51
|
Kuster N, Huet F, Dupuy AM, Akodad M, Battistella P, Agullo A, Leclercq F, Kalmanovich E, Meilhac A, Aguilhon S, Cristol JP, Roubille F. Multimarker approach including CRP, sST2 and GDF-15 for prognostic stratification in stable heart failure. ESC Heart Fail 2020; 7:2230-2239. [PMID: 32649062 PMCID: PMC7524044 DOI: 10.1002/ehf2.12680] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 02/11/2020] [Accepted: 02/19/2020] [Indexed: 12/16/2022] Open
Abstract
Aims Inflammation and cardiac remodelling are common and synergistic pathways in heart failure (HF). Emerging biomarkers such as soluble suppression of tumorigenicity 2 (sST2) and growth differentiation factor‐15 (GDF‐15), which are linked to inflammation and fibrosis process, have been proposed as prognosis factors. However, their potential additive values remain poorly investigated. Methods and results Here, we aimed at evaluating inflammatory and remodelling biomarkers to predict both short‐term and long‐term mortality in a population with chronic HF in comparison with other classical clinical or biological markers (i.e. N terminal pro brain natriuretic peptide, hs‐cTnT, C‐reactive protein) alone or using meta‐analysis global group in chronic HF risk score in a cohort of 182 patients followed during 80 months (interquartile range: 12.3–90.0). Proportional hazard assumption does not hold for sST2 and C‐reactive protein, and follow‐up was split into short term (less than 1 year), midterm (between 1 and 5 years), and long term (after 5 years). In univariate analysis, C‐reactive protein and sST2 were predictive of short‐term mortality but not of middle term and long term whereas GDF‐15 was predictive of short and mid‐term but not of long‐term mortality. In a multivariate model after adjustment for meta‐analysis global group in chronic HF score including the three markers, only sST2 was predictive of short‐term mortality (P = 0.0225), and only GDF‐15 was predictive of middle term mortality (P = 0.0375). None of the markers was predictive of long‐term mortality. Conclusions Our results demonstrate that both sST2 and GDF‐15 significantly improve the prognosis evaluation of HF patients and suggest that the value of GDF‐15 is more sustained overtime and could predict middle term events.
Collapse
Affiliation(s)
- Nils Kuster
- Department of Biochemistry, Centre Ressources Biologiques de Montpellier, University Hospital of Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Fabien Huet
- Cardiology Department, University Hospital of Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Anne-Marie Dupuy
- Department of Biochemistry, Centre Ressources Biologiques de Montpellier, University Hospital of Montpellier, Montpellier, France
| | - Mariama Akodad
- Cardiology Department, University Hospital of Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Pascal Battistella
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Audrey Agullo
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Florence Leclercq
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Eran Kalmanovich
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Alexandra Meilhac
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Sylvain Aguilhon
- Cardiology Department, University Hospital of Montpellier, Montpellier, France
| | - Jean-Paul Cristol
- Department of Biochemistry, Centre Ressources Biologiques de Montpellier, University Hospital of Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Francois Roubille
- Cardiology Department, University Hospital of Montpellier, Montpellier, France.,PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| |
Collapse
|
52
|
Wiese S, Voiosu A, Hove JD, Danielsen KV, Voiosu T, Grønbaek H, Møller HJ, Genovese F, Reese-Petersen AL, Mookerjee RP, Clemmesen JO, Gøtze JP, Andersen O, Møller S, Bendtsen F. Fibrogenesis and inflammation contribute to the pathogenesis of cirrhotic cardiomyopathy. Aliment Pharmacol Ther 2020; 52:340-350. [PMID: 32524673 DOI: 10.1111/apt.15812] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/05/2020] [Accepted: 05/04/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Fibrogenesis and inflammation contribute to the progression of cirrhosis. However, it is unknown if these processes also contribute to the development of cirrhotic cardiomyopathy (CCM). Novel magnetic resonance imaging with quantification of the extracellular volume (ECV) provides an estimate of the fibrotic remodelling in the liver and heart. AIM To investigate the relationship between liver and cardiac ECV in cirrhosis and their association with collagen turnover and inflammation. METHODS A prospective study of 52 patients with cirrhosis and 14 healthy controls. All patients underwent contrast-enhanced MRI with T1-mapping and quantification of myocardial and liver ECV, biochemical assessments of collagen turnover (PRO-C3, PRO-C5, PRO-C6, collagen type IV degradation fragment, collagen type V degradation fragment, LG1M) and inflammation (TNFα, IL-1β, IL-6, IL-8, IL-18, SDF1α, sCD163, sMR, soluble macrophage mannose receptor). RESULTS Myocardial and liver ECV were increased in patients compared with healthy controls (myocardial ECV 31.2 ± 5.5% vs 27.4 ± 2.9%, P = 0.037; liver ECV 44.1 ± 9.6% vs 33.7 ± 6.7%, P < 0.001). Myocardial ECV correlated strongly with liver ECV (r = 0.48, P = 0.001) and biomarkers of collagen formation and inflammation (P < 0.005). Similarly, liver ECV correlated with biomarkers of collagen formation and inflammation (P < 0.003). In a multivariate analysis, liver ECV was predicted by biomarkers of collagen formation (PRO-C3 and PRO-C6), whereas myocardial ECV was predicted by biomarkers of collagen formation (PRO-C6) and inflammation (IL-6 and sMR). CONCLUSION Structural myocardial changes seem closely related to liver fibrosis in patients with cirrhosis. The strong associations with biomarkers of collagen formation and inflammation provide new insight into the role of inflammation and fibrogenesis in the development of structural cardiac abnormalities, potentially leading to CCM.
Collapse
|
53
|
Kaempferol Prevents Against Ang II-induced Cardiac Remodeling Through Attenuating Ang II-induced Inflammation and Oxidative Stress. J Cardiovasc Pharmacol 2020; 74:326-335. [PMID: 31356553 PMCID: PMC6791499 DOI: 10.1097/fjc.0000000000000713] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Heart failure characterized by cardiac remodeling is a global problem. Angiotensin II (Ang II) induces cardiac inflammation and oxidative stress, which also is implicated in the pathophysiology of adverse collagen accumulation-induced remodeling. Kaempferol (KPF), a kind of flavonoid compounds, is capable of anti-inflammatory and antioxidant activities. However, the target of KPF still remains blurred. In this study, we investigated the effect of KPF on Ang II-induced collagen accumulation and explored the underlying mechanisms. Our results suggested that KPF prevented Ang II-induced cardiac fibrosis and dysfunction, in mice challenged with subcutaneous injection of Ang II. In culture cells, KPF significantly reduced Ang II-induced collagen accumulation. Furthermore, KPF remarkably decreased inflammation and oxidative stress in Ang II-stimulated cardiac fibroblasts by modulating NF-κB/mitogen-activated protein kinase and AMPK/Nrf2 pathways.
Collapse
|
54
|
Lopaschuk GD, Verma S. Mechanisms of Cardiovascular Benefits of Sodium Glucose Co-Transporter 2 (SGLT2) Inhibitors: A State-of-the-Art Review. JACC Basic Transl Sci 2020; 5:632-644. [PMID: 32613148 PMCID: PMC7315190 DOI: 10.1016/j.jacbts.2020.02.004] [Citation(s) in RCA: 423] [Impact Index Per Article: 105.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/05/2020] [Accepted: 02/05/2020] [Indexed: 12/20/2022]
Abstract
Recent clinical trials have shown that sodium glucose co-transport 2 (SGLT2) inhibitors have dramatic beneficial cardiovascular outcomes. These include a reduced incidence of cardiovascular death and heart failure hospitalization in people with and without diabetes, and those with and without prevalent heart failure. The actual mechanism(s) responsible for these beneficial effects are not completely clear. Several potential theses have been proposed to explain the cardioprotective effects of SGLT2 inhibition, which include diuresis/natriuresis, blood pressure reduction, erythropoiesis, improved cardiac energy metabolism, inflammation reduction, inhibition of the sympathetic nervous system, prevention of adverse cardiac remodeling, prevention of ischemia/reperfusion injury, inhibition of the Na+/H+-exchanger, inhibition of SGLT1, reduction in hyperuricemia, increasing autophagy and lysosomal degradation, decreasing epicardial fat mass, increasing erythropoietin levels, increasing circulating pro-vascular progenitor cells, decreasing oxidative stress, and improving vascular function. The strengths and weaknesses of these proposed mechanisms are reviewed in an effort to try to synthesize and prioritize the mechanisms as they relate to clinical event reduction.
Collapse
Affiliation(s)
- Gary D. Lopaschuk
- Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Subodh Verma
- Division of Cardiac Surgery, Li Ka Shing Knowledge Institute of St. Michael’s Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
55
|
Wang X, Wang Q, Li W, Zhang Q, Jiang Y, Guo D, Sun X, Lu W, Li C, Wang Y. TFEB-NF-κB inflammatory signaling axis: a novel therapeutic pathway of Dihydrotanshinone I in doxorubicin-induced cardiotoxicity. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:93. [PMID: 32448281 PMCID: PMC7245789 DOI: 10.1186/s13046-020-01595-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Doxorubicin is effective in a variety of solid and hematological malignancies. Unfortunately, clinical application of doxorubicin is limited due to a cumulative dose-dependent cardiotoxicity. Dihydrotanshinone I (DHT) is a natural product from Salvia miltiorrhiza Bunge with multiple anti-tumor activity and anti-inflammation effects. However, its anti-doxorubicin-induced cardiotoxicity (DIC) effect, either in vivo or in vitro, has not been elucidated yet. This study aims to explore the anti-inflammation effects of DHT against DIC, and to elucidate the potential regulatory mechanism. METHODS Effects of DHT on DIC were assessed in zebrafish, C57BL/6 mice and H9C2 cardiomyocytes. Echocardiography, histological examination, flow cytometry, immunochemistry and immunofluorescence were utilized to evaluate cardio-protective effects and anti-inflammation effects. mTOR agonist and lentivirus vector carrying GFP-TFEB were applied to explore the regulatory signaling pathway. RESULTS DHT improved cardiac function via inhibiting the activation of M1 macrophages and the excessive release of pro-inflammatory cytokines both in vivo and in vitro. The activation and nuclear localization of NF-κB were suppressed by DHT, and the effect was abolished by mTOR agonist with concomitant reduced expression of nuclear TFEB. Furthermore, reduced expression of nuclear TFEB is accompanied by up-regulated phosphorylation of IKKα/β and NF-κB, while TFEB overexpression reversed these changes. Intriguingly, DHT could upregulate nuclear expression of TFEB and reduce expressions of p-IKKα/β and p-NF-κB. CONCLUSIONS Our results demonstrated that DHT can be applied as a novel cardioprotective compound in the anti-inflammation management of DIC via mTOR-TFEB-NF-κB signaling pathway. The current study implicates TFEB-IKK-NF-κB signaling axis as a previously undescribed, druggable pathway for DIC.
