different radiation scenarios, time after exposure and experimental models. This review presents the evidence to build a qualitative omics-informed AOP and provides views on the potential benefits and challenges in using omics data to assess risk-related outcomes.
Collapse
Affiliation(s)
- Omid Azimzadeh
- Federal Office for Radiation Protection (BfS), Section Radiation Biology, 85764 Neuherberg, Germany
| | - Simone Moertl
- Federal Office for Radiation Protection (BfS), Section Radiation Biology, 85764 Neuherberg, Germany
| | - Raghda Ramadan
- Institute for Environment, Health and Safety, Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Bjorn Baselet
- Institute for Environment, Health and Safety, Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Evagelia C Laiakis
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA.,Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC 20057, USA
| | | | | | - Jaana M Hartikainen
- School of Medicine, Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland, Kuopio, Finland
| | - Jan Christian Kaiser
- Helmholtz Zentrum München, Institute of Radiation Medicine (HMGU-IRM), 85764 Neuherberg, Germany
| | - Afshin Beheshti
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Sisko Salomaa
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Vinita Chauhan
- Environmental Health Science Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Nobuyuki Hamada
- Biology and Environmental Chemistry Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Komae, Tokyo 201-8511, Japan
| |
Collapse
|
9
|
Huang C, Sharma A, Thakur R, Rai D, Katiki M, Germano JDF, Song Y, Singh S, Sin J, Sengstock D, Andres AM, Murali R, Mentzer RM, Gottlieb RA, Piplani H. Asporin, an extracellular matrix protein, is a beneficial regulator of cardiac remodeling. Matrix Biol 2022; 110:40-59. [DOI: 10.1016/j.matbio.2022.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 03/29/2022] [Accepted: 04/19/2022] [Indexed: 01/15/2023]
|
10
|
Hoermann H, Krueger I, Maurus N, Reusswig F, Sun Y, Kohlmorgen C, Grandoch M, Fischer JW, Elvers M. The Proteoglycan Biglycan Modulates Platelet Adhesion and Thrombus Formation in a GPVI-Dependent Manner. Int J Mol Sci 2021; 22:12168. [PMID: 34830059 PMCID: PMC8622445 DOI: 10.3390/ijms222212168] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/05/2021] [Accepted: 11/07/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Vascular injury induces the exposure of subendothelial extracellular matrix (ECM) important to serve as substrate for platelets to adhere to the injured vessel wall to avoid massive blood loss. Different ECM proteins are known to initiate platelet adhesion and activation. In atherosclerotic mice, the small, leucine-rich proteoglycan biglycan is important for the regulation of thrombin activity via heparin cofactor II. However, nothing is known about the role of biglycan for hemostasis and thrombosis under nonatherosclerotic conditions. METHODS The role of biglycan for platelet adhesion and thrombus formation was investigated using a recombinant protein and biglycan knockout mice. RESULTS The present study identified biglycan as important ECM protein for the adhesion and activation of platelets, and the formation of three-dimensional thrombi under flow conditions. Platelet adhesion to immobilized biglycan induces the reorganization of the platelet cytoskeleton. Mechanistically, biglycan binds and activates the major collagen receptor glycoprotein (GP)VI, because reduced platelet adhesion to recombinant biglycan was observed when GPVI was blocked and enhanced tyrosine phosphorylation in a GPVI-dependent manner was observed when platelets were stimulated with biglycan. In vivo, the deficiency of biglycan resulted in reduced platelet adhesion to the injured carotid artery and prolonged bleeding times. CONCLUSIONS Loss of biglycan in the vessel wall of mice but not in platelets led to reduced platelet adhesion at the injured carotid artery and prolonged bleeding times, suggesting a crucial role for biglycan as ECM protein that binds and activates platelets via GPVI upon vessel injury.
Collapse
Affiliation(s)
- Henrike Hoermann
- Department of Vascular and Endovascular Surgery, Experimental Vascular Medicine, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany; (H.H.); (I.K.); (N.M.); (F.R.)
| | - Irena Krueger
- Department of Vascular and Endovascular Surgery, Experimental Vascular Medicine, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany; (H.H.); (I.K.); (N.M.); (F.R.)
| | - Nadine Maurus
- Department of Vascular and Endovascular Surgery, Experimental Vascular Medicine, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany; (H.H.); (I.K.); (N.M.); (F.R.)
| | - Friedrich Reusswig
- Department of Vascular and Endovascular Surgery, Experimental Vascular Medicine, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany; (H.H.); (I.K.); (N.M.); (F.R.)
| | - Yi Sun
- Centre of Membrane Proteins and Receptors (COMPARE), Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK;
| | - Christina Kohlmorgen
- Institute for Pharmacology und Clinical Pharmacology, University Hospital of the Heinrich-Heine-University, 40225 Düsseldorf, Germany; (C.K.); (M.G.); (J.W.F.)
| | - Maria Grandoch
- Institute for Pharmacology und Clinical Pharmacology, University Hospital of the Heinrich-Heine-University, 40225 Düsseldorf, Germany; (C.K.); (M.G.); (J.W.F.)
| | - Jens W. Fischer
- Institute for Pharmacology und Clinical Pharmacology, University Hospital of the Heinrich-Heine-University, 40225 Düsseldorf, Germany; (C.K.); (M.G.); (J.W.F.)
| | - Margitta Elvers
- Department of Vascular and Endovascular Surgery, Experimental Vascular Medicine, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany; (H.H.); (I.K.); (N.M.); (F.R.)
| |
Collapse
|
11
|
van den Berg NWE, Neefs J, Kawasaki M, Nariswari FA, Wesselink R, Fabrizi B, Jongejan A, Klaver MN, Havenaar H, Hulsman EL, Wintgens LIS, Baalman SWE, Meulendijks ER, van Boven WJ, de Jong JSSG, van Putte BP, Driessen AHG, Boersma LVA, de Groot JR. Extracellular matrix remodeling precedes atrial fibrillation: Results of the PREDICT-AF trial. Heart Rhythm 2021; 18:2115-2125. [PMID: 34332113 DOI: 10.1016/j.hrthm.2021.07.059] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/08/2021] [Accepted: 07/23/2021] [Indexed: 01/18/2023]
Abstract
BACKGROUND To which extent atrial remodeling occurs before atrial fibrillation (AF) is unknown. OBJECTIVE The PREventive left atrial appenDage resection for the predICtion of fuTure Atrial Fibrillation (PREDICT-AF) study investigated such subclinical remodeling, which may be used for risk stratification and AF prevention. METHODS Patients (N = 150) without a history of AF with a CHA2DS2-VASc score of ≥2 at an increased risk of developing AF were included. The left atrial appendage was excised and blood samples were collected during elective cardiothoracic surgery for biomarker discovery. Participants were followed for 2 years with Holter monitoring to determine any atrial tachyarrhythmia after a 50-day blanking period. RESULTS Eighteen patients (12%) developed incident AF, which was associated with increased tissue gene expression of collagen I (COL1A1), collagen III (COL3A1), and collagen VIII (COL8A2), tenascin-C (TNC), thrombospondin-2 (THBS2), and biglycan (BGN). Furthermore, the fibroblast activating endothelin-1 (EDN1) and sodium voltage-gated channel β subunit 2 (SCN2B) were associated with incident AF whereas the Kir2.1 channel (KCNJ2) tended to downregulate. The plasma levels of COL8A2 and TNC correlated with tissue expression and predicted incident AF. A gene panel including tissue KCNJ2, COL1A1, COL8A2, and EDN1 outperformed clinical prediction models in discriminating incident AF. CONCLUSION The PREDICT-AF study demonstrates that atrial remodeling occurs long before incident AF and implies future potential for early patient identification and therapies to prevent AF (ClinicalTrials.gov identifier NCT03130985).
Collapse
Affiliation(s)
- Nicoline W E van den Berg
- Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jolien Neefs
- Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Makiri Kawasaki
- Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Fransisca A Nariswari
- Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Robin Wesselink
- Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Benedetta Fabrizi
- Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Aldo Jongejan
- Department of Epidemiology and Data Science, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Martijn N Klaver
- Department of Cardiology and Cardiothoracic Surgery, St. Antonius Ziekenhuis, Nieuwegein, The Netherlands
| | - Hanna Havenaar
- Department of Cardiology and Cardiothoracic Surgery, St. Antonius Ziekenhuis, Nieuwegein, The Netherlands
| | - Elise L Hulsman
- Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Lisette I S Wintgens
- Department of Cardiology and Cardiothoracic Surgery, St. Antonius Ziekenhuis, Nieuwegein, The Netherlands
| | - Sarah W E Baalman
- Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Eva R Meulendijks
- Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Wim Jan van Boven
- Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Bart P van Putte
- Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Department of Cardiology and Cardiothoracic Surgery, St. Antonius Ziekenhuis, Nieuwegein, The Netherlands
| | - Antoine H G Driessen
- Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Lucas V A Boersma
- Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Department of Cardiology and Cardiothoracic Surgery, St. Antonius Ziekenhuis, Nieuwegein, The Netherlands
| | - Joris R de Groot
- Department of Clinical and Experimental Cardiology and Cardiothoracic Surgery, Heart Center, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
| | | |
Collapse
|
12
|
Morimoto H, Hida Y, Maishi N, Nishihara H, Hatanaka Y, Li C, Matsuno Y, Nakamura T, Hirano S, Hida K. Biglycan, tumor endothelial cell secreting proteoglycan, as possible biomarker for lung cancer. Thorac Cancer 2021; 12:1347-1357. [PMID: 33709550 PMCID: PMC8088962 DOI: 10.1111/1759-7714.13907] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/09/2021] [Accepted: 02/09/2021] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES In lung cancer, surgery remains the most curative treatment and limited resection is beneficial for patients with low cardiopulmonary function and low malignancy tumors. However, there are no biomarkers of low malignancy to select candidates for limited resection without compromising the outcome of treatments. Recently we identified biglycan (BGN) as a tumor endothelial cell (TEC) marker that is associated with tumor progression in various cancers. In this study, we analyzed the association between BGN expression in TECs in lung cancer and cancer progression in patients. MATERIALS AND METHODS First, we performed immunohistochemistry of BGN with resected lung tumor tissues of 155 patients who had undergone thoracic surgery and analyzed the correlation between BGN-positive vessel density in primary lung tumors and clinicopathological factors. Second, we measured the BGN levels in preoperative serum of other 46 patients with lung cancer by ELISA, and analyzed the correlation between BGN expression in tumor tissues and blood BGN levels. RESULTS High BGN expression in the TECs was significantly associated with T factor, and was a significant negative predictor. BGN levels in preoperative serum of 46 patients with lung cancer was significantly correlated with BGN expression in the TECs. Preoperative serum BGN level was significantly lower in healthy volunteers and less invasive adenocarcinoma than in invasive adenocarcinoma and other lung carcinomas. These results suggest that low BGN level in preoperative serum in patients with lung cancer might indicate low malignancy. CONCLUSIONS BGN can be a potential biomarker for lung cancer.
Collapse
Affiliation(s)
- Hirofumi Morimoto
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan.,Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Yasuhiro Hida
- Department of Cardiovascular and Thoracic Surgery, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Nako Maishi
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Hiroshi Nishihara
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Yutaka Hatanaka
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan.,Research Division of Genome Companion Diagnostics, Hokkaido University Hospital, Sapporo, Japan
| | - Cong Li
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Yoshihiro Matsuno
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Toru Nakamura
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Satoshi Hirano
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Kyoko Hida
- Department of Vascular Biology and Molecular Pathology, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| |
Collapse
|
13
|
Tomin T, Schittmayer M, Sedej S, Bugger H, Gollmer J, Honeder S, Darnhofer B, Liesinger L, Zuckermann A, Rainer PP, Birner-Gruenberger R. Mass Spectrometry-Based Redox and Protein Profiling of Failing Human Hearts. Int J Mol Sci 2021; 22:ijms22041787. [PMID: 33670142 PMCID: PMC7916846 DOI: 10.3390/ijms22041787] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress contributes to detrimental functional decline of the myocardium, leading to the impairment of the antioxidative defense, dysregulation of redox signaling, and protein damage. In order to precisely dissect the changes of the myocardial redox state correlated with oxidative stress and heart failure, we subjected left-ventricular tissue specimens collected from control or failing human hearts to comprehensive mass spectrometry-based redox and quantitative proteomics, as well as glutathione status analyses. As a result, we report that failing hearts have lower glutathione to glutathione disulfide ratios and increased oxidation of a number of different proteins, including constituents of the contractile machinery as well as glycolytic enzymes. Furthermore, quantitative proteomics of failing hearts revealed a higher abundance of proteins responsible for extracellular matrix remodeling and reduced abundance of several ion transporters, corroborating contractile impairment. Similar effects were recapitulated by an in vitro cell culture model under a controlled oxygen atmosphere. Together, this study provides to our knowledge the most comprehensive report integrating analyses of protein abundance and global and peptide-level redox state in end-stage failing human hearts as well as oxygen-dependent redox and global proteome profiles of cultured human cardiomyocytes.
Collapse
Affiliation(s)
- Tamara Tomin
- Faculty of Technical Chemistry, Institute of Chemical Technologies and Analytics, Vienna University of Technology-TU Wien, Getreidemarkt 9/164, 1060 Vienna, Austria;
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Stiftingtalstrasse 6, 8010 Graz, Austria; (S.H.); (B.D.); (L.L.)
- BiotechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria;
| | - Matthias Schittmayer
- Faculty of Technical Chemistry, Institute of Chemical Technologies and Analytics, Vienna University of Technology-TU Wien, Getreidemarkt 9/164, 1060 Vienna, Austria;
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Stiftingtalstrasse 6, 8010 Graz, Austria; (S.H.); (B.D.); (L.L.)
- BiotechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria;
- Correspondence: (M.S.); (P.P.R.); (R.B.-G.)
| | - Simon Sedej
- BiotechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria;
- Division of Cardiology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria; (H.B.); (J.G.)
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
| | - Heiko Bugger
- Division of Cardiology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria; (H.B.); (J.G.)
| | - Johannes Gollmer
- Division of Cardiology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria; (H.B.); (J.G.)
| | - Sophie Honeder
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Stiftingtalstrasse 6, 8010 Graz, Austria; (S.H.); (B.D.); (L.L.)
- BiotechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria;
| | - Barbara Darnhofer
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Stiftingtalstrasse 6, 8010 Graz, Austria; (S.H.); (B.D.); (L.L.)
- BiotechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria;
| | - Laura Liesinger
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Stiftingtalstrasse 6, 8010 Graz, Austria; (S.H.); (B.D.); (L.L.)
- BiotechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria;
| | - Andreas Zuckermann
- Cardiac Transplantation, Department of Cardiac Surgery, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria;
| | - Peter P. Rainer
- BiotechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria;
- Division of Cardiology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria; (H.B.); (J.G.)
- Correspondence: (M.S.); (P.P.R.); (R.B.-G.)
| | - Ruth Birner-Gruenberger
- Faculty of Technical Chemistry, Institute of Chemical Technologies and Analytics, Vienna University of Technology-TU Wien, Getreidemarkt 9/164, 1060 Vienna, Austria;
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Stiftingtalstrasse 6, 8010 Graz, Austria; (S.H.); (B.D.); (L.L.)
- BiotechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria;
- Correspondence: (M.S.); (P.P.R.); (R.B.-G.)
| |
Collapse
|
14
|
Abstract
Preterm birth is a leading cause of neonatal mortality in the US and globally, with preterm premature rupture of fetal membranes (PPROM) accounting for one third of preterm births. Currently no predictive diagnostics are available to precisely assess risk and potentially reduce the incidence of PPROM. Bigycan and decorin, the main proteoglycans present in human fetal membranes, are involved in the physiological maturation of fetal membranes as well as in the pathophysiology of preterm birth. The serum protein sex hormone-binding globulin (SHBG) has recently been identified as a predictor of spontaneous preterm birth. We hypothesize that the balance between serum decorin and biglycan on one hand and SHBG on the other hand may provide insight into the status of the fetal membranes in early pregnancy, thereby predicting PPROM prior to symptoms. Using chart review, 18 patients with confirmed cases of PPROM were identified from 2013-2016. Second trimester residual serum was retreived from freezer storage for these cases along with 5 matched controls for each case. The biomarkers biglycan, decorin and SHBG were analyzed first separately, then in combination to determine their ability to predict PPROM. The predictive score for the combined model displays an AUC = 0.774. The ROC curve of the predicted score has an optimal threshold of 0.238 and a sensitivity and specificity of 0.72 and 0.84 respectively. This prenatal serum panel is a promising serum screening-based biochemical model to predict PPROM in asymptomatic women.
Collapse
|
15
|
Abstract
Myocardial fibrosis, the expansion of the cardiac interstitium through deposition of extracellular matrix proteins, is a common pathophysiologic companion of many different myocardial conditions. Fibrosis may reflect activation of reparative or maladaptive processes. Activated fibroblasts and myofibroblasts are the central cellular effectors in cardiac fibrosis, serving as the main source of matrix proteins. Immune cells, vascular cells and cardiomyocytes may also acquire a fibrogenic phenotype under conditions of stress, activating fibroblast populations. Fibrogenic growth factors (such as transforming growth factor-β and platelet-derived growth factors), cytokines [including tumour necrosis factor-α, interleukin (IL)-1, IL-6, IL-10, and IL-4], and neurohumoral pathways trigger fibrogenic signalling cascades through binding to surface receptors, and activation of downstream signalling cascades. In addition, matricellular macromolecules are deposited in the remodelling myocardium and regulate matrix assembly, while modulating signal transduction cascades and protease or growth factor activity. Cardiac fibroblasts can also sense mechanical stress through mechanosensitive receptors, ion channels and integrins, activating intracellular fibrogenic cascades that contribute to fibrosis in response to pressure overload. Although subpopulations of fibroblast-like cells may exert important protective actions in both reparative and interstitial/perivascular fibrosis, ultimately fibrotic changes perturb systolic and diastolic function, and may play an important role in the pathogenesis of arrhythmias. This review article discusses the molecular mechanisms involved in the pathogenesis of cardiac fibrosis in various myocardial diseases, including myocardial infarction, heart failure with reduced or preserved ejection fraction, genetic cardiomyopathies, and diabetic heart disease. Development of fibrosis-targeting therapies for patients with myocardial diseases will require not only understanding of the functional pluralism of cardiac fibroblasts and dissection of the molecular basis for fibrotic remodelling, but also appreciation of the pathophysiologic heterogeneity of fibrosis-associated myocardial disease.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, 1300 Morris Park Avenue Forchheimer G46B, Bronx, NY 10461, USA
| |
Collapse
|
16
|
Xia N, Lu Y, Gu M, Li N, Liu M, Jiao J, Zhu Z, Li J, Li D, Tang T, Lv B, Nie S, Zhang M, Liao M, Liao Y, Yang X, Cheng X. A Unique Population of Regulatory T Cells in Heart Potentiates Cardiac Protection From Myocardial Infarction. Circulation 2020; 142:1956-1973. [PMID: 32985264 DOI: 10.1161/circulationaha.120.046789] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Regulatory T cells (Tregs), traditionally recognized as potent suppressors of immune response, are increasingly attracting attention because of a second major function: residing in parenchymal tissues and maintaining local homeostasis. However, the existence, unique phenotype, and function of so-called tissue Tregs in the heart remain unclear. METHODS In mouse models of myocardial infarction (MI), myocardial ischemia/reperfusion injury, or cardiac cryoinjury, the dynamic accumulation of Tregs in the injured myocardium was monitored. The bulk RNA sequencing was performed to analyze the transcriptomic characteristics of Tregs from the injured myocardium after MI or ischemia/reperfusion injury. Photoconversion, parabiosis, single-cell T-cell receptor sequencing, and adoptive transfer were applied to determine the source of heart Tregs. The involvement of the interleukin-33/suppression of tumorigenicity 2 axis and Sparc (secreted acidic cysteine-rich glycoprotein), a molecule upregulated in heart Tregs, was further evaluated in functional assays. RESULTS We showed that Tregs were highly enriched in the myocardium of MI, ischemia/reperfusion injury, and cryoinjury mice. Transcriptomic data revealed that Tregs isolated from the injured hearts had plenty of differentially expressed transcripts in comparison with their lymphoid counterparts, including heart-draining lymphoid nodes, with a phenotype of promoting infarct repair, indicating a unique characteristic. The heart Tregs were accumulated mainly because of recruitment from the circulating Treg pool, whereas local proliferation also contributed to their expansion. Moreover, a remarkable case of repeatedly detected T-cell receptor of heart Tregs, more than that of spleen Tregs, suggests a model of clonal expansion. Besides, HelioshighNrp-1high phenotype proved the mainly thymic origin of heart Tregs, with a small contribution of phenotypic conversion of conventional CD4+ T cells, proved by the analysis of T-cell receptor repertoires and conventional CD4+ T cells adoptive transfer experiments. The interleukin-33/suppression of tumorigenicity 2 axis was essential for sustaining heart Treg populations. Last, we demonstrated that Sparc, which was highly expressed by heart Tregs, acted as a critical factor to protect the heart against MI by increasing collagen content and boosting maturation in the infarct zone. CONCLUSIONS We identified and characterized a phenotypically and functionally unique population of heart Tregs that may lay the foundation to harness Tregs for cardioprotection in MI and other cardiac diseases.
Collapse
Affiliation(s)
- Ni Xia
- Department of Cardiology, Union Hospital, and Key Laboratory of Biological Targeted Therapy of the Ministry of Education (N.X., Y. Lu, M.G., N.L., M.L., J.J., Z.Z., J.L., D.L., T.T., B.L., S.N., M.Z., M.L., Y. Liao, X.C.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuzhi Lu
- Department of Cardiology, Union Hospital, and Key Laboratory of Biological Targeted Therapy of the Ministry of Education (N.X., Y. Lu, M.G., N.L., M.L., J.J., Z.Z., J.L., D.L., T.T., B.L., S.N., M.Z., M.L., Y. Liao, X.C.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Muyang Gu
- Department of Cardiology, Union Hospital, and Key Laboratory of Biological Targeted Therapy of the Ministry of Education (N.X., Y. Lu, M.G., N.L., M.L., J.J., Z.Z., J.L., D.L., T.T., B.L., S.N., M.Z., M.L., Y. Liao, X.C.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nana Li
- Department of Cardiology, Union Hospital, and Key Laboratory of Biological Targeted Therapy of the Ministry of Education (N.X., Y. Lu, M.G., N.L., M.L., J.J., Z.Z., J.L., D.L., T.T., B.L., S.N., M.Z., M.L., Y. Liao, X.C.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meilin Liu
- Department of Cardiology, Union Hospital, and Key Laboratory of Biological Targeted Therapy of the Ministry of Education (N.X., Y. Lu, M.G., N.L., M.L., J.J., Z.Z., J.L., D.L., T.T., B.L., S.N., M.Z., M.L., Y. Liao, X.C.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiao Jiao
- Department of Cardiology, Union Hospital, and Key Laboratory of Biological Targeted Therapy of the Ministry of Education (N.X., Y. Lu, M.G., N.L., M.L., J.J., Z.Z., J.L., D.L., T.T., B.L., S.N., M.Z., M.L., Y. Liao, X.C.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhengfeng Zhu
- Department of Cardiology, Union Hospital, and Key Laboratory of Biological Targeted Therapy of the Ministry of Education (N.X., Y. Lu, M.G., N.L., M.L., J.J., Z.Z., J.L., D.L., T.T., B.L., S.N., M.Z., M.L., Y. Liao, X.C.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyong Li
- Department of Cardiology, Union Hospital, and Key Laboratory of Biological Targeted Therapy of the Ministry of Education (N.X., Y. Lu, M.G., N.L., M.L., J.J., Z.Z., J.L., D.L., T.T., B.L., S.N., M.Z., M.L., Y. Liao, X.C.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dan Li
- Department of Cardiology, Union Hospital, and Key Laboratory of Biological Targeted Therapy of the Ministry of Education (N.X., Y. Lu, M.G., N.L., M.L., J.J., Z.Z., J.L., D.L., T.T., B.L., S.N., M.Z., M.L., Y. Liao, X.C.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Tang
- Department of Cardiology, Union Hospital, and Key Laboratory of Biological Targeted Therapy of the Ministry of Education (N.X., Y. Lu, M.G., N.L., M.L., J.J., Z.Z., J.L., D.L., T.T., B.L., S.N., M.Z., M.L., Y. Liao, X.C.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bingjie Lv
- Department of Cardiology, Union Hospital, and Key Laboratory of Biological Targeted Therapy of the Ministry of Education (N.X., Y. Lu, M.G., N.L., M.L., J.J., Z.Z., J.L., D.L., T.T., B.L., S.N., M.Z., M.L., Y. Liao, X.C.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaofang Nie
- Department of Cardiology, Union Hospital, and Key Laboratory of Biological Targeted Therapy of the Ministry of Education (N.X., Y. Lu, M.G., N.L., M.L., J.J., Z.Z., J.L., D.L., T.T., B.L., S.N., M.Z., M.L., Y. Liao, X.C.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Zhang
- Department of Cardiology, Union Hospital, and Key Laboratory of Biological Targeted Therapy of the Ministry of Education (N.X., Y. Lu, M.G., N.L., M.L., J.J., Z.Z., J.L., D.L., T.T., B.L., S.N., M.Z., M.L., Y. Liao, X.C.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengyang Liao
- Department of Cardiology, Union Hospital, and Key Laboratory of Biological Targeted Therapy of the Ministry of Education (N.X., Y. Lu, M.G., N.L., M.L., J.J., Z.Z., J.L., D.L., T.T., B.L., S.N., M.Z., M.L., Y. Liao, X.C.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuhua Liao
- Department of Cardiology, Union Hospital, and Key Laboratory of Biological Targeted Therapy of the Ministry of Education (N.X., Y. Lu, M.G., N.L., M.L., J.J., Z.Z., J.L., D.L., T.T., B.L., S.N., M.Z., M.L., Y. Liao, X.C.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangping Yang
- Department of Immunology (X.Y.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, and Key Laboratory of Biological Targeted Therapy of the Ministry of Education (N.X., Y. Lu, M.G., N.L., M.L., J.J., Z.Z., J.L., D.L., T.T., B.L., S.N., M.Z., M.L., Y. Liao, X.C.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
Hanna A, Shinde AV, Frangogiannis NG. Validation of diagnostic criteria and histopathological characterization of cardiac rupture in the mouse model of nonreperfused myocardial infarction. Am J Physiol Heart Circ Physiol 2020; 319:H948-H964. [PMID: 32886000 DOI: 10.1152/ajpheart.00318.2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
In patients with myocardial infarction (MI), cardiac rupture is an uncommon but catastrophic complication. In the mouse model of nonreperfused MI, reported rupture rates are highly variable and depend not only on the genetic background and sex of animals but also on the method used for documentation of rupture. In most studies, diagnosis of cardiac rupture is based on visual inspection during autopsy; however, criteria are poorly defined. We performed systematic histopathological analysis of whole hearts from C57BL/6J mice dying after nonreperfused MI and evaluated the reliability of autopsy-based criteria in identification of rupture. Moreover, we compared the cell biological environment of the infarct between rupture-related and rupture-independent deaths. Histopathological analysis documented rupture in 50% of mice dying during the first week post-MI. Identification of a gross rupture site was highly specific but had low sensitivity; in contrast, hemothorax had high sensitivity but low specificity. Mice with rupture had lower myofibroblast infiltration, accentuated macrophage influx, and a trend toward reduced collagen content in the infarct. Male mice had increased mortality and higher incidence of rupture. However, infarct myeloid cells harvested from male and female mice at the peak of the incidence of rupture had comparable inflammatory gene expression. In conclusion, the reliability of autopsy in documentation of rupture in infarcted mice is dependent on the specific criteria used. Macrophage-driven inflammation and reduced activation of collagen-secreting reparative myofibroblasts may be involved in the pathogenesis of post-MI cardiac rupture.NEW & NOTEWORTHY We show that cardiac rupture accounts for 50% of deaths in C57BL/6J mice undergoing nonreperfused myocardial infarction protocols. Overestimation of rupture events in published studies likely reflects the low specificity of hemothorax as a criterion for documentation of rupture. In contrast, identification of a gross rupture site has high specificity and low sensitivity. We also show that mice dying of rupture have increased macrophage influx and attenuated myofibroblast infiltration in the infarct. These findings are consistent with a role for perturbations in the balance between inflammatory and reparative responses in the pathogenesis of postinfarction cardiac rupture. We also report that the male predilection for rupture in infarcted mice is not associated with increased inflammatory activation of myeloid cells.
