1
|
Zhang B, Enriquez-Sarano M, Schaff HV, Michelena HI, Roos CM, Hagler MA, Zhang H, Casaclang-Verzosa G, Huang R, Bartoo A, Ranadive S, Joyner MJ, Pislaru S, Nkomo VT, Kremers WK, Araoz PA, Singh G, Walters MA, Hawkinson J, Cunningham KY, Sung J, Dunagan B, Ye Z, Miller JD. Reactivation of Oxidized Soluble Guanylate Cyclase as a Novel Treatment Strategy to Slow Progression of Calcific Aortic Valve Stenosis: Preclinical and Randomized Clinical Trials to Assess Safety and Efficacy. Circulation 2025; 151:913-930. [PMID: 39989354 DOI: 10.1161/circulationaha.123.066523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 01/27/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND Pharmacological treatments for fibrocalcific aortic valve stenosis (FCAVS) have been elusive for >50 years. Here, we tested the hypothesis that reactivation of oxidized sGC (soluble guanylate cyclase), the primary receptor for nitric oxide, with ataciguat is a safe and efficacious strategy to slow progression of FCAVS. METHODS We used quantitative real-time reverse transcription polymerase chain reaction, Western blotting, and immunohistochemistry to characterize sGC signaling and the biological effects of ataciguat on signaling cascades related to nitric oxide, calcification, and fibrosis in excised human aortic valve tissue, aortic valve interstitial cells, and mouse aortic valves. We then conducted randomized, placebo-controlled phase I (14-day safety/tolerance) and phase II (6-month efficacy) trials in patients with moderate aortic valve stenosis. RESULTS In excised human tissue, we found robust losses in sGC signaling despite upregulation of sGC subunits. In vitro, ataciguat increased sGC signaling and reduced BMP2 (bone morphogenetic protein 2) signaling in aortic valve interstitial cells. In mice with established FCAVS, treatment with ataciguat attenuated BMP signaling and slowed progression of valve calcification and dysfunction. In a phase I, randomized, placebo-controlled trial, treatment with ataciguat for 2 weeks was safe and well tolerated in patients with moderate FCAVS (https://www.clinicaltrials.gov; Unique identifier: NCT02049203). In a separate phase II, randomized, placebo-controlled trial, treatment with ataciguat for 6 months slowed the progression of aortic valve calcification and tended to slow the progression of valvular and ventricular dysfunction in patients with moderate FCAVS (https://www.clinicaltrials.gov; Unique identifier: NCT02481258). CONCLUSIONS Collectively, this study highlights the therapeutic potential of the targeted restoration of the diseased/inactive form of sGC for treatment of FCAVS. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT02049203. URL: https://www.clinicaltrials.gov; Unique identifier: NCT02481258.
Collapse
Affiliation(s)
- Bin Zhang
- Departments of Surgery (B.Z., C.M.R., M.A.H., H.Z., G.C.-V., J.S., B.D., J.D.M.), Mayo Clinic, Rochester, MN
| | - Maurice Enriquez-Sarano
- Cardiovascular Diseases (M.E.-S., H.I.M., R.H., S.P., V.T.N., Z.Y.), Mayo Clinic, Rochester, MN
| | - Hartzell V Schaff
- Cardiovascular Surgery (H.V.S., B.D., J.D.M.), Mayo Clinic, Rochester, MN
| | - Hector I Michelena
- Cardiovascular Diseases (M.E.-S., H.I.M., R.H., S.P., V.T.N., Z.Y.), Mayo Clinic, Rochester, MN
| | - Carolyn M Roos
- Departments of Surgery (B.Z., C.M.R., M.A.H., H.Z., G.C.-V., J.S., B.D., J.D.M.), Mayo Clinic, Rochester, MN
| | - Michael A Hagler
- Departments of Surgery (B.Z., C.M.R., M.A.H., H.Z., G.C.-V., J.S., B.D., J.D.M.), Mayo Clinic, Rochester, MN
| | - Heyu Zhang
- Departments of Surgery (B.Z., C.M.R., M.A.H., H.Z., G.C.-V., J.S., B.D., J.D.M.), Mayo Clinic, Rochester, MN
| | - Grace Casaclang-Verzosa
- Departments of Surgery (B.Z., C.M.R., M.A.H., H.Z., G.C.-V., J.S., B.D., J.D.M.), Mayo Clinic, Rochester, MN
| | - Runqing Huang
- Cardiovascular Diseases (M.E.-S., H.I.M., R.H., S.P., V.T.N., Z.Y.), Mayo Clinic, Rochester, MN
| | | | | | | | - Sorin Pislaru
- Cardiovascular Diseases (M.E.-S., H.I.M., R.H., S.P., V.T.N., Z.Y.), Mayo Clinic, Rochester, MN
| | - Vuyisile T Nkomo
- Cardiovascular Diseases (M.E.-S., H.I.M., R.H., S.P., V.T.N., Z.Y.), Mayo Clinic, Rochester, MN
| | - Walter K Kremers
- Quantitative Health Sciences (W.K.K.), Mayo Clinic, Rochester, MN
| | | | - Gurpreet Singh
- Department of Medicinal Chemistry (G.S., M.A.W., J.H.), University of Minnesota, Minneapolis, MN
| | - Michael A Walters
- Department of Medicinal Chemistry (G.S., M.A.W., J.H.), University of Minnesota, Minneapolis, MN
| | - Jon Hawkinson
- Department of Medicinal Chemistry (G.S., M.A.W., J.H.), University of Minnesota, Minneapolis, MN
| | - Kevin Y Cunningham
- Bioinformatics and Computational Biology Program (K.Y.C.), University of Minnesota, Minneapolis, MN
| | - Jaeyun Sung
- Departments of Surgery (B.Z., C.M.R., M.A.H., H.Z., G.C.-V., J.S., B.D., J.D.M.), Mayo Clinic, Rochester, MN
| | - Brandon Dunagan
- Departments of Surgery (B.Z., C.M.R., M.A.H., H.Z., G.C.-V., J.S., B.D., J.D.M.), Mayo Clinic, Rochester, MN
- Cardiovascular Surgery (H.V.S., B.D., J.D.M.), Mayo Clinic, Rochester, MN
| | - Zi Ye
- Cardiovascular Diseases (M.E.-S., H.I.M., R.H., S.P., V.T.N., Z.Y.), Mayo Clinic, Rochester, MN
| | - Jordan D Miller
- Departments of Surgery (B.Z., C.M.R., M.A.H., H.Z., G.C.-V., J.S., B.D., J.D.M.), Mayo Clinic, Rochester, MN
- Cardiovascular Surgery (H.V.S., B.D., J.D.M.), Mayo Clinic, Rochester, MN
- Physiology and Biomedical Engineering (J.D.M.), Mayo Clinic, Rochester, MN
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN (J.D.M.)
| |
Collapse
|
2
|
Shelbaya K, Arthur V, Yang Y, Dorbala P, Buckley L, Claggett B, Skali H, Dufresne L, Yang TY, Engert JC, Thanassoulis G, Floyd J, Austin TR, Bortnick A, Kizer J, Freitas RCC, Singh SA, Aikawa E, Hoogeveen RC, Ballantyne C, Yu B, Coresh J, Blaha MJ, Matsushita K, Shah AM. Large-Scale Proteomics Identifies Novel Biomarkers and Circulating Risk Factors for Aortic Stenosis. J Am Coll Cardiol 2024; 83:577-591. [PMID: 38296402 DOI: 10.1016/j.jacc.2023.11.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 02/08/2024]
Abstract
BACKGROUND Limited data exist regarding risk factors for aortic stenosis (AS). The plasma proteome is a promising phenotype for discovery of novel biomarkers and potentially causative mechanisms. OBJECTIVES The aim of this study was to discover novel biomarkers with potentially causal associations with AS. METHODS We measured 4,877 plasma proteins (SomaScan aptamer-affinity assay) among ARIC (Atherosclerosis Risk In Communities) study participants in mid-life (visit 3 [V3]; n = 11,430; age 60 ± 6 years) and in late-life (V5; n = 4,899; age 76 ± 5 years). We identified proteins cross-sectionally associated with aortic valve (AV) peak velocity (AVmax) and dimensionless index by echocardiography at V5 and with incident AV-related hospitalization after V3 with the use of multivariable linear and Cox proportional hazard regression. We assessed associations of candidate proteins with changes in AVmax over 6 years and with AV calcification with the use of cardiac computed tomography, replicated analysis in an independent sample, performed Mendelian randomization, and evaluated gene expression in explanted human AV tissue. RESULTS Fifty-two proteins cross-sectionally were associated with AVmax and dimensionless index at V5 and with risk of incident AV-related hospitalization after V3. Among 3,413 participants in the Cardiovascular Health Study, 6 of those proteins were significantly associated with adjudicated moderate or severe AS, including matrix metalloproteinase 12 (MMP12), complement C1q tumor necrosis factor-related protein 1 (C1QTNF1), and growth differentiation factor-15. MMP12 was also associated with greater increase in AVmax over 6 years, greater degree of AV calcification, and greater expression in calcific compared with normal or fibrotic AV tissue. C1QTNF1 had consistent potential causal effects on both AS and AVmax according to Mendelian randomization analysis. CONCLUSIONS These findings identify MMP12 as a potential novel circulating biomarker of AS risk and C1QTNF1 as a new putative target to prevent AS progression.