Collapse
Affiliation(s)
- Xiaoping Wang
- grid.24695.3c0000 0001 1431 9176School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Qiyan Wang
- grid.24695.3c0000 0001 1431 9176School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Weili Li
- grid.24695.3c0000 0001 1431 9176School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Qian Zhang
- grid.24695.3c0000 0001 1431 9176School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Yanyan Jiang
- grid.24695.3c0000 0001 1431 9176School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Dongqing Guo
- grid.24695.3c0000 0001 1431 9176School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Xiaoqian Sun
- grid.24695.3c0000 0001 1431 9176School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Wenji Lu
- grid.24695.3c0000 0001 1431 9176School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Chun Li
- grid.24695.3c0000 0001 1431 9176Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029 China
| | - Yong Wang
- grid.24695.3c0000 0001 1431 9176School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029 China ,grid.24695.3c0000 0001 1431 9176School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029 China
| |
Collapse
|
56
|
Gu Q, Wang B, Zhao H, Wang W, Wang P, Deng Y. LncRNA promoted inflammatory response in ischemic heart failure through regulation of miR-455-3p/TRAF6 axis. Inflamm Res 2020; 69:667-681. [PMID: 32350569 DOI: 10.1007/s00011-020-01348-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 02/14/2020] [Accepted: 04/08/2020] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES Ischemic heart failure (IHF) is the most common cause of death globally. Growing evidence shows abnormal expression of long non-coding RNAs in heart failure patients. This study aims to investigate the effect of sex-determining region Y-box 2 (SOX2) overlapping transcript (SOX2-OT) on the regulation of the inflammatory response in ischemic heart failure. METHODS IHF rat and oxygen and glucose deprivation (OGD) cell models were established. qRT-PCR was employed to investigate the expression of SOX2-OT. ELISA, western blot and cell viability/apoptosis assays were performed to assess the effects of SOX2-OT. Online software program was used to identify miRNAs that target SOX2-OT, followed by validation using RNA pull-down. Potential targets of miRNAs were searched, and examined by immunoblotting, qRT-PCR and luciferase reporter assay. RESULTS SOX2-OT was up-regulated in IHF and OGD. Knockdown of SOX2-OT promoted cell proliferation, decreased apoptosis rate and cell oxidative damage, and ameliorated inflammatory response. SOX2-OT contains binding sites for miR-455-3p, miR-5586-3p and miR-1252-5p. RNA pull-down confirmed the binding ability between SOX2-OT and miR-455-3p. TRAF6 is a direct target of miR-455-3p. Moreover, the regulatory activity of SOX2-OT on inflammatory response was partially through its negative regulation of miR-455-3p, which directly regulates TRAF6. Down-regulation of SOX2-OT improved myocardial dysfunction in IHF rat. CONCLUSIONS Our results reveal that SOX2-OT may be a driver of IHF through repression of miR-455-3p, and miR-455-3p alleviates IHF by targeting TRAF6. Therefore, SOX2-OT/miR-455-3p/TRAF6 may be a potential target for advanced therapeutic strategy for IHF.
Collapse
Affiliation(s)
- Qianqian Gu
- Department of Geriatrics, Cangzhou Central Hospital, 16 Xinhua West Road, Cangzhou, 061000, Hebei, China.
| | - Bin Wang
- Department of Otorhinolaryngology, Cangzhou Central Hospital, Cangzhou, 061000, Hebei, China
| | - Hongying Zhao
- Department of Geriatrics, Cangzhou Central Hospital, 16 Xinhua West Road, Cangzhou, 061000, Hebei, China
| | - Wenjuan Wang
- Department of Geriatrics, Cangzhou Central Hospital, 16 Xinhua West Road, Cangzhou, 061000, Hebei, China
| | - Pengsheng Wang
- Department of Geriatrics, Cangzhou Central Hospital, 16 Xinhua West Road, Cangzhou, 061000, Hebei, China
| | - Yu Deng
- Department of Geriatrics, Cangzhou Central Hospital, 16 Xinhua West Road, Cangzhou, 061000, Hebei, China
| |
Collapse
|
57
|
Mourouzis K, Oikonomou E, Siasos G, Tsalamadris S, Vogiatzi G, Antonopoulos A, Fountoulakis P, Goliopoulou A, Papaioannou S, Tousoulis D. Pro-inflammatory Cytokines in Acute Coronary Syndromes. Curr Pharm Des 2020; 26:4624-4647. [PMID: 32282296 DOI: 10.2174/1381612826666200413082353] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 04/01/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Over the last decades, the role of inflammation and immune system activation in the initiation and progression of coronary artery disease (CAD) has been established. OBJECTIVES The study aimed to present the interplay between cytokines and their actions preceding and shortly after ACS. METHODS We searched in a systemic manner the most relevant articles to the topic of inflammation, cytokines, vulnerable plaque and myocardial infarction in MEDLINE, COCHRANE and EMBASE databases. RESULTS Different classes of cytokines (intereleukin [IL]-1 family, Tumor necrosis factor-alpha (TNF-α) family, chemokines, adipokines, interferons) are implicated in the entire process leading to destabilization of the atherosclerotic plaque, and consequently, to the incidence of myocardial infarction. Especially IL-1 and TNF-α family are involved in inflammatory cell accumulation, vulnerable plaque formation, platelet aggregation, cardiomyocyte apoptosis and adverse remodeling following the myocardial infarction. Several cytokines such as IL-6, adiponectin, interferon-γ, appear with significant prognostic value in ACS patients. Thus, research interest focuses on the modulation of inflammation in ACS to improve clinical outcomes. CONCLUSION Understanding the unique characteristics that accompany each cytokine-cytokine receptor interaction could illuminate the signaling pathways involved in plaque destabilization and indicate future treatment strategies to improve cardiovascular prognosis in ACS patients.
Collapse
Affiliation(s)
- Konstantinos Mourouzis
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Evangelos Oikonomou
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Gerasimos Siasos
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Sotiris Tsalamadris
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Georgia Vogiatzi
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Alexios Antonopoulos
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Petros Fountoulakis
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Athina Goliopoulou
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Spyridon Papaioannou
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Dimitris Tousoulis
- 1st Department of Cardiology, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| |
Collapse
|
58
|
Abstract
Neutrophil-lymphocyte ratio has been associated with clinical outcomes in several groups of cardiac patients, including patients with coronary artery disease, cardiac failure, and cardiac transplant recipients. We hypothesised that pre- and/or post-operative haematological cell counts are associated with clinical outcomes in children undergoing cardiac surgery for CHD. We performed a post hoc analysis of data collected as part of a prospective observational cohort study (n = 83, data available n = 47) of children evaluated for glucocorticoid receptor levels after cardiac surgery (July 2015-January 2016). The association of neutrophil-lymphocyte ratio with low cardiac output syndrome, time to inotrope free, and vasoactive-inotropic score was examined using proportional odds analysis, cox regression, and linear regression models, respectively. A majority (80%) of patients were infants (median/interquartile range 4.1/0.2-7.6 months) with conotruncal (36%) and left-sided obstructed lesions (28%). Two patients required mechanical circulatory support and three died. Higher pre-operative neutrophil-lymphocyte ratio was associated with higher cumulative odds of severe/moderate versus mild low cardiac output on post-operative day 1 (odds ratio 2.86; 95% confidence interval 1.18-6.93; p = 0.02). Pre-operative neutrophil-lymphocyte ratio was not significantly associated with time to inotrope free or vasoactive-inotrope score. Post-operative neutrophil-lymphocyte ratio was also not associated with outcomes. In children after congenital heart surgery, higher pre-operative neutrophil-lymphocyte ratio was associated with a higher chance of low cardiac output in the early post-operative period. Pre-operative neutrophil-lymphocyte ratio maybe a useful prognostic marker in children undergoing congenital heart surgery.
Collapse
|
59
|
Ghafourian K, Shapiro JS, Goodman L, Ardehali H. Iron and Heart Failure: Diagnosis, Therapies, and Future Directions. JACC Basic Transl Sci 2020; 5:300-313. [PMID: 32215351 PMCID: PMC7091506 DOI: 10.1016/j.jacbts.2019.08.009] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/29/2019] [Accepted: 08/29/2019] [Indexed: 12/11/2022]
Abstract
To date, 3 clinical trials have shown symptomatic benefit from the use of intravenous (IV) iron in patients with heart failure (HF) with low serum iron. This has led to recommendations in support of the use of IV iron in this population. However, the systemic and cellular mechanisms of iron homeostasis in cardiomyocyte health and disease are distinct, complex, and poorly understood. Iron metabolism in HF appears dysregulated, but it is still unclear whether the changes are maladaptive and pathologic or compensatory and protective for the cardiomyocytes. The serum markers of iron deficiency in HF do not accurately reflect cellular and mitochondrial iron levels, and the current definition based on the ferritin and transferrin saturation values is broad and inclusive of patients who do not need IV iron. This is particularly relevant in view of the potential risks that are associated with the use of IV iron. Reliable markers of cellular iron status may differentiate subgroups of HF patients who would benefit from cellular and mitochondrial iron chelation rather than IV iron.