Collapse
Affiliation(s)
- Anis Hanna
- Division of Cardiology, Department of Medicine, The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York
| | - Arti V Shinde
- Division of Cardiology, Department of Medicine, The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York
| | - Nikolaos G Frangogiannis
- Division of Cardiology, Department of Medicine, The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
18
|
Swiprosin-1/EFhD-2 Expression in Cardiac Remodeling and Post-Infarct Repair: Effect of Ischemic Conditioning. Int J Mol Sci 2020; 21:ijms21093359. [PMID: 32397496 PMCID: PMC7247556 DOI: 10.3390/ijms21093359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 02/08/2023] Open
Abstract
Swiprosin-1 (EFhD2) is a molecule that triggers structural adaptation of isolated adult rat cardiomyocytes to cell culture conditions by initiating a process known as cell spreading. This process mimics central aspects of cardiac remodeling, as it occurs subsequent to myocardial infarction. However, expression of swiprosin-1 in cardiac tissue and its regulation in vivo has not yet been addressed. The expression of swiprosin-1 was analyzed in mice, rat, and pig hearts undergoing myocardial infarction or ischemia/reperfusion with or without cardiac protection by ischemic pre- and postconditioning. In mouse hearts, swiprosin-1 protein expression was increased after 4 and 7 days in myocardial infarct areas specifically in cardiomyocytes as verified by immunoblotting and histology. In rat hearts, swiprosin-1 mRNA expression was induced within 7 days after ischemia/reperfusion but this induction was abrogated by conditioning. As in cultured cardiomyocytes, the expression of swiprosin-1 was associated with a coinduction of arrestin-2, suggesting a common mechanism of regulation. Rno-miR-32-3p and rno-miR-34c-3p were associated with the regulation pattern of both molecules. Moreover, induction of swiprosin-1 and ssc-miR-34c was also confirmed in the infarct zone of pigs. In summary, our data show that up-regulation of swiprosin-1 appears in the postischemic heart during cardiac remodeling and repair in different species.
Collapse
|
19
|
Wang X, Shen X, Weil BR, Young RF, Canty JM, Qu J. Quantitative proteomic and phosphoproteomic profiling of ischemic myocardial stunning in swine. Am J Physiol Heart Circ Physiol 2020; 318:H1256-H1271. [PMID: 32223553 DOI: 10.1152/ajpheart.00713.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Despite decades of research on the pathophysiology of myocardial stunning, protein changes and/or phosphorylation status underlying alterations in cardiac function/structure remain inadequately understood. Here, we utilized comprehensive and quantitative proteomic and phosphoproteomic approaches to explore molecular mechanisms of myocardial stunning in swine. The closed-chest swine (n = 5 pigs) were subjected to a 10-min left anterior descending coronary artery (LAD) occlusion producing regional myocardial stunning. Tissues from the ischemic LAD region and a remote nonischemic area of the left ventricle were collected 1 h after reperfusion. Ion current-based proteomics (IonStar) and quantitative phosphoproteomics were employed in parallel to identify alterations in protein level and site-specific phosphorylation changes. A novel swine heart protein database exhibiting high accuracy and low redundancy was developed here to facilitate comprehensive study. Further informatic investigations identified potential protein-protein interactions in stunned myocardium. In total, we quantified 2,099 protein groups and 4,699 phosphorylation sites with only 0.4% missing values. Proteomic analyses revealed downregulation of contractile function and extracellular matrix remodeling. Meanwhile, alterations in phosphorylation linked with contractile dysfunction and apoptotic cell death were uncovered. NetworKIN/STRING analysis predicted regulatory kinases responsible for altered phosphosites, such as protein kinase C-mediated phosphorylation of cardiac troponin I-S199 and CaMKII-mediated phosphorylation of phospholamban-T17. In summary, the ion current-based proteomics and phosphoproteomics reliably identified novel alterations in protein content and phosphorylation contributing to contractile dysfunction, extracellular matrix (ECM) damage, and programmed cell death in stunned myocardium, which corroborate well with our physiological observations. Moreover, this work developed a comprehensive database of the swine heart proteome, a highly valuable resource for future translational research in porcine models with cardiovascular diseases.NEW & NOTEWORTHY We first used ion current-based proteomics and phosphoproteomics to reliably identify novel alterations in protein expression and phosphorylation contributing to contractile dysfunction, extracellular matrix (ECM) damage, and programmed cell death in stunned myocardium and developed a comprehensive swine heart-specific proteome database, which provides a valuable resource for future research in porcine models of cardiovascular diseases.
Collapse
Affiliation(s)
- Xue Wang
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York.,New York State Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, Buffalo, New York
| | - Xiaomeng Shen
- New York State Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, Buffalo, New York.,Department of Biochemistry, University at Buffalo, Buffalo, New York
| | - Brian R Weil
- Department of Physiology and Biophysics, University at Buffalo, Buffalo, New York
| | - Rebeccah F Young
- Clinical and Translational Research Center, University at Buffalo, Buffalo, New York.,Division of Cardiovascular Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| | - John M Canty
- Department of Physiology and Biophysics, University at Buffalo, Buffalo, New York.,Veterans Affairs Western New York Healthcare System, Buffalo, New York.,Clinical and Translational Research Center, University at Buffalo, Buffalo, New York.,Division of Cardiovascular Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| | - Jun Qu
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York.,New York State Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, Buffalo, New York.,Department of Biochemistry, University at Buffalo, Buffalo, New York.,Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, New York
| |
Collapse
|
20
|
Petz A, Grandoch M, Gorski DJ, Abrams M, Piroth M, Schneckmann R, Homann S, Müller J, Hartwig S, Lehr S, Yamaguchi Y, Wight TN, Gorressen S, Ding Z, Kötter S, Krüger M, Heinen A, Kelm M, Gödecke A, Flögel U, Fischer JW. Cardiac Hyaluronan Synthesis Is Critically Involved in the Cardiac Macrophage Response and Promotes Healing After Ischemia Reperfusion Injury. Circ Res 2020; 124:1433-1447. [PMID: 30916618 DOI: 10.1161/circresaha.118.313285] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
RATIONALE Immediate changes in the ECM (extracellular matrix) microenvironment occur after myocardial ischemia and reperfusion (I/R) injury. OBJECTIVE Aim of this study was to unravel the role of the early hyaluronan (HA)-rich ECM after I/R. METHODS AND RESULTS Genetic deletion of Has2 and Has1 was used in a murine model of cardiac I/R. Chemical exchange saturation transfer imaging was adapted to image cardiac ECM post-I/R. Of note, the cardiac chemical exchange saturation transfer signal was severely suppressed by Has2 deletion and pharmacological inhibition of HA synthesis 24 hours after I/R. Has2 KO ( Has2 deficient) mice showed impaired hemodynamic function suggesting a protective role for endogenous HA synthesis. In contrast to Has2 deficiency, Has1-deficient mice developed no specific phenotype compared with control post-I/R. Importantly, in Has2 KO mice, cardiac macrophages were diminished after I/R as detected by 19F MRI (magnetic resonance imaging) of perfluorcarbon-labeled immune cells, Mac-2/Galectin-3 immunostaining, and FACS (fluorescence-activated cell sorting) analysis (CD45+CD11b+Ly6G-CD64+F4/80+cells). In contrast to macrophages, cardiac Ly6Chigh and Ly6Clow monocytes were unaffected post-I/R compared with control mice. Mechanistically, inhibition of HA synthesis led to increased macrophage apoptosis in vivo and in vitro. In addition, α-SMA (α-smooth muscle actin)-positive cells were reduced in the infarcted myocardium and in the border zone. In vitro, the myofibroblast response as measured by Acta2 mRNA expression was reduced by inhibition of HA synthesis and of CD44 signaling. Furthermore, Has2 KO fibroblasts were less able to contract collagen gels in vitro. The effects of HA/CD44 on fibroblasts and macrophages post-I/R might also affect intercellular cross talk because cardiac fibroblasts were activated by monocyte/macrophages and, in turn, protected macrophages from apoptosis. CONCLUSIONS Increased HA synthesis contributes to postinfarct healing by supporting macrophage survival and by promoting the myofibroblast response. Additionally, imaging of cardiac HA by chemical exchange saturation transfer post-I/R might have translational value.
Collapse
Affiliation(s)
- Anne Petz
- From the Institut für Pharmakologie und Klinische Pharmakologie (A.P., M.G., D.J.G., M.A., M.P., R.S., S.H., J.M., S.G., J.W.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany.,CARID, Cardiovascular Research Institute Düsseldorf (A.P., M.G., D.J.G., M.A., M.P., R.S., S.H., J.M., S.G., M. Kelm, A.G., U.F., J.W.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Maria Grandoch
- From the Institut für Pharmakologie und Klinische Pharmakologie (A.P., M.G., D.J.G., M.A., M.P., R.S., S.H., J.M., S.G., J.W.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany.,CARID, Cardiovascular Research Institute Düsseldorf (A.P., M.G., D.J.G., M.A., M.P., R.S., S.H., J.M., S.G., M. Kelm, A.G., U.F., J.W.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Daniel J Gorski
- From the Institut für Pharmakologie und Klinische Pharmakologie (A.P., M.G., D.J.G., M.A., M.P., R.S., S.H., J.M., S.G., J.W.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany.,CARID, Cardiovascular Research Institute Düsseldorf (A.P., M.G., D.J.G., M.A., M.P., R.S., S.H., J.M., S.G., M. Kelm, A.G., U.F., J.W.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Marcel Abrams
- From the Institut für Pharmakologie und Klinische Pharmakologie (A.P., M.G., D.J.G., M.A., M.P., R.S., S.H., J.M., S.G., J.W.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany.,CARID, Cardiovascular Research Institute Düsseldorf (A.P., M.G., D.J.G., M.A., M.P., R.S., S.H., J.M., S.G., M. Kelm, A.G., U.F., J.W.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Marco Piroth
- From the Institut für Pharmakologie und Klinische Pharmakologie (A.P., M.G., D.J.G., M.A., M.P., R.S., S.H., J.M., S.G., J.W.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany.,CARID, Cardiovascular Research Institute Düsseldorf (A.P., M.G., D.J.G., M.A., M.P., R.S., S.H., J.M., S.G., M. Kelm, A.G., U.F., J.W.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Rebekka Schneckmann
- From the Institut für Pharmakologie und Klinische Pharmakologie (A.P., M.G., D.J.G., M.A., M.P., R.S., S.H., J.M., S.G., J.W.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany.,CARID, Cardiovascular Research Institute Düsseldorf (A.P., M.G., D.J.G., M.A., M.P., R.S., S.H., J.M., S.G., M. Kelm, A.G., U.F., J.W.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Susanne Homann
- From the Institut für Pharmakologie und Klinische Pharmakologie (A.P., M.G., D.J.G., M.A., M.P., R.S., S.H., J.M., S.G., J.W.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany.,CARID, Cardiovascular Research Institute Düsseldorf (A.P., M.G., D.J.G., M.A., M.P., R.S., S.H., J.M., S.G., M. Kelm, A.G., U.F., J.W.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Julia Müller
- From the Institut für Pharmakologie und Klinische Pharmakologie (A.P., M.G., D.J.G., M.A., M.P., R.S., S.H., J.M., S.G., J.W.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany.,CARID, Cardiovascular Research Institute Düsseldorf (A.P., M.G., D.J.G., M.A., M.P., R.S., S.H., J.M., S.G., M. Kelm, A.G., U.F., J.W.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Sonja Hartwig
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, Germany (S.H., S.L.).,German Center for Diabetes Research, München-Neuherberg, Germany (S.H., S.L.)