Collapse
Affiliation(s)
| | | | - Yimin Yang
- Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Pranav Dorbala
- Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Leo Buckley
- Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Brian Claggett
- Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Hicham Skali
- Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Line Dufresne
- McGill University Health Centre, Montreal, Quebec, Canada
| | - Ta-Yu Yang
- McGill University Health Centre, Montreal, Quebec, Canada
| | - James C Engert
- McGill University Health Centre, Montreal, Quebec, Canada
| | | | - James Floyd
- Cardiovascular Health Research Unit, Seattle, Washington, USA
| | - Thomas R Austin
- Cardiovascular Health Research Unit, Seattle, Washington, USA
| | - Anna Bortnick
- Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jorge Kizer
- Veterans Affairs Medical Center, San Francisco, California, USA
| | | | - Sasha A Singh
- Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Elena Aikawa
- Brigham and Women's Hospital, Boston, Massachusetts, USA
| | | | | | - Bing Yu
- University of Texas Health Science School of Public Health, Houston, Texas, USA
| | - Josef Coresh
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Michael J Blaha
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | | | - Amil M Shah
- Brigham and Women's Hospital, Boston, Massachusetts, USA; University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| |
Collapse
|
3
|
Liu Z, Zhu G, Zhang Y, Zhang P, Zang W, Shen Z. Comprehensive comparative analysis of the prognostic impact of systemic inflammation biomarkers for patients underwent cardiac surgery. Front Immunol 2023; 14:1190380. [PMID: 37646036 PMCID: PMC10461628 DOI: 10.3389/fimmu.2023.1190380] [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: 03/20/2023] [Accepted: 07/31/2023] [Indexed: 09/01/2023] Open
Abstract
Background Inflammation plays an integral role in the development of cardiovascular disease, and few studies have identified different biomarkers to predict the prognosis of cardiac surgery. But there is a lack of reliable and valid evidence to determine the optimal systemic inflammatory biomarkers to predict prognosis. Methods From December 2015 and March 2021, we collected 10 systemic inflammation biomarkers among 820 patients who underwent cardiac surgery. Time-dependent receiver operating characteristic curves (ROC) curve at different time points and C-index was compared at different time points. Kaplan-Meier method was performed to analyze overall survival (OS). Cox proportional hazard regression analyses were used to assess independent risk factors for OS. A random internal validation was conducted to confirm the effectiveness of the biomarkers. Results The area under the ROC of lymphocyte-to-C-reactive protein ratio (LCR) was 0.655, 0.620 and 0.613 at 1-, 2- and 3-year respectively, and C-index of LCR for OS after cardiac surgery was 0.611, suggesting that LCR may serve as a favorable indicator for predicting the prognosis of cardiac surgery. Patients with low LCR had a higher risk of postoperative complications. Besides, Cox proportional hazard regression analyses indicated that LCR was considered as an independent risk factor of OS after cardiac surgery. Conclusion LCR shows promise as a noteworthy representative among the systemic inflammation biomarkers in predicting the prognosis of cardiac surgery. Screening for low LCR levels may help surgeons identify high-risk patients and guide perioperative management strategies.
Collapse
Affiliation(s)
- Zhang Liu
- Department of Cardio-Thoracic Surgery, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ge Zhu
- Department of Cardio-Thoracic Surgery, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yonggui Zhang
- Department of Cardio-Thoracic Surgery, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Peng Zhang
- Department of Cardio-Thoracic Surgery, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wangfu Zang
- Department of Cardio-Thoracic Surgery, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zile Shen
- Department of Gastrointestinal Surgery, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
4
|
Semenova D, Zabirnyk A, Lobov A, Boyarskaya N, Kachanova O, Uspensky V, Zainullina B, Denisov E, Gerashchenko T, Kvitting JPE, Kaljusto ML, Thiede B, Kostareva A, Stensløkken KO, Vaage J, Malashicheva A. Multi-omics of in vitro aortic valve calcification. Front Cardiovasc Med 2022; 9:1043165. [PMID: 36407442 PMCID: PMC9669078 DOI: 10.3389/fcvm.2022.1043165] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 09/23/2022] [Indexed: 09/10/2023] Open
Abstract
Heart valve calcification is an active cellular and molecular process that partly remains unknown. Osteogenic differentiation of valve interstitial cells (VIC) is a central mechanism in calcific aortic valve disease (CAVD). Studying mechanisms in CAVD progression is clearly needed. In this study, we compared molecular mechanisms of osteogenic differentiation of human VIC isolated from healthy donors or patients with CAVD by RNA-seq transcriptomics in early timepoint (48 h) and by shotgun proteomics at later timepoint (10th day). Bioinformatic analysis revealed genes and pathways involved in the regulation of VIC osteogenic differentiation. We found a high amount of stage-specific differentially expressed genes and good accordance between transcriptomic and proteomic data. Functional annotation of differentially expressed proteins revealed that osteogenic differentiation of VIC involved many signaling cascades such as: PI3K-Akt, MAPK, Ras, TNF signaling pathways. Wnt, FoxO, and HIF-1 signaling pathways were modulated only at the early timepoint and thus probably involved in the commitment of VIC to osteogenic differentiation. We also observed a significant shift of some metabolic pathways in the early stage of VIC osteogenic differentiation. Lentiviral overexpression of one of the most upregulated genes (ZBTB16, PLZF) increased calcification of VIC after osteogenic stimulation. Analysis with qPCR and shotgun proteomics suggested a proosteogenic role of ZBTB16 in the early stages of osteogenic differentiation.
Collapse
Affiliation(s)
- Daria Semenova
- Institute of Cytology Russian Academy of Science, St. Petersburg, Russia
- Almazov National Medical Research Center Russia, St. Petersburg, Russia
| | - Arsenii Zabirnyk
- Heart Physiology Research Group, Division of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Oslo University Hospital, Oslo, Norway
| | - Arseniy Lobov
- Institute of Cytology Russian Academy of Science, St. Petersburg, Russia
| | | | - Olga Kachanova
- Almazov National Medical Research Center Russia, St. Petersburg, Russia
| | - Vladimir Uspensky
- Almazov National Medical Research Center Russia, St. Petersburg, Russia
| | - Bozhana Zainullina
- Centre for Molecular and Cell Technologies, St. Petersburg State University, St. Petersburg, Russia
| | - Evgeny Denisov
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Tatiana Gerashchenko
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - John-Peder Escobar Kvitting
- Heart Physiology Research Group, Division of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Oslo University Hospital, Oslo, Norway
| | | | - Bernd Thiede
- Heart Physiology Research Group, Division of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Anna Kostareva
- Almazov National Medical Research Center Russia, St. Petersburg, Russia
| | - Kåre-Olav Stensløkken
- Heart Physiology Research Group, Division of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Jarle Vaage
- Heart Physiology Research Group, Division of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Oslo University Hospital, Oslo, Norway
| | - Anna Malashicheva
- Institute of Cytology Russian Academy of Science, St. Petersburg, Russia
| |
Collapse
|
5
|
Xiang J, He L, Li D, Wei S, Wu Z. Value of the systemic immune-inflammation index in predicting poor postoperative outcomes and the short-term prognosis of heart valve diseases: a retrospective cohort study. BMJ Open 2022; 12:e064171. [PMID: 36220322 PMCID: PMC9557268 DOI: 10.1136/bmjopen-2022-064171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVE Systemic immune-inflammation index (SII) is a novel biomarker that can predict poor outcomes in tumours, nervous system diseases and chronic heart failure. Here, we investigated the predictive value of SII on the poor postoperative outcomes and short-term prognosis of heart valve diseases (HVDs). DESIGN, SETTING AND PARTICIPANTS This retrospective cohort study enrolled all consecutive patients with HVDs (aortic stenosis, aortic regurgitation, mitral stenosis and mitral regurgitation) who underwent surgery (valve replacement or valve repair) at the Affiliated Hospital of North Sichuan Medical College between 2017 and 2020. MAIN OUTCOMES AND MEASURES Major complications in the perioperative period, all-cause mortality within 30 days and readmission within 30 days. RESULTS A total of 431 patients with HVDs were enrolled in this study, including 202 males and 229 females, aged 58.9±27.3 years. SII levels of patients in the poor outcomes group were significantly higher than those of patients in the favourable outcomes group (658.40±436.29 vs 335.72±174.76, respectively; p<0.001). Multivariate logistic regression analysis showed that age (OR 1.064, 95% CI 1.026 to 1.104, p=0.025), SII (OR 1.034, 95% CI 1.012 to 1.631, p=0.008) and aortic cross-clamping time (OR 1.013, 95% CI 1.004 to 1.023, p=0.006) were independent risk factors for poor outcomes and short-term prognosis in patients with HVD. The area under the curve of poor outcomes predicted by SII in patients with HVD was 0.806 (95% CI 0.763 to 0.848) and the optimised cut-off value 423.8×109 /L, with a sensitivity of 70.3% and specificity of 81.1%. The incidence of poor outcomes (p<0.001), 30-day mortality (p<0.001) and 30-day readmission rate (p=0.026) in the high SII group was significantly higher than that in the low SII group. CONCLUSIONS SII is closely related to poor postoperative outcomes and short-term prognosis of HVD and can serve as an independent predictive factor.
Collapse
Affiliation(s)
- Jun Xiang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Ling He
- Department of Paediatrics, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Donglin Li
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Shuliang Wei
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Zhong Wu
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
6
|
Trindade F, Ferreira AF, Saraiva F, Martins D, Mendes VM, Sousa C, Gavina C, Leite-Moreira A, Manadas B, Falcão-Pires I, Vitorino R. Optimization of a Protocol for Protein Extraction from Calcified Aortic Valves for Proteomics Applications: Development of a Standard Operating Procedure. Proteomes 2022; 10:proteomes10030030. [PMID: 36136308 PMCID: PMC9505568 DOI: 10.3390/proteomes10030030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/02/2022] [Accepted: 08/11/2022] [Indexed: 11/28/2022] Open
Abstract
The comprehension of the pathophysiological mechanisms, the identification of druggable targets, and putative biomarkers for aortic valve stenosis can be pursued through holistic approaches such as proteomics. However, tissue homogenization and protein extraction are made difficult by tissue calcification. The reproducibility of proteome studies is key in clinical translation of the findings. Thus, we aimed to optimize a protocol for aortic valve homogenization and protein extraction and to develop a standard operating procedure (SOP), which researchers can use to maximize protein yield while reducing inter-laboratory variability. We have compared the protein yield between conventional tissue grinding in nitrogen followed by homogenization with a Potter apparatus with a more advanced bead-beating system. Once we confirmed the superiority of the latter, we further optimized it by testing the effect of beads size, the number of homogenization cycles, tube capacity, lysis buffer/tissue mass ratio, and two different lysis buffers. Optimal protein extraction was achieved with 2.8 mm zirconium dioxide beads, in two homogenization cycles, in the presence of 20 µL RIPA buffer/mg tissue, using 2 mL O-ring cryotubes. As a proof of concept of the usefulness of this SOP for proteomics, the AV proteome of men and women with aortic stenosis was characterized, resulting in the quantification of proteins across six orders of magnitude and uncovering some putative proteins dysregulated by sex.
