1
|
Duque EJ, Giachelli C, Moysés RMA. The role of osteopontin in chronic kidney disease-mineral bone disorder. Curr Opin Nephrol Hypertens 2025; 34:291-296. [PMID: 40166833 DOI: 10.1097/mnh.0000000000001074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
PURPOSE OF REVIEW Chronic kidney disease-mineral bone disorder (CKD-MBD) is associated with several adverse outcomes, including bone fragility and sarcopenia. Identification of new agents mitigating systemic damage related to uremia is critical and needed to unveil pathways implicated in CKD-MBD. RECENT FINDINGS Osteopontin (OPN) is involved in different physiological and pathological processes and works as a bridge connecting several systems. It may serve as a biomarker for many diseases, including human cancers, neurodegenerative disorders and autoimmune diseases. OPN has been implicated in disturbances of bone mineralization and remodeling, and has an interplay with parathyroid hormone and FGF23 in experimental models. In patients with CKD and severe hyperparathyroidism, OPN expression in muscle tissue has been linked to worse functionality and local inflammation, which is partially reverted after parathyroidectomy. SUMMARY Future studies could confirm the role of OPN as a biomarker in nephrology. Greater understanding of its role in CKD-MBD will help us define a better therapeutic strategy in patients with CKD.
Collapse
Affiliation(s)
- Eduardo J Duque
- Division of Nephrology and Hypertension, Center for Translational Metabolism and Health, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Cecilia Giachelli
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Rosa M A Moysés
- LIM 16, Nephrology Department, Hospital das Clínicas da Faculdade de Medicina da USP (HCFMUSP), Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
2
|
Samaha J, Madhu S, Shehadeh LA, Martinez CA. Osteopontin as a potential mediator of inflammation in HIV and comorbid conditions. AIDS 2025; 39:483-495. [PMID: 40080169 DOI: 10.1097/qad.0000000000004112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/23/2024] [Indexed: 03/15/2025]
Abstract
INTRODUCTION Approximately 39 million people live with HIV globally, with 1.3 million new infections annually. Despite improved treatment, noncommunicable diseases (NCDs) such as cardiovascular disease (CVD), neurological disorders, chronic kidney disease (CKD), and cancer are now the leading causes of death among people with HIV (PWH). Osteopontin (OPN) has emerged as a notable mediator in the inflammatory response to HIV and related NCDs. Our aim is to review the current understanding of OPN's role in HIV-related inflammatory pathways to highlight potential therapeutic avenues for improved treatment and mitigation of comorbidities. METHODS We conducted a systematic review by searching relevant literature using specific keywords related to HIV, osteopontin, cardiovascular disease, inflammation, neurological disorders, cancer, and chronic kidney disease. The collected studies were organized and categorized by key themes, followed by a comprehensive analysis to identify patterns and draw conclusions regarding OPN's role in HIV-associated comorbidities. RESULTS The intricate interactions between OPN, its isoforms, and HIV-related illnesses suggest that OPN can exhibit both pro-inflammatory and anti-inflammatory roles, depending on the stage of the disease and the specific cell type involved. Its functions are diverse throughout the progression of HIV and its associated comorbidities, including CVD, CKD, cancer, and neurological disorders. CONCLUSION OPN's effects on the disease progression of HIV and related NCDs are highly variable due to its diverse functions. Therefore, further research is essential to fully understand its complex roles before considering OPN as a therapeutic target for HIV and its comorbidities.
Collapse
Affiliation(s)
- Jacklyn Samaha
- Department of Medicine
- Department of Public Health Sciences, University of Miami Miller School of Medicine
| | - Shashank Madhu
- Department of Medicine
- Department of Public Health Sciences, University of Miami Miller School of Medicine
| | - Lina A Shehadeh
- Department of Medicine
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | | |
Collapse
|
3
|
Bibi F, Aslam A, Naseem H, Khurshid H, Haider A, Ashraf H, Rafaqat S. Adipokines Pathogenesis in Heart Failure. CARDIOVASCULAR INNOVATIONS AND APPLICATIONS 2025; 10. [DOI: 10.15212/cvia.2024.0065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
Many studies have reported that obesity causes heart failure (HF) pathogenesis. The elevated circulating levels of angiopoietin-like protein 2 (ANGPTL2) observed in patients with HF suggest potential links among elevated ANGPTL2 levels, metabolic disturbances, and inflammation. C1q/TNF-related protein 3 and C1q/TNF-related protein 9 are diminished in patients with HF with reduced ejection fraction, in proportion to disease severity, and are associated with elevated morbidity and mortality. In addition, fibroblast growth factor 21 (FGF21) has been suggested to be involved in the pathophysiology of diastolic HF. Further studies are necessary to determine whether FGF21 plays a causal role in HF, and whether measuring circulating FGF21 might effectively improve HF prediction, diagnosis, and prognosis. Osteopontin has also been reported to be upregulated in patients with HF and therefore might potentially serve as a novel prognostic biomarker in patients with chronic HF. A considerable number of patients with community-acquired HF have elevated tumor necrosis factor-alpha, which is associated with significantly diminished survival and significantly elevated risk of HF.
Collapse
|
4
|
Soraci L, Ärnlöv J, Carlsson AC, Feldreich TR, Larsson A, Roller-Wirnsberger R, Wirnsberger G, Mattace-Raso F, Tap L, Formiga F, Moreno-González R, Soltysik B, Kostka J, Artzi-Medvedik R, Melzer I, Weingart C, Sieber C, Marcozzi S, Muglia L, Lattanzio F. Associations between plasma osteopontin, sex, and 2-year global and cardiorenal outcomes in older outpatients screened for CKD: a secondary analysis of the SCOPE study. Clin Kidney J 2024; 17:sfae336. [PMID: 39664995 PMCID: PMC11631127 DOI: 10.1093/ckj/sfae336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Indexed: 12/13/2024] Open
Abstract
Background Plasma osteopontin (pOPN) is a promising aging-related biomarker among individuals with and without kidney disease. The interaction between sex, pOPN levels, and global and cardiorenal outcomes among older individuals was not previously evaluated. Methods In this study we investigated the association of pOPN with 24-month global mortality, major cardiovascular events (MACEs), MACEs + cardiovascular (CV) mortality, and renal decline among older individuals; we also evaluated whether sex modified observed associations. pOPN levels were measured in a cohort of 2013 outpatients (908 men and 1105 women) aged 75 years or more enrolled in the context of a multicenter prospective cohort study in Europe. Multivariable linear regression, Cox and Fine Gray models, and linear mixed regression models were fitted to evaluate whether sex modified the associations between biomarkers and study outcomes. Results In total, 2013 older participants with a median age of 79 years, 54.9% of whom women, were included in the study; increased pOPN levels were associated with all-cause mortality specifically among women [reduced fully adjusted model resulting from backward selection, hazard ratio, 95% confidence interval (CI): 1.84, 1.20-2.89]. Addition of pOPN to models containing age, eGFR, and albumin-to-creatinine ratio (ACR) improved the time-dependent area under the curve (AUC) at 6, 12, and 24 months, among women only. No significant association was found between the biomarker levels, MACE, and MACE + CV mortality. Conversely, increased baseline pOPN was associated with eGFR decline in all patients (-0.45, 95%CI: -0.68 to -0.22 ml/min/1.73 m2 year) but with slightly steeper declines in women compared to men (-0.57, -0.99 to -0.15 vs -0.47, -0.88 to -0.07). Conclusions pOPN levels were significantly lower in women than in men but associated with all-cause mortality in women only; increase in serum pOPN was associated with eGFR decline over time in all patients, but with stronger associations among women. Assessment of pOPN may help identifying older female participants at risk of poor outcomes.
Collapse
Affiliation(s)
- Luca Soraci
- Italian National Research Center on Aging (IRCCS INRCA), Ancona, Fermo and Cosenza, Italy
| | - Johan Ärnlöv
- School of Health and Welfare, Dalarna University, Falun, Sweden
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden
| | - Axel C Carlsson
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden
- Academic Primary Health Care Centre, Stockholm Region, Stockholm, Sweden
| | - Tobias Rudholm Feldreich
- School of Health and Welfare, Dalarna University, Falun, Sweden
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden
| | - Anders Larsson
- Section of Clinical Chemistry, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | | | - Francesco Mattace-Raso
- Section of Geriatric Medicine, Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Lisanne Tap
- Section of Geriatric Medicine, Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Francesc Formiga
- Geriatric Unit, Internal Medicine Department and Nephrology Department, Bellvitge University Hospital – IDIBELL - L'Hospitalet de Llobregat, Barcelona, Spain
| | - Rafael Moreno-González
- Geriatric Unit, Internal Medicine Department and Nephrology Department, Bellvitge University Hospital – IDIBELL - L'Hospitalet de Llobregat, Barcelona, Spain
| | - Bartlomiej Soltysik
- Department of Geriatrics, Healthy Ageing Research Centre, Medical University of Lodz, Lodz, Poland
| | - Joanna Kostka
- Department of Gerontology, Medical University of Lodz, Lodz, Poland
| | - Rada Artzi-Medvedik
- The Recanati School for Community Health Professions at the faculty of Health Sciences at Ben-Gurion University of the Negev, Beersheba, Israel
- Maccabi Healthcare services southern region, Tel Aviv, Israel
| | - Itshak Melzer
- The Recanati School for Community Health Professions at the faculty of Health Sciences at Ben-Gurion University of the Negev, Beersheba, Israel
| | - Christian Weingart
- Institute for Biomedicine of Aging, Friedrich-Alexander-Universität Erlangen Nürnberg, Erlangen, Germany
| | - Cornel Sieber
- Institute for Biomedicine of Aging, Friedrich-Alexander-Universität Erlangen Nürnberg, Erlangen, Germany
- Cantonal Hospital Winterthur, Winterthur, Switzerland
| | - Serena Marcozzi
- Italian National Research Center on Aging (IRCCS INRCA), Ancona, Fermo and Cosenza, Italy
| | - Lucia Muglia
- Italian National Research Center on Aging (IRCCS INRCA), Ancona, Fermo and Cosenza, Italy
| | - Fabrizia Lattanzio
- Italian National Research Center on Aging (IRCCS INRCA), Ancona, Fermo and Cosenza, Italy
| |
Collapse
|
5
|
Torino C, Carbone F, Pizzini P, Mezzatesta S, D’Arrigo G, Gori M, Liberale L, Moriero M, Michelauz C, Frè F, Isoppo S, Gavoci A, Rosa FL, Scuricini A, Tirandi A, Ramoni D, Mallamaci F, Tripepi G, Montecucco F, Zoccali C. Osteopontin and Clinical Outcomes in Hemodialysis Patients. Biomedicines 2024; 12:2605. [PMID: 39595171 PMCID: PMC11592156 DOI: 10.3390/biomedicines12112605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/06/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Chronic kidney disease (CKD) and end-stage kidney disease (ESKD) are significant public health issues, with cardiovascular morbidity and mortality being the leading causes of death in hemodialysis patients. Osteopontin (OPN), a multifunctional glycoprotein, has emerged as a potential biomarker for vascular disease in CKD due to its role in inflammation, tissue remodeling, and calcification. METHODS This cohort study included 1124 hemodialysis patients from the PROGREDIRE study, a registry involving 35 dialysis units in Southern Italy. Serum osteopontin levels were measured using enzyme-linked immunosorbent assay (ELISA). The primary endpoints were all-cause and cardiovascular mortality. Multivariate Cox regression analyses were performed to assess the association between osteopontin levels and mortality, adjusting for traditional risk factors, biomarkers of inflammation, nutritional status, and ESKD-related factors. RESULTS During a mean follow-up of 2.8 years, 478 patients died, 271 from cardiovascular causes. Independent correlates of osteopontin included alkaline phosphatase and parathyroid hormone. Elevated osteopontin levels were significantly associated with increased all-cause mortality (HR 1.19, 95% CI 1.09-1.31, p < 0.001) and cardiovascular mortality (HR 1.22, 95% CI 1.08-1.38, p = 0.001) after adjusting for confounders. CONCLUSIONS Elevated osteopontin levels are associated with increased all-cause and cardiovascular mortality in hemodialysis patients. These findings implicate osteopontin in the high risk for death and cardiovascular disease in the hemodialysis population. Intervention studies are needed to definitively test this hypothesis.
Collapse
Affiliation(s)
- Claudia Torino
- Clinical Epidemiology of Renal Disease and Hypertension Unit, Reggio Cal CNR Unit of the Pisa CNR Institute of Clinical Physiology, 89124 Reggio Calabria, Italy; (C.T.); (P.P.); (S.M.); (G.D.); (F.M.); (G.T.)
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; (F.C.); (L.L.); (M.M.); (C.M.); (F.F.); (S.I.); (A.G.); (F.L.R.); (A.S.); (A.T.); (D.R.); (F.M.)
- IRCCS Ospedale Policlinico San Martino, Genoa-Italian Cardiovascular Network, 10 Largo Rosanna Benzi, 16132 Genoa, Italy
| | - Patrizia Pizzini
- Clinical Epidemiology of Renal Disease and Hypertension Unit, Reggio Cal CNR Unit of the Pisa CNR Institute of Clinical Physiology, 89124 Reggio Calabria, Italy; (C.T.); (P.P.); (S.M.); (G.D.); (F.M.); (G.T.)
| | - Sabrina Mezzatesta
- Clinical Epidemiology of Renal Disease and Hypertension Unit, Reggio Cal CNR Unit of the Pisa CNR Institute of Clinical Physiology, 89124 Reggio Calabria, Italy; (C.T.); (P.P.); (S.M.); (G.D.); (F.M.); (G.T.)
| | - Graziella D’Arrigo
- Clinical Epidemiology of Renal Disease and Hypertension Unit, Reggio Cal CNR Unit of the Pisa CNR Institute of Clinical Physiology, 89124 Reggio Calabria, Italy; (C.T.); (P.P.); (S.M.); (G.D.); (F.M.); (G.T.)
| | - Mercedes Gori
- CNR—Institute of Clinical Physiology, 00186 Rome, Italy;
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; (F.C.); (L.L.); (M.M.); (C.M.); (F.F.); (S.I.); (A.G.); (F.L.R.); (A.S.); (A.T.); (D.R.); (F.M.)