Collapse
Key Words
- 6MWT, 6-min walk test
- CKD, chronic kidney disease
- DMT1, divalent metal transporter 1 protein
- FCM, ferric carboxymaltose
- FGF, fibroblast growth factor
- Fpn1, ferroportin 1
- Hb, hemoglobin
- I/R, ischemia/reperfusion
- ID, iron deficiency
- IV, intravenous
- LVEF, left ventricular ejection fraction
- NTBI, non–transferrin-bound iron
- NYHA, New York Heart Association
- PGA, Patient Global Assessment
- RCT, randomized clinical trial
- ROS, reactive oxygen species
- TSAT, transferrin saturation
- TfR1, transferrin receptor protein 1
- VO2, peak oxygen uptake
- heart failure
- intravenous iron
- iron chelation
- iron deficiency
- sTfR, soluble transferrin receptor
Collapse
Affiliation(s)
| | | | | | - Hossein Ardehali
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, Illinois
| |
Collapse
|
60
|
Neutrophil-Lymphocyte Ratio in Patients with Acute Heart Failure Predicts In-Hospital and Long-Term Mortality. J Clin Med 2020; 9:jcm9020557. [PMID: 32085596 PMCID: PMC7073552 DOI: 10.3390/jcm9020557] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/11/2020] [Accepted: 02/13/2020] [Indexed: 01/18/2023] Open
Abstract
The application of a simple blood test to predict prognosis in acute heart failure (AHF) patients is not well established. Neutrophil-lymphocyte ratio (NLR) is inexpensive and easy to obtain in hospitalized patients using a routine blood test. We evaluate the prognostic implications of NLR as an independent predictor of in-hospital and long-term mortality in AHF patients. Among 5625 patients enrolled in the Korean Acute Heart Failure registry, 5580 patients were classified into quartiles by their NLR level, and analyzed for in-hospital and post-discharge three-year mortality. Patients in the highest NLR quartile had the highest in-hospital and post-discharge three-year mortality. The same results were seen by dividing the aggravating factor into the infection or ischemia group and the non-infection or non-ischemia group. For patients aggravated from infection or ischemia, a cut-off NLR value was 7.0 that increase the risk of in-hospital and post-discharge three-year mortality. In subgroups of patients not aggravated from infection or ischemia, a cut-off NLR value was 5.0 that increase the risk of in-hospital and post discharge three-year mortality. Elevated NLR in AHF patients at the index hospitalization is an independent predictor for in-hospital and post-discharge three-year mortality. Taken together, NLR is a marker for risk assessment of AHF patients.
Collapse
|
61
|
Chávez MN, Morales RA, López-Crisosto C, Roa JC, Allende ML, Lavandero S. Autophagy Activation in Zebrafish Heart Regeneration. Sci Rep 2020; 10:2191. [PMID: 32042056 PMCID: PMC7010704 DOI: 10.1038/s41598-020-59106-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 01/23/2020] [Indexed: 02/06/2023] Open
Abstract
Autophagy is an evolutionarily conserved process that plays a key role in the maintenance of overall cellular health. While it has been suggested that autophagy may elicit cardioprotective and pro-survival modulating functions, excessive activation of autophagy can also be detrimental. In this regard, the zebrafish is considered a hallmark model for vertebrate regeneration, since contrary to adult mammals, it is able to faithfully regenerate cardiac tissue. Interestingly, the role that autophagy may play in zebrafish heart regeneration has not been studied yet. In the present work, we hypothesize that, in the context of a well-established injury model of ventricular apex resection, autophagy plays a critical role during cardiac regeneration and its regulation can directly affect the zebrafish regenerative potential. We studied the autophagy events occurring upon injury using electron microscopy, in vivo tracking of autophagy markers, and protein analysis. Additionally, using pharmacological tools, we investigated how rapamycin, an inducer of autophagy, affects regeneration relevant processes. Our results show that a tightly regulated autophagic response is triggered upon injury and during the early stages of the regeneration process. Furthermore, treatment with rapamycin caused an impairment in the cardiac regeneration outcome. These findings are reminiscent of the pathophysiological description of an injured human heart and hence put forward the zebrafish as a model to study the poorly understood double-sword effect that autophagy has in cardiac homeostasis.
Collapse
Affiliation(s)
- Myra N Chávez
- Advanced Center for Chronic Diseases (ACCDiS) & Corporación Centro de Estudios Científicos de las Enfermedades Crónicas (CECEC), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Center for Genome Regulation (CGR), Department of Biology, Faculty of Sciences, University of Chile, Santiago, Chile
| | - Rodrigo A Morales
- Center for Genome Regulation (CGR), Department of Biology, Faculty of Sciences, University of Chile, Santiago, Chile
| | - Camila López-Crisosto
- Advanced Center for Chronic Diseases (ACCDiS) & Corporación Centro de Estudios Científicos de las Enfermedades Crónicas (CECEC), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Juan Carlos Roa
- Department of Pathology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Miguel L Allende
- Center for Genome Regulation (CGR), Department of Biology, Faculty of Sciences, University of Chile, Santiago, Chile.
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS) & Corporación Centro de Estudios Científicos de las Enfermedades Crónicas (CECEC), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile. .,Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Center, Dallas, USA.
| |
Collapse
|
62
|
Neutrophil to Lymphocyte Ratio Is Related to Thrombotic Complications and Survival in Continuous Flow Left Ventricular Assist Devices. ASAIO J 2020; 66:199-204. [DOI: 10.1097/mat.0000000000000971] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
63
|
Kirchhof P, Ezekowitz MD, Purmah Y, Schiffer S, Meng IL, Camm AJ, Hohnloser SH, Schulz A, Wosnitza M, Cappato R. Effects of Rivaroxaban on Biomarkers of Coagulation and Inflammation: A Post Hoc Analysis of the X-VeRT Trial. TH OPEN 2020; 4:e20-e32. [PMID: 31984306 PMCID: PMC6978177 DOI: 10.1055/s-0040-1701206] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/18/2019] [Indexed: 12/21/2022] Open
Abstract
Introduction
This X-VeRT (eXplore the efficacy and safety of once-daily oral riVaroxaban for the prevention of caRdiovascular events in patients with nonvalvular aTrial fibrillation scheduled for cardioversion) substudy evaluated the effects of treatment with rivaroxaban or a vitamin-K antagonist (VKA) on levels of biomarkers of coagulation (D-dimer, thrombin–antithrombin III complex [TAT] and prothrombin fragment [F1.2]) and inflammation (high sensitivity C-reactive protein [hs-CRP] and high-sensitivity interleukin-6 [hs-IL-6]) in patients with atrial fibrillation (AF) who were scheduled for cardioversion and had not received adequate anticoagulation at baseline (defined as, in the 21 days before randomization: no oral anticoagulant; international normalized ratio <2.0 with VKA treatment; or <80% compliance with non-VKA oral anticoagulant treatment).
Methods
Samples for biomarker analysis were taken at baseline (
n
= 958) and treatment completion (42 days after cardioversion;
n
= 918). The influence of clinical characteristics on baseline biomarker levels and the effect of treatment on changes in biomarker levels were evaluated using linear and logistic models.
Results
Baseline levels of some biomarkers were significantly associated with type of AF (D-dimer and hs-IL-6) and with history of congestive heart failure (hs-CRP, D-dimer, and hs-IL-6). Rivaroxaban and VKA treatments were associated with reductions from baseline in levels of D-dimer (−32.3 and −37.6%, respectively), TAT (−28.0 and −23.1%, respectively), hs-CRP (−12.5 and −17.9%, respectively), and hs-IL-6 (−9.2 and −9.8%, respectively). F1.2 levels were reduced from baseline in patients receiving a VKA (−53.0%) but not in those receiving rivaroxaban (2.7%).
Conclusion
Anticoagulation with rivaroxaban reduced levels of key inflammation and coagulation biomarkers to a similar extent as VKAs, with the exception of F1.2. Further investigation to confirm the value of these biomarkers in patients with AF is merited.
Collapse
Affiliation(s)
- Paulus Kirchhof
- Institute of Cardiovascular Sciences, University of Birmingham, Sandwell and West Birmingham Hospitals NHS Trust and University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom.,Department of Cardiology, University Heart Center Hamburg, Hamburg, Germany
| | - Michael D Ezekowitz
- Department of Cardiovascular Medicine, The Sidney Kimmel Medical College, Thomas Jefferson University, and the Lankenau Medical Center, Philadelphia, Pennsylvania, United States
| | - Yanish Purmah
- Institute of Cardiovascular Sciences, University of Birmingham, Sandwell and West Birmingham Hospitals NHS Trust and University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Sonja Schiffer
- Translational Medicine, Global R&D, Bayer AG, Wuppertal, Germany
| | - Isabelle L Meng
- Therapeutic Area Cardiovascular, Global Medical Affairs, Bayer AG, Berlin, Germany
| | - A John Camm
- Division of Clinical Sciences, St George's, University of London, London, United Kingdom
| | - Stefan H Hohnloser
- Division of Clinical Electrophysiology, Department of Cardiology, J. W. Goethe University, Frankfurt, Germany
| | - Anke Schulz
- Clinical Statistics EU, Global Clinical Development, Research and Clinical Sciences Statistics, Bayer AG, Berlin, Germany
| | - Melanie Wosnitza
- Therapeutic Area Cardiovascular, Global Medical Affairs, Bayer AG, Berlin, Germany
| | - Riccardo Cappato
- Arrhythmia and Electrophysiology Research Center, Humanitas Clinical and Research Center, Rozzano, Italy
| |
Collapse
|
64
|
Zhou P, Hua F, Wang X, Huang JL. Therapeutic potential of IKK-β inhibitors from natural phenolics for inflammation in cardiovascular diseases. Inflammopharmacology 2020; 28:19-37. [PMID: 31894515 DOI: 10.1007/s10787-019-00680-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/16/2019] [Indexed: 12/19/2022]
Abstract
Cardiovascular disease (CVDs) is a chronic disease with the highest morbidity and mortality in the world. Previous studies have suggested that preventing inflammation serves an efficient role in protection against cardiovascular diseases. Modulation of IKK-β activity can be used to treat and control CVDs associated with chronic inflammation, which targets the phosphorylation of IκB following the release of the RelA complex, and then translocates to the nucleus, eventually triggering the transcription of several genes that induce chemokines, cytokines, and adhesion molecules. Most importantly, the IκB kinase (IKK) complex is involved in transcriptional activation by phosphorylating the inhibitory molecule IkBα, enabling activation of NF-κB. Phenolic compounds possess cardioprotective potential that may be related to modulating inflammatory responses involved in CVDs. The SystemsDock analysis was used to explore whether 38 active compounds inhibit IKK-β activity based on literature. Docking results showed that the top docking score of three chemical compounds were icariin, salvianolic acid B, and plantainoside D in all compounds. Icariin, salvianolic acid B, and plantainoside D are the most promising IKKβ inhibitors. These phytochemicals could be helpful to find the lead compounds on designing and developing novel cardioprotective agents.