Collapse
Affiliation(s)
- Fábio Trindade
- Cardiovascular R&D Centre—UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Correspondence:
| | - Ana F. Ferreira
- Cardiovascular R&D Centre—UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Francisca Saraiva
- Cardiovascular R&D Centre—UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Diana Martins
- Cardiovascular R&D Centre—UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Vera M. Mendes
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Carla Sousa
- Cardiovascular R&D Centre—UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Cardiology Department, Centro Hospitalar Universitário de São João, 4200-319 Porto, Portugal
| | - Cristina Gavina
- Cardiovascular R&D Centre—UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Cardiology Department, Hospital Pedro Hispano, Unidade Local de Saúde de Matosinhos, 4464-513 Matosinhos, Portugal
| | - Adelino Leite-Moreira
- Cardiovascular R&D Centre—UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Department of Cardiothoracic Surgery, Centro Hospitalar Universitário de São João, 4200-319 Porto, Portugal
| | - Bruno Manadas
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Inês Falcão-Pires
- Cardiovascular R&D Centre—UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Rui Vitorino
- Cardiovascular R&D Centre—UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- iBiMED—Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
- LAQV/REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
7
|
Franzke B, Bileck A, Unterberger S, Aschauer R, Zöhrer PA, Draxler A, Strasser EM, Wessner B, Gerner C, Wagner KH. The plasma proteome is favorably modified by a high protein diet but not by additional resistance training in older adults: A 17-week randomized controlled trial. Front Nutr 2022; 9:925450. [PMID: 35990326 PMCID: PMC9389340 DOI: 10.3389/fnut.2022.925450] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundThe age-related loss of muscle mass significantly contributes to the development of chronic diseases, loss of mobility and dependency on others, yet could be improved by an optimized lifestyle.ObjectiveThe goal of this randomized controlled trial was to compare the influence of a habitual diet (CON) with either a diet containing the recommended protein intake (RP) or a high protein intake (HP), both with and without strength training, on the plasma proteome in older adults.MethodsOne hundred and thirty-six women and men (65–85 years) were randomly assigned to three intervention groups. CON continued their habitual diet; participants of the HP and RP group consumed either high protein or standard foods. After 6 weeks of dietary intervention, HP and RP groups additionally started a strength training intervention twice per week for 8 weeks. Twenty-four hours dietary recalls were performed every 7–10 days. Body composition was assessed and blood taken. Plasma proteomics were assessed with LC-MS.ResultsParticipants of the HP group doubled their baseline protein intake from 0.80 ± 0.31 to 1.63 ± 0.36 g/kg BW/d; RP increased protein intake from 0.89 ± 0.28 to 1.06 ± 0.26 g/kg BW/d. The CON group kept the protein intake stable throughout the study. Combined exercise and HP initiated notable changes, resulting in a reduction in bodyfat and increased muscle mass. Proteomics analyses revealed 14 significantly affected proteins by HP diet, regulating innate immune system, lipid transport and blood coagulation, yet the additional strength training did not elicit further changes.ConclusionsCombined HP and resistance exercise in healthy older adults seem to induce favorable changes in the body composition. Changes in the plasma proteome due to the high protein diet point to a beneficial impact for the innate immune system, lipid transport and blood coagulation system, all of which are involved in chronic disease development.Clinical trial registrationThe study was registered at ClinicalTrials.gov (NCT04023513).
Collapse
Affiliation(s)
- Bernhard Franzke
- Research Platform Active Ageing, University of Vienna, Vienna, Austria
- Department of Nutritional Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria
- *Correspondence: Bernhard Franzke
| | - Andrea Bileck
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Sandra Unterberger
- Research Platform Active Ageing, University of Vienna, Vienna, Austria
- Centre for Sport Science and University Sports, University of Vienna, Vienna, Austria
| | - Rudolf Aschauer
- Research Platform Active Ageing, University of Vienna, Vienna, Austria
- Centre for Sport Science and University Sports, University of Vienna, Vienna, Austria
| | - Patrick A. Zöhrer
- Research Platform Active Ageing, University of Vienna, Vienna, Austria
- Department of Nutritional Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Agnes Draxler
- Department of Nutritional Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Eva-Maria Strasser
- Karl Landsteiner Institute for Remobilization and Functional Health/Institute for Physical Medicine and Rehabilitation, Kaiser Franz Joseph Hospital, Social Medical Center South, Vienna, Austria
| | - Barbara Wessner
- Research Platform Active Ageing, University of Vienna, Vienna, Austria
- Centre for Sport Science and University Sports, University of Vienna, Vienna, Austria
| | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Karl-Heinz Wagner
- Research Platform Active Ageing, University of Vienna, Vienna, Austria
- Department of Nutritional Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| |
Collapse
|
8
|
Bogdanova M, Zabirnyk A, Malashicheva A, Semenova D, Kvitting JPE, Kaljusto ML, Perez MDM, Kostareva A, Stensløkken KO, Sullivan GJ, Rutkovskiy A, Vaage J. Models and Techniques to Study Aortic Valve Calcification in Vitro, ex Vivo and in Vivo. An Overview. Front Pharmacol 2022; 13:835825. [PMID: 35721220 PMCID: PMC9203042 DOI: 10.3389/fphar.2022.835825] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/29/2022] [Indexed: 11/23/2022] Open
Abstract
Aortic valve stenosis secondary to aortic valve calcification is the most common valve disease in the Western world. Calcification is a result of pathological proliferation and osteogenic differentiation of resident valve interstitial cells. To develop non-surgical treatments, the molecular and cellular mechanisms of pathological calcification must be revealed. In the current overview, we present methods for evaluation of calcification in different ex vivo, in vitro and in vivo situations including imaging in patients. The latter include echocardiography, scanning with computed tomography and magnetic resonance imaging. Particular emphasis is on translational studies of calcific aortic valve stenosis with a special focus on cell culture using human primary cell cultures. Such models are widely used and suitable for screening of drugs against calcification. Animal models are presented, but there is no animal model that faithfully mimics human calcific aortic valve disease. A model of experimentally induced calcification in whole porcine aortic valve leaflets ex vivo is also included. Finally, miscellaneous methods and aspects of aortic valve calcification, such as, for instance, biomarkers are presented.
Collapse
Affiliation(s)
- Maria Bogdanova
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Arsenii Zabirnyk
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Research and Development, Division of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway
| | - Anna Malashicheva
- Institute of Cytology, Russian Academy of Sciences, Saint Petersburg, Russia
| | - Daria Semenova
- Institute of Cytology, Russian Academy of Sciences, Saint Petersburg, Russia
| | | | - Mari-Liis Kaljusto
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | | | - Anna Kostareva
- Almazov National Medical Research Centre, Saint Petersburg, Russia.,Department of Woman and Children Health, Karolinska Institute, Stockholm, Sweden
| | - Kåre-Olav Stensløkken
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Gareth J Sullivan
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Norwegian Center for Stem Cell Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,Institute of Immunology, Oslo University Hospital, Oslo, Norway.,Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Pediatric Research, Oslo University Hospital, Oslo, Norway
| | - Arkady Rutkovskiy
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Pulmonary Diseases, Oslo University Hospital, Oslo, Norway
| | - Jarle Vaage
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Research and Development, Division of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
9
|
Greenberg HZE, Zhao G, Shah AM, Zhang M. Role of oxidative stress in calcific aortic valve disease and its therapeutic implications. Cardiovasc Res 2022; 118:1433-1451. [PMID: 33881501 PMCID: PMC9074995 DOI: 10.1093/cvr/cvab142] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 04/19/2021] [Indexed: 12/12/2022] Open
Abstract
Calcific aortic valve disease (CAVD) is the end result of active cellular processes that lead to the progressive fibrosis and calcification of aortic valve leaflets. In western populations, CAVD is a significant cause of cardiovascular morbidity and mortality, and in the absence of effective drugs, it will likely represent an increasing disease burden as populations age. As there are currently no pharmacological therapies available for preventing, treating, or slowing the development of CAVD, understanding the mechanisms underlying the initiation and progression of the disease is important for identifying novel therapeutic targets. Recent evidence has emerged of an important causative role for reactive oxygen species (ROS)-mediated oxidative stress in the pathophysiology of CAVD, inducing the differentiation of valve interstitial cells into myofibroblasts and then osteoblasts. In this review, we focus on the roles and sources of ROS driving CAVD and consider their potential as novel therapeutic targets for this debilitating condition.