- IRCCS Ospedale Policlinico San Martino, Genoa-Italian Cardiovascular Network, 10 Largo Rosanna Benzi, 16132 Genoa, Italy
| | - Margherita Moriero
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; (F.C.); (L.L.); (M.M.); (C.M.); (F.F.); (S.I.); (A.G.); (F.L.R.); (A.S.); (A.T.); (D.R.); (F.M.)
| | - Cristina Michelauz
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; (F.C.); (L.L.); (M.M.); (C.M.); (F.F.); (S.I.); (A.G.); (F.L.R.); (A.S.); (A.T.); (D.R.); (F.M.)
| | - Federica Frè
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; (F.C.); (L.L.); (M.M.); (C.M.); (F.F.); (S.I.); (A.G.); (F.L.R.); (A.S.); (A.T.); (D.R.); (F.M.)
| | - Simone Isoppo
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; (F.C.); (L.L.); (M.M.); (C.M.); (F.F.); (S.I.); (A.G.); (F.L.R.); (A.S.); (A.T.); (D.R.); (F.M.)
| | - Aurora Gavoci
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; (F.C.); (L.L.); (M.M.); (C.M.); (F.F.); (S.I.); (A.G.); (F.L.R.); (A.S.); (A.T.); (D.R.); (F.M.)
| | - Federica La Rosa
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; (F.C.); (L.L.); (M.M.); (C.M.); (F.F.); (S.I.); (A.G.); (F.L.R.); (A.S.); (A.T.); (D.R.); (F.M.)
| | - Alessandro Scuricini
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; (F.C.); (L.L.); (M.M.); (C.M.); (F.F.); (S.I.); (A.G.); (F.L.R.); (A.S.); (A.T.); (D.R.); (F.M.)
| | - Amedeo Tirandi
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; (F.C.); (L.L.); (M.M.); (C.M.); (F.F.); (S.I.); (A.G.); (F.L.R.); (A.S.); (A.T.); (D.R.); (F.M.)
| | - Davide Ramoni
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; (F.C.); (L.L.); (M.M.); (C.M.); (F.F.); (S.I.); (A.G.); (F.L.R.); (A.S.); (A.T.); (D.R.); (F.M.)
| | - Francesca Mallamaci
- Clinical Epidemiology of Renal Disease and Hypertension Unit, Reggio Cal CNR Unit of the Pisa CNR Institute of Clinical Physiology, 89124 Reggio Calabria, Italy; (C.T.); (P.P.); (S.M.); (G.D.); (F.M.); (G.T.)
- Nephrology, Hypertension and Renal Transplantation Unit, Grande Ospedale Metropolitano, 89124 Reggio Calabria, Italy
| | - Giovanni Tripepi
- Clinical Epidemiology of Renal Disease and Hypertension Unit, Reggio Cal CNR Unit of the Pisa CNR Institute of Clinical Physiology, 89124 Reggio Calabria, Italy; (C.T.); (P.P.); (S.M.); (G.D.); (F.M.); (G.T.)
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy; (F.C.); (L.L.); (M.M.); (C.M.); (F.F.); (S.I.); (A.G.); (F.L.R.); (A.S.); (A.T.); (D.R.); (F.M.)
- IRCCS Ospedale Policlinico San Martino, Genoa-Italian Cardiovascular Network, 10 Largo Rosanna Benzi, 16132 Genoa, Italy
| | - Carmine Zoccali
- Renal Research Institute, New York, NY 10065, USA
- IPNET, c/o Nefrologia del Grande Ospedale Metropolitano, 89124 Reggio Calabria, Italy
| |
Collapse
|
6
|
D'Arrigo G, Carbone F, Gori M, Torino C, Montecucco F, Liberale L, Ramoni D, Tirandi A, Tortorella C, Lisa A, Olivero C, Moriero M, Bertolotto M, Minetti S, Schiavetta E, Pizzini P, Cutrupi S, Mallamaci F, Tripepi G, Zoccali C. Osteopontin, death and cardiovascular events in stage G3-4 CKD patients: a joint model analysis. Nephrol Dial Transplant 2024; 39:1737-1739. [PMID: 38849311 DOI: 10.1093/ndt/gfae123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Indexed: 06/09/2024] Open
Affiliation(s)
- Graziella D'Arrigo
- Consiglio Nazionale delle Ricerche (CNR), Clinical Epidemiology of Renal Diseases and Hypertension Unit, Centro Fisiologia Clinica CNR, Reggio Calabria, Rome
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Mercedes Gori
- Nephrology, Dialysis and Transplantation Unit, Grande Ospedale Metropolitano
| | - Claudia Torino
- Consiglio Nazionale delle Ricerche (CNR), Clinical Epidemiology of Renal Diseases and Hypertension Unit, Centro Fisiologia Clinica CNR, Reggio Calabria, Rome
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Davide Ramoni
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Amedeo Tirandi
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Curzia Tortorella
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Anna Lisa
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Chiara Olivero
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Margherita Moriero
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Maria Bertolotto
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Silvia Minetti
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Elisa Schiavetta
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Patrizia Pizzini
- Consiglio Nazionale delle Ricerche (CNR), Clinical Epidemiology of Renal Diseases and Hypertension Unit, Centro Fisiologia Clinica CNR, Reggio Calabria, Rome
| | - Sebastiano Cutrupi
- Consiglio Nazionale delle Ricerche (CNR), Clinical Epidemiology of Renal Diseases and Hypertension Unit, Centro Fisiologia Clinica CNR, Reggio Calabria, Rome
| | - Francesca Mallamaci
- Consiglio Nazionale delle Ricerche (CNR), Clinical Epidemiology of Renal Diseases and Hypertension Unit, Centro Fisiologia Clinica CNR, Reggio Calabria, Rome
- Renal Research Institute, NY, USA
| | - Giovanni Tripepi
- Consiglio Nazionale delle Ricerche (CNR), Clinical Epidemiology of Renal Diseases and Hypertension Unit, Centro Fisiologia Clinica CNR, Reggio Calabria, Rome
| | - Carmine Zoccali
- Renal Research Institute, NY, USA
- Institute of Molecular Biology and Genetics (Biogem), Ariano Irpino, Italy
- Associazione Ipertensione Nefrologia Trapianto Renale (IPNET), c/o Nefrologia, Grande Ospedale Metropolitano, Reggio Calabria, Italy
| |
Collapse
|
7
|
Xu S, Zhang G, Tan X, Zeng Y, Jiang H, Jiang Y, Wang X, Song Y, Fan H, Zhou Y. Plasma Olink Proteomics Reveals Novel Biomarkers for Prediction and Diagnosis in Dilated Cardiomyopathy with Heart Failure. J Proteome Res 2024; 23:4139-4150. [PMID: 39129220 PMCID: PMC11385702 DOI: 10.1021/acs.jproteome.4c00522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
In this study, we utilized the Olink Cardiovascular III panel to compare the expression levels of 92 cardiovascular-related proteins between patients with dilated cardiomyopathy combined with heart failure (DCM-HF) (n = 20) and healthy normal people (Normal) (n = 18). The top five most significant proteins, including SPP1, IGFBP7, F11R, CHI3L1, and Plaur, were selected by Olink proteomics. These proteins were further validated using ELISA in plasma samples collected from an additional cohort. ELISA validation confirmed significant increases in SPP1, IGFBP7, F11R, CHI3L1, and Plaur in DCM-HF patients compared to healthy controls. GO and KEGG analysis indicated that NT-pro BNP, SPP1, IGFBP7, F11R, CHI3L1, Plaur, BLM hydrolase, CSTB, Gal-4, CCL15, CDH5, SR-PSOX, and CCL2 were associated with DCM-HF. Correlation analysis revealed that these 13 differentially expressed proteins have strong correlations with clinical indicators such as LVEF and NT-pro BNP, etc. Additionally, in the GEO-DCM data sets, the combined diagnostic value of these five core proteins AUC values of 0.959, 0.773, and 0.803, respectively indicating the predictive value of the five core proteins for DCM-HF. Our findings suggest that these proteins may be useful biomarkers for the diagnosis and prediction of DCM-HF, and further research is prompted to explore their potential as therapeutic targets.
Collapse
Affiliation(s)
- Shuai Xu
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University,Suzhou 215000, China
- Institute for Hypertension, Soochow University, Suzhou 215000, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou 450052, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou 450052, China
| | - Xin Tan
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University,Suzhou 215000, China
- Institute for Hypertension, Soochow University, Suzhou 215000, China
| | - Yiyao Zeng
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University,Suzhou 215000, China
- Institute for Hypertension, Soochow University, Suzhou 215000, China
| | - Hezi Jiang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University,Suzhou 215000, China
- Institute for Hypertension, Soochow University, Suzhou 215000, China
| | - Yufeng Jiang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University,Suzhou 215000, China
- Institute for Hypertension, Soochow University, Suzhou 215000, China
| | - Xiangyu Wang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University,Suzhou 215000, China
- Institute for Hypertension, Soochow University, Suzhou 215000, China
| | - Yahui Song
- Center of Translational Medicine and Clinical Laboratory, The Fourth Affiliated Hospital to Soochow University, Suzhou 215028, China
| | - Huimin Fan
- Center of Translational Medicine and Clinical Laboratory, The Fourth Affiliated Hospital to Soochow University, Suzhou 215028, China
| | - Yafeng Zhou
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University,Suzhou 215000, China
- Institute for Hypertension, Soochow University, Suzhou 215000, China
| |
Collapse
|
8
|
Huang K, Chen S, Yu LJ, Wu ZM, Chen QJ, Wang XQ, Li FF, Liu JM, Wang YX, Mao LS, Shen WF, Zhang RY, Shen Y, Lu L, Dai Y, Ding FH. Serum secreted phosphoprotein 1 level is associated with plaque vulnerability in patients with coronary artery disease. Front Immunol 2024; 15:1285813. [PMID: 38426091 PMCID: PMC10902157 DOI: 10.3389/fimmu.2024.1285813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 02/01/2024] [Indexed: 03/02/2024] Open
Abstract
Background Vulnerable plaque was associated with recurrent cardiovascular events. This study was designed to explore predictive biomarkers of vulnerable plaque in patients with coronary artery disease. Methods To reveal the phenotype-associated cell type in the development of vulnerable plaque and to identify hub gene for pathological process, we combined single-cell RNA and bulk RNA sequencing datasets of human atherosclerotic plaques using Single-Cell Identification of Subpopulations with Bulk Sample Phenotype Correlation (Scissor) and Weighted gene co-expression network analysis (WGCNA). We also validated our results in an independent cohort of patients by using intravascular ultrasound during coronary angiography. Results Macrophages were found to be strongly correlated with plaque vulnerability while vascular smooth muscle cell (VSMC), fibrochondrocyte (FC) and intermediate cell state (ICS) clusters were negatively associated with unstable plaque. Weighted gene co-expression network analysis showed that Secreted Phosphoprotein 1 (SPP1) in the turquoise module was highly correlated with both the gene module and the clinical traits. In a total of 593 patients, serum levels of SPP1 were significantly higher in patients with vulnerable plaques than those with stable plaque (113.21 [73.65 - 147.70] ng/ml versus 71.08 [20.64 - 135.68] ng/ml; P < 0.001). Adjusted multivariate regression analysis revealed that serum SPP1 was an independent determinant of the presence of vulnerable plaque. Receiver operating characteristic curve analysis indicated that the area under the curve was 0.737 (95% CI 0.697 - 0.773; P < 0.001) for adding serum SPP1 in predicting of vulnerable plaques. Conclusion Elevated serum SPP1 levels confer an increased risk for plaque vulnerability in patients with coronary artery disease.
Collapse
Affiliation(s)
- Ke Huang
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Shuai Chen
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Lin-Jun Yu
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Zhi-Ming Wu
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Qiu-Jing Chen
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Xiao-Qun Wang
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Fei-Fei Li
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Jing-Meng Liu
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Yi-Xuan Wang
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Lin-Shuang Mao
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Wei-Feng Shen
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Rui-Yan Zhang
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Ying Shen
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Lin Lu
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Yang Dai
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Feng-Hua Ding
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, China
| |
Collapse
|
9
|
Rafaqat S, Rafaqat S, Ijaz H. The Role of Biochemical Cardiac Markers in Atrial Fibrillation. J Innov Card Rhythm Manag 2023; 14:5611-5621. [PMID: 37927395 PMCID: PMC10621624 DOI: 10.19102/icrm.2023.14101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 04/13/2023] [Indexed: 11/07/2023] Open
Abstract
Atrial fibrillation (AF) is the most common type of cardiac arrhythmia. Proteins are a component of cardiac biomarkers containing cell structures that are released into the circulation when a myocardial injury occurs. They are essential in the diagnosis, risk assessment, and treatment of patients who have chest pain, are thought to have acute coronary syndrome, or are experiencing acute heart failure exacerbations. There are numerous biochemical cardiac markers, but this article summarizes the basic role of major biochemical cardiac markers, including cardiac natriuretic peptides, cardiac troponins, C-reactive protein (CRP), creatine kinase-MB, heart-type fatty acid-binding protein, ischemia-modified albumin, lipoprotein (a), osteopontin (OPN), and soluble suppression of tumorigenicity 2 (sST2), in AF. Atrial natriuretic peptide may serve as an indicator of atrial integrity, which may help to select appropriate treatment approaches for AF. Higher levels of N-terminal pro-B-type natriuretic peptide and brain natriuretic peptide are predictive of incidental AF. Increased troponin T release may indicate better clinical results following AF ablation. Similarly, CRP increases the risk of the AF-increasing calcium (Ca) influx in atrial myocytes, but not because of atrial fibrosis. Patients with postoperative AF have lower FABP3 gene expression in the atrium. Lipoprotein (a) (Lp[a]) may play a causative role in the onset of AF and impact various cardiac tissues. Clinical trials for Lp(a)-lowering drugs should assess their impact on preventing AF. Also, OPN was highly expressed in the circulation of AF patients and further increased with the progression of AF. sST2 was a reliable predictor of new-onset AF and can improve the accuracy of the AF risk model. There is a greater chance that these cardiac biomarkers might be employed to enhance clinical risk stratification in AF.
Collapse
Affiliation(s)
- Saira Rafaqat
- Department of Zoology, Lahore College for Women University, Lahore, Pakistan
| | - Sana Rafaqat
- Department of Biotechnology, Lahore College for Women University, Lahore, Pakistan
| | - Hafsa Ijaz
- Department of Zoology, Lahore College for Women University, Lahore, Pakistan
| |
Collapse
|
10
|
Wees I, Hendren NS, Kaur G, Roth LR, Grodin JL. Natriuretic Peptides and Cardiac Troponins: Markers of Disease Progression and Risk in Light Chain Cardiac Amyloidosis. Curr Heart Fail Rep 2023; 20:350-357. [PMID: 37428428 DOI: 10.1007/s11897-023-00616-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/21/2023] [Indexed: 07/11/2023]
Abstract
PURPOSE OF REVIEW Light chain (AL) amyloidosis can cause an infiltrative cardiomyopathy that can result in symptomatic heart failure. The vague, nonspecific onset of signs and symptoms may lead to a delay in diagnosis and treatment leading to poor outcomes. Cardiac biomarkers, such as troponins and natriuretic peptides, play a pivotal role in diagnosis, determining prognosis, and assessing treatment response in patients with AL amyloidosis. Because of the evolving landscape for both diagnosis and treatment of AL cardiac amyloidosis, we review the critical role these and other biomarkers play in the clinical management of this disease. RECENT FINDINGS A number of conventional cardiac and noncardiac serum biomarkers are commonly used in AL cardiac amyloidosis and may be surrogates for cardiac involvement and inform prognosis. These include typical heart failure biomarkers such as levels of circulating natriuretic peptides as well as cardiac troponins. Other noncardiac biomarkers frequently measured in AL cardiac amyloidosis included difference between the involved and uninvolved free light chains (dFLC) and markers of endothelial cell activation and damage such as von Willebrand factor antigen and matrix metalloproteinases. AL amyloidosis can lead to cardiac involvement which has been associated with poor outcomes, especially if not identified and treated early. Natriuretic peptides and cardiac troponins are cornerstones for the diagnosis and management of AL cardiac amyloidosis. Their levels may represent cardiac stress, injury, and possibly degree of cardiac involvement, and they play a key role in AL amyloidosis disease staging.