Collapse
Affiliation(s)
- Peng Zhou
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, People's Republic of China. .,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, People's Republic of China. .,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, People's Republic of China.
| | - Fang Hua
- Pharmacy School, Anhui Xinhua University, Hefei, 230088, People's Republic of China.,Natural Products Laboratory, International Joint Lab of Tea Chemistry and Health Effects, State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei, 230036, People's Republic of China
| | - Xiang Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, People's Republic of China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, People's Republic of China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, People's Republic of China
| | - Jin-Ling Huang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, People's Republic of China. .,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, People's Republic of China. .,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, People's Republic of China.
| |
Collapse
|
65
|
Wu Z, Zhao X, Miyamoto A, Zhao S, Liu C, Zheng W, Wang H. Effects of steroidal saponins extract from Ophiopogon japonicus root ameliorates doxorubicin-induced chronic heart failure by inhibiting oxidative stress and inflammatory response. PHARMACEUTICAL BIOLOGY 2019; 57:176-183. [PMID: 30860934 PMCID: PMC6419681 DOI: 10.1080/13880209.2019.1577467] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/08/2018] [Accepted: 01/28/2019] [Indexed: 05/25/2023]
Abstract
CONTEXT Ophiopogonis Radix, the root of Ophiopogon japonicus (Thunb.) Ker-Gawl (Liliaceae), is a Traditional Chinese Medicine, which has been investigated to possess effective treatment of cardiovascular diseases. OBJECTIVE This study evaluates the cardioprotective effects of steroidal saponins extract from Ophiopogon japonicus (SOJ) root against doxorubicin-induced chronic heart failure (CHF) through the amelioration of oxidative stress and inflammation. MATERIALS AND METHODS A Sprague-Dawley rat model of CHF was established by intraperitoneally injected with DOX. All rats were randomly divided into four groups: Control group, CHF group, CHF + SOJ (100 mg/kg) treatment group, SOJ (100 mg/kg) treatment group (n = 8/group). After six weeks administration, biometric and echocardiography were measured. The levels of biochemical parameters were measured using commercial kits. RESULTS The values of LVESP, +dP/dtmax, -dP/dtmax, EF and FS increased to 116.20 ± 1.68 mmHg, 2978.71 ± 168.26 mmHg/s, 3452.61 ± 286.09 mmHg/s, 68.26 ± 5.28% and 31.97 ± 3.79%, respectively; the values of LVEDP, LVESD and LVEDD decreased to 8.85 ± 0.84 mmHg, 8.39 ± 0.45 mm and 12.36 ± 0.87 mm in CHF + SOJ group. In addition, the levels of IL-6, TNF-α and IL-1β decreased to 154.41 ± 7.72 pg/mg protein, 110.02 ± 6.96 pg/mg protein and 39.39 ± 5.27 pg/mg protein, respectively; the relative activity of p38 MAPK decreased to 2.60 ± 0.40 in CHF + SOJ group. Furthermore, the activities of SOD, CAT and GSH-Px increased to 268.77 ± 6.20 U/mg protein, 13.68 ± 0.68 U/mg protein and 316.90 ± 8.08 µmol/mg protein, and the content of MDA decreased to 4.03 ± 0.43 nmol/mg protein in CHF + SOJ group. CONCLUSIONS SOJ exerts the cardioprotective effect against DOX-induced CHF through suppressing inflammatory and oxidative stress. These results provide evidence that SOJ might be an effective treatment for CHF.
Collapse
Affiliation(s)
- Zhongwei Wu
- Department of Cardiology, Hainan Western Central Hospital, Danzhou, China
| | - Xuekai Zhao
- Department of Cardiac Surgery, Zibo Central Hospital, Zibo, China
| | - Akira Miyamoto
- Department of Rehabilitation, Kobe International University, Kobe, Japan
| | - Shengji Zhao
- Department of Cardiology, Hainan Western Central Hospital, Danzhou, China
| | - Chaoquan Liu
- Department of Cardiology, Hainan Western Central Hospital, Danzhou, China
| | - Weimin Zheng
- Department of Cardiology, Hainan Western Central Hospital, Danzhou, China
| | - HongTao Wang
- Department of Cardiology, The Second Affiliated Hospital of Xi’an JiaoTong University, Xian, China
| |
Collapse
|
66
|
sST2 as a value-added biomarker in heart failure. Clin Chim Acta 2019; 501:120-130. [PMID: 31678574 DOI: 10.1016/j.cca.2019.10.029] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/22/2019] [Accepted: 10/22/2019] [Indexed: 01/09/2023]
Abstract
Soluble suppression of tumorigenicity-2 (sST2) is a biomarker widely investigated during the last few years. Its role has become clear in pathological conditions such as fibrosis and inflammation. From translational research to laboratory medicine, considerable efforts have been made to elucidate the features of sST2 biomarker and to consider its contribution to HF management. In this review, we summarized the results from recent works concerning sST2, and particularly we focused on the interest of sST2 in conditions for which classical biomarkers value interpretation is misleading. Indeed, despite other HF biomarkers, sST2 was proved to be independent from common comorbidities such as renal dysfunction and hypertension. Thus, sST2 showed promise for a combined strategy with natriuretic peptides, mainly for specific categories of patients. Particular attention was paid to findings on sST2 in HF with preserved ejection fraction (HFpEF), a form of HF for which reliable and specific biomarkers are awaited. Finally, a place is reserved to sST2 kinetics from basal to follow up values in order to improve clinical decision making and to customize patient treatments.
Collapse
|
67
|
Association between Atrial Fibrillation, Myocardial Infarction, Heart Failure and Mortality in Patients with Nontuberculous Mycobacterial Infection: a nationwide population-based study. Sci Rep 2019; 9:15503. [PMID: 31664094 PMCID: PMC6820717 DOI: 10.1038/s41598-019-51801-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 10/03/2019] [Indexed: 12/30/2022] Open
Abstract
NTM infection demonstrates an increasing incidence and prevalence. We studied the impact of NTM in cardiovascular events. Using the Korean nationwide database, we included newly diagnosed 1,730 NTM patients between 2005 and 2008 and followed up for new-onset atrial fibrillation (AF), myocardial infarction (MI), heart failure (HF), ischemic stroke (IS), and death. Covariates-matched non-NTM subjects (1:5, n = 8,650) were selected and analyzed. Also, NTM infection was classified into indolent or progressive NTM for risk stratification. During 4.16 ± 1.15 years of the follow-up period, AF, MI, HF, IS, and death were newly diagnosed in 87, 125, 121, 162, and 468 patients. In multivariate analysis, NTM group showed an increased risk of AF (hazard ratio [HR] 2.307, 95% confidence interval [CI] 1.560-3.412) and all-cause death (HR 1.751, 95% CI 1.412-2.172) compared to non-NTM subjects, whereas no significant difference in MI (HR 0.868, 95% CI 0.461-1.634), HF (HR 1.259, 95% CI 0.896-2.016), and IS (HR 1.429, 95% CI 0.981-2.080). After stratification, 1,730 NTM patients were stratified into 1,375 (79.5%) indolent NTM group and 355 (20.5%) progressive NTM group. Progressive NTM showed an increased risk of AF and mortality than indolent NTM group. Screening for AF and IS prevention would be appropriate in these high-risk patients.
Collapse
|
68
|
Turcato G, Sanchis-Gomar F, Cervellin G, Zorzi E, Sivero V, Salvagno GL, Tenci A, Lippi G. Evaluation of Neutrophil-lymphocyte and Platelet-lymphocyte Ratios as Predictors of 30-day Mortality in Patients Hospitalized for an Episode of Acute Decompensated Heart Failure. J Med Biochem 2019; 38:452-460. [PMID: 31496909 PMCID: PMC6708303 DOI: 10.2478/jomb-2018-0044] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/21/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND To investigate the association between both neutrophil to lymphocyte ratio (NLR) and platelet to lymphocyte ratio (PLR) and 30-day mortality in patients hospitalized for an episode of acute decompensated heart failure (ADHF). METHODS 439 patients admitted to emergency department (ED) for an episode of ADHF. Clinical history, demographic, clinical and laboratory data recorded at ED admission and then correlated with 30-day mortality. RESULTS 45/439 (10.3%) patients died within 30 days from ED admission. The median values of NLR (4.1 vs 11.7) and PLR (159.1 vs 285.9) were significantly lower in survivors than in patients who died. The area under the ROC curve of NLR was significantly higher than that of the neutrophil count (0.76 vs 0.59; p<0.001), whilst the AUC of PLR was significantly better than that of the platelet count (0.71 vs 0.51; p<0.001). In univariate analysis, both NLR and PLR were significantly associated with 30-day. In the fully-adjusted multivariate model, NLR (odds ratio, 3.63) and PLR (odds ratio, 3.22) remained independently associated with 30-day mortality after ED admission. CONCLUSIONS Routine assessment of NLR and PLR at ED admission may be a valuable aid to complement other conventional measures for assessing the medium-short risk of ADHF patients.
Collapse
Affiliation(s)
- Gianni Turcato
- Department of Emergency Medicine, G. Fracastoro Hospital of San Bonifacio, Azienda Ospedaliera Scaligera, San Bonifacio, Verona, Italy
| | - Fabian Sanchis-Gomar
- Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, USA
- Department of Physiology, Faculty of Medicine, University of Valencia and INCLIVA Biomedical Research Institute, Valencia, Spain
| | | | - Elisabetta Zorzi
- Department of Cardiology and Intensive Care Cardiology, G. Fracastoro Hospital of San Bonifacio, Azienda Ospedaliera Scaligera, San Bonifacio, Verona, Italy
| | - Valentina Sivero
- Department of Emergency Medicine, G. Fracastoro Hospital of San Bonifacio, Azienda Ospedaliera Scaligera, San Bonifacio, Verona, Italy
| | | | - Andrea Tenci
- Department of Emergency Medicine, G. Fracastoro Hospital of San Bonifacio, Azienda Ospedaliera Scaligera, San Bonifacio, Verona, Italy
| | - Giuseppe Lippi
- Section of Clinical Biochemistry, University of Verona, Verona, Italy
| |
Collapse
|
69
|
Yue LJ, Zhu XY, Li RS, Chang HJ, Gong B, Tian CC, Liu C, Xue YX, Zhou Q, Xu TS, Wang DJ. S‑allyl‑cysteine sulfoxide (alliin) alleviates myocardial infarction by modulating cardiomyocyte necroptosis and autophagy. Int J Mol Med 2019; 44:1943-1951. [PMID: 31573046 PMCID: PMC6777694 DOI: 10.3892/ijmm.2019.4351] [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: 02/20/2019] [Accepted: 09/02/2019] [Indexed: 12/14/2022] Open
Abstract
S-allyl-cysteine sulfoxide (alliin) is the main organosulfur component of garlic and its preparations. The present study aimed to examine the protective effect of alliin on cardiac function and the underlying mechanism in a mouse model of myocardial infarction (MI). Notably, alliin treatment preserved heart function, attenuated the area of infarction in the myocardium of mice and reduced lesions in the myocardium, including cardiomyocyte fibrosis and death. Further mechanistic experiments revealed that alliin inhibited necroptosis but promoted autophagy in vitro and in vivo. Cell viability assays showed that alliin dose-dependently reduced the necroptotic index and inhibited the expression of necroptosis-related receptor-interacting protein 1, receptor-interacting protein 3 and tumor necrosis factor receptor-associated factor 2, whereas the levels of Beclin 1 and microtubule-associated protein 1 light chain 3, which are associated with autophagy, exhibited an opposite trend upon treatment with alliin. In addition, the level of peroxisome proliferator-activated receptor γ was increased by alliin. Collectively, these findings demonstrate that alliin has the potential to protect cardiomyocytes from necroptosis following MI and that this protective effect occurs via the enhancement of autophagy.