| | - Stefan Lehr
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at the Heinrich-Heine-University Duesseldorf, Leibniz Center for Diabetes Research, Germany (S.H., S.L.).,German Center for Diabetes Research, München-Neuherberg, Germany (S.H., S.L.)
| | - Yu Yamaguchi
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA (Y.Y.)
| | - Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA (T.N.W.)
| | - Simone Gorressen
- From the Institut für Pharmakologie und Klinische Pharmakologie (A.P., M.G., D.J.G., M.A., M.P., R.S., S.H., J.M., S.G., J.W.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany.,CARID, Cardiovascular Research Institute Düsseldorf (A.P., M.G., D.J.G., M.A., M.P., R.S., S.H., J.M., S.G., M. Kelm, A.G., U.F., J.W.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Zhaoping Ding
- Institut für Molekulare Kardiologie (Z.D., U.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Sebastian Kötter
- Institut für Herz- und Kreislaufphysiologie (S.K., M. Krüger, A.H., A.G.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Martina Krüger
- Institut für Herz- und Kreislaufphysiologie (S.K., M. Krüger, A.H., A.G.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Andre Heinen
- Institut für Herz- und Kreislaufphysiologie (S.K., M. Krüger, A.H., A.G.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Malte Kelm
- CARID, Cardiovascular Research Institute Düsseldorf (A.P., M.G., D.J.G., M.A., M.P., R.S., S.H., J.M., S.G., M. Kelm, A.G., U.F., J.W.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany.,Klinik für Kardiologie, Pneumologie und Angiologie (M. Kelm, U.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Axel Gödecke
- CARID, Cardiovascular Research Institute Düsseldorf (A.P., M.G., D.J.G., M.A., M.P., R.S., S.H., J.M., S.G., M. Kelm, A.G., U.F., J.W.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany.,Institut für Herz- und Kreislaufphysiologie (S.K., M. Krüger, A.H., A.G.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Ulrich Flögel
- CARID, Cardiovascular Research Institute Düsseldorf (A.P., M.G., D.J.G., M.A., M.P., R.S., S.H., J.M., S.G., M. Kelm, A.G., U.F., J.W.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany.,Institut für Molekulare Kardiologie (Z.D., U.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany.,Klinik für Kardiologie, Pneumologie und Angiologie (M. Kelm, U.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| | - Jens W Fischer
- From the Institut für Pharmakologie und Klinische Pharmakologie (A.P., M.G., D.J.G., M.A., M.P., R.S., S.H., J.M., S.G., J.W.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany.,CARID, Cardiovascular Research Institute Düsseldorf (A.P., M.G., D.J.G., M.A., M.P., R.S., S.H., J.M., S.G., M. Kelm, A.G., U.F., J.W.F.), University Hospital, Heinrich-Heine-University Düsseldorf, Germany
| |
Collapse
|
21
|
Affiliation(s)
- Thomas N Wight
- From the Matrix Biology Program, Benaroya Research Institute, Seattle, WA
| | - Robert B Vernon
- From the Matrix Biology Program, Benaroya Research Institute, Seattle, WA
| |
Collapse
|
22
|
The Non-Fibrillar Side of Fibrosis: Contribution of the Basement Membrane, Proteoglycans, and Glycoproteins to Myocardial Fibrosis. J Cardiovasc Dev Dis 2019; 6:jcdd6040035. [PMID: 31547598 PMCID: PMC6956278 DOI: 10.3390/jcdd6040035] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 02/07/2023] Open
Abstract
The extracellular matrix (ECM) provides structural support and a microenvironmentfor soluble extracellular molecules. ECM is comprised of numerous proteins which can be broadly classified as fibrillar (collagen types I and III) and non-fibrillar (basement membrane, proteoglycans, and glycoproteins). The basement membrane provides an interface between the cardiomyocytes and the fibrillar ECM, while proteoglycans sequester soluble growth factors and cytokines. Myocardial fibrosis was originally only linked to accumulation of fibrillar collagens, but is now recognized as the expansion of the ECM including the non-fibrillar ECM proteins. Myocardial fibrosis can be reparative to replace the lost myocardium (e.g., ischemic injury or myocardial infarction), or can be reactive resulting from pathological activity of fibroblasts (e.g., dilated or hypertrophic cardiomyopathy). Contribution of fibrillar collagens to fibrosis is well studied, but the role of the non-fibrillar ECM proteins has remained less explored. In this article, we provide an overview of the contribution of the non-fibrillar components of the extracellular space of the heart to highlight the potential significance of these molecules in fibrosis, with direct evidence for some, although not all of these molecules in their direct contribution to fibrosis.
Collapse
|
23
|
Fang D, Lai Z, Wang Y. Overexpression of Biglycan is Associated with Resistance to Rapamycin in Human WERI-Rb-1 Retinoblastoma Cells by Inducing the Activation of the Phosphatidylinositol 3-Kinases (PI3K)/Akt/Nuclear Factor kappa B (NF-κB) Signaling Pathway. Med Sci Monit 2019; 25:6639-6648. [PMID: 31483776 PMCID: PMC6743380 DOI: 10.12659/msm.915075] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Biglycan (BGN) is an extracellular matrix (ECM) protein that regulates the growth of epithelial cells. The mammalian target of rapamycin (mTOR) inhibitor, rapamycin, is a treatment for advanced retinoblastoma. This study aimed to investigate the effects of expression of BGN on the response of human WERI-Rb-1 retinoblastoma cells to rapamycin and to investigate the associated signaling pathways. Material/Methods BGN gene expression was induced in human WERI-Rb-1 retinoblastoma cells, which were incubated with rapamycin at doses of 0, 5, 10, 20, 30, and 50 μg/ml. Cells were treated with the PI3K/Akt pathway inhibitor, LY294002. The MTT assay determined the rate of cell inhibition. Real-time polymerase chain reaction (RT-PCR) was performed to measure BGN gene expression using RT2-PCR. Western blot detected the protein levels of BGN, p-PI3K, p-Akt, nuclear NF-κB, and p65. Results Rapamycin impaired cell growth, induced cell apoptosis, and suppressed the expression levels of p-PI3K, p-Akt, nuclear NF-κB, and p65. Overexpression of the BGN gene restored growth potential and inhibited apoptosis and was associated with the activation of the PI3K/Akt-mediated NF-κB pathway. In cells that overexpressed BGN, inhibition of the PI3K/Akt pathway by LY294002 increased the sensitivity of human WERI-Rb-1 retinoblastoma cells to rapamycin. Conclusions Overexpression of BGN induced rapamycin resistance in WERI-Rb-1 retinoblastoma cells by activating PI3K/Akt/NF-κB signaling.
Collapse
Affiliation(s)
- Dong Fang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Zhaoguang Lai
- Department of Ophthalmology, Peoples' Hospital of Guangxi Autonomous Region, Nanning, Guangxi, China (mainland)
| | - Yan Wang
- Department of Ophthalmology, 521 Hospital of Xi'an Weapon Industry, Xi'an, Shaanxi, China (mainland)
| |
Collapse
|
24
|
Underhill LA, Avalos N, Tucker R, Zhang Z, Messerlian G, Lechner B. Serum Decorin and Biglycan as Potential Biomarkers to Predict PPROM in Early Gestation. Reprod Sci 2019:1933719119831790. [PMID: 30895897 DOI: 10.1177/1933719119831790] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Preterm birth is a leading cause of neonatal mortality in the US and globally, with preterm premature rupture of fetal membranes (PPROM) accounting for one third of preterm births. Currently no predictive diagnostics are available to precisely assess risk and potentially reduce the incidence of PPROM. Bigycan and decorin, the main proteoglycans present in human fetal membranes, are involved in the physiological maturation of fetal membranes as well as in the pathophysiology of preterm birth. The serum protein sex hormone-binding globulin (SHBG) has recently been identified as a predictor of spontaneous preterm birth. We hypothesize that the balance between serum decorin and biglycan on one hand and SHBG on the other hand may provide insight into the status of the fetal membranes in early pregnancy, thereby predicting PPROM prior to symptoms. Using chart review, 18 patients with confirmed cases of PPROM were identified from 2013-2016. Second trimester residual serum was retreived from freezer storage for these cases along with 5 matched controls for each case. The biomarkers biglycan, decorin and SHBG were analyzed first separately, then in combination to determine their ability to predict PPROM. The predictive score for the combined model displays an AUC = 0.774. The ROC curve of the predicted score has an optimal threshold of 0.238 and a sensitivity and specificity of 0.72 and 0.84 respectively. This prenatal serum panel is a promising serum screening-based biochemical model to predict PPROM in asymptomatic women.
Collapse
Affiliation(s)
- Lori A Underhill
- 1 Warren Alpert Medical School at Brown University, Providence, RI ,USA
- 2 Department of Pediatrics, Women and Infants Hospital, Providence, RI, USA
| | - Nora Avalos
- 2 Department of Pediatrics, Women and Infants Hospital, Providence, RI, USA
| | - Richard Tucker
- 2 Department of Pediatrics, Women and Infants Hospital, Providence, RI, USA
| | - Zheng Zhang
- 3 School of Public Health, Brown University, Providence, RI, USA
| | - Geralyn Messerlian
- 2 Department of Pediatrics, Women and Infants Hospital, Providence, RI, USA
| | - Beatrice Lechner
- 1 Warren Alpert Medical School at Brown University, Providence, RI ,USA
- 2 Department of Pediatrics, Women and Infants Hospital, Providence, RI, USA
| |
Collapse
|
25
|
Inflammation and fibrosis in murine models of heart failure. Basic Res Cardiol 2019; 114:19. [PMID: 30887214 DOI: 10.1007/s00395-019-0722-5] [Citation(s) in RCA: 238] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 02/12/2019] [Indexed: 02/07/2023]
Abstract
Heart failure is a consequence of various cardiovascular diseases and associated with poor prognosis. Despite progress in the treatment of heart failure in the past decades, prevalence and hospitalisation rates are still increasing. Heart failure is typically associated with cardiac remodelling. Here, inflammation and fibrosis are thought to play crucial roles. During cardiac inflammation, immune cells invade the cardiac tissue and modulate tissue-damaging responses. Cardiac fibrosis, however, is characterised by an increased amount and a disrupted composition of extracellular matrix proteins. As evidence exists that cardiac inflammation and fibrosis are potentially reversible in experimental and clinical set ups, they are interesting targets for innovative heart failure treatments. In this context, animal models are important as they mimic clinical conditions of heart failure patients. The advantages of mice in this respect are short generation times and genetic modifications. As numerous murine models of heart failure exist, the selection of a proper disease model for a distinct research question is demanding. To facilitate this selection, this review aims to provide an overview about the current understanding of the pathogenesis of cardiac inflammation and fibrosis in six frequently used murine models of heart failure. Hence, it compares the models of myocardial infarction with or without reperfusion, transverse aortic constriction, chronic subjection to angiotensin II or deoxycorticosterone acetate, and coxsackievirus B3-induced viral myocarditis in this context. It furthermore provides information about the clinical relevance and the limitations of each model, and, if applicable, about the recent advancements in their methodological proceedings.
Collapse
|
26
|
Functional and structural studies of tolloid-like 1 mutants associated with atrial-septal defect 6. Biosci Rep 2019; 39:BSR20180270. [PMID: 30538173 PMCID: PMC6328869 DOI: 10.1042/bsr20180270] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 11/07/2018] [Accepted: 11/29/2018] [Indexed: 11/23/2022] Open
Abstract
Inactive mammalian tolloid-like 1 (tll1) and mutations detected in tolloid-like 1 (TLL1) have been linked to the lack of the heart septa formation in mice and to a similar human inborn condition called atrial-septal defect 6 (ASD6; OMIM 613087, formerly ASD II). Previously, we reported four point mutations in TLL1 found in approximately 20% of ASD6 patients. Three mutations in the coding sequence were M182L, V238A, and I629V. In this work, we present the effects of these mutations on TLL1 function. Three recombinant cDNA constructs carrying the mutations and one wild-type construct were prepared and then expressed in HT-1080 cells. Corresponding recombinant proteins were analyzed for their metalloendopeptidase activity using a native substrate, chordin. The results of these assays demonstrated that in comparison with the native TLL1, mutants cleaved chordin and procollagen I at significantly lower rates. CD analyses revealed significant structural differences between the higher order structure of wild-type and mutant variants. Moreover, biosensor-based assays of binding interactions between TLL1 variants and chordin demonstrated a significant decrease in the binding affinities of the mutated variants. The results from this work indicate that mutations detected in TLL1 of ASD6 patients altered its metalloendopeptidase activity, structure, and substrate-binding properties, thereby suggesting a possible pathomechanism of ASD6.