Collapse
Affiliation(s)
- Harry Z E Greenberg
- Department of Cardiology, Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Guoan Zhao
- Department of Cardiology, The First Affiliated Hospital of Xinxiang Medical University, Heart Center of Xinxiang Medical University, Henan, China
| | - Ajay M Shah
- Department of Cardiology, Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Min Zhang
- Department of Cardiology, Cardiovascular Division, King's College London British Heart Foundation Centre of Research Excellence, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
10
|
Apolipoproteins and lipoprotein(a) as factors modulating fibrin clot properties in patients with severe aortic stenosis. Atherosclerosis 2022; 344:49-56. [DOI: 10.1016/j.atherosclerosis.2022.01.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/15/2021] [Accepted: 01/14/2022] [Indexed: 01/29/2023]
|
11
|
Mourino-Alvarez L, Corbacho-Alonso N, Sastre-Oliva T, Corros-Vicente C, Solis J, Tejerina T, Padial LR, Barderas MG. Diabetes Mellitus and Its Implications in Aortic Stenosis Patients. Int J Mol Sci 2021; 22:ijms22126212. [PMID: 34207517 PMCID: PMC8227301 DOI: 10.3390/ijms22126212] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 12/18/2022] Open
Abstract
Aortic stenosis (AS) and diabetes mellitus (DM) are both progressive diseases that if left untreated, result in significant morbidity and mortality. Several studies revealed that the prevalence of DM is substantially higher in patients with AS and, thus, the progression from mild to severe AS is greater in those patients with DM. DM and common comorbidities associated with both diseases, DM and AS, increase patient management complexity and make aortic valve replacement the only effective treatment. For that reason, a better understanding of the pathogenesis underlying both these diseases and the relationships between them is necessary to design more appropriate preventive and therapeutic approaches. In this review, we provided an overview of the main aspects of the relationship between AS and DM, including common comorbidities and risk factors. We also discuss the established treatments/therapies in patients with AS and DM.
Collapse
Affiliation(s)
- Laura Mourino-Alvarez
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos (HNP), SESCAM, 45071 Toledo, Spain; (L.M.-A.); (N.C.-A.); (T.S.-O.); (C.C.-V.)
| | - Nerea Corbacho-Alonso
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos (HNP), SESCAM, 45071 Toledo, Spain; (L.M.-A.); (N.C.-A.); (T.S.-O.); (C.C.-V.)
| | - Tamara Sastre-Oliva
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos (HNP), SESCAM, 45071 Toledo, Spain; (L.M.-A.); (N.C.-A.); (T.S.-O.); (C.C.-V.)
| | - Cecilia Corros-Vicente
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos (HNP), SESCAM, 45071 Toledo, Spain; (L.M.-A.); (N.C.-A.); (T.S.-O.); (C.C.-V.)
| | - Jorge Solis
- Department of Cardiology, Hospital Universitario 12 de Octubre and Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Atria Clinic, 28009 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (J.S.); or (M.G.B.); Fax: +34-925247745 (M.G.B.)
| | - Teresa Tejerina
- Department of Pharmacology, School of Medicine, Universidad Complutense, 28040 Madrid, Spain;
| | - Luis R. Padial
- Department of Cardiology, Hospital Virgen de la Salud, SESCAM, 45004 Toledo, Spain;
| | - Maria G. Barderas
- Department of Vascular Physiopathology, Hospital Nacional de Paraplejicos (HNP), SESCAM, 45071 Toledo, Spain; (L.M.-A.); (N.C.-A.); (T.S.-O.); (C.C.-V.)
- Correspondence: (J.S.); or (M.G.B.); Fax: +34-925247745 (M.G.B.)
| |
Collapse
|
12
|
Tosu AR, Kalyoncuoglu M, Biter Hİ, Cakal S, Selcuk M, Çinar T, Belen E, Can MM. Prognostic Value of Systemic Immune-Inflammation Index for Major Adverse Cardiac Events and Mortality in Severe Aortic Stenosis Patients after TAVI. ACTA ACUST UNITED AC 2021; 57:medicina57060588. [PMID: 34201104 PMCID: PMC8228319 DOI: 10.3390/medicina57060588] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/03/2021] [Indexed: 12/20/2022]
Abstract
Background and objectives: In this study, we aimed to evaluate whether the systemic immune-inflammation index (SII) has a prognostic value for major adverse cardiac events (MACEs), including stroke, re-hospitalization, and short-term all-cause mortality at 6 months, in aortic stenosis (AS) patients who underwent transcatheter aortic valve implantation (TAVI). Materials and Methods: A total of 120 patients who underwent TAVI due to severe AS were retrospectively included in our study. The main outcome of the study was MACEs and short-term all-cause mortality at 6 months. Results: The SII was found to be higher in TAVI patients who developed MACEs than in those who did not develop them. Multivariate Cox regression analysis revealed that the SII (HR: 1.002, 95%CI: 1.001–1.003, p < 0.01) was an independent predictor of MACEs in AS patients after TAVI. The optimal value of the SII for MACEs in AS patients following TAVI was >1.056 with 94% sensitivity and 96% specificity (AUC (the area under the curve): 0.960, p < 0.01). We noted that the AUC value of SII in predicting MACEs was significantly higher than the AUC value of the C-reactive protein (AUC: 0.960 vs. AUC: 0.714, respectively). Conclusions: This is the first study to show that high pre-procedural SII may have a predictive value for MACEs and short-term mortality in AS patients undergoing TAVI.
Collapse
Affiliation(s)
- Aydin Rodi Tosu
- Department of Cardiology, Haseki Training and Research Hospital, University of Health Sciences, 34130 Istanbul, Turkey; (A.R.T.); (M.K.); (H.İ.B.); (S.C.); (E.B.); (M.M.C.)
| | - Muhsin Kalyoncuoglu
- Department of Cardiology, Haseki Training and Research Hospital, University of Health Sciences, 34130 Istanbul, Turkey; (A.R.T.); (M.K.); (H.İ.B.); (S.C.); (E.B.); (M.M.C.)
| | - Halil İbrahim Biter
- Department of Cardiology, Haseki Training and Research Hospital, University of Health Sciences, 34130 Istanbul, Turkey; (A.R.T.); (M.K.); (H.İ.B.); (S.C.); (E.B.); (M.M.C.)
| | - Sinem Cakal
- Department of Cardiology, Haseki Training and Research Hospital, University of Health Sciences, 34130 Istanbul, Turkey; (A.R.T.); (M.K.); (H.İ.B.); (S.C.); (E.B.); (M.M.C.)
| | - Murat Selcuk
- Department of Cardiology, Sultan II. Abdülhamid Han Training and Research Hospital, University of Health Sciences, 34668 Istanbul, Turkey;
| | - Tufan Çinar
- Department of Cardiology, Sultan II. Abdülhamid Han Training and Research Hospital, University of Health Sciences, 34668 Istanbul, Turkey;
- Correspondence: ; Tel.: +90-216-542-2020; Fax: +90-216-542-2010
| | - Erdal Belen
- Department of Cardiology, Haseki Training and Research Hospital, University of Health Sciences, 34130 Istanbul, Turkey; (A.R.T.); (M.K.); (H.İ.B.); (S.C.); (E.B.); (M.M.C.)
| | - Mehmet Mustafa Can
- Department of Cardiology, Haseki Training and Research Hospital, University of Health Sciences, 34130 Istanbul, Turkey; (A.R.T.); (M.K.); (H.İ.B.); (S.C.); (E.B.); (M.M.C.)
| |
Collapse
|
13
|
Abstract
Calcific aortic valve disease sits at the confluence of multiple world-wide epidemics of aging, obesity, diabetes, and renal dysfunction, and its prevalence is expected to nearly triple over the next 3 decades. This is of particularly dire clinical relevance, as calcific aortic valve disease can progress rapidly to aortic stenosis, heart failure, and eventually premature death. Unlike in atherosclerosis, and despite the heavy clinical toll, to date, no pharmacotherapy has proven effective to halt calcific aortic valve disease progression, with invasive and costly aortic valve replacement representing the only treatment option currently available. This substantial gap in care is largely because of our still-limited understanding of both normal aortic valve biology and the key regulatory mechanisms that drive disease initiation and progression. Drug discovery is further hampered by the inherent intricacy of the valvular microenvironment: a unique anatomic structure, a complex mixture of dynamic biomechanical forces, and diverse and multipotent cell populations collectively contributing to this currently intractable problem. One promising and rapidly evolving tactic is the application of multiomics approaches to fully define disease pathogenesis. Herein, we summarize the application of (epi)genomics, transcriptomics, proteomics, and metabolomics to the study of valvular heart disease. We also discuss recent forays toward the omics-based characterization of valvular (patho)biology at single-cell resolution; these efforts promise to shed new light on cellular heterogeneity in healthy and diseased valvular tissues and represent the potential to efficaciously target and treat key cell subpopulations. Last, we discuss systems biology- and network medicine-based strategies to extract meaning, mechanisms, and prioritized drug targets from multiomics datasets.
Collapse
Affiliation(s)
- Mark C. Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Schlieren, CH
| | - Thomas F. Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, CH
- Heart Division, Royal Brompton & Harefield Hospitals, London, UK
- National Heart and Lung Institute, Imperial College, London, UK
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
14
|
Metabolomics in Severe Aortic Stenosis Reveals Intermediates of Nitric Oxide Synthesis as Most Distinctive Markers. Int J Mol Sci 2021; 22:ijms22073569. [PMID: 33808189 PMCID: PMC8037707 DOI: 10.3390/ijms22073569] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Calcific aortic valve disease (CAVD) is a rapidly growing global health problem with an estimated 12.6 million cases globally in 2017 and a 112% increase of deaths since 1990 due to aging and population growth. CAVD may develop into aortic stenosis (AS) by progressive narrowing of the aortic valve. AS is underdiagnosed, and if treatment by aortic valve replacement (AVR) is delayed, this leads to poor recovery of cardiac function, absence of symptomatic improvement and marked increase of mortality. Considering the current limitations to define the stage of AS-induced cardiac remodeling, there is need for a novel method to aid in the diagnosis of AS and timing of intervention, which may be found in metabolomics profiling of patients. METHODS Serum samples of nine healthy controls and 10 AS patients before and after AVR were analyzed by untargeted mass spectrometry. Multivariate modeling was performed to determine a metabolic profile of 30 serum metabolites which distinguishes AS patients from controls. Human cardiac microvascular endothelial cells (CMECs) were incubated with serum of the AS patients and then stained for ICAM-1 with Western Blot to analyze the effect of AS patient serum on endothelial cell activation. RESULTS The top 30 metabolic profile strongly distinguishes AS patients from healthy controls and includes 17 metabolites related to nitric oxide metabolism and 12 metabolites related to inflammation, in line with the known pathomechanism for calcific aortic valve disease. Nine metabolites correlate strongly with left ventricular mass, of which three show reversal back to control values after AVR. Western blot analysis of CMECs incubated with AS patient sera shows a significant reduction (14%) in ICAM-1 in AS samples taken after AVR compared to AS patient sera before AVR. CONCLUSION Our study defined a top 30 metabolic profile with biological and clinical relevance, which may be used as blood biomarker to identify AS patients in need of cardiac surgery. Future studies are warranted in patients with mild-to-moderate AS to determine if these metabolites reflect disease severity and can be used to identify AS patients in need of cardiac surgery.