Collapse
Affiliation(s)
- Isabel Wees
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Parkland Health and Hospital System, Dallas, TX, USA
| | - Nicholas S Hendren
- Parkland Health and Hospital System, Dallas, TX, USA
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gurbakhash Kaur
- Division of Hematology and Oncology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Lori R Roth
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Justin L Grodin
- Parkland Health and Hospital System, Dallas, TX, USA.
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
11
|
Nolze A, Matern S, Grossmann C. Calcineurin Is a Universal Regulator of Vessel Function-Focus on Vascular Smooth Muscle Cells. Cells 2023; 12:2269. [PMID: 37759492 PMCID: PMC10528183 DOI: 10.3390/cells12182269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Calcineurin, a serine/threonine phosphatase regulating transcription factors like NFaT and CREB, is well known for its immune modulatory effects and role in cardiac hypertrophy. Results from experiments with calcineurin knockout animals and calcineurin inhibitors indicate that calcineurin also plays a crucial role in vascular function, especially in vascular smooth muscle cells (VSMCs). In the aorta, calcineurin stimulates the proliferation and migration of VSMCs in response to vascular injury or angiotensin II administration, leading to pathological vessel wall thickening. In the heart, calcineurin mediates coronary artery formation and VSMC differentiation, which are crucial for proper heart development. In pulmonary VSMCs, calcineurin/NFaT signaling regulates the release of Ca2+, resulting in increased vascular tone followed by pulmonary arterial hypertension. In renal VSMCs, calcineurin regulates extracellular matrix secretion promoting fibrosis development. In the mesenteric and cerebral arteries, calcineurin mediates a phenotypic switch of VSMCs leading to altered cell function. Gaining deeper insights into the underlying mechanisms of calcineurin signaling will help researchers to understand developmental and pathogenetical aspects of the vasculature. In this review, we provide an overview of the physiological function and pathophysiology of calcineurin in the vascular system with a focus on vascular smooth muscle cells in different organs. Overall, there are indications that under certain pathological settings reduced calcineurin activity seems to be beneficial for cardiovascular health.
Collapse
Affiliation(s)
| | | | - Claudia Grossmann
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany
| |
Collapse
|
12
|
Wang H, Shi J, Wang J, Hu Y. MicroRNA‑378: An important player in cardiovascular diseases (Review). Mol Med Rep 2023; 28:172. [PMID: 37503766 PMCID: PMC10436248 DOI: 10.3892/mmr.2023.13059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 05/31/2023] [Indexed: 07/29/2023] Open
Abstract
Cardiovascular disease (CVD) is a common chronic clinical condition and is the main cause of death in humans worldwide. Understanding the genetic and molecular mechanisms involved in the development of CVD is essential to develop effective prevention strategies and therapeutic measures. An increasing number of CVD‑related genetic studies have been conducted, including those on the potential roles of microRNAs (miRs). These studies have demonstrated that miR‑378 is involved in the pathological processes of CVD, including those of myocardial infarction, heart failure and coronary heart disease. Despite the potential importance of miR‑378 CVD, a comprehensive summary of the related literature is lacking. Thus, the present review aimed to summarize the findings of previous studies on the roles and mechanisms of miR‑378 in a variety of CVDs and provide an up‑to date basis for further r research targeting the prevention and treatment of CVDs.
Collapse
Affiliation(s)
- Huan Wang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P.R. China
| | - Jingjing Shi
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P.R. China
| | - Jiuchong Wang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P.R. China
| | - Yuanhui Hu
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P.R. China
| |
Collapse
|
13
|
Cicekli I, Saglam D, Takar N. A New Perspective on Metabolic Syndrome with Osteopontin: A Comprehensive Review. Life (Basel) 2023; 13:1608. [PMID: 37511983 PMCID: PMC10381599 DOI: 10.3390/life13071608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/12/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Metabolic syndrome (MetS) imposes a substantial burden on the healthcare systems and economies of countries and is a major public health concern worldwide. MetS is mainly caused by an imbalance between calorie intake and energy expenditure; however, it is recognized that additional variables, such as chronic inflammation, may have the same predictive potential as insulin resistance or MetS components in the genesis of type 2 diabetes and cardiovascular events. More importantly, the early diagnosis or treatment of MetS may significantly reduce the burden on the health systems of the disease with any prevention or biomarker and should not be underestimated. Osteopontin (OPN), also called secreted phosphoprotein 1, is a soluble protein found mostly in body fluids. Studies suggest that serum OPN levels may be an early and new biomarker to predict metabolic and cardiovascular complications significantly associated with some diseases. This review aims to provide specific insight into the new biomarker OPN in MetS. With this purpose, it is examined the link between the MetS cornerstones and OPN. In addition, the interaction between the microbiota and MetS is predicted to be bidirectional, and the microbiota may act as a bridge in this interaction process. Increased OPN levels may have unfavourable consequences for cardiovascular diseases, diabetes, and obesity, all of which are components of MetS. Further studies are required to evaluate the use of OPN levels as a clinical biomarker risk of MetS.
Collapse
Affiliation(s)
- Ipek Cicekli
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
| | - Duygu Saglam
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
| | - Nadir Takar
- Department of Family Medicine, Kartal Dr. Lutfi Kirdar City Hospital, Istanbul Provincial Directorate of Health, Istanbul 34865, Turkey
| |
Collapse
|
14
|
Mamazhakypov A, Maripov A, Sarybaev AS, Schermuly RT, Sydykov A. Osteopontin in Pulmonary Hypertension. Biomedicines 2023; 11:biomedicines11051385. [PMID: 37239056 DOI: 10.3390/biomedicines11051385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/01/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Pulmonary hypertension (PH) is a pathological condition with multifactorial etiology, which is characterized by elevated pulmonary arterial pressure and pulmonary vascular remodeling. The underlying pathogenetic mechanisms remain poorly understood. Accumulating clinical evidence suggests that circulating osteopontin may serve as a biomarker of PH progression, severity, and prognosis, as well as an indicator of maladaptive right ventricular remodeling and dysfunction. Moreover, preclinical studies in rodent models have implicated osteopontin in PH pathogenesis. Osteopontin modulates a plethora of cellular processes within the pulmonary vasculature, including cell proliferation, migration, apoptosis, extracellular matrix synthesis, and inflammation via binding to various receptors such as integrins and CD44. In this article, we provide a comprehensive overview of the current understanding of osteopontin regulation and its impact on pulmonary vascular remodeling, as well as consider research issues required for the development of therapeutics targeting osteopontin as a potential strategy for the management of PH.
Collapse
Affiliation(s)
- Argen Mamazhakypov
- Department of Internal Medicine, Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, 35392 Giessen, Germany
| | - Abdirashit Maripov
- Department of Mountain and Sleep Medicine and Pulmonary Hypertension, National Center of Cardiology and Internal Medicine, Bishkek 720040, Kyrgyzstan
| | - Akpay S Sarybaev
- Department of Mountain and Sleep Medicine and Pulmonary Hypertension, National Center of Cardiology and Internal Medicine, Bishkek 720040, Kyrgyzstan
| | - Ralph Theo Schermuly
- Department of Internal Medicine, Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, 35392 Giessen, Germany
| | - Akylbek Sydykov
- Department of Internal Medicine, Excellence Cluster Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, 35392 Giessen, Germany
| |
Collapse
|
15
|
Cheong KI, Leu HB, Wu CC, Yin WH, Wang JH, Lin TH, Tseng WK, Chang KC, Chu SH, Yeh HI, Chen JW, Wu YW. The clinical significance of osteopontin on the cardiovascular outcomes in patients with stable coronary artery disease. J Formos Med Assoc 2023; 122:328-337. [PMID: 36494313 DOI: 10.1016/j.jfma.2022.11.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 10/06/2022] [Accepted: 11/20/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Osteopontin (OPN) is a noncollagenous matricellular protein which is mainly present in bone matrix. A high OPN level has been associated with heart failure and acute coronary syndrome, however data on patients with chronic coronary syndrome (CCS) are lacking. The present study aimed to evaluate the association between OPN and the prognosis of Taiwanese patients with CCS. METHODS We enrolled participants from the Biosignature Registry, a nationwide prospective cohort study conducted at nine different medical centers throughout Taiwan. The inclusion criteria were participants who had received successful percutaneous coronary intervention at least once previously, and stable under medical therapy for at least 1 month before enrollment. They were followed for at least 72 months. Logistic regression and Cox proportional hazard model were used to investigate the association between OPN and clinical outcomes. The outcomes of this study were the first occurrence of hard cardiovascular events and composite cardiovascular outcomes including cardiovascular mortality, revascularization, hospitalization for acute myocardial infarction (AMI) or heart failure. RESULTS A total of 666 patients with both hs-CRP and osteopontin measurements were enrolled and followed for 72 months. OPN was correlated positively with AMI-related hospitalization, where the highest tertile (Tertile 3) of baseline OPN had the highest risk of AMI-related hospitalization, which remained significant after multivariate adjustments (HR 3.20, p = 0.017). In contrast, combining OPN and hs-CRP did not improve the prediction of CV outcomes. CONCLUSION OPN may be a potentially valuable biomarker in predicting CV outcomes. During 6 years of follow-up period, an OPN level >4810 pg/ml was associated with a significantly higher incidence of AMI-related hospitalization in CCS patients who received successful PCI before the enrollment.
Collapse
Affiliation(s)
- Kei-Ip Cheong
- Division of Cardiology, Cardiovascular Medical Center, Far Eastern Memorial Hospital, New Taipei City, Taiwan; College of General Education, Chihlee University of Technology, New Taipei City, Taiwan
| | - Hsin-Bang Leu
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chao Tung University, Taipei, Taiwan
| | - Chau-Chung Wu
- University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan; Graduate Institute of Medical Education & Bioethics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wei-Hsian Yin
- Division of Cardiology, Heart Center, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Ji-Hung Wang
- Division of Cardiology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Tsung-Hsien Lin
- Kaohsiung Medical University Hospital and Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wei-Kung Tseng
- Division of Cardiology, Department of Internal Medicine, E-Da Hospital, Kaohsiung, Taiwan
| | - Kuan-Cheng Chang
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung 404332, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404333, Taiwan
| | | | - Hung-I Yeh
- Department of Internal Medicine and Medical Research, Mackay Memorial Hospital, Taipei, Taiwan; Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Jaw-Wen Chen
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chao Tung University, Taipei, Taiwan; Healthcare and Services Center, Taipei Veterans General Hospital, Taipei, Taiwan; Cardiovascular Research Center, National Yang Ming Chao Tung University, Taipei, Taiwan; Institute of Pharmacology, National Yang Ming Chao Tung University, Taipei, Taiwan.
| | - Yen-Wen Wu
- Division of Cardiology, Cardiovascular Medical Center, Far Eastern Memorial Hospital, New Taipei City, Taiwan; School of Medicine, National Yang Ming Chao Tung University, Taipei, Taiwan; Graduate Institute of Medicine, Yuan Ze University, Taoyuan, Taiwan.
| | | |
Collapse
|
16
|
Robinson JA, Toribio M, Quinaglia T, Awadalla M, Talathi R, Durbin CG, Alhallak I, Alagpulinsa DA, Fourman LT, Suero-Abreu GA, Nelson MD, Stanley TL, Longenecker CT, Szczepaniak LS, Jerosch-Herold M, Neilan TG, Zanni MV, Burdo TH. Plasma osteopontin relates to myocardial fibrosis and steatosis and to immune activation among women with HIV. AIDS 2023; 37:305-310. [PMID: 36541642 PMCID: PMC9782710 DOI: 10.1097/qad.0000000000003417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Women with HIV (WWH) have heightened heart failure risk. Plasma OPN (osteopontin) is a powerful predictor of heart failure outcomes in the general population. Limited data exist on relationships between plasma OPN and surrogates of HIV-associated heart failure risk. DESIGN Prospective, cross-sectional. METHODS We analyzed relationships between plasma OPN and cardiac structure/function (assessed using cardiovascular magnetic resonance imaging) and immune activation (biomarkers and flow cytometry) among 20 WWH and 14 women without HIV (WWOH). RESULTS Plasma OPN did not differ between groups. Among WWH, plasma OPN related directly to the markers of cardiac fibrosis, growth differentiation factor-15 (ρ = 0.51, P = 0.02) and soluble interleukin 1 receptor-like 1 (ρ = 0.45, P = 0.0459). Among WWH (but not among WWOH or the whole group), plasma OPN related directly to both myocardial fibrosis (ρ = 0.49, P = 0.03) and myocardial steatosis (ρ = 0.46, P = 0.0487). Among the whole group and WWH (and not among WWOH), plasma OPN related directly to the surface expression of C-X3-C motif chemokine receptor 1 (CX3CR1) on nonclassical (CD14-CD16+) monocytes (whole group: ρ = 0.36, P = 0.04; WWH: ρ = 0.46, P = 0.04). Further, among WWH and WWOH (and not among the whole group), plasma OPN related directly to the surface expression of CC motif chemokine receptor 2 (CCR2) on inflammatory (CD14+CD16+) monocytes (WWH: ρ = 0.54, P = 0.01; WWOH: ρ = 0.60, P = 0.03), and in WWH, this held even after controlling for HIV-specific parameters. CONCLUSION Among WWH, plasma OPN, a powerful predictor of heart failure outcomes, related to myocardial fibrosis and steatosis and the expression of CCR2 and CX3CR1 on select monocyte subpopulations. OPN may play a role in heart failure pathogenesis among WWH. CLINICALTRIALSGOV REGISTRATION NCT02874703.
Collapse
Affiliation(s)
- Jake A Robinson
- Department of Microbiology, Immunology, and Inflammation, Center for Neurovirology and Gene Editing, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | | | - Thiago Quinaglia
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology
| | - Magid Awadalla
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology
| | | | | | | | - David A Alagpulinsa
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | | | | | - Michael D Nelson
- Applied Physiology and Advanced Imaging Laboratory, Department of Kinesiology, University of Texas at Arlington, Arlington, TX
| | | | | | | | - Michael Jerosch-Herold
- Division of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Tomas G Neilan
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology
| | | | - Tricia H Burdo
- Department of Microbiology, Immunology, and Inflammation, Center for Neurovirology and Gene Editing, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| |
Collapse
|
17
|
Schroeder ME, Batan D, Gonzalez Rodriguez A, Speckl KF, Peters DK, Kirkpatrick BE, Hach GK, Walker CJ, Grim JC, Aguado BA, Weiss RM, Anseth KS. Osteopontin activity modulates sex-specific calcification in engineered valve tissue mimics. Bioeng Transl Med 2023; 8:e10358. [PMID: 36684107 PMCID: PMC9842038 DOI: 10.1002/btm2.10358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/29/2022] [Accepted: 05/13/2022] [Indexed: 02/06/2023] Open
Abstract
Patients with aortic valve stenosis (AVS) have sexually dimorphic phenotypes in their valve tissue, where male valvular tissue adopts a calcified phenotype and female tissue becomes more fibrotic. The molecular mechanisms that regulate sex-specific calcification in valvular tissue remain poorly understood. Here, we explored the role of osteopontin (OPN), a pro-fibrotic but anti-calcific bone sialoprotein, in regulating the calcification of female aortic valve tissue. Recognizing that OPN mediates calcification processes, we hypothesized that aortic valvular interstitial cells (VICs) in female tissue have reduced expression of osteogenic markers in the presence of elevated OPN relative to male VICs. Human female valve leaflets displayed reduced and smaller microcalcifications, but increased OPN expression relative to male leaflets. To understand how OPN expression contributes to observed sex dimorphisms in valve tissue, we employed enzymatically degradable hydrogels as a 3D cell culture platform to recapitulate male or female VIC interactions with the extracellular matrix. Using this system, we recapitulated sex differences observed in human tissue, specifically demonstrating that female VICs exposed to calcifying medium have smaller mineral deposits within the hydrogel relative to male VICs. We identified a change in OPN dynamics in female VICs in the presence of calcification stimuli, where OPN deposition localized from the extracellular matrix to perinuclear regions. Additionally, exogenously delivered endothelin-1 to encapsulated VICs increased OPN gene expression in male cells, which resulted in reduced calcification. Collectively, our results suggest that increased OPN in female valve tissue may play a sex-specific role in mitigating mineralization during AVS progression.