Collapse
|
27
|
Christensen G, Herum KM, Lunde IG. Sweet, yet underappreciated: Proteoglycans and extracellular matrix remodeling in heart disease. Matrix Biol 2019; 75-76:286-299. [DOI: 10.1016/j.matbio.2018.01.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/22/2017] [Accepted: 01/01/2018] [Indexed: 12/20/2022]
|
28
|
Emerging roles of proteoglycans in cardiac remodeling. Int J Cardiol 2018; 278:192-198. [PMID: 30528626 DOI: 10.1016/j.ijcard.2018.11.125] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 11/12/2018] [Accepted: 11/27/2018] [Indexed: 02/07/2023]
Abstract
Cardiac remodeling is the response of the heart to a range of pathological stimuli. Cardiac remodeling is initially adaptive; however, if sustained, it ultimately causes adverse clinical outcomes. Cardiomyocyte loss or hypertrophy, inflammation and fibrosis are hallmarks of cardiac remodeling. Proteoglycans, which are composed of glycosaminoglycans and a core protein, are a non-structural component of the extracellular matrix. The lack of proteoglycans results in cardiovascular defects during development. Moreover, emerging evidence has indicated that proteoglycans act as significant modifiers in ischemia and pressure overload-related cardiac remodeling. Proteoglycans may also provide novel therapeutic strategies for further improvement in the prognosis of cardiovascular diseases.
Collapse
|
29
|
Precursor proadrenomedullin influences cardiomyocyte survival and local inflammation related to myocardial infarction. Proc Natl Acad Sci U S A 2018; 115:E8727-E8736. [PMID: 30166452 DOI: 10.1073/pnas.1721635115] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Increased adrenomedullin (ADM) levels are associated with various cardiac diseases such as myocardial infarction (MI). ADM is cleaved off from the full-length precursor protein proadrenomedullin (ProADM) during its posttranslational processing. To date, no biological effect of ProADM is reported, while ADM infusion leads to antiapoptotic effects and improved cardiac function. Using an MI mouse model, we found an induction of ProADM gene as well as protein expression during the early phase of MI. This was accompanied by apoptosis and increasing inflammation, which substantially influence the post-MI remodeling processes. Simulating ischemia in vitro, we demonstrate that ProADM expression was increased in cardiomyocytes and cardiac fibroblasts. Subsequently, we treated ischemic cardiomyocytes with either ProADM or ADM and found that both proteins increased survival. This effect was diminishable by blocking the ADM1 receptor. To investigate whether ProADM and ADM play a role in the regulation of cardiac inflammation, we analyzed chemokine expression after treatment of cells with both proteins. While ProADM induced an expression of proinflammatory cytokines, thus promoting inflammation, ADM reduced chemokine expression. On leukocytes, both proteins repressed chemokine expression, revealing antiinflammatory effects. However, ProADM but not ADM dampened concurrent activation of leukocytes. Our data show that the full-length precursor ProADM is biologically active by reducing apoptosis to a similar extent as ADM. We further assume that ProADM induces local inflammation in affected cardiac tissue but attenuates exaggerated inflammation, whereas ADM has low impact. Our data suggest that both proteins are beneficial during MI by influencing apoptosis and inflammation.
Collapse
|
30
|
Frangogiannis NG. Cardiac fibrosis: Cell biological mechanisms, molecular pathways and therapeutic opportunities. Mol Aspects Med 2018; 65:70-99. [PMID: 30056242 DOI: 10.1016/j.mam.2018.07.001] [Citation(s) in RCA: 521] [Impact Index Per Article: 86.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 07/23/2018] [Indexed: 12/13/2022]
Abstract
Cardiac fibrosis is a common pathophysiologic companion of most myocardial diseases, and is associated with systolic and diastolic dysfunction, arrhythmogenesis, and adverse outcome. Because the adult mammalian heart has negligible regenerative capacity, death of a large number of cardiomyocytes results in reparative fibrosis, a process that is critical for preservation of the structural integrity of the infarcted ventricle. On the other hand, pathophysiologic stimuli, such as pressure overload, volume overload, metabolic dysfunction, and aging may cause interstitial and perivascular fibrosis in the absence of infarction. Activated myofibroblasts are the main effector cells in cardiac fibrosis; their expansion following myocardial injury is primarily driven through activation of resident interstitial cell populations. Several other cell types, including cardiomyocytes, endothelial cells, pericytes, macrophages, lymphocytes and mast cells may contribute to the fibrotic process, by producing proteases that participate in matrix metabolism, by secreting fibrogenic mediators and matricellular proteins, or by exerting contact-dependent actions on fibroblast phenotype. The mechanisms of induction of fibrogenic signals are dependent on the type of primary myocardial injury. Activation of neurohumoral pathways stimulates fibroblasts both directly, and through effects on immune cell populations. Cytokines and growth factors, such as Tumor Necrosis Factor-α, Interleukin (IL)-1, IL-10, chemokines, members of the Transforming Growth Factor-β family, IL-11, and Platelet-Derived Growth Factors are secreted in the cardiac interstitium and play distinct roles in activating specific aspects of the fibrotic response. Secreted fibrogenic mediators and matricellular proteins bind to cell surface receptors in fibroblasts, such as cytokine receptors, integrins, syndecans and CD44, and transduce intracellular signaling cascades that regulate genes involved in synthesis, processing and metabolism of the extracellular matrix. Endogenous pathways involved in negative regulation of fibrosis are critical for cardiac repair and may protect the myocardium from excessive fibrogenic responses. Due to the reparative nature of many forms of cardiac fibrosis, targeting fibrotic remodeling following myocardial injury poses major challenges. Development of effective therapies will require careful dissection of the cell biological mechanisms, study of the functional consequences of fibrotic changes on the myocardium, and identification of heart failure patient subsets with overactive fibrotic responses.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, 1300 Morris Park Avenue, Forchheimer G46B, Bronx, NY, 10461, USA.
| |
Collapse
|
31
|
Liu B, Xu T, Xu X, Cui Y, Xing X. Biglycan promotes the chemotherapy resistance of colon cancer by activating NF-κB signal transduction. Mol Cell Biochem 2018; 449:285-294. [PMID: 29761248 DOI: 10.1007/s11010-018-3365-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 05/02/2018] [Indexed: 01/05/2023]
Abstract
Biglycan (BGN) is overexpressed in cancer stem cells of colon cancer and induces the activation of NF-κB pathway which contributes to the chemotherapy resistance of diverse cancer types. Therefore, we hypothesized that the overexpression of BGN also promoted the development of multiple drug resistance (MDR) in colon cancer via NF-κB pathway. The expression of BGN was bilaterally modulated in colon cancer cell lines HT-29 and SW-480 and the effect of treatments on the cell proliferation and resistance to 5-FU was assessed. Moreover, the role of NF-κB signaling in the BGN-mediated formation of MDR was further investigated by subjecting BGN-overexpressed SW-480 cells to the co-treatment of chemo-agents and NF-κB inhibitor, PDTC. The inhibition of BGN expression decreased the proliferation potential of HT-29 cells while the induction of BGN expression increased the potential of SW-480 cells. BGN knockdown increased HT-29 cells' sensitivity to 5-FU, represented by the lower colony number and higher apoptotic rate. To the contrary, BGN overexpression promoted the resistance of SW-480 cells to 5-FU. The effect of BGN modulation on colon cancer cells was associated with the changes in apoptosis and NF-κB pathways: BGN inhibition increased the expressions of pro-apoptosis indicators and suppressed NF-κB pathway activity while BGN overexpression had the opposite effect. It was also found that the BGN-mediated formation of MDR was impaired when NF-κB pathway was blocked. Findings outlined in the current study showed that BGN contributed to the formation of chemotherapy resistance in colon cancer cells by activating NF-κB signaling.
Collapse
Affiliation(s)
- Bin Liu
- Department of Hematology and Breast Cancer, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, 44 Xiaoheyan Road, Shenyang, 110042, People's Republic of China
| | - Tonghong Xu
- Department of Hematology and Breast Cancer, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, 44 Xiaoheyan Road, Shenyang, 110042, People's Republic of China
| | - Xinning Xu
- Department of Hematology and Breast Cancer, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, 44 Xiaoheyan Road, Shenyang, 110042, People's Republic of China
| | - Yuzhu Cui
- Department of Hematology and Breast Cancer, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, 44 Xiaoheyan Road, Shenyang, 110042, People's Republic of China
| | - Xiaojing Xing
- Department of Hematology and Breast Cancer, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, 44 Xiaoheyan Road, Shenyang, 110042, People's Republic of China.
| |
Collapse
|
32
|
Fischer JW. Role of hyaluronan in atherosclerosis: Current knowledge and open questions. Matrix Biol 2018; 78-79:324-336. [PMID: 29510229 DOI: 10.1016/j.matbio.2018.03.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 02/20/2018] [Accepted: 03/01/2018] [Indexed: 12/21/2022]
Abstract
Hyaluronan (HA), HA synthases (HAS) and HA receptors are expressed during the progression of atherosclerotic plaques. HA is thought to promote the activated phenotype of local vascular smooth muscle cells characterized by increased migration, proliferation and matrix synthesis. Furthermore, HA may modulate the immune response by increasing macrophage retention and by promoting the polarization of Th1 cells that enhance macrophage driven inflammation as well. The pro-atherosclerotic functions of HA are opposed by the presence of HA in the glycocalyx where it critically contributes to anti-thrombotic and anti-inflammatory function of the glycocalyx. Patients with atherosclerosis often are affected by comorbidities among them diabetes mellitus type 2 and inflammatory comorbidities. Diabetes mellitus type 2 likely has close interrelations to HA synthesis in atherosclerosis because the activity and transcription of HA synthases are sensitive to the intracellular glucose metabolism, which determines the substrate availability and the posttranslational modifications of HA synthases. The pro-inflammatory comorbidities aggravate the course of atherosclerosis and will affect the expression of the genes related to HA biosynthesis, -degradation, HA-matrix assembly or signaling. One example being the induction of HAS3 by interleukin-1β and other cytokines. Furthermore complications of atherosclerosis such as the healing after myocardial infarction also involve HA responses.
Collapse
Affiliation(s)
- Jens W Fischer
- Institut für Pharmakologie und Klinische Pharmakologie, University Hospital, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany; CARID, Cardiovascular Research Institute Düsseldorf, University Hospital, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
33
|
Azimzadeh O, Tapio S. Proteomics landscape of radiation-induced cardiovascular disease: somewhere over the paradigm. Expert Rev Proteomics 2017; 14:987-996. [PMID: 28976223 DOI: 10.1080/14789450.2017.1388743] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Epidemiological studies clearly show that thoracic or whole body exposure to ionizing radiation increases the risk of cardiac morbidity and mortality. Radiation-induced cardiovascular disease (CVD) has been intensively studied during the last ten years but the underlying molecular mechanisms are still poorly understood. Areas covered: Heart proteomics is a powerful tool holding promise for the future research. The central focus of this review is to compare proteomics data on radiation-induced CVD with data arising from proteomics of healthy and diseased cardiac tissue in general. In this context we highlight common and unique features of radiation-related and other heart pathologies. Future prospects and challenges of the field are discussed. Expert commentary: Data from comprehensive cardiac proteomics have deepened the knowledge of molecular mechanisms involved in radiation-induced cardiac dysfunction. State-of-the-art proteomics has the potential to identify novel diagnostic and therapeutic markers of this disease.
Collapse
Affiliation(s)
- Omid Azimzadeh
- a Institute of Radiation Biology , Helmholtz Zentrum München, German Research Center for Environmental Health GmbH , Neuherberg , Germany
| | - Soile Tapio
- a Institute of Radiation Biology , Helmholtz Zentrum München, German Research Center for Environmental Health GmbH , Neuherberg , Germany
| |
Collapse
|
34
|
Osteoglycin prevents the development of age-related diastolic dysfunction during pressure overload by reducing cardiac fibrosis and inflammation. Matrix Biol 2017; 66:110-124. [PMID: 28958774 DOI: 10.1016/j.matbio.2017.09.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 09/15/2017] [Accepted: 09/17/2017] [Indexed: 12/17/2022]
Abstract
The small leucine-rich proteoglycan osteoglycin has been implicated in matrix homeostasis in different organs, including the ischemic heart. However, whether osteoglycin modulates cardiac hypertrophy, fibrosis or inflammation in hypertensive heart disease and during aging remains unknown. Angiotensin-II-induced pressure overload increases cardiac osteoglycin expression, concomitant with the onset of inflammation and extracellular matrix deposition. Interestingly aging led to decreased cardiac levels of osteoglycin, yet absence of osteoglycin did not affect organ structure or cardiac function up to the age of 18months. However, Angiotensin-II infusion in combination with aging resulted in exaggerated cardiac fibrosis and inflammation in the osteoglycin null mice as compared to wild-type mice, resulting in increased diastolic dysfunction as determined by magnetic resonance imaging. In vitro, stimulation of bone marrow derived macrophages from osteoglycin null mice with Angiotensin-II resulted in significantly higher levels of ICAM-1 as well as pro-inflammatory cytokines and chemokines IL-1β and MCP-1 as compared to WT cells. Further, stimulation of human cardiac fibroblasts with osteoglycin reduced cell proliferation and inhibited TGF-β induced collagen gene expression. In mouse cardiac tissue, osteoglycin expression inversely correlated with TGF-β expression and in cardiac biopsies of aortic stenosis patients, osteoglycin expression is significantly higher than in control biopsies. Interestingly, osteoglycin levels were higher in patients with less severe myocardial fibrosis and overall in the aortic stenosis patients osteoglycin levels negatively correlated with collagen content in the myocardium. In conclusion, osteoglycin expression is increased in the heart in response to pressure overload and its absence results in increased cardiac inflammation and fibrosis resulting in increased diastolic dysfunction.