Collapse
Affiliation(s)
- Li-Jun Yue
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Xi-Yu Zhu
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Rui-Sha Li
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Hui-Jing Chang
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Bing Gong
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Chong-Chong Tian
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Chang Liu
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Yun-Xing Xue
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Qing Zhou
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Tian-Shu Xu
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Dong-Jin Wang
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| |
Collapse
|
70
|
Bansal A, Uriel N, Colombo PC, Narisetty K, Long JW, Bhimaraj A, Cleveland JC, Goldstein DJ, Stulak JM, Najjar SS, Lanfear DE, Adler ED, Dembitsky WP, Somo SI, Crandall DL, Chen D, Connors JM, Mehra MR. Effects of a fully magnetically levitated centrifugal-flow or axial-flow left ventricular assist device on von Willebrand factor: A prospective multicenter clinical trial. J Heart Lung Transplant 2019; 38:806-816. [DOI: 10.1016/j.healun.2019.05.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 01/13/2023] Open
|
71
|
Liu ZJ, Liu H, Wu C, Xue K. Effect of sepsis on the action potential and cardiac serotonin response in rats. Exp Ther Med 2019; 18:2207-2212. [PMID: 31452710 DOI: 10.3892/etm.2019.7810] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/16/2019] [Indexed: 01/22/2023] Open
Abstract
The current study aimed to investigate the effect of sepsis on rat serotonin (5-HT) responses and cardiac action potentials. A total of 20 rats were randomly divided into a sepsis and control group (each, n=10). Rat hearts were harvested and perfused using the Langendorff method 18-h after the induction of sepsis, which was assessed using cecal puncture. Cardiac action potential was subsequently measured using a multichannel electrophysiology instrument. Immunohistochemistry and quantitative analysis were performed to identify the effect of sepsis on myocardial 5-HT expression. The results revealed that mitochondrial changes were present in septic rat hearts. Heart rate (361.10±12.29 bpm vs. 348.60±12.38 bpm; P<0.05) was significantly higher, atrial action potential duration (106.40±2.95 ms vs. 86.60±4.12 ms; P<0.01) was significantly longer and the area (0.62±0.06 µm2 vs. 0.39±0.05 µm2; P<0.05) and number (0.92±0.02/field vs. 0.46±0.01/field; P<0.01) of myocardial cells were significantly higher in the septic compared with the control group. These results demonstrated that 5-HT prolongs the atrial action potential, increases heart rate and aggravates myocardial injury, indicating that it may therefore be one of the factors that leads to cardiac dysfunction in sepsis.
Collapse
Affiliation(s)
- Zheng-Jiang Liu
- Department of Cardiology, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong 511500, P.R. China
| | - Hua Liu
- Department of Cardiology, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong 511500, P.R. China
| | - Changdong Wu
- Intensive Medicine Unit, The People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region 830001, P.R. China
| | - Kedong Xue
- Intensive Medicine Unit, The People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region 830001, P.R. China
| |
Collapse
|
72
|
Abstract
Advanced heart failure (HF) is a progressive disease characterized by recurrent hospitalizations and high risk of mortality. Indeed, outcomes in late stages of HF approximate those seen in patients with various aggressive malignancies. Clinical trials assessing beneficial outcomes of new treatments in patients with cancer have used innovative approaches to measure impact on total disease burden or surrogates to assess treatment efficacy. Although most cardiovascular outcomes trials continue to use time-to-first event analyses to assess the primary efficacy end point, such analyses do not adequately reflect the impact of new treatments on the totality of the chronic disease burden. Consequently, patient enrichment and other strategies for ongoing clinical trial design, as well as new statistical methodologies, are important considerations, particularly when studying a population with advanced chronic HF. The DREAM-HF trial (Double-Blind Randomized Assessment of Clinical Events With Allogeneic Mesenchymal Precursor Cells in Advanced Heart Failure) is an ongoing, randomized, sham-controlled phase 3 study of the efficacy and safety of mesenchymal precursor cells as immunotherapy in patients with advanced chronic HF with reduced ejection fraction. Mesenchymal precursor cells have a unique multimodal mechanism of action that is believed to result in polarization of proinflammatory type 1 macrophages in the heart to an anti-inflammatory type 2 macrophage state, inhibition of maladaptive adverse left ventricular remodeling, reversal of cardiac and peripheral endothelial dysfunction, and recovery of deranged vasculature. The objective of DREAM-HF is to confirm earlier phase 2 results and evaluate whether mesenchymal precursor cells will reduce the rate of nonfatal recurrent HF-related major adverse cardiac events while delaying or preventing progression of HF to terminal cardiac events. DREAM-HF is an example of an ongoing contemporary events-driven cardiovascular cell-based immunotherapy study that has utilized the concepts of baseline disease enrichment, prognostic enrichment, and predictive enrichment to improve its efficiency by using accumulating data from within as well as external to the trial. Adaptive enrichment designs and strategies are important components of a rational approach to achieve clinical research objectives in shorter clinical trial timelines and with increased cost-effectiveness without compromising ethical standards or the overall statistical integrity of the study. The DREAM-HF trial also presents an alternative approach to traditional composite time-to-first event primary efficacy end points. Statistical methodologies such as the joint frailty model provide opportunities to expand the scope of events-driven HF with reduced ejection fraction clinical trials to utilize time to recurrent nonfatal HF-related major adverse cardiac events as the primary efficacy end point without compromising the integrity of the statistical analyses for terminal cardiac events. In advanced chronic HF with reduced ejection fraction studies, the joint frailty model is utilized to reflect characteristics of the high-risk patient population with important unmet therapeutic needs. In some cases, use of the joint frailty model may substantially reduce sample size requirements. In addition, using an end point that is acceptable to the Food and Drug Administration and the European Medicines Agency, such as recurrent nonfatal HF-related major adverse cardiac events, enables generation of clinically relevant pharmacoeconomic data while providing comprehensive views of the patient's overall cardiovascular disease burden. The major goal of this review is to provide lessons learned from the ongoing DREAM-HF trial that relate to biologic plausibility and flexible clinical trial design and are potentially applicable to other development programs of innovative therapies for patients with advanced cardiovascular disease. Clinical Trial Registration: URL: https://www.clinicaltrials.gov. Unique identifier: NCT02032004.
Collapse
Affiliation(s)
| | | | - Barry Greenberg
- University of California, San Diego School of Medicine, La Jolla (B.G.)
- Advanced Heart Failure Treatment Program, Sulpizio Cardiovascular Center, University of California, San Diego Healthcare System, La Jolla (B.G.)
| | - Emerson C. Perin
- Stem Cell Center and Adult Cardiology, Texas Heart Institute, Houston (E.C.P.)
| |
Collapse
|
73
|
Lieben Louis X, Meikle Z, Chan L, DeGagne G, Cummer R, Meikle S, Krishnan S, Yu L, Netticadan T, Wigle JT. Divergent Effects of Resveratrol on Rat Cardiac Fibroblasts and Cardiomyocytes. Molecules 2019; 24:molecules24142604. [PMID: 31319579 PMCID: PMC6680709 DOI: 10.3390/molecules24142604] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/11/2019] [Accepted: 07/12/2019] [Indexed: 11/27/2022] Open
Abstract
In this study, we tested the potential cardioprotective effects of the phytoalexin resveratrol (Rsv) on primary adult rat cardiac fibroblasts (CF), myofibroblasts (MF) and cardiomyocytes. Adult rat CF and cardiomyocytes were isolated from male 10-week old Sprague–Dawley rats, cultured for either 24 h (cardiomyocytes) or 48 h (CF) before treatments. To isolate MF, CF were trypsinized after 48 h in culture, seeded in fresh plates and cultured for 24 h prior to treatment. All three cells were then treated for a further 24 h with a range of Rsv doses. In CF and MF, cell proliferation, viability, apoptosis assays were performed with or without Rsv treatment for 24 h. In cardiomyocytes, cell viability and apoptosis assay were performed 24 h after treatment. In separate experiments, CF was pre-incubated with estrogen, tamoxifen and fulvestrant for 30 min prior to Rsv treatment. Rsv treatment decreased proliferation of both fibroblasts and myofibroblasts. Rsv treatment also increased the proportion of dead CF and MF in a dose dependent manner. However, treatment with Rsv did not induce cell death in adult cardiomyocytes. There was an increase in the percentage of cells with condensed nuclei with Rsv treatment in both CF and MF, but not in cardiomyocytes. Treatment with estrogen, tamoxifen and fulvestrant alone or in combination with Rsv did not have any additional effects on CF survival. Our results demonstrate that treatment with Rsv can inhibit cell proliferation and induce cell death in rat CF and MF, while not affecting cardiomyocyte survival. We also demonstrated that the induction of cell death in CF with Rsv treatment was independent of estrogen receptor alpha (ERα) signaling.
Collapse
Affiliation(s)
- Xavier Lieben Louis
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Zach Meikle
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Laura Chan
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Garret DeGagne
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Rebecca Cummer
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
| | - Shannon Meikle
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
| | - Sampath Krishnan
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
| | - Liping Yu
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Agriculture and Agri-Food Canada, Winnipeg Winnipeg, MB R2H 2A6, Canada
| | - Thomas Netticadan
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada.
- Agriculture and Agri-Food Canada, Winnipeg Winnipeg, MB R2H 2A6, Canada.
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| | - Jeffrey T Wigle
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada.