Collapse
|
15
|
Qian Y, Li L, Sun Z, Liu J, Yuan W, Wang Z. A multi-omics view of the complex mechanism of vascular calcification. Biomed Pharmacother 2021; 135:111192. [PMID: 33401220 DOI: 10.1016/j.biopha.2020.111192] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 12/19/2020] [Accepted: 12/26/2020] [Indexed: 02/07/2023] Open
Abstract
Vascular calcification is a high incidence and high risk disease with increasing morbidity and high mortality, which is considered the consequence of smooth muscle cell transdifferentiation initiating the mechanism of accumulation of hydroxyl calcium phosphate. Vascular calcification is also thought to be strongly associated with poor outcomes in diabetes and chronic kidney disease. Numerous studies have been accomplished; however, the specific mechanism of the disease remains unclear. Development of the genome project enhanced the understanding of life science and has entered the post-genomic era resulting in a variety of omics techniques used in studies and a large amount of available data; thus, a new perspective on data analysis has been revealed. Omics has a broader perspective and is thus advantageous over a single pathway analysis in the study of complex vascular calcification mechanisms. This paper reviews in detail various omics studies including genomics, proteomics, transcriptomics, metabolomics and multiple group studies on vascular calcification. Advances and deficiencies in the use of omics to study vascular calcification are presented in a comprehensive view. We also review the methodology of the omics studies and omics data analysis and processing. In addition, the methodology and data processing presented here can be applied to other areas. An omics landscape perspective across the boundaries between genomics, transcriptomics, proteomics and metabolomics is used to examine the mechanisms of vascular calcification. The perspective combined with various technologies also provides a direction for the subsequent exploration of clinical significance.
Collapse
Affiliation(s)
- Yongjiang Qian
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, China
| | - Lihua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, China
| | - Zhen Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, China
| | - Jia Liu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, China
| | - Wei Yuan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, China.
| |
Collapse
|
16
|
Aikawa E, Blaser MC. 2020 Jeffrey M. Hoeg Award Lecture: Calcifying Extracellular Vesicles as Building Blocks of Microcalcifications in Cardiovascular Disorders. Arterioscler Thromb Vasc Biol 2021; 41:117-127. [PMID: 33115271 PMCID: PMC7832175 DOI: 10.1161/atvbaha.120.314704] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/11/2020] [Indexed: 12/12/2022]
Abstract
Cardiovascular calcification is an insidious form of ectopic tissue mineralization that presents as a frequent comorbidity of atherosclerosis, aortic valve stenosis, diabetes, renal failure, and chronic inflammation. Calcification of the vasculature and heart valves contributes to mortality in these diseases. An inability to clinically image or detect early microcalcification coupled with an utter lack of pharmaceutical therapies capable of inhibiting or regressing entrenched and detectable macrocalcification has led to a prominent and deadly gap in care for a growing portion of our rapidly aging population. Recognition of this mounting concern has arisen over the past decade and led to a series of revolutionary works that has begun to pull back the curtain on the pathogenesis, mechanistic basis, and causative drivers of cardiovascular calcification. Central to this progress is the discovery that calcifying extracellular vesicles act as active precursors of cardiovascular microcalcification in diverse vascular beds. More recently, the omics revolution has resulted in the collection and quantification of vast amounts of molecular-level data. As the field has become poised to leverage these resources for drug discovery, new means of deriving relevant biological insights from these rich and complex datasets have come into focus through the careful application of systems biology and network medicine approaches. As we look onward toward the next decade, we envision a growing need to standardize approaches to study this complex and multifaceted clinical problem and expect that a push to translate mechanistic findings into therapeutics will begin to finally provide relief for those impacted by this disease.
Collapse
Affiliation(s)
- Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark C. Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Mansour A, Darwiche W, Yaker L, Da Nascimento S, Gomila C, Rossi C, Jung V, Sonnet P, Kamel S, Guerrera IC, Boullier A, Ausseil J. GFOGER Peptide Modifies the Protein Content of Extracellular Vesicles and Inhibits Vascular Calcification. Front Cell Dev Biol 2020; 8:589761. [PMID: 33330469 PMCID: PMC7734313 DOI: 10.3389/fcell.2020.589761] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/26/2020] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE Vascular calcification (VC) is an active process during which vascular smooth muscle cells (VSMCs) undergo an osteogenic switch and release extracellular vesicles (EVs). In turn, the EVs serve as calcification foci via interaction with type 1 collagen (COL1). We recently showed that a specific, six-amino-acid repeat (GFOGER) in the sequence of COL1 was involved in the latter's interaction with integrins expressed on EVs. Our main objective was to test the GFOGER ability to inhibit VC. APPROACH We synthesized the GFOGER peptide and tested its ability to inhibit the inorganic phosphate (Pi)-induced calcification of VSMCs and aortic rings. Using mass spectrometry, we studied GFOGER's effect on the protein composition of EVs released from Pi-treated VSMCs. RESULTS Calcification of mouse VSMCs (MOVAS-1 cells), primary human VSMCs, and rat aortic rings was lower in the presence of GFOGER than with Pi alone (with relative decreases of 66, 58, and 91%, respectively; p < 0.001 for all) (no effect was observed with the scramble peptide GOERFG). A comparative proteomic analysis of EVs released from MOVAS-1 cells in the presence or absence of Pi highlighted significant differences in EVs' protein content. Interestingly, the expression of some of the EVs' proteins involved in the calcification process (such as osteogenic markers, TANK-binding kinase 1, and casein kinase II) was diminished in the presence of GFOGER peptide (data are available via ProteomeXchange with identifier PXD018169∗). The decrease of osteogenic marker expression observed in the presence of GFOGER was confirmed by q-RT-PCR analysis. CONCLUSION GFOGER peptide reduces vascular calcification by modifying the protein content of the subsequently released EVs, in particular by decreasing osteogenicswitching in VSMCs.
Collapse
Affiliation(s)
- Ali Mansour
- MP3CV-UR7517, CURS-Université de Picardie Jules Verne, Amiens, France
| | - Walaa Darwiche
- MP3CV-UR7517, CURS-Université de Picardie Jules Verne, Amiens, France
| | - Linda Yaker
- MP3CV-UR7517, CURS-Université de Picardie Jules Verne, Amiens, France
| | - Sophie Da Nascimento
- AGIR, UR4294, UFR de Pharmacie, Université de Picardie Jules Verne, Amiens, France
| | - Cathy Gomila
- MP3CV-UR7517, CURS-Université de Picardie Jules Verne, Amiens, France
| | - Claire Rossi
- Alliance Sorbonne Université, Université de Technologie de Compiègne, UMR7025 CNRS Enzyme and Cell Engineering Laboratory, Compiègne, France
| | - Vincent Jung
- Plateforme protéomique Necker, Faculté de Médecine Paris Descartes, Université de Paris – Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris, France
| | - Pascal Sonnet
- AGIR, UR4294, UFR de Pharmacie, Université de Picardie Jules Verne, Amiens, France
| | - Saïd Kamel
- MP3CV-UR7517, CURS-Université de Picardie Jules Verne, Amiens, France
- Laboratoire de Biochimie, Centre Hospitalier Universitaire d’ Amiens, Amiens, France
| | - Ida Chiara Guerrera
- Plateforme protéomique Necker, Faculté de Médecine Paris Descartes, Université de Paris – Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris, France
| | - Agnès Boullier
- MP3CV-UR7517, CURS-Université de Picardie Jules Verne, Amiens, France
- Laboratoire de Biochimie, Centre Hospitalier Universitaire d’ Amiens, Amiens, France
| | - Jérôme Ausseil
- MP3CV-UR7517, CURS-Université de Picardie Jules Verne, Amiens, France
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR1043 – CNRS UMR5282 – Université Toulouse III, Toulouse, France
- CHU Toulouse – Institut Fédératif de Biologie, Laboratoire de Biochimie, Toulouse, France
| |
Collapse
|
18
|
Miranda-Silva D, G Rodrigues P, Alves E, Rizo D, Fonseca ACRG, Lima T, Baganha F, Conceição G, Sousa C, Gonçalves A, Miranda I, Vasques-Nóvoa F, Magalhães J, Leite-Moreira A, Falcão-Pires I. Mitochondrial Reversible Changes Determine Diastolic Function Adaptations During Myocardial (Reverse) Remodeling. Circ Heart Fail 2020; 13:e006170. [PMID: 33176457 DOI: 10.1161/circheartfailure.119.006170] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Often, pressure overload-induced myocardial remodeling does not undergo complete reverse remodeling after decreasing afterload. Recently, mitochondrial abnormalities and oxidative stress have been successively implicated in the pathogenesis of several chronic pressure overload cardiac diseases. Therefore, we aim to clarify the myocardial energetic dysregulation in (reverse) remodeling, mainly focusing on the mitochondria. METHODS Thirty-five Wistar Han male rats randomly underwent sham or ascending (supravalvular) aortic banding procedure. Echocardiography revealed that banding induced concentric hypertrophy and diastolic dysfunction (early diastolic transmitral flow velocity to peak early-diastolic annular velocity ratio, E/E': sham, 13.6±2.1, banding, 18.5±4.1, P=0.014) accompanied by increased oxidative stress (dihydroethidium fluorescence: sham, 1.6×108±6.1×107, banding, 2.6×108±4.5×107, P<0.001) and augmented mitochondrial function. After 8 to 9 weeks, half of the banding animals underwent overload relief by an aortic debanding surgery (n=10). RESULTS Two weeks later, hypertrophy decreased with the decline of oxidative stress (dihydroethidium fluorescence: banding, 2.6×108±4.5×107, debanding, 1.96×108±6.8×107, P<0.001) and diastolic dysfunction improved simultaneously (E/E': banding, 18.5±4.1, debanding, 15.1±1.8, P=0.029). The reduction of energetic demands imposed by overload relief allowed the mitochondria to reduce its activity and myocardial levels of phosphocreatine, phosphocreatine/ATP, and ATP/ADP to normalize in debanding towards sham values (phosphocreatine: sham, 38.4±7.4, debanding, 35.6±8.7, P=0.71; phosphocreatine/ATP: sham, 1.22±0.23 debanding, 1.11±0.24, P=0.59; ATP/ADP: sham, 6.2±0.9, debanding, 5.6±1.6, P=0.66). Despite the decreased mitochondrial area, complex III and V expression increased in debanding compared with sham or banding. Autophagy and mitophagy-related markers increased in banding and remained higher in debanding rats. CONCLUSIONS During compensatory and maladaptive hypertrophy, mitochondria become more active. However, as the disease progresses, the myocardial energetic demands increase and the myocardium becomes energy deficient. During reverse remodeling, the concomitant attenuation of cardiac hypertrophy and oxidative stress allowed myocardial energetics, left ventricle hypertrophy, and diastolic dysfunction to recover. Autophagy and mitophagy are probably involved in the myocardial adaptation to overload and to unload. We conclude that these mitochondrial reversible changes underlie diastolic function adaptations during myocardial (reverse) remodeling.