Collapse
Affiliation(s)
- Megan E. Schroeder
- Department of Chemical and Biological EngineeringUniversity of Colorado BoulderBoulderColoradoUSA
- The BioFrontiers InstituteUniversity of Colorado BoulderBoulderColoradoUSA
| | - Dilara Batan
- The BioFrontiers InstituteUniversity of Colorado BoulderBoulderColoradoUSA
- Department of BiochemistryUniversity of Colorado BoulderBoulderColoradoUSA
| | - Andrea Gonzalez Rodriguez
- Department of Chemical and Biological EngineeringUniversity of Colorado BoulderBoulderColoradoUSA
- The BioFrontiers InstituteUniversity of Colorado BoulderBoulderColoradoUSA
| | - Kelly F. Speckl
- Department of Chemical and Biological EngineeringUniversity of Colorado BoulderBoulderColoradoUSA
- The BioFrontiers InstituteUniversity of Colorado BoulderBoulderColoradoUSA
| | - Douglas K. Peters
- The BioFrontiers InstituteUniversity of Colorado BoulderBoulderColoradoUSA
- Department of Molecular, Cellular, and Developmental BiologyUniversity of Colorado BoulderBoulderColoradoUSA
| | - Bruce E. Kirkpatrick
- Department of Chemical and Biological EngineeringUniversity of Colorado BoulderBoulderColoradoUSA
- The BioFrontiers InstituteUniversity of Colorado BoulderBoulderColoradoUSA
- Medical Scientist Training ProgramUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Grace K. Hach
- Department of Chemical and Biological EngineeringUniversity of Colorado BoulderBoulderColoradoUSA
- The BioFrontiers InstituteUniversity of Colorado BoulderBoulderColoradoUSA
| | - Cierra J. Walker
- The BioFrontiers InstituteUniversity of Colorado BoulderBoulderColoradoUSA
- Materials Science and Engineering ProgramUniversity of Colorado BoulderBoulderColoradoUSA
| | - Joseph C. Grim
- Department of Chemical and Biological EngineeringUniversity of Colorado BoulderBoulderColoradoUSA
- The BioFrontiers InstituteUniversity of Colorado BoulderBoulderColoradoUSA
| | - Brian A. Aguado
- Department of Chemical and Biological EngineeringUniversity of Colorado BoulderBoulderColoradoUSA
- The BioFrontiers InstituteUniversity of Colorado BoulderBoulderColoradoUSA
- Department of BioengineeringUniversity of California San DiegoLa JollaCaliforniaUSA
- Sanford Consortium for Regenerative MedicineLa JollaCaliforniaUSA
| | - Robert M. Weiss
- Department of Internal MedicineUniversity of IowaIowa CityIowaUSA
| | - Kristi S. Anseth
- Department of Chemical and Biological EngineeringUniversity of Colorado BoulderBoulderColoradoUSA
- The BioFrontiers InstituteUniversity of Colorado BoulderBoulderColoradoUSA
- Materials Science and Engineering ProgramUniversity of Colorado BoulderBoulderColoradoUSA
| |
Collapse
|
18
|
Screening for Biomarkers Associated with Left Ventricular Function During Follow-up After Acute Coronary Syndrome. J Cardiovasc Transl Res 2023; 16:244-254. [PMID: 35727504 PMCID: PMC9944718 DOI: 10.1007/s12265-022-10285-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 06/01/2022] [Indexed: 10/18/2022]
Abstract
A proportion of patients with the acute coronary syndrome (ACS) will suffer progressive remodeling of the left ventricular (LV). The aim was to screen for important biomarkers from a large-scale protein profiling in 420 ACS patients and define biomarkers associated with reduced LV function early and 1 year after the ACS. Transferrin receptor protein 1 and NT-proBNP were associated with LV function early and after 1 year, whereas osteopontin and soluble ST2 were associated with LV function in the early phase and, tissue-type plasminogen activator after 1 year. Fatty-acid-binding protein and galectin 3 were related to worse GLS but not to LVEF 1 year after the ACS. Proteins involved in remodeling and iron transport in cardiomyocytes were related to worse LV function after ACS. Biomarkers for energy metabolism and fibrosis were exclusively related to worse LV function by GLS. Studies on the functions of these proteins might add knowledge to the biological processes involved in heart failure in long term after ACS.
Collapse
|
19
|
Ni SH, OuYang XL, Liu X, Lin JH, Li Y, Sun SN, Deng JP, Han XW, Zhang XJ, Li H, Huang YS, Chen ZX, Lian ZM, Wang ZK, Long WJ, Wang LJ, Yang ZQ, Lu L. A molecular phenotypic screen reveals that lobetyolin alleviates cardiac dysfunction in 5/6 nephrectomized mice by inhibiting osteopontin. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 107:154412. [PMID: 36191549 DOI: 10.1016/j.phymed.2022.154412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 08/01/2022] [Accepted: 08/19/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Cardiovascular diseases are the major cause of mortality in patients with advanced chronic kidney diseases. The predominant abnormality observed among this population is cardiac dysfunction secondary to myocardial remodelings, such as hypertrophy and fibrosis, emphasizing the need to develop potent therapies that maintain cardiac function in patients with end-stage renal disease. AIMS To identify potential compounds and their targets as treatments for cardiorenal syndrome type 4 (CRS) using molecular phenotyping and in vivo/in vitro experiments. METHODS Gene expression was assessed using bioinformatics and verified in animal experiments using 5/6 nephrectomized mice (NPM). Based on this information, a molecular phenotyping strategy was pursued to screen potential compounds. Picrosirius red staining, wheat germ agglutinin staining, Echocardiography, immunofluorescence staining, and real-time quantitative PCR (qPCR) were utilized to evaluate the effects of compounds on CRS in vivo. Furthermore, qPCR, immunofluorescence staining and flow cytometry were applied to assess the effects of these compounds on macrophages/cardiac fibroblasts/cardiomyocytes. RNA-Seq analysis was performed to locate the targets of the selected compounds. Western blotting was performed to validate the targets and mechanisms. The reversibility of these effects was tested by overexpressing Osteopontin (OPN). RESULTS OPN expression increased more remarkably in individuals with uremia-induced cardiac dysfunction than in other cardiomyopathies. Lobetyolin (LBT) was identified in the compound screen, and it improved cardiac dysfunction and suppressed remodeling in NPM mice. Additionally, OPN modulated the effect of LBT on cardiac dysfunction in vivo and in vitro. Further experiments revealed that LBT suppressed OPN expression via the phosphorylation of c-Jun N-terminal protein kinase (JNK) signaling pathway. CONCLUSIONS LBT improved CRS by inhibiting OPN expression through the JNK pathway. This study is the first to describe a cardioprotective effect of LBT and provides new insights into CRS drug discovery.
Collapse
Affiliation(s)
- Shi-Hao Ni
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Xiao-Lu OuYang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Xin Liu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Jin-Hai Lin
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Yue Li
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Shu-Ning Sun
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Jian-Ping Deng
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Xiao-Wei Han
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Xiao-Jiao Zhang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Huan Li
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Yu-Sheng Huang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Zi-Xin Chen
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Zhi-Ming Lian
- Guangzhou integrated traditional Chinese and Western Medicine Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Zhen-Kui Wang
- Guangzhou integrated traditional Chinese and Western Medicine Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China
| | - Wen-Jie Long
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China.
| | - Ling-Jun Wang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China.
| | - Zhong-Qi Yang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China.
| | - Lu Lu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou 510407, China.
| |
Collapse
|
20
|
Hada Y, Uchida HA, Umebayashi R, Yoshida M, Wada J. Cilostazol Attenuates AngII-Induced Cardiac Fibrosis in apoE Deficient Mice. Int J Mol Sci 2022; 23:ijms23169065. [PMID: 36012328 PMCID: PMC9408896 DOI: 10.3390/ijms23169065] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
Cardiac fibrosis is characterized by the net accumulation of extracellular matrix in the myocardium and is an integral component of most pathological cardiac conditions. Cilostazol, a selective inhibitor of phosphodiesterase type III with anti-platelet, anti-mitogenic, and vasodilating properties, is widely used to treat the ischemic symptoms of peripheral vascular disease. Here, we investigated whether cilostazol has a protective effect against Angiotensin II (AngII)-induced cardiac fibrosis. Male apolipoprotein E-deficient mice were fed either a normal diet or a diet containing cilostazol (0.1% wt/wt). After 1 week of diet consumption, the mice were infused with saline or AngII (1000 ng kg−1 min−1) for 28 days. AngII infusion increased heart/body weight ratio (p < 0.05), perivascular fibrosis (p < 0.05), and interstitial cardiac fibrosis (p < 0.0001), but were significantly attenuated by cilostazol treatment (p < 0.05, respectively). Cilostazol also reduced AngII-induced increases in fibrotic and inflammatory gene expression (p < 0.05, respectively). Furthermore, cilostazol attenuated both protein and mRNA abundance of osteopontin induced by AngII in vivo. In cultured human cardiac myocytes, cilostazol reduced mRNA expression of AngII-induced osteopontin in dose-dependent manner. This reduction was mimicked by forskolin treatment but was cancelled by co-treatment of H-89. Cilostazol attenuates AngII-induced cardiac fibrosis in mice through activation of the cAMP−PKA pathway.
Collapse
Affiliation(s)
- Yoshiko Hada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Haruhito A. Uchida
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
- Department of Chronic Kidney Disease and Cardiovascular Disease, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
- Correspondence: ; Tel.: +81-86-235-7235
| | - Ryoko Umebayashi
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Masashi Yoshida
- Department of Chronic Kidney Disease and Cardiovascular Disease, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
- Department of Cardiovascular Medicine, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| |
Collapse
|
21
|
Mamazhakypov A, Sartmyrzaeva M, Sarybaev AS, Schermuly R, Sydykov A. Clinical and Molecular Implications of Osteopontin in Heart Failure. Curr Issues Mol Biol 2022; 44:3573-3597. [PMID: 36005141 PMCID: PMC9406846 DOI: 10.3390/cimb44080245] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/04/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
The matricellular protein osteopontin modulates cell-matrix interactions during tissue injury and healing. A complex multidomain structure of osteopontin enables it not only to bind diverse cell receptors but also to interact with various partners, including other extracellular matrix proteins, cytokines, and growth factors. Numerous studies have implicated osteopontin in the development and progression of myocardial remodeling in diverse cardiac diseases. Osteopontin influences myocardial remodeling by regulating extracellular matrix production, the activity of matrix metalloproteinases and various growth factors, inflammatory cell recruitment, myofibroblast differentiation, cardiomyocyte apoptosis, and myocardial vascularization. The exploitation of osteopontin loss- and gain-of-function approaches in rodent models provided an opportunity for assessment of the cell- and disease-specific contribution of osteopontin to myocardial remodeling. In this review, we summarize the recent knowledge on osteopontin regulation and its impact on various cardiac diseases, as well as delineate complex disease- and cell-specific roles of osteopontin in cardiac pathologies. We also discuss the current progress of therapeutics targeting osteopontin that may facilitate the development of a novel strategy for heart failure treatment.
Collapse
Affiliation(s)
- Argen Mamazhakypov
- Department of Internal Medicine, German Center for Lung Research (DZL), Justus Liebig University of Giessen, 35392 Giessen, Germany
| | - Meerim Sartmyrzaeva
- Department of Mountain and Sleep Medicine and Pulmonary Hypertension, National Center of Cardiology and Internal Medicine, Bishkek 720040, Kyrgyzstan
| | - Akpay Sh. Sarybaev
- Department of Mountain and Sleep Medicine and Pulmonary Hypertension, National Center of Cardiology and Internal Medicine, Bishkek 720040, Kyrgyzstan
| | - Ralph Schermuly
- Department of Internal Medicine, German Center for Lung Research (DZL), Justus Liebig University of Giessen, 35392 Giessen, Germany
| | - Akylbek Sydykov
- Department of Internal Medicine, German Center for Lung Research (DZL), Justus Liebig University of Giessen, 35392 Giessen, Germany
- Correspondence:
| |
Collapse
|
22
|
Saleh S, George J, Kott KA, Meikle PJ, Figtree GA. The Translation and Commercialisation of Biomarkers for Cardiovascular Disease—A Review. Front Cardiovasc Med 2022; 9:897106. [PMID: 35722087 PMCID: PMC9201254 DOI: 10.3389/fcvm.2022.897106] [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: 03/15/2022] [Accepted: 05/11/2022] [Indexed: 11/19/2022] Open
Abstract
As a leading cause of mortality and morbidity worldwide, cardiovascular disease and its diagnosis, quantification, and stratification remain significant health issues. Increasingly, patients present with cardiovascular disease in the absence of known risk factors, suggesting the presence of yet unrecognized pathological processes and disease predispositions. Fortunately, a host of emerging cardiovascular biomarkers characterizing and quantifying ischaemic heart disease have shown great promise in both laboratory settings and clinical trials. These have demonstrated improved predictive value additional to widely accepted biomarkers as well as providing insight into molecular phenotypes beneath the broad umbrella of cardiovascular disease that may allow for further personalized treatment regimens. However, the process of translation into clinical practice – particularly navigating the legal and commercial landscape – poses a number of challenges. Practical and legal barriers to the biomarker translational pipeline must be further considered to develop strategies to bring novel biomarkers into the clinical sphere and apply these advances at the patient bedside. Here we review the progress of emerging biomarkers in the cardiovascular space, with particular focus on those relevant to the unmet needs in ischaemic heart disease.