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| |
Collapse
|
74
|
Iravani Saadi M, Babaee Beigi MA, Ghavipishe M, Tahamtan M, Geramizadeh B, Zare A, Yaghoobi R. The circulating level of interleukins 6 and 18 in ischemic and idiopathic dilated cardiomyopathy. J Cardiovasc Thorac Res 2019; 11:132-137. [PMID: 31384408 PMCID: PMC6669430 DOI: 10.15171/jcvtr.2019.23] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 06/19/2019] [Indexed: 01/09/2023] Open
Abstract
Introduction: By aging population, the heart failure and its life-threatening complications have become an enormous issue in public health. Regarding the inflammation as a major contributing pathological factor, the determination of most important inflammatory targets for immunomodulation is a problematic puzzle in the treatment of heart failure patients and the inflammatory pathways primarily involved in different underlying conditions contributing to heart failure can be an area which is worthy of focused research. Considering the dilated cardiomyopathy (DCM) as a relatively high-incident disease leading to heart failure, the aim of this study is to determine the difference in the expression level of interleukin (IL)-6 and IL-18 in patients with ischemic and idiopathic DCM. Methods: 39 non-diabetic patients with ischemic and 37 ones with idiopathic DCM were enrolled in the study. 48 healthy individuals were also considered as control group. For quantitative determination of the mRNA expression level of IL-6 and IL-18 genes, an in-house- SYBR Green real-time PCR was used and Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was considered as internal control gene. The left ventricular end-diastolic volume (LVEDV) and left ventricular ejection fraction (LVEF) was calculated by 2D echocardiographic assessment. Data were finally analyzed via SPSS statistical software version 19.0 using independent t test and 2-∆∆Ct method and P<0.05 were considered statistically significant. Results: The IL-6 was significantly higher expressed in patients with ischemic and idiopathic DCM than in healthy controls (274.3 and 168.8 times, respectively, both P values <0.001). The same higher expression of IL-18 was observed in ischemic DCM (48.5 times) and idiopathic DCM (45.2 times) compared with healthy individuals (both P values <0.001). Conclusion: Both ischemic and idiopathic DCM associates with IL-6 and IL-18 overexpression. However, no significant difference was observed between these two subtypes of DCM in either interleukin expression level. There is certainly need to further studies for evaluating the uniformity of results and also assessing other molecules in determining their roles in pathophysiology and probable utility for management.
Collapse
Affiliation(s)
- Mahdiyar Iravani Saadi
- Hematology Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Transplant Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Ali Babaee Beigi
- Cardiovascular Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Ghavipishe
- Cardiovascular Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Tahamtan
- Cardiovascular Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bita Geramizadeh
- Department of Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abdolhossein Zare
- Transplant Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ramin Yaghoobi
- Transplant Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
75
|
Abstract
Inflammatory processes underlie many diseases associated with injury of the heart muscle, including conditions without an obvious inflammatory pathogenic component such as hypertensive and diabetic cardiomyopathy. Persistence of cardiac inflammation can cause irreversible structural and functional deficits. Some are induced by direct damage of the heart muscle by cellular and soluble mediators but also by metabolic adaptations sustained by the inflammatory microenvironment. It is well established that both cardiomyocytes and immune cells undergo metabolic reprogramming in the site of inflammation, which allow them to deal with decreased availability of nutrients and oxygen. However, like in cancer, competition for nutrients and increased production of signalling metabolites such as lactate initiate a metabolic cross-talk between immune cells and cardiomyocytes which, we propose, might tip the balance between resolution of the inflammation versus adverse cardiac remodeling. Here we review our current understanding of the metabolic reprogramming of both heart tissue and immune cells during inflammation, and we discuss potential key mechanisms by which these metabolic responses intersect and influence each other and ultimately define the prognosis of the inflammatory process in the heart.
Collapse
Affiliation(s)
- Federica M Marelli-Berg
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom.,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Dunja Aksentijevic
- School of Biological and Chemical Sciences, Queen Mary University of London, G.E. Fogg Building, Mile End Road, London E1 4NS, United Kingdom.,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| |
Collapse
|
76
|
Lourenço P, Pereira J, Ribeiro A, Ferreira-Coimbra J, Barroso I, Guimarães JT, Leite-Moreira A, Bettencourt P. C-reactive protein decrease associates with mortality reduction only in heart failure with preserved ejection fraction. J Cardiovasc Med (Hagerstown) 2019; 20:23-29. [PMID: 30407267 DOI: 10.2459/jcm.0000000000000726] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
AIMS The prognostic role of high-sensitivity C-reactive protein (hsCRP) in acute heart failure is less well established than for chronic heart failure and the impact of its variation is unknown. We studied the impact of hsCRP variation in acute heart failure and whether it differed according to left ventricular function. METHODS We analyzed patients prospectively included in an acute heart failure registry. Admission and discharge hsCRP were evaluated as part of the registry's protocol and its relative variation (ΔhsCRP) was assessed. ΔhsCRP during hospitalization = [(admission hsCRP - discharge hsCRP)/admission hsCRP] × 100. Endpoint: all-cause death; follow-up: 3 years. A multivariate Cox-regression model was used to assess the prognostic value of ΔhsCRP (continuous and categorical variable: cut-off 40% decrease); analysis was stratified according to ventricular function. RESULTS We studied 439 patients: mean age 75 years, 50.1% men and 69.2% had heart failure with reduced ejection fraction (HFrEF). Median discharge hsCRP was 12.4 mg/l and median ΔhsCRP was ∼40%. During follow-up 247 patients (56.3%) died: 73 (54.1%) heart failure with preserved ejection fraction (HFpEF) patients and 174 (57.2%) HFrEF patients. The multivariate-adjusted hazard ratio of 3-year mortality in HFpEF patients with hsCRP decrease of at least 40% during hospitalization was 0.56 (95% CI 0.32-0.99). A decrease of at least 40% in hsCRP was not mortality-associated in HFrEF patients. There was interaction between ΔhsCRP and left ventricular ejection fraction. CONCLUSION A decrease of at least 40% in hsCRP in acute heart failure was associated with a 44% decrease in 3-year death risk in HFpEF patients. No association between ΔhsCRP and prognosis existed in HFrEF patients. Inflammation appears to play a different role according to left ventricular function.
Collapse
Affiliation(s)
| | | | | | | | - Isaac Barroso
- Serviço de Patologia Clínica, Centro Hospitalar São João
| | - João-Tiago Guimarães
- Serviço de Patologia Clínica, Centro Hospitalar São João.,Faculdade de Medicina da Universidade do Porto, Unidade I&D Cardiovascular do Porto
| | - Adelino Leite-Moreira
- Faculdade de Medicina da Universidade do Porto, Unidade I&D Cardiovascular do Porto.,Serviço de Cirurgia Cardiotorácica, Centro Hospitalar São João
| | - Paulo Bettencourt
- Faculdade de Medicina da Universidade do Porto, Unidade I&D Cardiovascular do Porto.,Serviço de Medicina Interna, Hospital CUF Porto, Porto, Portugal
| |
Collapse
|
77
|
Inflammation and Heart Failure: Therapeutic or Diagnostic Opportunity? J Am Coll Cardiol 2019; 69:1286-1287. [PMID: 28279295 DOI: 10.1016/j.jacc.2017.01.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 01/18/2017] [Indexed: 11/20/2022]
|
78
|
Fiordelisi A, Iaccarino G, Morisco C, Coscioni E, Sorriento D. NFkappaB is a Key Player in the Crosstalk between Inflammation and Cardiovascular Diseases. Int J Mol Sci 2019; 20:ijms20071599. [PMID: 30935055 PMCID: PMC6480579 DOI: 10.3390/ijms20071599] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/21/2019] [Accepted: 03/26/2019] [Indexed: 02/08/2023] Open
Abstract
Inflammation is a key mechanism of cardiovascular diseases. It is an essential component of atherosclerosis and a significant risk factor for the development of cardiovascular events. In the crosstalk between inflammation and cardiovascular diseases, the transcription factor NFκB seems to be a key player since it is involved in the development and progression of both inflammation and cardiac and vascular damage. In this review, we deal with the recent findings of the role of inflammation in cardiac diseases, focusing, in particular, on NFκB as a functional link. We describe strategies for the therapeutic targeting of NFκB as a potential strategy for the failing heart.
Collapse
Affiliation(s)
- Antonella Fiordelisi
- Department of Advanced Biomedical Sciences, Federico II University of Naples, 80131 Napoli, Italy.
| | - Guido Iaccarino
- Department of Advanced Biomedical Sciences, Federico II University of Naples, 80131 Napoli, Italy.
| | - Carmine Morisco
- Department of Advanced Biomedical Sciences, Federico II University of Naples, 80131 Napoli, Italy.
| | - Enrico Coscioni
- Division of Cardiac Surgery, AOU San Giovanni di Dio e Ruggi d'Aragona, 84131 Salerno, Italy.
| | - Daniela Sorriento
- Department of Advanced Biomedical Sciences, Federico II University of Naples, 80131 Napoli, Italy.
| |
Collapse
|
79
|
Effect of aerobic and resistance training on inflammatory markers in heart failure patients: systematic review and meta-analysis. Heart Fail Rev 2019; 23:209-223. [PMID: 29392623 DOI: 10.1007/s10741-018-9677-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Elevated levels of pro-inflammatory markers are evident in patients with heart failure and are associated with disease severity and prognosis. Exercise training has been shown to reduce circulating levels of pro-inflammatory cytokines and other pro-inflammatory markers in healthy and clinical populations. The aim of the systematic review and meta-analysis was to investigate the effect of aerobic (AT) and resistance training (RT) interventions on circulating concentrations of inflammatory markers; tumour necrosis factor-alpha (TNF-α), interleukin 6 (IL-6), C-reactive protein (CRP), fibrinogen, soluble intercellular adhesion molecule (sICAM) and soluble vascular adhesion molecule (sVCAM) in heart failure patients. We conducted database searches (PubMed, EMBASE and Cochrane Trials Register to 30 June 2017) for exercise-based trials in heart failure, using the following search terms: exercise training, inflammation, tumour necrosis factor-alpha, interleukin 6, C-reactive protein, fibrinogen, soluble intercellular adhesions molecule-1, soluble vascular adhesion molecule-1. Twenty studies, representing 18 independent trials, were included in the review. Pooled data of six studies indicated a minimally favourable effect of exercise training on circulating TNF-α [SMD 0.42 (95% CI 0.15, 0.68), p = 0.002)]. However, together the pooled and descriptive analyses failed to provide strong evidence for a reduction in other pro-inflammatory markers. However, given the complexity of heart failure and the pathways involved in the immune and inflammatory process, large prospective trials considering aetiology, comorbidities and local skeletal muscle inflammation are required to elucidate on the anti-inflammatory effect of exercise in this population.