Collapse
Affiliation(s)
- Daniela Miranda-Silva
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Patrícia G Rodrigues
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Estela Alves
- LaMetEX, Laboratory of Metabolism and Exercise (E.A., D.R., J.M.).,CIAFEL, Research Centre in Physical Activity, Health and Leisure, Faculty of Sports, Portugal (E.A., D.R., J.M.)
| | - David Rizo
- LaMetEX, Laboratory of Metabolism and Exercise (E.A., D.R., J.M.).,CIAFEL, Research Centre in Physical Activity, Health and Leisure, Faculty of Sports, Portugal (E.A., D.R., J.M.)
| | - Ana Catarina R G Fonseca
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Portugal (A.C.R.G.F.)
| | - Tânia Lima
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Fabiana Baganha
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Gloria Conceição
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Cláudia Sousa
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Alexandre Gonçalves
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Isabel Miranda
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Francisco Vasques-Nóvoa
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - José Magalhães
- LaMetEX, Laboratory of Metabolism and Exercise (E.A., D.R., J.M.).,CIAFEL, Research Centre in Physical Activity, Health and Leisure, Faculty of Sports, Portugal (E.A., D.R., J.M.)
| | - Adelino Leite-Moreira
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Inês Falcão-Pires
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| |
Collapse
|
19
|
Integrative Multi-Omics Analysis in Calcific Aortic Valve Disease Reveals a Link to the Formation of Amyloid-Like Deposits. Cells 2020; 9:cells9102164. [PMID: 32987857 PMCID: PMC7600313 DOI: 10.3390/cells9102164] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 09/16/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
Calcific aortic valve disease (CAVD) is the most prevalent valvular heart disease in the developed world, yet no pharmacological therapy exists. Here, we hypothesize that the integration of multiple omic data represents an approach towards unveiling novel molecular networks in CAVD. Databases were searched for CAVD omic studies. Differentially expressed molecules from calcified and control samples were retrieved, identifying 32 micro RNAs (miRNA), 596 mRNAs and 80 proteins. Over-representation pathway analysis revealed platelet degranulation and complement/coagulation cascade as dysregulated pathways. Multi-omics integration of overlapping proteome/transcriptome molecules, with the miRNAs, identified a CAVD protein–protein interaction network containing seven seed genes (apolipoprotein A1 (APOA1), hemoglobin subunit β (HBB), transferrin (TF), α-2-macroglobulin (A2M), transforming growth factor β-induced protein (TGFBI), serpin family A member 1 (SERPINA1), lipopolysaccharide binding protein (LBP), inter-α-trypsin inhibitor heavy chain 3 (ITIH3) and immunoglobulin κ constant (IGKC)), four input miRNAs (miR-335-5p, miR-3663-3p, miR-21-5p, miR-93-5p) and two connector genes (amyloid beta precursor protein (APP) and transthyretin (TTR)). In a metabolite–gene–disease network, Alzheimer’s disease exhibited the highest degree of betweenness. To further strengthen the associations based on the multi-omics approach, we validated the presence of APP and TTR in calcified valves from CAVD patients by immunohistochemistry. Our study suggests a novel molecular CAVD network potentially linked to the formation of amyloid-like structures. Further investigations on the associated mechanisms and therapeutic potential of targeting amyloid-like deposits in CAVD may offer significant health benefits.
Collapse
|
20
|
Liu Y, Li Y, Zhang T, Zhao H, Fan S, Cai X, Liu Y, Li Z, Gao S, Li Y, Yu C. Analysis of biomarkers and metabolic pathways in patients with unstable angina based on ultra‑high‑performance liquid chromatography‑quadrupole time‑of‑flight mass spectrometry. Mol Med Rep 2020; 22:3862-3872. [PMID: 32901869 PMCID: PMC7533448 DOI: 10.3892/mmr.2020.11476] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 06/26/2020] [Indexed: 12/29/2022] Open
Abstract
Unstable angina (UA) is a coronary disease with a high mortality and morbidity worldwide. The present study aimed to use non-invasive techniques to identify urine biomarkers in patients with UA, so as to provide more information for the early diagnosis and treatment of the disease. Based on metabolomics, urine samples from 28 patients with UA and 28 healthy controls (HCs) were analyzed using ultra-high-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF/MS). A total of 16 significant biomarkers that could distinguish between patients with UA and HCs, including D-glucuronic acid, creatinine, succinic acid and N-acetylneuraminic acid, were identified. The major metabolic pathways associated with UA were subsequently analyzed by non-targeted metabolomics. The results demonstrated that amino acid and energy metabolism, fatty acid metabolism, purine metabolism and steroid hormone biosynthetic metabolism may serve important roles in UA. The results of the current study may provide a theoretical basis for the early diagnosis of UA and novel treatment strategies for clinicians. The trial was registered with the Chinese Clinical Trial Registration Center (registration no. ChiCTR-ROC-17013957) at Tianjin University of Traditional Chinese Medicine.
Collapse
Affiliation(s)
- Yuechen Liu
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Yue Li
- Research Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Tianpu Zhang
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Huan Zhao
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Simiao Fan
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Xuemeng Cai
- Research Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Yijia Liu
- Research Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Zhu Li
- Research Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Shan Gao
- Research Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Yubo Li
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Chunquan Yu
- Research Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| |
Collapse
|
21
|
Patient Management in Aortic Stenosis: Towards Precision Medicine Through Protein Analysis, Imaging and Diagnostic Tests. J Clin Med 2020; 9:jcm9082421. [PMID: 32731585 PMCID: PMC7463596 DOI: 10.3390/jcm9082421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/22/2020] [Accepted: 07/24/2020] [Indexed: 01/12/2023] Open
Abstract
Aortic stenosis is the most frequent valvular disease in developed countries. It progresses from mild fibrocalcific leaflet changes to a more severe leaflet calcification at the end stages of the disease. Unfortunately, symptoms of aortic stenosis are unspecific and only appear when it is too late, complicating patients' management. The global impact of aortic stenosis is increasing due to the growing elderly population. The disease supposes a great challenge because of the multiple comorbidities of these patients. Nowadays, the only effective treatment is valve replacement, which has a high cost in both social and economic terms. For that reason, it is crucial to find potential diagnostic, prognostic and therapeutic indicators that could help us to detect this disease in its earliest stages. In this article, we comprehensively review several key observations and translational studies related to protein markers that are promising for being implemented in the clinical field as well as a discussion about the role of precision medicine in aortic stenosis.
Collapse
|
22
|
Impaired Fibrinolysis in Patients with Isolated Aortic Stenosis is Associated with Enhanced Oxidative Stress. J Clin Med 2020; 9:jcm9062002. [PMID: 32630544 PMCID: PMC7355626 DOI: 10.3390/jcm9062002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/18/2020] [Accepted: 06/23/2020] [Indexed: 11/17/2022] Open
Abstract
Aortic stenosis (AS) has been associated with impaired fibrinolysis and increased oxidative stress. This study aimed to investigate whether oxidative stress could alter fibrin clot properties in AS. We studied 173 non-diabetic patients, aged 51–79 years, with isolated AS. We measured plasma protein carbonylation (PC) and thiobarbituric acid reactive substances (TBARS), along with plasma clot permeability (Ks), thrombin generation, and fibrinolytic efficiency, which were evaluated by two assays: clot lysis time (CLT) and lysis time (Lys50). Coagulation factors and fibrinolytic proteins were also determined. Plasma PC showed an association with AS severity, reflected by the aortic valve area and the mean and maximum aortic gradients. Plasma PC was positively correlated with CLT, Lys50, plasminogen activator inhibitor-1 (PAI-1), and tissue factor (TF) antigens. TBARS were positively correlated with maximum aortic gradient, Lys50, and TF antigen. Regression analysis showed that PC predicted prolonged CLT (>104 min; odds ratio (OR) 6.41, 95% confidence interval (CI) 2.58–17.83, p < 0.001) and Lys50 (>565 s; OR 5.83, 95% CI 2.23–15.21, p < 0.001). Multivariate regression analysis showed that mean aortic gradient, PC, α2-antiplasmin, PAI-1, and triglycerides were predictors of prolonged CLT, while PC, α2-antiplasmin, and fibrinogen were predictors of Lys50. Our findings suggest that elevated oxidative stress contributes to impaired fibrinolysis in AS and is associated with AS severity.