Collapse
Affiliation(s)
- Soloman Saleh
- Cardiothoracic and Vascular Health, Kolling Institute of Medical Research, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Jacob George
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Katharine A. Kott
- Cardiothoracic and Vascular Health, Kolling Institute of Medical Research, Sydney, NSW, Australia
- Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Peter J. Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Gemma A. Figtree
- Cardiothoracic and Vascular Health, Kolling Institute of Medical Research, Sydney, NSW, Australia
- Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- *Correspondence: Gemma A. Figtree
| |
Collapse
|
23
|
González A, Richards AM, de Boer RA, Thum T, Arfsten H, Hülsmann M, Falcao-Pires I, Díez J, Foo RSY, Chan MY, Aimo A, Anene-Nzelu CG, Abdelhamid M, Adamopoulos S, Anker SD, Belenkov Y, Ben Gal T, Cohen-Solal A, Böhm M, Chioncel O, Delgado V, Emdin M, Jankowska EA, Gustafsson F, Hill L, Jaarsma T, Januzzi JL, Jhund PS, Lopatin Y, Lund LH, Metra M, Milicic D, Moura B, Mueller C, Mullens W, Núñez J, Piepoli MF, Rakisheva A, Ristić AD, Rossignol P, Savarese G, Tocchetti CG, Van Linthout S, Volterrani M, Seferovic P, Rosano G, Coats AJS, Bayés-Genís A. Cardiac remodelling - Part 1: From cells and tissues to circulating biomarkers. A review from the Study Group on Biomarkers of the Heart Failure Association of the European Society of Cardiology. Eur J Heart Fail 2022; 24:927-943. [PMID: 35334137 DOI: 10.1002/ejhf.2493] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/09/2022] [Accepted: 03/21/2022] [Indexed: 11/10/2022] Open
Abstract
Cardiac remodelling refers to changes in left ventricular structure and function over time, with a progressive deterioration that may lead to heart failure (HF) development (adverse remodelling) or vice versa a recovery (reverse remodelling) in response to HF treatment. Adverse remodelling predicts a worse outcome, whilst reverse remodelling predicts a better prognosis. The geometry, systolic and diastolic function and electric activity of the left ventricle are affected, as well as the left atrium and on the long term even right heart chambers. At a cellular and molecular level, remodelling involves all components of cardiac tissue: cardiomyocytes, fibroblasts, endothelial cells and leucocytes. The molecular, cellular and histological signatures of remodelling may differ according to the cause and severity of cardiac damage, and clearly to the global trend toward worsening or recovery. These processes cannot be routinely evaluated through endomyocardial biopsies, but may be reflected by circulating levels of several biomarkers. Different classes of biomarkers (e.g. proteins, non-coding RNAs, metabolites and/or epigenetic modifications) and several biomarkers of each class might inform on some aspects on HF development, progression and long-term outcomes, but most have failed to enter clinical practice. This may be due to the biological complexity of remodelling, so that no single biomarker could provide great insight on remodelling when assessed alone. Another possible reason is a still incomplete understanding of the role of biomarkers in the pathophysiology of cardiac remodelling. Such role will be investigated in the first part of this review paper on biomarkers of cardiac remodelling.
Collapse
Affiliation(s)
- Arantxa González
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, and IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - A Mark Richards
- Department of medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore
- Christchurch Heart Institute, University of Otago, Dunedin, New Zealand
| | - Rudolf A de Boer
- University Medical Center Groningen, University of Groningen, Department of Cardiology, Groningen, The Netherlands
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS) and Rebirth Center for Translational Regenerative Therapies, Hannover Medical School, Hannover, Germany
- Fraunhofer Institute of Toxicology and Experimental Medicine, Hannover, Germany
| | - Henrike Arfsten
- Clinical Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Martin Hülsmann
- Clinical Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Inês Falcao-Pires
- Department od Surgery and Physiology, Cardiovascular Research and Development Center, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Javier Díez
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, and IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
- Departments of Cardiology and Cardiac Surgery, and Nephrology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Roger S Y Foo
- Department of medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore
| | - Mark Y Chan
- Department of medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore
| | - Alberto Aimo
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Chukwuemeka G Anene-Nzelu
- Department of medicine, Yong Loo-Lin School of Medicine, National University of Singapore, Singapore
- Montreal Heart Institute, Montreal, Canada
| | | | - Stamatis Adamopoulos
- 2nd Department of Cardiovascular Medicine, Onassis Cardiac Surgery Center, Athens, Greece
| | - Stefan D Anker
- Department of Cardiology (CVK), and Berlin Institute of Health Center for Regenerative Therapies (BCRT), German Centre for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitätsmedizin, Berlin, Germany
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | | | - Tuvia Ben Gal
- Cardiology Department, Rabin Medical Center, Beilinson, Israel
| | | | - Michael Böhm
- Universitätsklinikum des Saarlandes, Klinik für Innere Medizin III, Kardiologie, Angiologie und Internistische Intensivmedizin, Saarland University, Homburg/Saar, Germany
| | - Ovidiu Chioncel
- Emergency Institute for Cardiovascular Diseases 'Prof. C.C. Iliescu' Bucharest, University of Medicine Carol Davila, Bucharest, Romania
| | - Victoria Delgado
- Institut del Cor, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Michele Emdin
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Ewa A Jankowska
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Finn Gustafsson
- Rigshospitalet-Copenhagen University Hospital, Heart Centre, Department of Cardiology, Copenhagen, Denmark
| | | | | | - James L Januzzi
- Massachusetts General Hospital and Baim Institute for Clinical Research, Boston, MA, USA
| | - Pardeep S Jhund
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland
| | - Yuri Lopatin
- Volgograd State Medical University, Volgograd, Russia
| | - Lars H Lund
- Department of Medicine, Karolinska Institutet, and Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Marco Metra
- Cardiology, ASST Spedali Civili; Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Davor Milicic
- University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Brenda Moura
- Faculty of Medicine, University of Porto, Porto, Portugal
- Cardiology Department, Porto Armed Forces Hospital, Portugal
| | | | | | - Julio Núñez
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
- Hospital Clínico Universitario de Valencia, INCLIVA, Universidad de Valencia, Valencia, Spain
| | - Massimo F Piepoli
- Cardiology Division, Castelsangiovanni Hospital, Castelsangiovanni, Italy
| | - Amina Rakisheva
- Scientific Research Institute of Cardiology and Internal Medicine, Almaty, Kazakhstan
| | - Arsen D Ristić
- Department of Cardiology, University Clinical Center of Serbia, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Patrick Rossignol
- Université de Lorraine, Centre d'Investigations Cliniques- Plurithématique 1433, and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT, Nancy, France
| | - Gianluigi Savarese
- Department of Medicine, Karolinska Institutet, and Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Carlo G Tocchetti
- Cardio-Oncology Unit, Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | - Sophie Van Linthout
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | | | - Petar Seferovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - Giuseppe Rosano
- St. George's Hospitals, NHS Trust, University of London, London, UK
| | | | - Antoni Bayés-Genís
- CIBERCV, Carlos III Institute of Health, Madrid, Spain
- Institut del Cor, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
24
|
Khamissi FZ, Ning L, Kefaloyianni E, Dun H, Arthanarisami A, Keller A, Atkinson JJ, Li W, Wong B, Dietmann S, Lavine K, Kreisel D, Herrlich A. Identification of kidney injury released circulating osteopontin as causal agent of respiratory failure. SCIENCE ADVANCES 2022; 8:eabm5900. [PMID: 35213222 PMCID: PMC8880785 DOI: 10.1126/sciadv.abm5900] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/30/2021] [Indexed: 05/08/2023]
Abstract
Tissue injury can drive secondary organ injury; however, mechanisms and mediators are not well understood. To identify interorgan cross-talk mediators, we used acute kidney injury (AKI)-induced acute lung injury (ALI) as a clinically important example. Using kidney and lung single-cell RNA sequencing after AKI in mice followed by ligand-receptor pairing analysis across organs, kidney ligands to lung receptors, we identify kidney-released circulating osteopontin (OPN) as a novel AKI-ALI mediator. OPN release from kidney tubule cells triggered lung endothelial leakage, inflammation, and respiratory failure. Pharmacological or genetic OPN inhibition prevented AKI-ALI. Transplantation of ischemic wt kidneys caused AKI-ALI, but not of ischemic OPN-global knockout kidneys, identifying kidney-released OPN as necessary interorgan signal to cause AKI-ALI. We show that OPN serum levels are elevated in patients with AKI and correlate with kidney injury. Our results demonstrate feasibility of using ligand-receptor analysis across organs to identify interorgan cross-talk mediators and may have important therapeutic implications in human AKI-ALI and multiorgan failure.
Collapse
Affiliation(s)
| | | | | | - Hao Dun
- Washington University School in St. Louis School of Medicine, 660 S Euclid Avenue, St. Louis, MO 63110, USA
| | | | - Amy Keller
- Washington University School in St. Louis School of Medicine, 660 S Euclid Avenue, St. Louis, MO 63110, USA
| | - Jeffrey J. Atkinson
- Washington University School in St. Louis School of Medicine, 660 S Euclid Avenue, St. Louis, MO 63110, USA
| | - Wenjun Li
- Washington University School in St. Louis School of Medicine, 660 S Euclid Avenue, St. Louis, MO 63110, USA
| | - Brian Wong
- Washington University School in St. Louis School of Medicine, 660 S Euclid Avenue, St. Louis, MO 63110, USA
| | - Sabine Dietmann
- Washington University School in St. Louis School of Medicine, 660 S Euclid Avenue, St. Louis, MO 63110, USA
| | - Kory Lavine
- Washington University School in St. Louis School of Medicine, 660 S Euclid Avenue, St. Louis, MO 63110, USA
| | - Daniel Kreisel
- Washington University School in St. Louis School of Medicine, 660 S Euclid Avenue, St. Louis, MO 63110, USA
| | | |
Collapse
|
25
|
Fotiou D, Theodorakakou F, Kastritis E. Biomarkers in AL Amyloidosis. Int J Mol Sci 2021; 22:ijms222010916. [PMID: 34681575 PMCID: PMC8536050 DOI: 10.3390/ijms222010916] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/02/2021] [Accepted: 10/07/2021] [Indexed: 01/01/2023] Open
Abstract
Systemic AL amyloidosis is a rare complex hematological disorder caused by clonal plasma cells which produce amyloidogenic immunoglobulins. Outcome and prognosis is the combinatory result of the extent and pattern of organ involvement secondary to amyloid fibril deposition and the biology and burden of the underlying plasma cell clone. Prognosis, as assessed by overall survival, and early outcomes is determined by degree of cardiac dysfunction and current staging systems are based on biomarkers that reflect the degree of cardiac damage. The risk of progression to end-stage renal disease requiring dialysis is assessed by renal staging systems. Longer-term survival and response to treatment is affected by markers of the underlying plasma cell clone; the genetic background of the clonal disease as evaluated by interphase fluorescence in situ hybridization in particular has predictive value and may guide treatment selection. Free light chain assessment forms the basis of hematological response criteria and minimal residual disease as assessed by sensitive methods is gradually being incorporated into clinical practice. However, sensitive biomarkers that could aid in the early diagnosis and that could reflect all aspects of organ damage and disease biology are needed and efforts to identify them are continuous.
Collapse
|
26
|
Shirakawa K, Sano M. T Cell Immunosenescence in Aging, Obesity, and Cardiovascular Disease. Cells 2021; 10:cells10092435. [PMID: 34572084 PMCID: PMC8464832 DOI: 10.3390/cells10092435] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 12/24/2022] Open
Abstract
Although advances in preventive medicine have greatly improved prognosis, cardiovascular disease (CVD) remains the leading cause of death worldwide. This clearly indicates that there remain residual cardiovascular risks that have not been targeted by conventional therapies. The results of multiple animal studies and clinical trials clearly indicate that inflammation is the most important residual risk and a potential target for CVD prevention. The immune cell network is intricately regulated to maintain homeostasis. Ageing associated changes to the immune system occurs in both innate and adaptive immune cells, however T cells are most susceptible to this process. T-cell changes due to thymic degeneration and homeostatic proliferation, metabolic abnormalities, telomere length shortening, and epigenetic changes associated with aging and obesity may not only reduce normal immune function, but also induce inflammatory tendencies, a process referred to as immunosenescence. Since the disruption of biological homeostasis by T cell immunosenescence is closely related to the development and progression of CVD via inflammation, senescent T cells are attracting attention as a new therapeutic target. In this review, we discuss the relationship between CVD and T cell immunosenescence associated with aging and obesity.
Collapse
Affiliation(s)
- Kohsuke Shirakawa
- Department of Cardiovascular Medicine, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo 1138421, Japan;
| | - Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 1608582, Japan
- Correspondence: ; Tel.: +81-(3)-5363-3874
| |
Collapse
|
27
|
Zhu Z, He Y, Shi M, Guo D, Zhang K, Ren L, Peng Y, Yang P, Chen J, Zang Y, Wang A, Xu T, Li Q, Ju Z, Geng D, Zhang Y, He J. Plasma osteopontin levels and adverse clinical outcomes after ischemic stroke. Atherosclerosis 2021; 332:33-40. [PMID: 34375911 DOI: 10.1016/j.atherosclerosis.2021.07.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 06/07/2021] [Accepted: 07/29/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND AIMS Osteopontin is implicated in atherosclerosis, and its expression is upregulated in response to brain injury. The aim of this study was to prospectively investigate the associations between plasma osteopontin levels and adverse clinical outcomes in ischemic stroke patients. METHODS We measured baseline plasma osteopontin levels in 3545 ischemic stroke patients from the China Antihypertensive Trial in Acute Ischemic Stroke (CATIS). The primary outcome was the composite outcome of death and major disability (modified Rankin scale score ≥3) at 1 year after ischemic stroke, and secondary outcomes included major disability, death, and the composite outcome of death and vascular events. RESULTS During 1 year of follow-up, patients in the fourth quartile of plasma osteopontin had the highest risks of primary outcome, major disability, death, and the composite outcome of death and vascular events. After multivariate adjustment, the odds ratios or hazard ratios (95 % confidence intervals) associated with each standard deviation increase in log-transformed osteopontin were 1.20 (1.09-1.33) for primary outcome, 1.11 (1.00-1.23) for major disability, 1.29 (1.10-1.52) for death, and 1.15 (1.01-1.30) for the composite outcome of death and vascular events. The addition of plasma osteopontin to conventional risk factors significantly improved the risk reclassification for the primary outcome (net reclassification improvement: 16.91%, p < 0.001; integrated discrimination improvement: 0.43%, p = 0.002). CONCLUSIONS Elevated plasma osteopontin levels at baseline were associated with increased risks of adverse clinical outcomes at 1 year after ischemic stroke, suggesting that osteopontin is a promising prognostic biomarker for ischemic stroke.
Collapse
Affiliation(s)
- Zhengbao Zhu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China; Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Yan He
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Mengyao Shi
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Daoxia Guo
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Kaixin Zhang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Liyun Ren
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Yanbo Peng
- Department of Neurology, Affiliated Hospital of North China University of Science and Technology, Hebei, China
| | - Pinni Yang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Jing Chen
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA; Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Yuhan Zang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Aili Wang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Tan Xu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Qunwei Li
- Department of Epidemiology, School of Public Health, Taishan Medical College, Shandong, China
| | - Zhong Ju
- Department of Neurology, Kerqin District First People's Hospital of Tongliao City, Inner Mongolia, China
| | - Deqin Geng
- Department of Neurology, Affiliated Hospital of Xuzhou Medical College, Jiangsu, China
| | - Yonghong Zhang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China.