Collapse
|
35
|
Nagaraju CK, Dries E, Popovic N, Singh AA, Haemers P, Roderick HL, Claus P, Sipido KR, Driesen RB. Global fibroblast activation throughout the left ventricle but localized fibrosis after myocardial infarction. Sci Rep 2017; 7:10801. [PMID: 28883544 PMCID: PMC5589875 DOI: 10.1038/s41598-017-09790-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/28/2017] [Indexed: 11/09/2022] Open
Abstract
Fibroblast (Fb) differentiation and interstitial fibrosis contribute to cardiac remodeling and loss of function after myocardial infarction (MI). We investigated regional presence and regulation of fibrosis in a pig MI model. In vivo analysis of regional function and perfusion defined three regions: the scar, the myocardium adjacent to the scar (MIadjacent, reduced function, reduced perfusion reserve), and the remote myocardium (MIremote, minimal functional deficit, maintained perfusion). Interstitial and perivascular fibrosis, and increase of collagen type I, was only observed in the MIadjacent. Fb activated protein-alpha (FAP-α) was enriched in MIadjacent compared to MIremote. TGF-β1, which triggers Fb differentiation, was upregulated in both MIadjacent and MIremote, whereas lysyl oxidase, a regulator of collagen cross-linking, and the proteoglycans decorin and biglycan were only increased in the MIadjacent. Fb isolated and cultured for 4 days had myoFb characteristics with little difference between MIremote and MIadjacent, although RNA sequencing revealed differences in gene expression profiles. Fbs from all regions maintained proliferative capacity, and induced contraction of 3-D collagen matrices but scar myoFb was more effective. These data suggest that after MI, signaling through TGF-β1, possibly related to increased mechanical load, drives Fb activation throughout the left ventricle while regional signaling determines further maturation and extracellular matrix remodeling after MI.
Collapse
Affiliation(s)
- Chandan K Nagaraju
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, Leuven, 3000, Belgium
| | - Eef Dries
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, Leuven, 3000, Belgium
| | - Natasa Popovic
- Cardiovascular Imaging and Dynamics, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, Leuven, 3000, Belgium
| | - Abhishek A Singh
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, Leuven, 3000, Belgium
| | - Peter Haemers
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, Leuven, 3000, Belgium
| | - H Llewelyn Roderick
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, Leuven, 3000, Belgium
| | - Piet Claus
- Cardiovascular Imaging and Dynamics, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, Leuven, 3000, Belgium
| | - Karin R Sipido
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, Leuven, 3000, Belgium.
| | - Ronald B Driesen
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, Leuven, 3000, Belgium
| |
Collapse
|
36
|
Suthahar N, Meijers WC, Silljé HHW, de Boer RA. From Inflammation to Fibrosis-Molecular and Cellular Mechanisms of Myocardial Tissue Remodelling and Perspectives on Differential Treatment Opportunities. Curr Heart Fail Rep 2017; 14:235-250. [PMID: 28707261 PMCID: PMC5527069 DOI: 10.1007/s11897-017-0343-y] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE OF REVIEW In this review, we highlight the most important cellular and molecular mechanisms that contribute to cardiac inflammation and fibrosis. We also discuss the interplay between inflammation and fibrosis in various precursors of heart failure (HF) and how such mechanisms can contribute to myocardial tissue remodelling and development of HF. RECENT FINDINGS Recently, many research articles attempt to elucidate different aspects of the interplay between inflammation and fibrosis. Cardiac inflammation and fibrosis are major pathophysiological mechanisms operating in the failing heart, regardless of HF aetiology. Currently, novel therapeutic options are available or are being developed to treat HF and these are discussed in this review. A progressive disease needs an aggressive management; however, existing therapies against HF are insufficient. There is a dynamic interplay between inflammation and fibrosis in various precursors of HF such as myocardial infarction (MI), myocarditis and hypertension, and also in HF itself. There is an urgent need to identify novel therapeutic targets and develop advanced therapeutic strategies to combat the syndrome of HF. Understanding and describing the elements of the inflammatory and fibrotic pathways are essential, and specific drugs that target these pathways need to be evaluated.
Collapse
Affiliation(s)
- Navin Suthahar
- Department of Cardiology, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB, Groningen, The Netherlands
| | - Wouter C Meijers
- Department of Cardiology, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB, Groningen, The Netherlands
| | - Herman H W Silljé
- Department of Cardiology, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB, Groningen, The Netherlands
| | - Rudolf A de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB, Groningen, The Netherlands.
| |
Collapse
|
37
|
Subramanian V, Seemann I, Merl-Pham J, Hauck SM, Stewart FA, Atkinson MJ, Tapio S, Azimzadeh O. Role of TGF Beta and PPAR Alpha Signaling Pathways in Radiation Response of Locally Exposed Heart: Integrated Global Transcriptomics and Proteomics Analysis. J Proteome Res 2016; 16:307-318. [DOI: 10.1021/acs.jproteome.6b00795] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Vikram Subramanian
- Helmholtz Zentrum München - German Research Center for Environmental Health GmbH, Institute of Radiation Biology, 85764 Neuherberg, Germany
| | - Ingar Seemann
- Division
of Biological Stress Response, Netherlands Cancer Institute, 1006 BE Amsterdam, The Netherlands
| | - Juliane Merl-Pham
- Helmholtz Zentrum Muenchen - German Research Centre for Environmental Health GmbH, Research Unit Protein Science, 80939 Munich, Germany
| | - Stefanie M. Hauck
- Helmholtz Zentrum Muenchen - German Research Centre for Environmental Health GmbH, Research Unit Protein Science, 80939 Munich, Germany
| | - Fiona A. Stewart
- Division
of Biological Stress Response, Netherlands Cancer Institute, 1006 BE Amsterdam, The Netherlands
| | - Michael J. Atkinson
- Helmholtz Zentrum München - German Research Center for Environmental Health GmbH, Institute of Radiation Biology, 85764 Neuherberg, Germany
- Chair
of Radiation Biology, Technical University of Munich, 81675 Munich, Germany
| | - Soile Tapio
- Helmholtz Zentrum München - German Research Center for Environmental Health GmbH, Institute of Radiation Biology, 85764 Neuherberg, Germany
| | - Omid Azimzadeh
- Helmholtz Zentrum München - German Research Center for Environmental Health GmbH, Institute of Radiation Biology, 85764 Neuherberg, Germany
| |
Collapse
|
38
|
Beetz N, Rommel C, Schnick T, Neumann E, Lother A, Monroy-Ordonez EB, Zeeb M, Preissl S, Gilsbach R, Melchior-Becker A, Rylski B, Stoll M, Schaefer L, Beyersdorf F, Stiller B, Hein L. Ablation of biglycan attenuates cardiac hypertrophy and fibrosis after left ventricular pressure overload. J Mol Cell Cardiol 2016; 101:145-155. [PMID: 27789290 DOI: 10.1016/j.yjmcc.2016.10.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 10/20/2016] [Accepted: 10/22/2016] [Indexed: 11/16/2022]
Abstract
AIMS Biglycan, a small leucine-rich proteoglycan, has been shown to play an important role in stabilizing fibrotic scars after experimental myocardial infarction. However, the role of biglycan in the development and regression of cardiomyocyte hypertrophy and fibrosis during cardiac pressure overload and unloading remains elusive. Thus, the aim of the present study was to assess the effect of biglycan on cardiac remodeling in a mouse model of left ventricular pressure overload and unloading. METHODS AND RESULTS Left ventricular pressure overload induced by transverse aortic constriction (TAC) in mice resulted in left ventricular dysfunction, fibrosis and increased biglycan expression. Fluorescence- and magnetic-assisted sorting of cardiac cell types revealed upregulation of biglycan in the fibroblast population, but not in cardiomyocytes, endothelial cells or leukocytes after TAC. Removal of the aortic constriction (rTAC) after short-term pressure overload (3weeks) improved cardiac contractility and reversed ventricular hypertrophy but not fibrosis in wild-type (WT) mice. Biglycan ablation (KO) enhanced functional recovery but did not resolve cardiac fibrosis. After long-term TAC for 9weeks, ablation of biglycan attenuated the development of cardiac hypertrophy and fibrosis. In vitro, biglycan induced hypertrophy of neonatal rat cardiomyocytes and led to activation of a hypertrophic gene program. Putative downstream mediators of biglycan signaling include Rcan1, Abra and Tnfrsf12a. These genes were concordantly induced by TAC in WT but not in biglycan KO mice. CONCLUSIONS Left ventricular pressure overload induces biglycan expression in cardiac fibroblasts. Ablation of biglycan improves cardiac function and attenuates left ventricular hypertrophy and fibrosis after long-term pressure overload. In vitro biglycan induces hypertrophy of cardiomyocytes, suggesting that biglycan may act as a signaling molecule between cell types to modulate cardiac remodeling.
Collapse
Affiliation(s)
- Nadine Beetz
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Carolin Rommel
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Tilman Schnick
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Heart Center, Department of Congenital Heart Defects and Pediatric Cardiology, University of Freiburg, Freiburg, Germany
| | - Elena Neumann
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Heart Center, Department of Congenital Heart Defects and Pediatric Cardiology, University of Freiburg, Freiburg, Germany
| | - Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Heart Center, Department of Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Elsa Beatriz Monroy-Ordonez
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martin Zeeb
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sebastian Preissl
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ralf Gilsbach
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ariane Melchior-Becker
- Institute for Pharmacology and Clinical Pharmacology, University of Düsseldorf, Düsseldorf, Germany
| | - Bartosz Rylski
- Heart Center, Department of Cardiovascular Surgery, University of Freiburg, Freiburg, Germany
| | - Monika Stoll
- Institute of Human Genetics, Genetic Epidemiology, University of Münster, Münster, Germany
| | - Liliana Schaefer
- Pharmazentrum, Allgemeine Pharmakologie und Toxikologie, Goethe Universität, Frankfurt, Germany
| | - Friedhelm Beyersdorf
- Heart Center, Department of Cardiovascular Surgery, University of Freiburg, Freiburg, Germany
| | - Brigitte Stiller
- Heart Center, Department of Congenital Heart Defects and Pediatric Cardiology, University of Freiburg, Freiburg, Germany
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg, Germany; BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
39
|
Barallobre-Barreiro J, Lynch M, Yin X, Mayr M. Systems biology-opportunities and challenges: the application of proteomics to study the cardiovascular extracellular matrix. Cardiovasc Res 2016; 112:626-636. [PMID: 27635058 PMCID: PMC5157133 DOI: 10.1093/cvr/cvw206] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 08/31/2016] [Accepted: 09/09/2016] [Indexed: 12/29/2022] Open
Abstract
Systems biology approaches including proteomics are becoming more widely used in cardiovascular research. In this review article, we focus on the application of proteomics to the cardiac extracellular matrix (ECM). ECM remodelling is a hallmark of many cardiovascular diseases. Proteomic techniques using mass spectrometry (MS) provide a platform for the comprehensive analysis of ECM proteins without a priori assumptions. Proteomics overcomes various constraints inherent to conventional antibody detection. On the other hand, studies that use whole tissue lysates for proteomic analysis mask the identification of the less abundant ECM constituents. In this review, we first discuss decellularization-based methods that enrich for ECM proteins in cardiac tissue, and how targeted MS allows for accurate protein quantification. The second part of the review will focus on post-translational modifications including hydroxylation and glycosylation and on the release of matrix fragments with biological activity (matrikines), all of which can be interrogated by proteomic techniques.