Collapse
|
23
|
Kontostathi G, Makridakis M, Zoidakis J, Vlahou A. Applications of multiple reaction monitoring targeted proteomics assays in human plasma. Expert Rev Mol Diagn 2019; 19:499-515. [PMID: 31057016 DOI: 10.1080/14737159.2019.1615448] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Multiple (or selected) reaction monitoring-mass spectrometry (MRM/SRM) is a targeted proteomic method that can be used for relative and absolute quantification. Multiple reports exist supporting the potential of the approach in proteomic biomarker validation. Areas covered: To get an overview of the applications of MRM in protein quantification in plasma, a search in MedLine/PubMed was performed using the keywords: 'MRM/SRM plasma proteomic/proteomics/proteome'. The retrieved studies were further filtered to focus on disease biomarkers and the main results are summarized. Expert opinion: MRM is increasingly employed for the quantification of both well-established but also newly discovered putative biomarkers and occasionally their post-translationally modified forms in plasma. Fractionation is regularly required for the detection of low abundance proteins. Standardized procedures to facilitate assay establishment and marker quantification have been proposed and, in few cases, implemented. Nevertheless, in most cases, absolute quantification is not performed. To advance, multiple technical issues including the regular use of standard labeled peptides and appropriate quality controls to monitor assay performance should be considered. Additionally, clinical aspects involving careful study design to address biomarker clinical use should also be considered.
Collapse
Affiliation(s)
- Georgia Kontostathi
- a Biotechnology Division , Biomedical Research Foundation, Academy of Athens (BRFAA) , Athens , Greece
| | - Manousos Makridakis
- a Biotechnology Division , Biomedical Research Foundation, Academy of Athens (BRFAA) , Athens , Greece
| | - Jerome Zoidakis
- a Biotechnology Division , Biomedical Research Foundation, Academy of Athens (BRFAA) , Athens , Greece
| | - Antonia Vlahou
- a Biotechnology Division , Biomedical Research Foundation, Academy of Athens (BRFAA) , Athens , Greece
| |
Collapse
|
24
|
Opdebeeck B, Maudsley S, Azmi A, De Maré A, De Leger W, Meijers B, Verhulst A, Evenepoel P, D'Haese PC, Neven E. Indoxyl Sulfate and p-Cresyl Sulfate Promote Vascular Calcification and Associate with Glucose Intolerance. J Am Soc Nephrol 2019; 30:751-766. [PMID: 30940651 DOI: 10.1681/asn.2018060609] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 02/13/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Protein-bound uremic toxins indoxyl sulfate (IS) and p-cresyl sulfate (PCS) have been associated with cardiovascular morbidity and mortality in patients with CKD. However, direct evidence for a role of these toxins in CKD-related vascular calcification has not been reported. METHODS To study early and late vascular alterations by toxin exposure, we exposed CKD rats to vehicle, IS (150 mg/kg per day), or PCS (150 mg/kg per day) for either 4 days (short-term exposure) or 7 weeks (long-term exposure). We also performed unbiased proteomic analyses of arterial samples coupled to functional bioinformatic annotation analyses to investigate molecular signaling events associated with toxin-mediated arterial calcification. RESULTS Long-term exposure to either toxin at serum levels similar to those experienced by patients with CKD significantly increased calcification in the aorta and peripheral arteries. Our analyses revealed an association between calcification events, acute-phase response signaling, and coagulation and glucometabolic signaling pathways, whereas escape from toxin-induced calcification was linked with liver X receptors and farnesoid X/liver X receptor signaling pathways. Additional metabolic linkage to these pathways revealed that IS and PCS exposure engendered a prodiabetic state evidenced by elevated resting glucose and reduced GLUT1 expression. Short-term exposure to IS and PCS (before calcification had been established) showed activation of inflammation and coagulation signaling pathways in the aorta, demonstrating that these signaling pathways are causally implicated in toxin-induced arterial calcification. CONCLUSIONS In CKD, both IS and PCS directly promote vascular calcification via activation of inflammation and coagulation pathways and were strongly associated with impaired glucose homeostasis.
Collapse
Affiliation(s)
- Britt Opdebeeck
- Laboratory of Pathophysiology, Department of Biomedical Sciences
| | - Stuart Maudsley
- Receptor Biology Lab, Department of Biomedical Sciences, and.,Translational Neurobiology Group, Flanders Institute of Biotechnology Center for Molecular Neurology, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Abdelkrim Azmi
- Translational Neurobiology Group, Flanders Institute of Biotechnology Center for Molecular Neurology, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Annelies De Maré
- Laboratory of Pathophysiology, Department of Biomedical Sciences
| | - Wout De Leger
- Division of Molecular Design and Synthesis, Department of Chemistry and
| | - Bjorn Meijers
- Division of Internal Medicine, Nephrology, University Hospitals Leuven, Leuven, Belgium.,Laboratory of Nephrology, Department of Immunology and Microbiology, Catholic University of Leuven, Leuven, Belgium; and
| | - Anja Verhulst
- Laboratory of Pathophysiology, Department of Biomedical Sciences
| | - Pieter Evenepoel
- Division of Internal Medicine, Nephrology, University Hospitals Leuven, Leuven, Belgium.,Laboratory of Nephrology, Department of Immunology and Microbiology, Catholic University of Leuven, Leuven, Belgium; and
| | | | - Ellen Neven
- Laboratory of Pathophysiology, Department of Biomedical Sciences
| |
Collapse
|
25
|
Jiang L, Wang J, Li R, Fang ZM, Zhu XH, Yi X, Lan H, Wei X, Jiang DS. Disturbed energy and amino acid metabolism with their diagnostic potential in mitral valve disease revealed by untargeted plasma metabolic profiling. Metabolomics 2019; 15:57. [PMID: 30937548 DOI: 10.1007/s11306-019-1518-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/25/2019] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Mitral valve disease (MVD), including mitral valve regurgitation (MR) and mitral valve stenosis (MS), is a chronic and progressive cardiac malady. However, the metabolic alterations in MVD is not well-understood till now. The current gold standard diagnostic test, transthoracic echocardiography, has limitations on high-throughput measurement and lacks molecular information for early diagnosis of the disease. OBJECTIVE The present study aimed to investigate the biochemical alterations and to explore their diagnostic potential for MVD. METHODS Plasma metabolic profile derangements and their diagnostic potential were non-invasively explored in 34 MR and 20 MS patients against their corresponding controls, using high-throughput NMR-based untargeted metabolomics. RESULTS Eighteen differential metabolites were identified for MR and MS patients respectively, on the basis of multivariate and univariate data analysis, which were mainly involved in energy metabolism, amino acid metabolism, calcium metabolism and inflammation. These differential metabolites, notably the significantly down-regulated formate and lactate, showed high diagnostic potential for MVD by using Spearman's rank-order correlation analysis and ROC analysis. CONCLUSIONS To the best of our knowledge, the present study is the first one that explores the metabolic derangements and their diagnostic values in MVD patients using metabolomics. The findings indicated that metabolic disturbance occurred in MVD patients, with plasma formate and lactate emerged as important candidate biomarkers for MVD.
Collapse
Affiliation(s)
- Limiao Jiang
- Department of Epidemiology and Biostatistics, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China.
| | - Jing Wang
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Rui Li
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Ze-Min Fang
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Xue-Hai Zhu
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China
- NHC Key Laboratory of Organ Transplantation, Wuhan, China
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Xin Yi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Hongwen Lan
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, China
| | - Xiang Wei
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China
- NHC Key Laboratory of Organ Transplantation, Wuhan, China
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Ding-Sheng Jiang
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave., Wuhan, 430030, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.
- NHC Key Laboratory of Organ Transplantation, Wuhan, China.
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
26
|
Hioki H, Watanabe Y, Kozuma K, Yamamoto M, Naganuma T, Araki M, Tada N, Shirai S, Yamanaka F, Higashimori A, Mizutani K, Tabata M, Takagi K, Ueno H, Hayashida K. Effect of Serum C-Reactive Protein Level on Admission to Predict Mortality After Transcatheter Aortic Valve Implantation. Am J Cardiol 2018; 122:294-301. [PMID: 29735216 DOI: 10.1016/j.amjcard.2018.04.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/27/2018] [Accepted: 04/02/2018] [Indexed: 11/16/2022]
Abstract
The relation between C-reactive protein (CRP) level on admission and mortality after transcatheter aortic valve implantation (TAVI) remains unclear. To evaluate the impact of serum CRP level on mortality after TAVI, we assessed 1,016 patients with CRP who underwent TAVI and 538 patients with high-sensitive CRP (hs-CRP) level who underwent TAVI on admission in the OCEAN (Optimized Transcatheter Valvular Intervention)-TAVI registry. Study population was stratified into 2 groups (high/low), according to the median of CRP and hs-CRP on admission. We assessed the impact of high CRP and hs-CRP level on all-cause death after TAVI. During 2-year follow-up, all-cause death after TAVI was 9.4% in patients with CRP and 11.9% in patients with hs-CRP. Median value of serum CRP was 0.10 mg/dl in both CRP and hs-CRP. Patients with high CRP (>0.10 mg/dl) had significantly higher incidence of all-cause death compared with those with low CRP (11.5% vs 7.6%, log-rank p = 0.015). Multivariate Cox regression analysis with a time-varying covariate demonstrated that high CRP was an independent predictor of all-cause death within the first 3 months (hazard ratio 2.78, 95% CI 1.30 to 5.95) compared with from 3 months to 2 years (hazard ratio 0.80, 95% CI 0.47 to 1.36) (P for interaction = 0.008). Inversely, these results were not observed in the stratification using hs-CRP on admission. In conclusion, high CRP on admission was significantly associated with an increased risk of all-cause death after TAVI, particularly within the first 3 months after TAVI. Risk stratification using CRP may be a simple and useful strategy to identify high-risk patients who undergo TAVI.