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA; Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
28
|
Keranov S, Dörr O, Jafari L, Liebetrau C, Keller T, Troidl C, Riehm J, Rutsatz W, Bauer P, Kriechbaum S, Voss S, Richter MJ, Tello K, Gall H, Ghofrani HA, Guth S, Seeger W, Hamm CW, Nef H. Osteopontin and galectin-3 as biomarkers of maladaptive right ventricular remodeling in pulmonary hypertension. Biomark Med 2021; 15:1021-1034. [PMID: 34289706 DOI: 10.2217/bmm-2021-0009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: This study assessed the utility of osteopontin (OPN) and galectin-3 (Gal-3) as biomarkers of maladaptive right ventricular remodeling in pulmonary hypertension (PH). Materials & methods: We examined plasma levels of OPN and Gal-3 in patients with PH (n = 62), dilated cardiomyopathy (n = 34), left ventricular hypertrophy (LVH; n = 47), and controls without right ventricle (RV) or LV abnormalities (n = 38). Results: OPN and Gal-3 levels were higher in PH, dilated cardiomyopathy and LVH than in the controls. OPN concentrations in PH patients with maladaptive RV were significantly higher than in those with adaptive RV. Gal-3 did not differentiate between adaptive and maladaptive RV remodeling in PH. OPN and Gal-3 levels did not correlate with parameters of LV remodeling. Conclusion: OPN is a potential biomarker of RV maladaptation.
Collapse
Affiliation(s)
- Stanislav Keranov
- Department of Cardiology & Angiology, University of Giessen, Giessen, 35392, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Rhein Main, Bad Nauheim, 61231, Germany
| | - Oliver Dörr
- Department of Cardiology & Angiology, University of Giessen, Giessen, 35392, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Rhein Main, Bad Nauheim, 61231, Germany
| | - Leili Jafari
- Department of Cardiology, Kerckhoff Heart & Lung Center, Bad Nauheim, 61231, Germany
| | - Christoph Liebetrau
- Department of Cardiology & Angiology, University of Giessen, Giessen, 35392, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Rhein Main, Bad Nauheim, 61231, Germany.,Department of Cardiology, Kerckhoff Heart & Lung Center, Bad Nauheim, 61231, Germany
| | - Till Keller
- DZHK (German Center for Cardiovascular Research), Partner Site Rhein Main, Bad Nauheim, 61231, Germany.,Department of Cardiology, Kerckhoff Heart & Lung Center, Bad Nauheim, 61231, Germany
| | - Christian Troidl
- DZHK (German Center for Cardiovascular Research), Partner Site Rhein Main, Bad Nauheim, 61231, Germany.,Department of Cardiology, Kerckhoff Heart & Lung Center, Bad Nauheim, 61231, Germany
| | - Jessica Riehm
- Department of Cardiology & Angiology, University of Giessen, Giessen, 35392, Germany
| | - Wiebke Rutsatz
- Department of Cardiology & Angiology, University of Giessen, Giessen, 35392, Germany
| | - Pascal Bauer
- Department of Cardiology & Angiology, University of Giessen, Giessen, 35392, Germany
| | - Steffen Kriechbaum
- DZHK (German Center for Cardiovascular Research), Partner Site Rhein Main, Bad Nauheim, 61231, Germany.,Department of Cardiology, Kerckhoff Heart & Lung Center, Bad Nauheim, 61231, Germany
| | - Sandra Voss
- DZHK (German Center for Cardiovascular Research), Partner Site Rhein Main, Bad Nauheim, 61231, Germany.,Department of Cardiology, Kerckhoff Heart & Lung Center, Bad Nauheim, 61231, Germany
| | - Manuel J Richter
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen & Marburg Lung Center (UGMLC), Institute for Lung Health (ILH), Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Giessen, 35392, Germany
| | - Khodr Tello
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen & Marburg Lung Center (UGMLC), Institute for Lung Health (ILH), Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Giessen, 35392, Germany
| | - Henning Gall
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen & Marburg Lung Center (UGMLC), Institute for Lung Health (ILH), Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Giessen, 35392, Germany
| | - Hossein A Ghofrani
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen & Marburg Lung Center (UGMLC), Institute for Lung Health (ILH), Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Giessen, 35392, Germany
| | - Stefan Guth
- Department of Thoracic Surgery, Kerckhoff Heart & Lung Center, Bad Nauheim, 61231, Germany
| | - Werner Seeger
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen & Marburg Lung Center (UGMLC), Institute for Lung Health (ILH), Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), Giessen, 35392, Germany
| | - Christian W Hamm
- Department of Cardiology & Angiology, University of Giessen, Giessen, 35392, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Rhein Main, Bad Nauheim, 61231, Germany.,Department of Cardiology, Kerckhoff Heart & Lung Center, Bad Nauheim, 61231, Germany
| | - Holger Nef
- Department of Cardiology & Angiology, University of Giessen, Giessen, 35392, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Rhein Main, Bad Nauheim, 61231, Germany.,Department of Cardiology, Kerckhoff Heart & Lung Center, Bad Nauheim, 61231, Germany
| |
Collapse
|
29
|
Osteopontin in Cardiovascular Diseases. Biomolecules 2021; 11:biom11071047. [PMID: 34356671 PMCID: PMC8301767 DOI: 10.3390/biom11071047] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/14/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022] Open
Abstract
Unprecedented advances in secondary prevention have greatly improved the prognosis of cardiovascular diseases (CVDs); however, CVDs remain a leading cause of death globally. These findings suggest the need to reconsider cardiovascular risk and optimal medical therapy. Numerous studies have shown that inflammation, pro-thrombotic factors, and gene mutations are focused not only on cardiovascular residual risk but also as the next therapeutic target for CVDs. Furthermore, recent clinical trials, such as the Canakinumab Anti-inflammatory Thrombosis Outcomes Study trial, showed the possibility of anti-inflammatory therapy for patients with CVDs. Osteopontin (OPN) is a matricellular protein that mediates diverse biological functions and is involved in a number of pathological states in CVDs. OPN has a two-faced phenotype that is dependent on the pathological state. Acute increases in OPN have protective roles, including wound healing, neovascularization, and amelioration of vascular calcification. By contrast, chronic increases in OPN predict poor prognosis of a major adverse cardiovascular event independent of conventional cardiovascular risk factors. Thus, OPN can be a therapeutic target for CVDs but is not clinically available. In this review, we discuss the role of OPN in the development of CVDs and its potential as a therapeutic target.
Collapse
|
30
|
Khan S, Rasool ST. Current Use of Cardiac Biomarkers in Various Heart Conditions. Endocr Metab Immune Disord Drug Targets 2021; 21:980-993. [PMID: 32867665 DOI: 10.2174/1871530320999200831171748] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 08/07/2020] [Accepted: 08/07/2020] [Indexed: 01/08/2023]
Abstract
Biomarkers are increasingly recognized to have significant clinical value in early identification and progression of various cardiovascular diseases. There are many heart conditions, such as congestive heart failure (CHF), ischemic heart diseases (IHD), and diabetic cardiomyopathy (DCM), and cardiac remodeling, in which the severity of the cardiac pathology can be mirrored through these cardiac biomarkers. From the emergency department (ED) evaluation of acute coronary syndromes (ACS) or suspected acute myocardial infarction (AMI) with cardiac marker Troponin to the diagnosis of chronic conditions like Heart Failure (HF) with natriuretic peptides, like B-type natriuretic peptide (BNP), N-terminal pro-B- type natriuretic peptide (Nt-proBNP) and mid regional pro-atrial natriuretic peptide (MR- proANP), their use is continuously increasing. Their clinical importance has led to the discovery of newer biomarkers, such as the soluble source of tumorigenicity 2 (sST2), galectin-3 (Gal-3), growth differentiation factor-15 (GDF-15), and various micro ribonucleic acids (miRNAs). Since cardiac pathophysiology involves a complex interplay between inflammatory, genetic, neurohormonal, and biochemical levels, these biomarkers could be enzymes, hormones, and biologic substances showing cardiac injury, stress, and malfunction. Therefore, multi-marker approaches with different combinations of novel cardiac biomarkers, and continual assessment of cardiac biomarkers are likely to improve cardiac risk prediction, stratification, and overall patient wellbeing. On the other hand, these biomarkers may reflect coexisting or isolated disease processes in different organ systems other than the cardiovascular system. Therefore, knowledge of cardiac biomarkers is imperative. In this article, we have reviewed the role of cardiac biomarkers and their use in the diagnosis and prognosis of various cardiovascular diseases from different investigations conducted in recent years.
Collapse
Affiliation(s)
- Shahzad Khan
- Department of Pathophysiology, Wuhan University School of Medicine, Hubei, Wuhan 4300711, China
| | | |
Collapse
|
31
|
Cediel G, Codina P, Spitaleri G, Domingo M, Santiago-Vacas E, Lupón J, Bayes-Genis A. Gender-Related Differences in Heart Failure Biomarkers. Front Cardiovasc Med 2021; 7:617705. [PMID: 33469552 PMCID: PMC7813809 DOI: 10.3389/fcvm.2020.617705] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/09/2020] [Indexed: 12/13/2022] Open
Abstract
Important differences in comorbidities and clinical characteristics exist between women and men with heart failure (HF). In particular, differences in the kinetics of biological circulating biomarkers—a critical component of cardiovascular care—are highly relevant. Most circulating HF biomarkers are assessed daily by clinicians without taking sex into account, despite the multiple gender-related differences observed in plasma concentrations. Even in health, compared to men, women tend to exhibit higher levels of natriuretic peptides and galectin-3 and lower levels of cardiac troponins and the cardiac stress marker, soluble ST2. Many biological factors can provide a reliable explanation for these differences, like body composition, fat distribution, or menopausal status. Notwithstanding, these sex-specific differences in biomarker levels do not reflect different pathobiological mechanisms in HF between women and men, and they do not necessarily imply a need to use different diagnostic cut-off levels in clinical practice. To date, the sex-specific prognostic value of HF biomarkers for risk stratification is an unresolved issue that future research must elucidate. This review outlines current evidence regarding gender-related differences in circulating biomarkers widely used in HF, the pathophysiological mechanisms underlying these differences, and their clinical relevance.
Collapse
Affiliation(s)
- Germán Cediel
- Heart Institute, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Department of Medicine, CIBERCV, Autonomous University of Barcelona, Barcelona, Spain
| | - Pau Codina
- Heart Institute, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Department of Medicine, CIBERCV, Autonomous University of Barcelona, Barcelona, Spain
| | - Giosafat Spitaleri
- Heart Institute, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Department of Medicine, CIBERCV, Autonomous University of Barcelona, Barcelona, Spain
| | - Mar Domingo
- Heart Institute, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Department of Medicine, CIBERCV, Autonomous University of Barcelona, Barcelona, Spain
| | - Evelyn Santiago-Vacas
- Heart Institute, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Department of Medicine, CIBERCV, Autonomous University of Barcelona, Barcelona, Spain
| | - Josep Lupón
- Heart Institute, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Department of Medicine, CIBERCV, Autonomous University of Barcelona, Barcelona, Spain
| | - Antoni Bayes-Genis
- Heart Institute, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Department of Medicine, CIBERCV, Autonomous University of Barcelona, Barcelona, Spain
| |
Collapse
|
32
|
Topf A, Mirna M, Ohnewein B, Jirak P, Kopp K, Fejzic D, Haslinger M, Motloch LJ, Hoppe UC, Berezin A, Lichtenauer M. The Diagnostic and Therapeutic Value of Multimarker Analysis in Heart Failure. An Approach to Biomarker-Targeted Therapy. Front Cardiovasc Med 2020; 7:579567. [PMID: 33344515 PMCID: PMC7746655 DOI: 10.3389/fcvm.2020.579567] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 11/02/2020] [Indexed: 12/12/2022] Open
Abstract
Background: Heart failure is a pathophysiological state, which is still associated with high morbidity and mortality despite established therapies. Diverse well-known biomarkers fail to assess the variety of individual pathophysiology in the context of heart failure. Methods: An analysis of prospective, multimarker-specific therapeutic approaches to heart failure based on studies in current literature was performed. A total of 159 screened publications in the field of biomarkers in heart failure were hand-selected and found to be eligible for this study by a team of experts. Results: Established biomarkers of the inflammatory axis, matrix remodeling, fibrosis and oxidative stress axis, as well as potential therapeutic interventions were investigated. Interaction with end organs, such as cardio-hepatic, cardio-renal and cardio-gastrointestinal interactions show the complexity of the syndrome and could be of further therapeutic value. MicroRNAs are involved in a wide variety of physiologic and pathophysiologic processes in heart failure and could be useful in diagnostic as well as therapeutic setting. Conclusion: Based on our analysis by a biomarker-driven approach in heart failure therapy, patients could be treated more specifically in long term with a consideration of different aspects of heart failure. New studies evaluating a multimarker - based therapeutic approach could lead in a decrease in the morbidity and mortality of heart failure patients.
Collapse
Affiliation(s)
- Albert Topf
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Moritz Mirna
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Bernhard Ohnewein
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Peter Jirak
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Kristen Kopp
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Dzeneta Fejzic
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Michael Haslinger
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Lukas J. Motloch
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Uta C. Hoppe
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Alexander Berezin
- Internal Medicine Department, State Medical University, Zaporozhye, Ukraine
| | - Michael Lichtenauer
- Department of Cardiology, Clinic of Internal Medicine II, Paracelsus Medical University of Salzburg, Salzburg, Austria
| |
Collapse
|
33
|
Shirakawa K, Sano M. Sodium-Glucose Co-Transporter 2 Inhibitors Correct Metabolic Maladaptation of Proximal Tubular Epithelial Cells in High-Glucose Conditions. Int J Mol Sci 2020; 21:ijms21207676. [PMID: 33081406 PMCID: PMC7589591 DOI: 10.3390/ijms21207676] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/07/2020] [Accepted: 10/14/2020] [Indexed: 01/10/2023] Open
Abstract
Glucose filtered in the glomerulus is actively reabsorbed by sodium-glucose co-transporter 2 (SGLT2) in proximal tubular epithelial cells (PTEC) and passively returned to the blood via glucose transporter 2 (GLUT2). Healthy PTEC rely primarily on fatty acid beta-oxidation (FAO) for energy. In phase III trials, SGLT2 inhibitors improved outcomes in diabetic kidney disease (DKD). Tubulointerstitial renal fibrosis due to altered metabolic reprogramming of PTEC might be at the root of the pathogenesis of DKD. Here, we investigated the molecular mechanism of SGLT2 inhibitors’ renoprotective effect by examining transcriptional activity of Spp1, which encodes osteopontin, a key mediator of tubulointerstitial renal fibrosis. With primary cultured PTEC from Spp1-enhanced green fluorescent protein knock-in mice, we proved that in high-glucose conditions, increased SGLT2- and GLUT-mediated glucose uptake is causatively involved in aberrant activation of the glycolytic pathway in PTEC, thereby increasing mitochondrial reactive oxygen species (ROS) formation and transcriptional activation of Spp1. FAO activation did not play a direct role in these processes, but elevated expression of a tubular-specific enzyme, myo-inositol oxygenase, was at least partly involved. Notably, canagliflozin blocked overexpression of myo-inositol oxygenase. In conclusion, SGLT2 inhibitors exerted renoprotective effects by inhibiting aberrant glycolytic metabolism and mitochondrial ROS formation in PTEC in high-glucose conditions.