Collapse
|
80
|
Radley G, Pieper IL, Ali S, Bhatti F, Thornton CA. The Inflammatory Response to Ventricular Assist Devices. Front Immunol 2018; 9:2651. [PMID: 30498496 PMCID: PMC6249332 DOI: 10.3389/fimmu.2018.02651] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 10/26/2018] [Indexed: 12/27/2022] Open
Abstract
The therapeutic use of ventricular assist devices (VADs) for end-stage heart failure (HF) patients who are ineligible for transplant has increased steadily in the last decade. In parallel, improvements in VAD design have reduced device size, cost, and device-related complications. These complications include infection and thrombosis which share underpinning contribution from the inflammatory response and remain common risks from VAD implantation. An added and underappreciated difficulty in designing a VAD that supports heart function and aids the repair of damaged myocardium is that different types of HF are accompanied by different inflammatory profiles that can affect the response to the implanted device. Circulating inflammatory markers and changes in leukocyte phenotypes receive much attention as biomarkers for mortality and disease progression. However, they are seldom used to monitor progress during and outcomes from VAD therapy or during the design phase for new devices. Even the partial reversal of heart damage associated with heart failure is a desirable outcome from VAD use. Therefore, improved understanding of the interplay between VADs and the recipient's inflammatory response would potentially increase their uptake, improve patient lives, and fuel research related to other blood-contacting medical devices. Here we provide a review of what is currently known about inflammation in heart failure and how this inflammatory profile is altered in heart failure patients receiving VAD therapy.
Collapse
Affiliation(s)
- Gemma Radley
- Swansea University Medical School, Swansea, United Kingdom.,Calon Cardio-Technology Ltd, Institute of Life Science, Swansea, United Kingdom
| | - Ina Laura Pieper
- Swansea University Medical School, Swansea, United Kingdom.,Scandinavian Real Heart AB, Västerås, Sweden
| | - Sabrina Ali
- Calon Cardio-Technology Ltd, Institute of Life Science, Swansea, United Kingdom
| | - Farah Bhatti
- Department of Cardiology, Morriston Hospital, Abertawe Bro Morgannwg University Health Board, Swansea, United Kingdom
| | | |
Collapse
|
81
|
Dolati S, Yousefi M, Mahdipour M, Afrasiabi Rad A, Pishgahi A, Nouri M, Jodati AR. Mesenchymal stem cell and bone marrow mononuclear cell therapy for cardiomyopathy: From bench to bedside. J Cell Biochem 2018; 120:45-55. [DOI: 10.1002/jcb.27531] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 08/01/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Sanam Dolati
- Aging Research Institute, Tabriz University of Medical Sciences Tabriz Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Student’s Research Committee, Tabriz University of Medical Sciences Tabriz Iran
| | - Mehdi Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Department of Immunology Tabriz University of Medical Sciences Tabriz Iran
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Department of Reproductive Biology Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences Tabriz Iran
| | - Abbas Afrasiabi Rad
- Cardiovascular Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Department of Cardiac Surgery Tabriz University of Medical Tabriz Iran
| | - Alireza Pishgahi
- Department of Physical Medicine and Rehabilitation Physical Medicine and Rehabilitation Research Center, Tabriz University of Medical Science Tabriz Iran
| | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Department of Reproductive Biology Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences Tabriz Iran
| | - Ahmad Reza Jodati
- Cardiovascular Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Department of Cardiac Surgery Tabriz University of Medical Tabriz Iran
| |
Collapse
|
82
|
Kittel-Schneider S, Kaspar M, Berliner D, Weber H, Deckert J, Ertl G, Störk S, Angermann C, Reif A. CRP genetic variants are associated with mortality and depressive symptoms in chronic heart failure patients. Brain Behav Immun 2018; 71:133-141. [PMID: 29627531 DOI: 10.1016/j.bbi.2018.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 03/07/2018] [Accepted: 04/04/2018] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE Heart failure (HF) is a complex medical condition with a multitude of genetic and other factors being involved in the pathogenesis. Emerging evidence points to an involvement of inflammatory mechanisms at least in subgroups of patients. The same is true for depression and depressive symptoms, which have a high prevalence in HF patients and are risk factors for the development and outcomes of cardiovascular disease. METHODS In 936 patients of the Interdisciplinary Network Heart Failure (INH) program, CRP and IL-6 protein blood levels were measured and genetic variants (single nucleotide polymorphisms) of the CRP and IL6 gene analyzed regarding their influence on mortality. RESULTS Less common recessive genotypes of two single nucleotide polymorphisms in the CRP gene (rs1800947 and rs11265263) were associated with significantly higher mortality risk (p < 0.006), higher CRP levels (p = 0.029, p = 0.006) and increased depressive symptoms in the PHQ-9 (p = 0.005, p = 0.003). Variants in the IL-6 gene were not associated with mortality. CONCLUSION Our results hint towards an association of less common CRP genetic variants with increased mortality risk, depressive symptoms and peripheral CRP levels in this population of HF patients thereby suggesting a possible role of the inflammatory system as link between poor prognosis in HF and depressive symptoms.
Collapse
Affiliation(s)
- S Kittel-Schneider
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, Frankfurt, Germany; Comprehensive Heart Failure Center, University Hospital of Würzburg, Würzburg, Germany.
| | - M Kaspar
- Comprehensive Heart Failure Center, University Hospital of Würzburg, Würzburg, Germany
| | - D Berliner
- Comprehensive Heart Failure Center, University Hospital of Würzburg, Würzburg, Germany; Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - H Weber
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, Frankfurt, Germany; Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Würzburg, Würzburg, Germany
| | - J Deckert
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Würzburg, Würzburg, Germany
| | - G Ertl
- Comprehensive Heart Failure Center, University Hospital of Würzburg, Würzburg, Germany; Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - S Störk
- Comprehensive Heart Failure Center, University Hospital of Würzburg, Würzburg, Germany; Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - C Angermann
- Comprehensive Heart Failure Center, University Hospital of Würzburg, Würzburg, Germany; Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - A Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, Frankfurt, Germany; Comprehensive Heart Failure Center, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
83
|
Influences of Ivabradine treatment on serum levels of cardiac biomarkers sST2, GDF-15, suPAR and H-FABP in patients with chronic heart failure. Acta Pharmacol Sin 2018; 39:1189-1196. [PMID: 29239349 PMCID: PMC6289366 DOI: 10.1038/aps.2017.167] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 09/29/2017] [Indexed: 12/18/2022] Open
Abstract
Chronic heart failure (CHF) represents a major cause of hospitalization and death. Recent evidence shows that novel biomarkers such as soluble suppression of tumorigenicity (sST2), growth-differentiation factor-15 (GDF-15), soluble urokinase plasminogen activator receptor (suPAR) and heart-type fatty acid binding protein (H-FABP) are correlated with inflammatory and ischemic responses in CHF patients. In this study we examined the effects of Ivabradine that inhibited the hyperpolarization-activated cyclic nucleotide-gated channel (HCN channel, also called funny current If), thereby leading to selective heart rate reduction and improved myocardial oxygen supply on the cardiac biomarkers sST2, GDF-15, suPAR and H-FABP in 50 CHF patients at the University Hospital of Jena. Patients were divided into three groups based on the etiology of CHF: dilated cardiomyopathy (DCM, n=20), ischemic cardiomyopathy (ICM, n=20) and hypertensive cardiomyopathy (HCM, n=10). The patients were administered Ivabradine (5 mg, bid for 3 months, and 7.5 mg bid for further 3 months). Analyses of cardiovascular biomarkers were performed at baseline as well as at 3- and 6-month follow-ups. At 6-month follow-up, GDF-15 levels were significantly reduced compared to baseline levels (P=0.0215), indicating a reduction in the progress of cardiac remodeling. H-FABP concentration was significantly lower in DCM patients compared to ICM (1.89 vs 3.24 μg/mL) and HCM patients (1.89 vs 3.80 μg/mL), and decreased over the 6-month follow-up (P=0.0151). suPAR median levels remained elevated, implying major ongoing inflammatory processes. As shown by significant decreases in GDF-15 and H-FABP levels, a reduction in ventricular remodeling and sub-clinical ischemia could be assumed. However, markers of hemodynamic stress (sST2) and inflammation (suPAR) showed no change or progression after 6 months of Ivabradine treatment in CHF patients. Further studies are necessary to validate the clinical applicability of these novel cardiovascular biomarkers.
Collapse
|
84
|
Carrizales-Sepúlveda EF, Ordaz-Farías A, Vera-Pineda R, Flores-Ramírez R. Periodontal Disease, Systemic Inflammation and the Risk of Cardiovascular Disease. Heart Lung Circ 2018; 27:1327-1334. [PMID: 29903685 DOI: 10.1016/j.hlc.2018.05.102] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Accepted: 05/11/2018] [Indexed: 01/08/2023]
Abstract
Periodontal and cardiovascular disease are both major health issues. Poor oral health has long been associated with the development of systemic diseases, with the typical example being the risk of endocarditis posterior to dental procedures. Through the years, the association of periodontal disease with other non-infectious systemic diseases has been brought to attention. One of the most interesting associations is the one that exists with the development of cardiovascular disease. Many studies, including systematic reviews and meta-analyses, suggest an important association between periodontal disease and ischaemic heart disease, cerebrovascular disease, heart failure, atrial fibrillation and peripheral artery disease. Among the proposed mechanisms of this relationship, systemic inflammation appears to play a major role. Evidence suggests that periodontal inflammation triggers a systemic inflammatory state that, added to the damage mediated by antibodies that cross react between periodontal pathogens and components of the intimal wall, and the direct lesion of the intima by bacteria entering the circulation, promotes atheroma plaque development and progression. There are other studies that show a clear relationship between periodontal disease severity, elevations of inflammatory markers, and the presence of atherosclerosis. Here, we give a review of the available evidence supporting this association, and the possible mechanisms involved.