Collapse
Affiliation(s)
| | - Marc Lynch
- King's British Heart Foundation Centre, King's College London, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Xiaoke Yin
- King's British Heart Foundation Centre, King's College London, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Manuel Mayr
- King's British Heart Foundation Centre, King's College London, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
40
|
Gáspár R, Pipicz M, Hawchar F, Kovács D, Djirackor L, Görbe A, Varga ZV, Kiricsi M, Petrovski G, Gácser A, Csonka C, Csont T. The cytoprotective effect of biglycan core protein involves Toll-like receptor 4 signaling in cardiomyocytes. J Mol Cell Cardiol 2016; 99:138-150. [PMID: 27515282 DOI: 10.1016/j.yjmcc.2016.08.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 07/15/2016] [Accepted: 08/08/2016] [Indexed: 02/06/2023]
Abstract
AIMS Exogenously administered biglycan (core protein with high-molecular weight glycosaminoglycan chains) has been shown to protect neonatal cardiomyocytes against simulated ischemia/reperfusion injury (SI/R), however, the mechanism of action is not clear. In this study we aimed to investigate, which structural component of biglycan is responsible for its cardiocytoprotective effect and to further explore the molecular mechanisms involved in the cytoprotection. METHODS AND RESULTS A pilot study was conducted to demonstrate that both native (glycanated) and deglycanated biglycan can attenuate cell death induced by SI/R in a dose-dependent manner in primary neonatal cardiomyocytes isolated from Wistar rats. In separate experiments, we have shown that similarly to glycanated biglycan, recombinant human biglycan core protein (rhBGNc) protects cardiomyocytes against SI/R injury. In contrast, the glycosaminoglycan component dermatan sulfate had no significant effect on cell viability, while chondroitin sulfate further enhanced cell death induced by SI/R. Treatment of cardiomyocytes with rhBGNc reverses the effect of SI/R upon markers of necrosis, apoptosis, mitochondrial membrane potential, and autophagy. We have also shown that pharmacological blockade of Toll-like receptor 4 (TLR4) signaling or its downstream mediators (IRAK1/4, ERK, JNK and p38 MAP kinases) abolished the cytoprotective effect of rhBGNc against SI/R injury. Pretreatment of cardiomyocytes with rhBGNc for 20h resulted in increased Akt phosphorylation and NO production without having significant effect on phosphorylation of ERK1/2, STAT3, and on the production of superoxide. Treatment over 10min and 1h with rhBGNc increased ERK1 phosphorylation, while the SI/R-induced increase in superoxide production was attenuated by rhBGNc. Blockade of NO synthesis also prevented the cardiocytoprotective effect of rhBGNc. CONCLUSIONS The core protein of exogenous biglycan protects myocardial cells from SI/R injury via TLR4-mediated mechanisms involving activation of ERK, JNK and p38 MAP kinases and increased NO production. The cytoprotective effect of rhBGNc is due to modulation of SI/R-induced changes in necrosis, apoptosis and autophagy.
Collapse
Affiliation(s)
- Renáta Gáspár
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Márton Pipicz
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Fatime Hawchar
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Dávid Kovács
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Luna Djirackor
- Stem Cells and Eye Research Laboratory, Department of Ophthalmology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Anikó Görbe
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Zoltán V Varga
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Mónika Kiricsi
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Goran Petrovski
- Stem Cells and Eye Research Laboratory, Department of Ophthalmology, Faculty of Medicine, University of Szeged, Szeged, Hungary; Centre of Eye Research, Department of Ophthalmology, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Attila Gácser
- Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Csaba Csonka
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Tamás Csont
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary.
| |
Collapse
|
41
|
Liu S, Ji Y, Yao J, Zhao X, Xu H, Guan Y, Breyer RM, Sheng H, Zhu J. Knockout of the Prostaglandin E2 Receptor Subtype 3 Promotes Eccentric Cardiac Hypertrophy and Fibrosis in Mice. J Cardiovasc Pharmacol Ther 2016; 22:71-82. [PMID: 27093953 DOI: 10.1177/1074248416642520] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background: Prostaglandin E2 receptor subtype 3 (EP3), a Gi protein-coupled receptor activated by prostaglandin E2, plays a particular role in cardioprotection. This study aimed to investigate the impact of EP3 deletion on cardiac remodeling and further elucidate the related involvement of possible signaling pathways. Methods and Results: The animals used were EP3 receptor knockout (EP3KO) mice and wild-type (WT) litter mate controls at 16-18 weeks old. The high-resolution echocardiography and weight index indicated that eccentric cardiac hypertrophy might occur in EP3KO mice, which were having worse cardiac function than WT litter mates. Isolated adult myocytes from EP3KO hearts showed spontaneous lengthening. Cardiac fibrosis was observed in EP3KO mice through Masson trichrome staining. The elevated messenger RNA (mRNA) level in matrix genes and the reduced mRNA, protein, and activity levels of matrix metalloproteinase 2 (MMP-2) indicated an increased synthesis and suppressed degradation of matrix collagen in EP3KO mice. The phosphorylation level of extracellular signal-regulated kinase (ERK) 1/2 protein was reduced in the cardiac tissue of EP3KO mice, accompanied by no significant change in the protein level of total ERK1/2, total p38, phospho-p38, glycogen synthase kinase-3β (GSK3β), phospho-GSK3β, and calcineurin (CaN) as well as CaN activity. Conclusion: EP3 knockout in cardiac tissues could induce eccentric cardiac hypertrophy and cardiac fibrosis at 16-18 weeks old. These effects of EP3 knockout might be regulated through inactivating MAPK/ERK pathway and affecting the MMP-2 expression. Overall, PGE2-EP3 is necessary to maintain the normal growth and development of the heart.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yawei Ji
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Jian Yao
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Xiaodan Zhao
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Hu Xu
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Youfei Guan
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Richard M. Breyer
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hongzhuan Sheng
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Jianhua Zhu
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
42
|
Tumour endothelial cells in high metastatic tumours promote metastasis via epigenetic dysregulation of biglycan. Sci Rep 2016; 6:28039. [PMID: 27295191 PMCID: PMC4904795 DOI: 10.1038/srep28039] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 05/27/2016] [Indexed: 11/15/2022] Open
Abstract
Tumour blood vessels are gateways for distant metastasis. Recent studies have revealed that tumour endothelial cells (TECs) demonstrate distinct phenotypes from their normal counterparts. We have demonstrated that features of TECs are different depending on tumour malignancy, suggesting that TECs communicate with surrounding tumour cells. However, the contribution of TECs to metastasis has not been elucidated. Here, we show that TECs actively promote tumour metastasis through a bidirectional interaction between tumour cells and TECs. Co-implantation of TECs isolated from highly metastatic tumours accelerated lung metastases of low metastatic tumours. Biglycan, a small leucine-rich repeat proteoglycan secreted from TECs, activated tumour cell migration via nuclear factor-κB and extracellular signal–regulated kinase 1/2. Biglycan expression was upregulated by DNA demethylation in TECs. Collectively, our results demonstrate that TECs are altered in their microenvironment and, in turn, instigate tumour cells to metastasize, which is a novel mechanism for tumour metastasis.
Collapse
|
43
|
Altara R, Manca M, Sabra R, Eid AA, Booz GW, Zouein FA. Temporal cardiac remodeling post-myocardial infarction: dynamics and prognostic implications in personalized medicine. Heart Fail Rev 2015; 21:25-47. [PMID: 26498937 DOI: 10.1007/s10741-015-9513-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Despite dramatic improvements in short-term mortality rates following myocardial infarction (MI), long-term survival for MI patients who progress to heart failure remains poor. MI occurs when the left ventricle (LV) is deprived of oxygen for a sufficient period of time to induce irreversible necrosis of the myocardium. The LV response to MI involves significant tissue, cellular, and molecular level modifications, as well as substantial hemodynamic changes that feedback negatively to amplify the response. Inflammation to remove necrotic myocytes and fibroblast activation to form a scar are key wound healing responses that are highly variable across individuals. Few biomarkers of early remodeling stages are currently clinically adopted. The discovery of underlying pathophysiological mechanisms and associated novel biomarkers has the potential of improving prognostic capability and therapeutic monitoring. Combining these biomarkers with other prominent ones could constitute a powerful diagnostic and prognostic tool that directly reflects the pathophysiological remodeling of the LV. Understanding temporal remodeling at the tissue, cellular, and molecular level and its link to a well-defined set of biomarkers at early stages post-MI is a prerequisite for improving personalized care and devising more successful therapeutic interventions. Here we summarize the integral mechanisms that occur during early cardiac remodeling in the post-MI setting and highlight the most prominent biomarkers for assessing disease progression.
Collapse
Affiliation(s)
- Raffaele Altara
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA.,Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Marco Manca
- DG-DI, Medical Applications, CERN, Geneva, Switzerland
| | - Ramzi Sabra
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Assaad A Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - George W Booz
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Fouad A Zouein
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, USA. .,Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
44
|
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: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
45
|
Richardson WJ, Clarke SA, Quinn TA, Holmes JW. Physiological Implications of Myocardial Scar Structure. Compr Physiol 2015; 5:1877-909. [PMID: 26426470 DOI: 10.1002/cphy.c140067] [Citation(s) in RCA: 173] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Once myocardium dies during a heart attack, it is replaced by scar tissue over the course of several weeks. The size, location, composition, structure, and mechanical properties of the healing scar are all critical determinants of the fate of patients who survive the initial infarction. While the central importance of scar structure in determining pump function and remodeling has long been recognized, it has proven remarkably difficult to design therapies that improve heart function or limit remodeling by modifying scar structure. Many exciting new therapies are under development, but predicting their long-term effects requires a detailed understanding of how infarct scar forms, how its properties impact left ventricular function and remodeling, and how changes in scar structure and properties feed back to affect not only heart mechanics but also electrical conduction, reflex hemodynamic compensations, and the ongoing process of scar formation itself. In this article, we outline the scar formation process following a myocardial infarction, discuss interpretation of standard measures of heart function in the setting of a healing infarct, then present implications of infarct scar geometry and structure for both mechanical and electrical function of the heart and summarize experiences to date with therapeutic interventions that aim to modify scar geometry and structure. One important conclusion that emerges from the studies reviewed here is that computational modeling is an essential tool for integrating the wealth of information required to understand this complex system and predict the impact of novel therapies on scar healing, heart function, and remodeling following myocardial infarction.
Collapse
Affiliation(s)
- William J Richardson
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| | - Samantha A Clarke
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - T Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jeffrey W Holmes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA.,Department of Medicine, University of Virginia, Charlottesville, Virginia, USA.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
46
|
Neill T, Schaefer L, Iozzo RV. Decoding the Matrix: Instructive Roles of Proteoglycan Receptors. Biochemistry 2015; 54:4583-98. [PMID: 26177309 DOI: 10.1021/acs.biochem.5b00653] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The extracellular matrix is a dynamic repository harboring instructive cues that embody substantial regulatory dominance over many evolutionarily conserved intracellular activities, including proliferation, apoptosis, migration, motility, and autophagy. The matrix also coordinates and parses hierarchical information, such as angiogenesis, tumorigenesis, and immunological responses, typically providing the critical determinants driving each outcome. We provide the first comprehensive review focused on proteoglycan receptors, that is, signaling transmembrane proteins that use secreted proteoglycans as ligands, in addition to their natural ligands. The majority of these receptors belong to an exclusive subset of receptor tyrosine kinases and assorted cell surface receptors that specifically bind, transduce, and modulate fundamental cellular processes following interactions with proteoglycans. The class of small leucine-rich proteoglycans is the most studied so far and constitutes the best understood example of proteoglycan-receptor interactions. Decorin and biglycan evoke autophagy and immunological responses that deter, suppress, or exacerbate pathological conditions such as tumorigenesis, angiogenesis, and chronic inflammatory disease. Basement membrane-associated heparan sulfate proteoglycans (perlecan, agrin, and collagen XVIII) represent a unique cohort and provide proteolytically cleaved bioactive fragments for modulating cellular behavior. The receptors that bind the genuinely multifactorial and multivalent proteoglycans represent a nexus in understanding basic biological pathways and open new avenues for therapeutic and pharmacological intervention.
Collapse
Affiliation(s)
- Thomas Neill
- †Department of Pathology, Anatomy and Cell Biology and Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, United States
| | - Liliana Schaefer
- ‡Department of Pharmacology, Goethe University, 60590 Frankfurt, Germany
| | - Renato V Iozzo
- †Department of Pathology, Anatomy and Cell Biology and Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, United States
| |
Collapse
|
47
|
Gorressen S, Stern M, van de Sandt AM, Cortese-Krott MM, Ohlig J, Rassaf T, Gödecke A, Fischer JW, Heusch G, Merx MW, Kelm M. Circulating NOS3 modulates left ventricular remodeling following reperfused myocardial infarction. PLoS One 2015; 10:e0120961. [PMID: 25875863 PMCID: PMC4397096 DOI: 10.1371/journal.pone.0120961] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 01/27/2015] [Indexed: 12/21/2022] Open
Abstract
Purpose Nitric oxide (NO) is constitutively produced and released from the endothelium and several blood cell types by the isoform 3 of the NO synthase (NOS3). We have shown that NO protects against myocardial ischemia/reperfusion (I/R) injury and that depletion of circulating NOS3 increases within 24h of ischemia/reperfusion the size of myocardial infarction (MI) in chimeric mice devoid of circulating NOS3. In the current study we hypothesized that circulating NOS3 also affects remodeling of the left ventricle following reperfused MI. Methods To analyze the role of circulating NOS3 we transplanted bone marrow of NOS3−/− and wild type (WT) mice into WT mice, producing chimerae expressing NOS3 only in vascular endothelium (BC−/EC+) or in both, blood cells and vascular endothelium (BC+/EC+). Both groups underwent 60 min of coronary occlusion in a closed-chest model of reperfused MI. During the 3 weeks post MI, structural and functional LV remodeling was serially assessed (24h, 4d, 1w, 2w and 3w) by echocardiography. At 72 hours post MI, gene expression of several extracellular matrix (ECM) modifying molecules was determined by quantitative RT-PCR analysis. At 3 weeks post MI, hemodynamics were obtained by pressure catheter, scar size and collagen content were quantified post mortem by Gomori’s One-step trichrome staining. Results Three weeks post MI, LV end-systolic (53.2±5.9μl;***p≤0.001;n = 5) and end-diastolic volumes (82.7±5.6μl;*p<0.05;n = 5) were significantly increased in BC−/EC+, along with decreased LV developed pressure (67.5±1.8mmHg;n = 18;***p≤0.001) and increased scar size/left ventricle (19.5±1.5%;n = 13;**p≤0.01) compared to BC+/EC+ (ESV:35.6±2.2μl; EDV:69.1±2.6μl n = 8; LVDP:83.2±3.2mmHg;n = 24;scar size/LV13.8±0.7%;n = 16). Myocardial scar of BC−/EC+ was characterized by increased total collagen content (20.2±0.8%;n = 13;***p≤0.001) compared to BC+/EC+ (15.9±0.5;n = 16), and increased collagen type I and III subtypes. Conclusion Circulating NOS3 ameliorates maladaptive left ventricular remodeling following reperfused myocardial infarction.