Collapse
Affiliation(s)
- Kohsuke Shirakawa
- Department of Cardiovascular Medicine, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo 113-8431, Japan;
| | - Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Correspondence: ; Tel.: +81-(3)-5363-3874
| |
Collapse
|
34
|
Tsang HG, Clark EL, Markby GR, Bush SJ, Hume DA, Corcoran BM, MacRae VE, Summers KM. Expression of Calcification and Extracellular Matrix Genes in the Cardiovascular System of the Healthy Domestic Sheep ( Ovis aries). Front Genet 2020; 11:919. [PMID: 33101359 PMCID: PMC7506100 DOI: 10.3389/fgene.2020.00919] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/23/2020] [Indexed: 12/31/2022] Open
Abstract
The maintenance of a healthy cardiovascular system requires expression of genes that contribute to essential biological activities and repression of those that are associated with functions likely to be detrimental to cardiovascular homeostasis. Vascular calcification is a major disruption to cardiovascular homeostasis, where tissues of the cardiovascular system undergo ectopic calcification and consequent dysfunction, but little is known about the expression of calcification genes in the healthy cardiovascular system. Large animal models are of increasing importance in cardiovascular disease research as they demonstrate more similar cardiovascular features (in terms of anatomy, physiology and size) to humans than do rodent species. We used RNA sequencing results from the sheep, which has been utilized extensively to examine calcification of prosthetic cardiac valves, to explore the transcriptome of the heart and cardiac valves in this large animal, in particular looking at expression of calcification and extracellular matrix genes. We then examined genes implicated in the process of vascular calcification in a wide array of cardiovascular tissues and across multiple developmental stages, using RT-qPCR. Our results demonstrate that there is a balance between genes that promote and those that suppress mineralization during development and across cardiovascular tissues. We show extensive expression of genes encoding proteins involved in formation and maintenance of the extracellular matrix in cardiovascular tissues, and high expression of hematopoietic genes in the cardiac valves. Our analysis will support future research into the functions of implicated genes in the development of valve calcification, and increase the utility of the sheep as a large animal model for understanding ectopic calcification in cardiovascular disease. This study provides a foundation to explore the transcriptome of the developing cardiovascular system and is a valuable resource for the fields of mammalian genomics and cardiovascular research.
Collapse
Affiliation(s)
- Hiu-Gwen Tsang
- The Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, United Kingdom
| | - Emily L. Clark
- The Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, United Kingdom
| | - Greg R. Markby
- The Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, United Kingdom
| | - Stephen J. Bush
- The Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, United Kingdom
- Nuffield Department of Clinical Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - David A. Hume
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - Brendan M. Corcoran
- The Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh, United Kingdom
| | - Vicky E. MacRae
- The Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, United Kingdom
| | - Kim M. Summers
- The Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, United Kingdom
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| |
Collapse
|
35
|
The evolving systemic biomarker milieu in obese ZSF1 rat model of human cardiometabolic syndrome: Characterization of the model and cardioprotective effect of GDF15. PLoS One 2020; 15:e0231234. [PMID: 32804947 PMCID: PMC7430742 DOI: 10.1371/journal.pone.0231234] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 08/03/2020] [Indexed: 12/22/2022] Open
Abstract
Cardiometabolic syndrome has become a global health issue. Heart failure is a common comorbidity of cardiometabolic syndrome. Successful drug development to prevent cardiometabolic syndrome and associated comorbidities requires preclinical models predictive of human conditions. To characterize the heart failure component of cardiometabolic syndrome, cardiometabolic, metabolic, and renal biomarkers were evaluated in lean and obese ZSF1 19- to 32-week-old male rats. Histopathological assessment of kidneys and hearts was performed. Cardiac function, exercise capacity, and left ventricular gene expression were also analyzed. Obese ZSF1 rats exhibited multiple features of human cardiometabolic syndrome by pathological changes in systemic renal, metabolic, and cardiovascular disease circulating biomarkers. Hemodynamic assessment, echocardiography, and decreased exercise capacity confirmed heart failure with preserved ejection fraction. RNA-seq results demonstrated changes in left ventricular gene expression associated with fatty acid and branched chain amino acid metabolism, cardiomyopathy, cardiac hypertrophy, and heart failure. Twelve weeks of growth differentiation factor 15 (GDF15) treatment significantly decreased body weight, food intake, blood glucose, and triglycerides and improved exercise capacity in obese ZSF1 males. Systemic cardiovascular injury markers were significantly lower in GDF15-treated obese ZSF1 rats. Obese ZSF1 male rats represent a preclinical model for human cardiometabolic syndrome with established heart failure with preserved ejection fraction. GDF15 treatment mediated dietary response and demonstrated a cardioprotective effect in obese ZSF1 rats.
Collapse
|
36
|
Hitscherich PG, Xie LH, Del Re D, Lee EJ. The effects of macrophages on cardiomyocyte calcium-handling function using in vitro culture models. Physiol Rep 2020; 7:e14137. [PMID: 31301118 PMCID: PMC6640591 DOI: 10.14814/phy2.14137] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 04/18/2019] [Accepted: 04/23/2019] [Indexed: 12/11/2022] Open
Abstract
Following myocardial infarction (MI), myocardial inflammation plays a crucial role in the pathogenesis of MI injury and macrophages are among the key cells activated during the initial phases of the host response regulating the healing process. While macrophages have emerged as attractive effectors in tissue injury and repair, the contribution of macrophages on cardiac cell function and survival is not fully understood due to complexity of the in vivo inflammatory microenvironment. Understanding the key cells involved and how they communicate with one another is of paramount importance for the development of effective clinical treatments. Here, novel in vitro myocardial inflammation models were developed to examine how both direct and indirect interactions with polarized macrophage subsets present in the post-MI microenvironment affect cardiomyocyte function. The indirect model using conditioned medium from polarized macrophage subsets allowed examination of the effects of macrophage-derived factors on stem cell-derived cardiomyocyte function for up to 3 days. The results from the indirect model demonstrated that pro-inflammatory macrophage-derived factors led to a significant downregulation of cardiac troponin T (cTnT) and sarcoplasmic/endoplasmic reticulum calcium ATPase (Serca2) gene expression. It also demonstrated that inhibition of macrophage-secreted matricellular protein, osteopontin (OPN), led to a significant decrease in cardiomyocyte store-operated calcium entry (SOCE). In the direct model, stem cell-derived cardiomyocytes were co-cultured with polarized macrophage subsets for up to 3 days. It was demonstrated that anti-inflammatory macrophages significantly increased cardiomyocyte Ca2+ fractional release while macrophages independent of their subtypes led to significant downregulation of SOCE response in cardiomyocytes. This study describes simplified and controlled in vitro myocardial inflammation models, which allow examination of potential beneficial and deleterious effects of macrophages on cardiomyocytes and vise versa. This can lead to our improved understanding of the inflammatory microenvironment post-MI, otherwise difficult to directly investigate in vivo or by using currently available in vitro models.
Collapse
Affiliation(s)
- Pamela G Hitscherich
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey
| | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Dominic Del Re
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Eun Jung Lee
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey
| |
Collapse
|
37
|
Suthahar N, Meems LMG, Ho JE, de Boer RA. Sex-related differences in contemporary biomarkers for heart failure: a review. Eur J Heart Fail 2020; 22:775-788. [PMID: 32220046 PMCID: PMC7319414 DOI: 10.1002/ejhf.1771] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 01/28/2020] [Accepted: 01/28/2020] [Indexed: 12/28/2022] Open
Abstract
The use of circulating biomarkers for heart failure (HF) is engrained in contemporary cardiovascular practice and provides objective information about various pathophysiological pathways associated with HF syndrome. However, biomarker profiles differ considerably among women and men. For instance, in the general population, markers of cardiac stretch (natriuretic peptides) and fibrosis (galectin‐3) are higher in women, whereas markers of cardiac injury (cardiac troponins) and inflammation (sST2) are higher in men. Such differences may reflect sex‐specific pathogenic processes associated with HF risk, but may also arise as a result of differences in sex hormone profiles and fat distribution. From a clinical perspective, sex‐related differences in biomarker levels may affect the objectivity of biomarkers in HF management because what is considered to be ‘normal’ in one sex may not be so in the other. The objectives of this review are, therefore: (i) to examine the sex‐specific dynamics of clinically relevant HF biomarkers in the general population, as well as in HF patients; (ii) to discuss the overlap between sex‐related and obesity‐related effects, and (iii) to identify knowledge gaps to stimulate research on sex‐related differences in
HF.
Collapse
Affiliation(s)
- Navin Suthahar
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, The Netherlands
| | - Laura M G Meems
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, The Netherlands
| | - Jennifer E Ho
- Division of Cardiology, Department of Medicine, and Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rudolf A de Boer
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, The Netherlands
| |
Collapse
|
38
|
Chemokines profile in patients with chronic heart failure treated with cardiac resynchronization therapy. Adv Med Sci 2020; 65:102-110. [PMID: 31923769 DOI: 10.1016/j.advms.2019.11.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 07/18/2019] [Accepted: 11/25/2019] [Indexed: 12/20/2022]
Abstract
PURPOSE Inflammatory mechanisms have been suggested to play a role in the heart failure with reduced ejection fraction (HF-REF) development, but the role of chemokines is largely unknown. Cardiac resynchronization therapy (CRT) may reverse the HF-REF course. We aimed to evaluate selected chemokines concentrations in HF-REF patients and their relationship with disease severity and clinical response to CRT. MATERIALS AND METHODS The study included 37 patients (64.1 ± 11.04 years, 6 females) with HF-REF subjected to CRT, controlled prior to implantation and after 6 months. The control population included 26 healthy volunteers (63.9 ± 8.1 years, 8 females). Serum chemokines concentrations were determined using multiplex method. RESULTS HF-REF patients were characterized by the higher baseline MIF, NAP-2 and PF4 concentrations and lower Axl, BTC, IL-9, and IL-18 BPa concentrations comparing to controls. After 6 months of CRT only NAP-2 concentration decreased significantly in comparison to the baseline values. CONCLUSIONS HF-REF patients present altered chemokines profile compared to the control group. The CRT-related alleviation of HF-REF causes only slight changes in the chemokines concentrations especially in the platelet-associated ones. The precise chemokines role in the HF-REF pathogenesis and their prognostic value remains to be established.
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW To highlight the advances in the diagnosis and management of acute heart failure (AHF) from recent clinical trials and observational studies. RECENT FINDINGS We highlight date from recent studies examining the use of novel biomarkers (micro RNAs, osteopontin, insulin-like growth-factor binding protein-7) for the diagnosis of AHF. In addition, we summarize the evidence for novel pharmacological therapies including: serelaxin, ularitide, clevidipine, urocortins, BMS-986231, TRB027, vericiguat, omecamtiv, and torsemide. Finally, we explore the evidence behind the use of novel management algorithms. SUMMARY Despite many advances in understanding the pathophysiology of AHF, there is currently insufficient evidence to suggest changes to standards of care.
Collapse
|
40
|
Kwee LC, Neely ML, Grass E, Gregory SG, Roe MT, Ohman EM, Fox KAA, White HD, Armstrong PW, Bowsman LM, Haas JV, Duffin KL, Chan MY, Shah SH. Associations of osteopontin and NT-proBNP with circulating miRNA levels in acute coronary syndrome. Physiol Genomics 2019; 51:506-515. [PMID: 31530226 PMCID: PMC7054637 DOI: 10.1152/physiolgenomics.00033.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The genomic regulatory networks underlying the pathogenesis of non-ST-segment elevation acute coronary syndrome (NSTE-ACS) are incompletely understood. As intermediate traits, protein biomarkers report on underlying disease severity and prognosis in NSTE-ACS. We hypothesized that integration of dense microRNA (miRNA) profiling with biomarker measurements would highlight potential regulatory pathways that underlie the relationships between prognostic biomarkers, miRNAs, and cardiovascular phenotypes. We performed miRNA sequencing using whole blood from 186 patients from the TRILOGY-ACS trial. Seven circulating prognostic biomarkers were measured: NH2-terminal pro-B-type natriuretic peptide (NT-proBNP), high-sensitivity C-reactive protein, osteopontin (OPN), myeloperoxidase, growth differentiation factor 15, monocyte chemoattractant protein, and neopterin. We tested miRNAs for association with each biomarker with generalized linear models and controlled the false discovery rate at 0.05. Ten miRNAs, including known cardiac-related miRNAs 25-3p and 423-3p, were associated with NT-proBNP levels (min. P = 7.5 × 10−4) and 48 miRNAs, including cardiac-related miRNAs 378a-3p, 20b-5p and 320a, -b, and -d, were associated with OPN levels (min. P = 1.6 × 10−6). NT-proBNP and OPN were also associated with time to cardiovascular death, myocardial infarction (MI), or stroke in the sample. By integrating large-scale miRNA profiling with circulating biomarkers as intermediate traits, we identified associations of known cardiac-related and novel miRNAs with two prognostic biomarkers and identified potential genomic networks regulating these biomarkers. These results, highlighting plausible biological pathways connecting miRNAs with biomarkers and outcomes, may inform future studies seeking to delineate genomic pathways underlying NSTE-ACS outcomes.
Collapse
Affiliation(s)
| | - Megan L Neely
- Duke Clinical Research Institute, Durham, North Carolina
| | | | - Simon G Gregory
- Duke Molecular Physiology Institute, Durham, North Carolina.,Department of Neurology, Duke University School of Medicine, Durham, North Carolina
| | - Matthew T Roe
- Duke Clinical Research Institute, Durham, North Carolina.,Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - E Magnus Ohman
- Duke Clinical Research Institute, Durham, North Carolina.,Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Keith A A Fox
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Harvey D White
- Green Lane Cardiovascular Service, Auckland City Hospital, Auckland, New Zealand
| | - Paul W Armstrong
- Canadian VIGOUR Centre and Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Lenden M Bowsman
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Joseph V Haas
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Kevin L Duffin
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Mark Y Chan
- Division of Cardiology, Department of Medicine, National University of Singapore, Singapore
| | - Svati H Shah
- Duke Molecular Physiology Institute, Durham, North Carolina.,Duke Clinical Research Institute, Durham, North Carolina.,Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
41
|
Yang Y, Wang Y, Gao PJ. Osteopontin associated with left ventricular hypertrophy and diastolic dysfunction in essential hypertension. J Hum Hypertens 2019; 34:388-396. [DOI: 10.1038/s41371-019-0246-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/27/2019] [Accepted: 07/05/2019] [Indexed: 12/16/2022]
|
42
|
Kim D, Lee GY, Choi JO, Kim K, Kim SJ, Ju ES, Jeon ES. Prognostic values of novel biomarkers in patients with AL amyloidosis. Sci Rep 2019; 9:12200. [PMID: 31434944 PMCID: PMC6704139 DOI: 10.1038/s41598-019-48513-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 08/01/2019] [Indexed: 11/17/2022] Open
Abstract
As cardiac involvement is the most important prognostic marker in light-chain amyloidosis (AL), revised Mayo staging for AL incorporated N-terminal pro-brain natriuretic peptide (NTproBNP) and troponin T (TnT). However, prognostic value of novel biomarkers, such as soluble suppression of tumorigenicity 2 (sST2), growth differentiation factor 15 (GDF15), or osteopontin (OPN) is unknown in AL amyloidosis. We aimed to investigate additive predictive effects of novel biomarkers for overall mortality rates of AL amyloidosis patients. Levels of sST2, GDF15, and OPN were quantified at diagnosis in a total of 73 AL amyloidosis patients at Samsung Medical Center from 2010 to 2016. The median follow-up duration of the censored cases was 18.0 (12.4–28.1) months. A total of 25 deaths occurred during the follow-up period. Two novel biomarkers, sST2 and GDF-15 showed satisfactory predictive performances for both one-year and overall survival from ROC analysis. Best cut-off values for predicting one-year mortality were selected. Elevated sST2 and GDF-15 levels showed significant incremental prognostic values in addition to NT-ProBNP and TnT for overall mortality. Patients were assigned 1 point for elevated sST2 or GDF-15. The mean values of NT-proBNP, TnT, mean LV wall thickness, and septal e′ velocity differed significantly according to the scores. Patients with higher scores showed significantly worse prognosis even in patients with advanced revised Mayo staging. Two novel biomarkers, sST2 and GDF-15, showed satisfactory prognostic value for overall survival of AL amyloidosis patients. Furthermore, sST2 and GDF-15 showed additive incremental values over conventional biomarkers and further discriminated prognosis of patients in advanced stages.