Collapse
Affiliation(s)
- Hirofumi Hioki
- Division of Cardiology, Department of Internal Medicine, Teikyo University Hospital, Tokyo, Japan.
| | - Yusuke Watanabe
- Division of Cardiology, Department of Internal Medicine, Teikyo University Hospital, Tokyo, Japan
| | - Ken Kozuma
- Division of Cardiology, Department of Internal Medicine, Teikyo University Hospital, Tokyo, Japan
| | | | - Toru Naganuma
- Interventional Cardiology Unit, New Tokyo Hospital, Chiba, Japan
| | - Motoharu Araki
- Department of Cardiology, Saiseikai Yokohama City Eastern Hospital, Yokohama, Japan
| | - Norio Tada
- Department of Cardiovascular Center, Sendai Kosei Hospital, Sendai, Japan
| | - Shinichi Shirai
- Department of Cardiology, Kokura Memorial Hospital, Fukuoka, Japan
| | - Futoshi Yamanaka
- Department of Cardiovascular Medicine, Shonan Kamakura General Hospital, Kanmakura, Japan
| | | | - Kazuki Mizutani
- Department of Cardiovascular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Minoru Tabata
- Department of Cardiovascular Surgery, Tokyo Bay Urayasu Ichikawa Medical Center, Urayasu, Japan
| | - Kensuke Takagi
- Department of Cardiology, Ogaki Municipal Hospital, Ogaki, Japan
| | - Hiroshi Ueno
- Second Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Kentaro Hayashida
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
27
|
Mourino-Alvarez L, Baldan-Martin M, Sastre-Oliva T, Martin-Lorenzo M, Maroto AS, Corbacho-Alonso N, Rincon R, Martin-Rojas T, Lopez-Almodovar LF, Alvarez-Llamas G, Vivanco F, Padial LR, de la Cuesta F, Barderas MG. A comprehensive study of calcific aortic stenosis: from rabbit to human samples. Dis Model Mech 2018; 11:dmm.033423. [PMID: 29752279 PMCID: PMC6031362 DOI: 10.1242/dmm.033423] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 05/03/2018] [Indexed: 12/22/2022] Open
Abstract
The global incidence of calcific aortic stenosis (CAS) is increasing owing, in part, to a growing elderly population. The condition poses a great challenge to public health, because of the multiple comorbidities of these older patients. Using a rabbit model of CAS, we sought to characterize protein alterations associated with calcified valve tissue that can be ultimately measured in plasma as non-invasive biomarkers of CAS. Aortic valves from healthy and mild stenotic rabbits were analyzed by two-dimensional difference gel electrophoresis, and selected reaction monitoring was used to directly measure the differentially expressed proteins in plasma from the same rabbits to corroborate their potential as diagnostic indicators. Similar analyses were performed in plasma from human subjects, to examine the suitability of these diagnostic indicators for transfer to the clinical setting. Eight proteins were found to be differentially expressed in CAS tissue, but only three were also altered in plasma samples from rabbits and humans: transitional endoplasmic reticulum ATPase, tropomyosin α-1 chain and L-lactate dehydrogenase B chain. Results of receiver operating characteristic curves showed the discriminative power of the scores, which increased when the three proteins were analyzed as a panel. Our study shows that a molecular panel comprising three proteins related to osteoblastic differentiation could have utility as a serum CAS indicator and/or therapeutic target. Summary: Using a rabbit model of calcific aortic stenosis, we have defined a molecular panel of three proteins related to osteoblastic differentiation. Additionally, this panel has been confirmed in human samples.
Collapse
Affiliation(s)
- Laura Mourino-Alvarez
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain
| | - Montserrat Baldan-Martin
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain
| | - Tamara Sastre-Oliva
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain
| | | | - Aroa Sanz Maroto
- Department of Immunology, IIS-Fundacion Jimenez Diaz, 28040 Madrid, Spain
| | - Nerea Corbacho-Alonso
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain
| | - Raul Rincon
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain
| | - Tatiana Martin-Rojas
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain
| | | | - Gloria Alvarez-Llamas
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain
| | - Fernando Vivanco
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain
| | | | - Fernando de la Cuesta
- Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Maria Gonzalez Barderas
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain
| |
Collapse
|
28
|
|
29
|
Beneficial Effects of Galectin-3 Blockade in Vascular and Aortic Valve Alterations in an Experimental Pressure Overload Model. Int J Mol Sci 2017; 18:ijms18081664. [PMID: 28758988 PMCID: PMC5578054 DOI: 10.3390/ijms18081664] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 07/28/2017] [Accepted: 07/28/2017] [Indexed: 12/19/2022] Open
Abstract
Galectin-3 (Gal-3) is involved in cardiovascular fibrosis and aortic valve (AV) calcification. We hypothesized that Gal-3 pharmacological inhibition with modified citrus pectin (MCP) could reduce aortic and AV remodeling in normotensive rats with pressure overload (PO). Six weeks after aortic constriction, vascular Gal-3 expression was up-regulated in male Wistar rats. Gal-3 overexpression was accompanied by an increase in the aortic media layer thickness, enhanced total collagen, and augmented expression of fibrotic mediators. Further, vascular inflammatory markers as well as inflammatory cells content were greater in aorta from PO rats. MCP treatment (100 mg/kg/day) prevented the increase in Gal-3, media thickness, fibrosis, and inflammation in the aorta of PO rats. Gal-3 levels were higher in AVs from PO rats. This paralleled enhanced AV fibrosis, inflammation, as well as greater expression of calcification markers. MCP treatment prevented the increase in Gal-3 as well as fibrosis, inflammation, and calcification in AVs. Overall, Gal-3 is overexpressed in aorta and AVs from PO rats. Gal-3 pharmacological inhibition blocks aortic and AV remodeling in experimental PO. Gal-3 could be a new therapeutic approach to delay the progression and the development of aortic remodeling and AV calcification in PO.
Collapse
|
30
|
Thomassen HK, Cioffi G, Gerdts E, Einarsen E, Midtbø HB, Mancusi C, Cramariuc D. Echocardiographic aortic valve calcification and outcomes in women and men with aortic stenosis. Heart 2017; 103:1619-1624. [PMID: 28698175 PMCID: PMC5739834 DOI: 10.1136/heartjnl-2016-311040] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 04/02/2017] [Accepted: 04/03/2017] [Indexed: 01/20/2023] Open
Abstract
Objective Sex differences in risk factors of aortic valve calcification (AVC) by echocardiography have not been reported from a large prospective study in aortic stenosis (AS). Methods AVC was assessed using a prognostically validated visual score and grouped into none/mild or moderate/severe AVC in 1725 men and women with asymptomatic AS in the Simvastatin Ezetimibe in Aortic Stenosis study. The severity of AS was assessed by the energy loss index (ELI) taking pressure recovery in the aortic root into account. Results More men than women had moderate/severe AVC at baseline despite less severe AS by ELI (p<0.01). Moderate/severe AVC at baseline was independently associated with lower aortic compliance and more severe AS in both sexes, and with increased high-sensitive C reactive protein (hs-CRP) only in men (all p<0.01). In Cox regression analyses, moderate/severe AVC at baseline was associated with a 2.5-fold (95% CI 1.64 to 3.80) higher hazard rate of major cardiovascular events in women, and a 2.2-fold higher hazard rate in men (95% CI 1.54 to 3.17) (both p<0.001), after adjustment for age, hypertension, study treatment, aortic compliance, left ventricular (LV) mass and systolic function, AS severity and hs-CRP. Moderate/severe AVC at baseline also predicted a 1.8-fold higher hazard rate of all-cause mortality in men (95% CI 1.04 to 3.06, p<0.05) independent of age, AS severity, LV mass and aortic compliance, but not in women. Conclusion In conclusion, AVC scored by echocardiography has sex-specific characteristics in AS. Moderate/severe AVC is associated with higher cardiovascular morbidity in both sexes, and with higher all-cause mortality in men. Trial registration number ClinicalTrials.gov identifier: NCT00092677
Collapse
Affiliation(s)
| | - Giovanni Cioffi
- Department of Cardiology, Cura Villa Bianca Hospital, Trento, Italy
| | - Eva Gerdts
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Eigir Einarsen
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - Costantino Mancusi
- Department of Translational Medical Sciences, Federico II University Hospital, Naples, Italy
| | - Dana Cramariuc
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Mass spectrometry is an ever evolving technology that is equipped with a variety of tools for protein research. Some lipoprotein studies, especially those pertaining to HDL biology, have been exploiting the versatility of mass spectrometry to understand HDL function through its proteome. Despite the role of mass spectrometry in advancing research as a whole, however, the technology remains obscure to those without hands on experience, but still wishing to understand it. In this review, we walk the reader through the coevolution of common mass spectrometry workflows and HDL research, starting from the basic unbiased mass spectrometry methods used to profile the HDL proteome to the most recent targeted methods that have enabled an unprecedented view of HDL metabolism. RECENT FINDINGS Unbiased global proteomics have demonstrated that the HDL proteome is organized into subgroups across the HDL size fractions providing further evidence that HDL functional heterogeneity is in part governed by its varying protein constituents. Parallel reaction monitoring, a novel targeted mass spectrometry method, was used to monitor the metabolism of HDL apolipoproteins in humans and revealed that apolipoproteins contained within the same HDL size fraction exhibit diverse metabolic properties. SUMMARY Mass spectrometry provides a variety of tools and strategies to facilitate understanding, through its proteins, the complex biology of HDL.
Collapse
Affiliation(s)
- Sasha A. Singh
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Center for Excellence in Vascular Biology, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|