Collapse
Affiliation(s)
- Simone Gorressen
- Medical Faculty, Division of Cardiology, Pulmonology & Vascular Medicine, Heinrich-Heine-University, Düsseldorf, Germany
| | - Manuel Stern
- Medical Faculty, Division of Cardiology, Pulmonology & Vascular Medicine, Heinrich-Heine-University, Düsseldorf, Germany
| | - Annette M. van de Sandt
- Medical Faculty, Division of Cardiology, Pulmonology & Vascular Medicine, Heinrich-Heine-University, Düsseldorf, Germany
| | - Miriam M. Cortese-Krott
- Medical Faculty, Division of Cardiology, Pulmonology & Vascular Medicine, Heinrich-Heine-University, Düsseldorf, Germany
| | - Jan Ohlig
- Medical Faculty, Division of Cardiology, Pulmonology & Vascular Medicine, Heinrich-Heine-University, Düsseldorf, Germany
| | - Tienush Rassaf
- Medical Faculty, Division of Cardiology, Pulmonology & Vascular Medicine, Heinrich-Heine-University, Düsseldorf, Germany
| | - Axel Gödecke
- Medical Faculty, Department of Cardiovascular Physiology, Heinrich-Heine-University, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, Düsseldorf, Germany
| | - Jens W. Fischer
- CARID, Cardiovascular Research Institute Düsseldorf, Düsseldorf, Germany
- Medical Faculty, Institute of Pharmacology und Clinical Pharmacology, Heinrich Heine University, Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center Essen, University of Essen Medical School, Essen, Germany
| | - Marc W. Merx
- Medical Faculty, Division of Cardiology, Pulmonology & Vascular Medicine, Heinrich-Heine-University, Düsseldorf, Germany
- Department of Cardiology, Vascular Medicine and Intensive Care Medicine, Robert Koch Krankenhaus, Klinikum Region Hannover, Hannover, Germany
| | - Malte Kelm
- Medical Faculty, Division of Cardiology, Pulmonology & Vascular Medicine, Heinrich-Heine-University, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, Düsseldorf, Germany
- * E-mail:
| |
Collapse
|
48
|
Takawale A, Sakamuri SS, Kassiri Z. Extracellular Matrix Communication and Turnover in Cardiac Physiology and Pathology. Compr Physiol 2015; 5:687-719. [DOI: 10.1002/cphy.c140045] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
49
|
Markers of fibrosis, inflammation, and remodeling pathways in heart failure. Clin Chim Acta 2015; 443:29-38. [DOI: 10.1016/j.cca.2014.09.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 08/22/2014] [Accepted: 09/03/2014] [Indexed: 01/13/2023]
|
50
|
Van Aelst LN, Voss S, Carai P, Van Leeuwen R, Vanhoutte D, Sanders-van Wijk S, Eurlings L, Swinnen M, Verheyen FK, Verbeken E, Nef H, Troidl C, Cook SA, Brunner-La Rocca HP, Möllmann H, Papageorgiou AP, Heymans S. Osteoglycin Prevents Cardiac Dilatation and Dysfunction After Myocardial Infarction Through Infarct Collagen Strengthening. Circ Res 2015; 116:425-36. [DOI: 10.1161/circresaha.116.304599] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rationale:
To maintain cardiac mechanical and structural integrity after an ischemic insult, profound alterations occur within the extracellular matrix. Osteoglycin is a small leucine-rich proteoglycan previously described as a marker of cardiac hypertrophy.
Objective:
To establish whether osteoglycin may play a role in cardiac integrity and function after myocardial infarction (MI).
Methods and Results:
Osteoglycin expression is associated with collagen deposition and scar formation in mouse and human MI. Absence of osteoglycin in mice resulted in significantly increased rupture-related mortality with tissue disruption, intramyocardial bleeding, and increased cardiac dysfunction, despite equal infarct sizes. Surviving osteoglycin null mice had greater infarct expansion in comparison with wild-type mice because of impaired collagen fibrillogenesis and maturation in the infarcts as revealed by electron microscopy and collagen polarization. Absence of osteoglycin did not affect cardiomyocyte hypertrophy in the remodeling remote myocardium. In cultured fibroblasts, osteoglycin knockdown or supplementation did not alter transforming growth factor-β signaling. Adenoviral overexpression of osteoglycin in wild-type mice significantly improved collagen quality, thereby blunting cardiac dilatation and dysfunction after MI. In osteoglycin null mice, adenoviral overexpression of osteoglycin was unable to prevent rupture-related mortality because of insufficiently restoring osteoglycin protein levels in the heart. Finally, circulating osteoglycin levels in patients with heart failure were significantly increased in the patients with a previous history of MI compared with those with nonischemic heart failure and correlated with survival, left ventricular volumes, and other markers of fibrosis.
Conclusions:
Increased osteoglycin expression in the infarct scar promotes proper collagen maturation and protects against cardiac disruption and adverse remodeling after MI. In human heart failure, osteoglycin is a promising biomarker for ischemic heart failure.
Collapse
Affiliation(s)
- Lucas N.L. Van Aelst
- From the Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, Catholic University of Leuven, Leuven, Belgium (L.N.L.V.A., P.C., A.-P.P., S.H.); Department of Cardiology (L.N.L.V.A., M.S.) and Department of Pathology (E.V.), University Hospitals Leuven, Leuven, Belgium; Department of Cardiology, Kerckhoff Heart Center, Bad Nauheim, Germany (S.V., H.N., C.T., H.M.); Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), University Hospital
| | - Sandra Voss
- From the Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, Catholic University of Leuven, Leuven, Belgium (L.N.L.V.A., P.C., A.-P.P., S.H.); Department of Cardiology (L.N.L.V.A., M.S.) and Department of Pathology (E.V.), University Hospitals Leuven, Leuven, Belgium; Department of Cardiology, Kerckhoff Heart Center, Bad Nauheim, Germany (S.V., H.N., C.T., H.M.); Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), University Hospital
| | - Paolo Carai
- From the Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, Catholic University of Leuven, Leuven, Belgium (L.N.L.V.A., P.C., A.-P.P., S.H.); Department of Cardiology (L.N.L.V.A., M.S.) and Department of Pathology (E.V.), University Hospitals Leuven, Leuven, Belgium; Department of Cardiology, Kerckhoff Heart Center, Bad Nauheim, Germany (S.V., H.N., C.T., H.M.); Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), University Hospital
| | - Rick Van Leeuwen
- From the Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, Catholic University of Leuven, Leuven, Belgium (L.N.L.V.A., P.C., A.-P.P., S.H.); Department of Cardiology (L.N.L.V.A., M.S.) and Department of Pathology (E.V.), University Hospitals Leuven, Leuven, Belgium; Department of Cardiology, Kerckhoff Heart Center, Bad Nauheim, Germany (S.V., H.N., C.T., H.M.); Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), University Hospital
| | - Davy Vanhoutte
- From the Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, Catholic University of Leuven, Leuven, Belgium (L.N.L.V.A., P.C., A.-P.P., S.H.); Department of Cardiology (L.N.L.V.A., M.S.) and Department of Pathology (E.V.), University Hospitals Leuven, Leuven, Belgium; Department of Cardiology, Kerckhoff Heart Center, Bad Nauheim, Germany (S.V., H.N., C.T., H.M.); Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), University Hospital
| | - Sandra Sanders-van Wijk
- From the Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, Catholic University of Leuven, Leuven, Belgium (L.N.L.V.A., P.C., A.-P.P., S.H.); Department of Cardiology (L.N.L.V.A., M.S.) and Department of Pathology (E.V.), University Hospitals Leuven, Leuven, Belgium; Department of Cardiology, Kerckhoff Heart Center, Bad Nauheim, Germany (S.V., H.N., C.T., H.M.); Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), University Hospital
| | - Luc Eurlings
- From the Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, Catholic University of Leuven, Leuven, Belgium (L.N.L.V.A., P.C., A.-P.P., S.H.); Department of Cardiology (L.N.L.V.A., M.S.) and Department of Pathology (E.V.), University Hospitals Leuven, Leuven, Belgium; Department of Cardiology, Kerckhoff Heart Center, Bad Nauheim, Germany (S.V., H.N., C.T., H.M.); Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), University Hospital
| | - Melissa Swinnen
- From the Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, Catholic University of Leuven, Leuven, Belgium (L.N.L.V.A., P.C., A.-P.P., S.H.); Department of Cardiology (L.N.L.V.A., M.S.) and Department of Pathology (E.V.), University Hospitals Leuven, Leuven, Belgium; Department of Cardiology, Kerckhoff Heart Center, Bad Nauheim, Germany (S.V., H.N., C.T., H.M.); Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), University Hospital
| | - Fons K. Verheyen
- From the Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, Catholic University of Leuven, Leuven, Belgium (L.N.L.V.A., P.C., A.-P.P., S.H.); Department of Cardiology (L.N.L.V.A., M.S.) and Department of Pathology (E.V.), University Hospitals Leuven, Leuven, Belgium; Department of Cardiology, Kerckhoff Heart Center, Bad Nauheim, Germany (S.V., H.N., C.T., H.M.); Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), University Hospital
| | - Eric Verbeken
- From the Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, Catholic University of Leuven, Leuven, Belgium (L.N.L.V.A., P.C., A.-P.P., S.H.); Department of Cardiology (L.N.L.V.A., M.S.) and Department of Pathology (E.V.), University Hospitals Leuven, Leuven, Belgium; Department of Cardiology, Kerckhoff Heart Center, Bad Nauheim, Germany (S.V., H.N., C.T., H.M.); Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), University Hospital
| | - Holger Nef
- From the Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, Catholic University of Leuven, Leuven, Belgium (L.N.L.V.A., P.C., A.-P.P., S.H.); Department of Cardiology (L.N.L.V.A., M.S.) and Department of Pathology (E.V.), University Hospitals Leuven, Leuven, Belgium; Department of Cardiology, Kerckhoff Heart Center, Bad Nauheim, Germany (S.V., H.N., C.T., H.M.); Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), University Hospital
| | - Christian Troidl
- From the Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, Catholic University of Leuven, Leuven, Belgium (L.N.L.V.A., P.C., A.-P.P., S.H.); Department of Cardiology (L.N.L.V.A., M.S.) and Department of Pathology (E.V.), University Hospitals Leuven, Leuven, Belgium; Department of Cardiology, Kerckhoff Heart Center, Bad Nauheim, Germany (S.V., H.N., C.T., H.M.); Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), University Hospital
| | - Stuart A. Cook
- From the Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, Catholic University of Leuven, Leuven, Belgium (L.N.L.V.A., P.C., A.-P.P., S.H.); Department of Cardiology (L.N.L.V.A., M.S.) and Department of Pathology (E.V.), University Hospitals Leuven, Leuven, Belgium; Department of Cardiology, Kerckhoff Heart Center, Bad Nauheim, Germany (S.V., H.N., C.T., H.M.); Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), University Hospital
| | - Hans-Peter Brunner-La Rocca
- From the Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, Catholic University of Leuven, Leuven, Belgium (L.N.L.V.A., P.C., A.-P.P., S.H.); Department of Cardiology (L.N.L.V.A., M.S.) and Department of Pathology (E.V.), University Hospitals Leuven, Leuven, Belgium; Department of Cardiology, Kerckhoff Heart Center, Bad Nauheim, Germany (S.V., H.N., C.T., H.M.); Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), University Hospital
| | - Helge Möllmann
- From the Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, Catholic University of Leuven, Leuven, Belgium (L.N.L.V.A., P.C., A.-P.P., S.H.); Department of Cardiology (L.N.L.V.A., M.S.) and Department of Pathology (E.V.), University Hospitals Leuven, Leuven, Belgium; Department of Cardiology, Kerckhoff Heart Center, Bad Nauheim, Germany (S.V., H.N., C.T., H.M.); Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), University Hospital
| | - Anna-Pia Papageorgiou
- From the Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, Catholic University of Leuven, Leuven, Belgium (L.N.L.V.A., P.C., A.-P.P., S.H.); Department of Cardiology (L.N.L.V.A., M.S.) and Department of Pathology (E.V.), University Hospitals Leuven, Leuven, Belgium; Department of Cardiology, Kerckhoff Heart Center, Bad Nauheim, Germany (S.V., H.N., C.T., H.M.); Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), University Hospital
| | - Stephane Heymans
- From the Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, Catholic University of Leuven, Leuven, Belgium (L.N.L.V.A., P.C., A.-P.P., S.H.); Department of Cardiology (L.N.L.V.A., M.S.) and Department of Pathology (E.V.), University Hospitals Leuven, Leuven, Belgium; Department of Cardiology, Kerckhoff Heart Center, Bad Nauheim, Germany (S.V., H.N., C.T., H.M.); Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), University Hospital
| |
Collapse
|