Collapse
Affiliation(s)
- Darae Kim
- Division of Cardiology, Department of Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ga Yeon Lee
- Division of Cardiology, Department of Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jin-Oh Choi
- Division of Cardiology, Department of Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kihyun Kim
- Divsion of Hematology and Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seok Jin Kim
- Divsion of Hematology and Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Eun-Seon Ju
- Division of Cardiology, Department of Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Eun-Seok Jeon
- Division of Cardiology, Department of Medicine, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
43
|
Plasma osteopontin levels, but not its myocardial expression, reflect heart failure severity in recently diagnosed dilated cardiomyopathy. Herz 2019; 45:105-110. [PMID: 31289910 DOI: 10.1007/s00059-019-4829-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 04/27/2019] [Accepted: 06/03/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Elevated levels of the extracellular matrix glycoprotein osteopontin (OPN) may be detected in both myocardium and plasma under various pathological conditions affecting the heart. Several studies demonstrated increased plasma OPN levels in patients with heart failure due to dilated cardiomyopathy (DCM), while other studies showed high OPN expression levels in the myocardium of such patients. However, very little is known about OPN levels in both plasma and myocardium of the same individual with DCM. Therefore, we aimed to compare plasma OPN levels and levels of myocardial OPN expression in patients with recent-onset DCM (Ro-DCM). METHODS We examined plasma OPN as well as creatinine, C‑reactive protein (CRP), brain natriuretic peptide (BNP), and troponin I levels in 25 patients with Ro-DCM. Furthermore, all subjects underwent transthoracic echocardiography, selective coronary angiography, and endomyocardial biopsy (EMB) for the assessment of myocardial OPN expression. RESULTS No significant correlation between myocardial OPN expression and clinical, biochemical, or echocardiographic parameters was found. In log transformation analysis, plasma OPN levels correlated significantly with BNP levels (r = 0.46, p = 0.031), with CRP levels (r = 0.52, p = 0.015), and with early diastolic mitral annular velocity (r = -0.57, p = 0.009). There was a borderline association between the plasma OPN log value and New York Heart Association class (p = 0.053). CONCLUSION Plasma OPN levels reflect heart failure severity in patients with Ro-DCM. Myocardial OPN expression is not associated with either plasma OPN levels or markers of heart failure in these individuals.
Collapse
|
44
|
Yan X, Wang J, Zhu Y, Feng W, Zhai C, Liu L, Shi W, Wang Q, Zhang Q, Chai L, Li M. S1P induces pulmonary artery smooth muscle cell proliferation by activating calcineurin/NFAT/OPN signaling pathway. Biochem Biophys Res Commun 2019; 516:921-927. [PMID: 31277946 DOI: 10.1016/j.bbrc.2019.06.160] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 06/28/2019] [Indexed: 02/07/2023]
Abstract
The upregulation of osteopontin(OPN) has been found to contribute to the proliferation of pulmonary artery smooth muscle cells(PASMCs), and activation of PPARγ has been shown to suppress OPN expression in THP-1 cells. However, the molecular mechanisms underlying the upregulation of OPN expression and PPARγ agonist modulation of OPN expression in PASMCs remain largely unclear. Here we found that S1P stimulated PASMCs proliferation and up-regulated OPN expression in rat PASMCs, which was accompanied with the activation of phospholipase C(PLC), calcineurin and translocation of NFATc3 to nucleus. Further study showed that inhibition of PLC by U73122, suppression of calcineurin activity by cyclosporine A(CsA) or knockdown of NFATc3 using small interfering RNA suppressed S1P-induced OPN up-regulation. Activation of PPARγ by pioglitazone suppressed S1P-induced activation of calcineurin/NFATc3 signaling pathway and followed OPN up-regulation. Taken together, our study indicates that S1P stimulates OPN expression by activation of PLC/calcineurin/NFATc3 signaling pathway, and activation of PPARγ suppresses calcineurin/NFATc3-mediated OPN expression in PASMCs.
Collapse
Affiliation(s)
- Xin Yan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yanting Zhu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Wei Feng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Cui Zhai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Lu Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Wenhua Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Qingting Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Limin Chai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
45
|
Coculescu BI, Manole G, Dincă GV, Coculescu EC, Berteanu C, Stocheci CM. Osteopontin - a biomarker of disease, but also of stage stratification of the functional myocardial contractile deficit by chronic ischaemic heart disease. J Enzyme Inhib Med Chem 2019; 34:783-788. [PMID: 30843743 PMCID: PMC6407584 DOI: 10.1080/14756366.2019.1587418] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
The study analyses the significance of the plasmatic values of the OPN dosed to 91 people suffering from diastolic cardiac dysfunction with preserved ejection fraction, thus revealing significant growths of its level compared to the normal value. Despite being a clinical research, its conclusions are a breakthrough, differing from the results of other studies published in the relevant medical literature. We can make this assertion because this study analyses the clinical information given by the circulating values of the OPN, based on experimental models (animals), or on patients with congestive heart failure, which can be identified with the existence of a low systolic flow. The results of our study allow us to assert that the plasmatic values of this glycoprotein lead to its acceptance in the medical practice as a new biomarker that provides indicators regarding the stratification of risk with the patients suffering from heart failure of the diastolic dysfunction type, but whose systolic flow is preserved.
Collapse
Affiliation(s)
- Bogdan-Ioan Coculescu
- a Faculty of Medicine , Titu Maiorescu University , Bucharest , Romania.,b Center for Military Medical Scientific Research , Bucharest , Romania.,c Faculty of General Nursing , Bioterra University , Bucharest , Romania
| | - Gheorghe Manole
- c Faculty of General Nursing , Bioterra University , Bucharest , Romania.,d Clinical Hospital Colentina , Bucharest , Romania
| | | | - Elena Claudia Coculescu
- e Faculty of Dental Medicine , Carol Davila University of Medicine and Pharmacy , Bucharest , Romania
| | | | | |
Collapse
|
46
|
Predictors of cardiac arrhythmic events in non coronary artery disease patients. BMC Cardiovasc Disord 2019; 19:104. [PMID: 31046686 PMCID: PMC6498690 DOI: 10.1186/s12872-019-1083-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 04/16/2019] [Indexed: 11/29/2022] Open
Abstract
Arrhythmic sudden cardiac death (SCD) represents a major worldwide public health problem accounting for 15–20% of deaths. Risk stratification to identify patients at risk of SCD is crucial in order to implement preventive measures in the general population. Several biomarkers have been tested exploring different pathophysiological mechanisms of cardiac conditions. Conflicting results have been described limiting so far their use in clinical practice. The use of new biomarkers such as microRNAs and sex hormones and the emerging role of genetic on risk prediction of SCD is a current research topic showing promising results. This review outlines the role of plasma biomarkers to predict ventricular arrhythmias and SCD in non coronary artery disease with a special focus on their relationship with the genetic biomarkers.
Collapse
|
47
|
Luedde M, Spehlmann ME, Frey N. Progress in heart failure treatment in Germany. Clin Res Cardiol 2018; 107:105-113. [DOI: 10.1007/s00392-018-1317-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 06/27/2018] [Indexed: 12/29/2022]
|
48
|
Luedde M, Roy S, Hippe HJ, Cardenas DV, Spehlmann M, Vucur M, Hoening P, Loosen S, Frey N, Trautwein C, Luedde T, Koch A, Tacke F, Roderburg C. Elevated serum levels of bone sialoprotein during ICU treatment predict long-term mortality in critically ill patients. Sci Rep 2018; 8:9750. [PMID: 29950701 PMCID: PMC6021423 DOI: 10.1038/s41598-018-28201-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 06/19/2018] [Indexed: 12/19/2022] Open
Abstract
Bone sialoprotein (BSP), a member of the SIBLINGs (for Small Integrin-Binding LIgand, N-linked Glycoproteins) family, has recently be associated to inflammatory and infectious diseases. We therefore measured BSP concentrations in 136 patients at admission to the intensive care unit (ICU) and 3 days of ICU. BSP levels were compared to 36 healthy blood donors and correlated to clinical data. In these analysis, BSP serum levels were strongly elevated at the time point of admission to the ICU when compared to healthy controls. Moreover BSP concentrations were significantly elevated after 3 days of treatment on the intensive care unit. A further increase in BSP levels was detected in patients with higher APACHE-II-scores and in patients with septic disease. While in most patients, BSP levels decreased during the first three days of treatment on a medical ICU, patients with persistently elevated BSP levels displayed an unfavorable outcome. In these patients, persistently elevated BSP concentrations were a superior predictor of mortality than established indicators of patient´ prognosis such as the SAPS2 or the APACHE-II score. In summary, our data argue for a novel utility for BSP as a biomarker in patients treated on a medical ICU.
Collapse
Affiliation(s)
- Mark Luedde
- Department of Internal Medicine III, University Hospital of Schleswig Holstein, Campus Kiel, Rosalind-Franklin-Str. 12, 24105, Kiel, Germany
| | - Sanchari Roy
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Hans-Joerg Hippe
- Department of Internal Medicine III, University Hospital of Schleswig Holstein, Campus Kiel, Rosalind-Franklin-Str. 12, 24105, Kiel, Germany
| | - David Vargas Cardenas
- Department of Internal Medicine III, University Hospital of Schleswig Holstein, Campus Kiel, Rosalind-Franklin-Str. 12, 24105, Kiel, Germany
| | - Martina Spehlmann
- Department of Internal Medicine III, University Hospital of Schleswig Holstein, Campus Kiel, Rosalind-Franklin-Str. 12, 24105, Kiel, Germany
| | - Mihael Vucur
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Pia Hoening
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Sven Loosen
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Norbert Frey
- Department of Internal Medicine III, University Hospital of Schleswig Holstein, Campus Kiel, Rosalind-Franklin-Str. 12, 24105, Kiel, Germany
| | - Christian Trautwein
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Tom Luedde
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Alexander Koch
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Frank Tacke
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Christoph Roderburg
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
| |
Collapse
|
49
|
Zhang L, Xu Z, Wu Y, Liao J, Zeng F, Shi L. Akt/eNOS and MAPK signaling pathways mediated the phenotypic switching of thoracic aorta vascular smooth muscle cells in aging/hypertensive rats. Physiol Res 2018; 67:543-553. [PMID: 29750880 DOI: 10.33549/physiolres.933779] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Considerable evidence demonstrates that phenotypic switching of vascular smooth muscle cells (VSMCs) is influenced by aging and hypertension. During phenotypic switching, VSMCs undergo a switch to a proliferative and migratory phenotype, with this switch being a common pathology in cardiovascular diseases. The aim of this study was to explore the joint influence of age and hypertension on thoracic aortic smooth muscle phenotypic switching and the balance of Akt and mitogen-activated protein kinase (MAPK) signaling during this switch. Different ages of spontaneously hypertensive rats (SHR) and Wistar-Kyoto rats (WKY) were used to establish hypertension and aging models. The phenotypic state was determined by detecting the marker proteins alpha-SM-actin, calponin, and osteopontin (OPN) via immunohistochemical staining and Western blot. Signaling proteins associated with the Akt and MAPK pathways were detected in rat thoracic aorta using Western blot. Both aging and hypertension caused a decrease in contractile (differentiated) phenotype markers (alpha-SM-actin and calponin), while the synthetic (proliferative or de-differentiated) phenotype maker was elevated (OPN). When combining hypertension and aging, this effect was enhanced, with Akt signaling decreased, while MAPK signaling was increased. These results suggested that VSMCs phenotype switching is modulated by a balance between Akt and MAPK signaling in the process of aging and hypertension.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Exercise Physiology, Beijing Sport University, Beijing, P. R. China.
| | | | | | | | | | | |
Collapse
|
50
|
Zwakenberg SR, van der Schouw YT, Schalkwijk CG, Spijkerman AMW, Beulens JWJ. Bone markers and cardiovascular risk in type 2 diabetes patients. Cardiovasc Diabetol 2018; 17:45. [PMID: 29571288 PMCID: PMC5866514 DOI: 10.1186/s12933-018-0691-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 03/19/2018] [Indexed: 12/11/2022] Open
Abstract
Background Vascular calcifications are associated with a three- to fourfold increased risk of cardiovascular disease (CVD) and are highly prevalent in type 2 diabetes patients. Emerging evidence indicates that vascular calcification is a process of active bone formation regulated by stimulators and inhibitors of calcification. Therefore, this study aimed to evaluate whether six bone markers are associated with CVD risk in patients with type 2 diabetes. Methods We used data of a case-cohort study, nested in the EPIC-NL cohort, comprising 134 CVD cases and a random subcohort of 218 participants, all with type 2 diabetes at baseline. Six bone markers (osteocalcin, osteopontin, osteonectin, osteoprotegerin, alkaline phosphatase and sclerostin) were measured in baseline plasma samples with multiplex assays and information on CVD events was obtained. The association of bone makers with CVD risk was evaluated using Cox proportional hazard analyses. Results Higher concentrations of plasma osteopontin were associated (ptrend < 0.01) with an increased CVD risk with a hazard ratio of 2.00 (95%-CI 1.20–3.35) for the highest versus the lowest quartile in a multivariable adjusted model. The other bone markers were not associated with CVD risk. Conclusions Higher osteopontin concentrations were associated with an increased CVD risk in type 2 diabetes patients. No consistent associations were found for the other five bone markers and risk of CVD in type 2 diabetes patients. Electronic supplementary material The online version of this article (10.1186/s12933-018-0691-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sabine R Zwakenberg
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, PO box 85500, 3508 GA, Utrecht, The Netherlands.
| | - Yvonne T van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, PO box 85500, 3508 GA, Utrecht, The Netherlands
| | - Casper G Schalkwijk
- Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands.,CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Annemieke M W Spijkerman
- Center for Nutrition, Prevention and Health Services, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Joline W J Beulens
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, PO box 85500, 3508 GA, Utrecht, The Netherlands.,Department of Epidemiology & Biostatistics, EMGO+ Institute for Health and Care Research, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|