Collapse
Affiliation(s)
| | - Alejandro Ordaz-Farías
- Echocardiography Laboratory, Cardiology Service, Hospital Universitario, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México
| | - Raymundo Vera-Pineda
- Internal Medicine Department, Hospital Universitario, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México
| | - Ramiro Flores-Ramírez
- Internal Medicine Department, Hospital Universitario, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México; Echocardiography Laboratory, Cardiology Service, Hospital Universitario, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, México
| |
Collapse
|
85
|
Girerd N, Zannad F, Rossignol P. Reply: Carbohydrate Antigen-125 in Heart Failure: An Overlooked Biomarker of Congestion. JACC. HEART FAILURE 2018; 6:442-443. [PMID: 29724372 DOI: 10.1016/j.jchf.2018.02.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 02/22/2018] [Indexed: 10/17/2022]
|
86
|
Alvarez-Alvarez B, García-Seara J, Rodríguez-Mañero M, Iglesias-Alvarez D, Martínez-Sande JL, Agra-Bermejo RM, Fernández López XA, González-Melchor L, Gude Sampedro F, Díaz-Louzao C, González-Juanatey JR. Prognostic value of nutrition status in the response of cardiac resynchronization therapy. Indian Pacing Electrophysiol J 2018; 18:133-139. [PMID: 29649579 PMCID: PMC6090074 DOI: 10.1016/j.ipej.2018.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 03/03/2018] [Accepted: 04/05/2018] [Indexed: 11/17/2022] Open
Abstract
Background Cardiac resynchronization therapy (CRT) is indicated in symptomatic heart failure (HF) patients after achieving optimal medical therapy. However, there are still a large percentage of patients who do not respond to CRT. Malnutrition is a frequent comorbidity in patients with HF, and it is associated with a poorer prognosis. Here, we evaluate the nutritional status of patients assessed by Controlling Nutritional Status (CONUT) score and its association with structural remodeling and cardiovascular events. Methods We investigated the effect of CONUT on HF/death in 302 consecutive patients with a CRT device implanted between 2005 and 2015 in a single tertiary center. We categorized the patients into three groups: normal nutritional status (CONUT 0–1), mild malnutrition (CONUT 2–4) and moderate-severe malnutrition (CONUT ≥ 5). Changes in nutritional status were assessed in patients with mild-to-severe malnutrition prior to CRT. Results One hundred and forty-eight patients exhibited normal nutritional status (49.0%), 99 patients exhibited mild malnutrition (32.8%) and 55 patients exhibited moderate-severe malnutrition (18.2%). CONUT scores of at least 2 were associated with higher risk of HF/death compared with CONUT 0–1. Significant left ventricular (LV) reverse remodeling was noted in patients with better nutritional status. In addition, those malnutrition patients at baseline that improved nutritional state exhibited fewer HF/death events at follow-up. Conclusion CONUT score prior to CRT was an independent risk factor of death/HF and was correlated with LV reverse remodeling. Improvements in CONUT score during long-term follow-up were associated with a reduction in the rate of HF/death.
Collapse
Affiliation(s)
- Belén Alvarez-Alvarez
- Cardiology Department, Clinical University Hospital of Santiago de Compostela, CIBERCV, Spain.
| | - Javier García-Seara
- Arrhythmia Unit, Clinical University Hospital of Santiago de Compostela, CIBERCV, Spain
| | | | - Diego Iglesias-Alvarez
- Cardiology Department, Clinical University Hospital of Santiago de Compostela, CIBERCV, Spain
| | - Jose L Martínez-Sande
- Arrhythmia Unit, Clinical University Hospital of Santiago de Compostela, CIBERCV, Spain
| | - Rosa M Agra-Bermejo
- Cardiology Department, Clinical University Hospital of Santiago de Compostela, CIBERCV, Spain
| | | | | | | | - Carla Díaz-Louzao
- Epidemiology Department, Clinical University Hospital of Santiago de Compostela, Spain
| | | |
Collapse
|
87
|
Impact of reduced forced expiratory volume on cardiac prognosis in patients with chronic heart failure. Heart Vessels 2018; 33:1037-1045. [PMID: 29556692 DOI: 10.1007/s00380-018-1153-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 03/16/2018] [Indexed: 01/08/2023]
Abstract
In patients with chronic heart failure (CHF), comorbidity of airflow limitation is associated with poor outcomes. The forced expiratory volume in 1 s (FEV1) is used to evaluate the severity of airflow limitation. However, the impact of FEV1 severity on prognosis has only been partially elucidated in patients with CHF. In total, 248 consecutive patients with CHF who successfully fulfilled spirometric measurement criteria were enrolled and prospectively followed. Percent predicted FEV1 (FEV1%predicted) was associated with the New York Heart Association Functional Classification. FEV1%predicted was significantly associated with diastolic dysfunction, evaluated using echocardiography; elevated inflammation markers; and increased pulmonary arterial pressure. There were 60 cardiac events, including 9 cardiac-related deaths and 51 re-hospitalizations due to the exacerbation of CHF during a follow-up period. Kaplan-Meier analysis revealed that the lowest FEV1%predicted group had the highest event rate, irrespective of the presence of smoking history. Multivariate Cox proportional hazard analysis showed that FEV1%predicted was an independent predictor of cardiac events after adjusting for confounders. The net reclassification improvement and integrated discrimination improvement were improved by the addition of FEV1%predicted to other cardiac risk factors. Decreased FEV1%predicted was independently associated with the poor cardiac outcomes in patients with CHF.
Collapse
|
88
|
Ma T, Zhu D, Chen D, Zhang Q, Dong H, Wu W, Lu H, Wu G. Sulforaphane, a Natural Isothiocyanate Compound, Improves Cardiac Function and Remodeling by Inhibiting Oxidative Stress and Inflammation in a Rabbit Model of Chronic Heart Failure. Med Sci Monit 2018. [PMID: 29527002 PMCID: PMC5859672 DOI: 10.12659/msm.906123] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Background The aim of this study was to investigate the effects of sulforaphane (SFN), a natural isothiocyanate compound, in a rabbit ascending aortic cerclage model of chronic heart failure (CHF). Material/Methods Thirty New Zealand White rabbits were divided into the sham operation group (n=10), the CHF group (n=10), and the CHF + SFN group (n=10) treated with subcutaneous SFN (0.5 mg/kg) for five days per week for 12 weeks. After 12 weeks, echocardiography and biometric analysis were performed, followed by the examination of the rabbit hearts. Enzyme-linked immunosorbent assay (ELISA) and Western blot were used to detect levels of inflammatory cytokines, superoxide dismutase (SOD), and malondialdehyde (MDA). Results In the CHF group, compared with the sham operation group, there was an increase in the heart weight to body weight ratio (HW/BW), the left ventricular weight to body weight ratio (LVW/BW), the left ventricular end diastolic diameter (LVEDD), the left ventricular end systolic diameter (LVESD), plasma brain natriuretic peptide (BNP) and atrial natriuretic peptide (ANP) levels, the cardiac collagen volume fraction (CVF), apoptotic index, expression levels of collagen I, collagen III, tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and malondialdehyde (MDA) in the myocardial tissue, and a decrease in the left ventricular shortening fraction (LVFS) and left ventricular ejection fraction (LVEF), and cardiac superoxide dismutase (SOD) activity. These changes were corrected in the SFN-treated group. Conclusions In a rabbit model of CHF, treatment with SFN improved cardiac function and remodeling by inhibiting oxidative stress and inflammation.
Collapse
Affiliation(s)
- Tongliang Ma
- Department of Emergency Internal Medicine, The People's Hospital of Bozhou, Bozhou, Anhui, China (mainland)
| | - Decai Zhu
- Department of Emergency Internal Medicine, The People's Hospital of Bozhou, Bozhou, Anhui, China (mainland)
| | - Duoxue Chen
- Department of Cardiology, The People's Hospital of Bozhou, Bozhou, Anhui, China (mainland)
| | - Qiaoyun Zhang
- Department of Emergency Internal Medicine, The People's Hospital of Bozhou, Bozhou, Anhui, China (mainland)
| | - Huifang Dong
- Department of Emergency Internal Medicine, The People's Hospital of Bozhou, Bozhou, Anhui, China (mainland)
| | - Wenwu Wu
- Department of Emergency Internal Medicine, The People's Hospital of Bozhou, Bozhou, Anhui, China (mainland)
| | - Huihe Lu
- Department of Cardiology, Nantong First People's Hospital, Nantong, Jiangsu, China (mainland)
| | - Guangfu Wu
- Department of Emergency Internal Medicine, The People's Hospital of Bozhou, Bozhou, Anhui, China (mainland)
| |
Collapse
|
89
|
Influence of LGALS3 gene polymorphisms on susceptibility and prognosis of dilated cardiomyopathy in a Northern Han Chinese population. Gene 2018; 642:293-298. [DOI: 10.1016/j.gene.2017.11.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 10/07/2017] [Accepted: 11/08/2017] [Indexed: 12/22/2022]
|
90
|
Vulesevic B, Sirois MG, Allen BG, de Denus S, White M. Subclinical Inflammation in Heart Failure: A Neutrophil Perspective. Can J Cardiol 2018; 34:717-725. [PMID: 29801737 DOI: 10.1016/j.cjca.2018.01.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 01/18/2018] [Accepted: 01/18/2018] [Indexed: 12/18/2022] Open
Abstract
Although it is widely recognized that inflammation plays a critical role in the development and pathology of heart failure (HF), very little is known about the involvement of one of the most abundant immune cells in the blood, a primary immune response cell: the neutrophil. This review summarizes the current literature on the role of subclinical inflammation, with a focus on the neutrophil in the pathophysiology of the HF syndrome. Some emerging therapeutic strategies are also discussed.
Collapse
Affiliation(s)
- Branka Vulesevic
- Research Center, Montreal Heart Institute, Université de Montréal, Montreal, Québec, Canada; Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, Québec, Canada
| | - Martin G Sirois
- Research Center, Montreal Heart Institute, Université de Montréal, Montreal, Québec, Canada; Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, Québec, Canada
| | - Bruce G Allen
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Québec, Canada
| | - Simon de Denus
- Research Center, Montreal Heart Institute, Université de Montréal, Montreal, Québec, Canada; Faculty of Pharmacy, Université de Montréal, Montreal, Québec, Canada
| | - Michel White
- Research Center, Montreal Heart Institute, Université de Montréal, Montreal, Québec, Canada; Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, Québec, Canada; Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Québec, Canada.
| |
Collapse
|
91
|
Lee RH, Sloane R, Pieper C, Lyles KW, Adler RA, Van Houtven C, LaFleur J, Colón-Emeric C. Clinical Fractures Among Older Men With Diabetes Are Mediated by Diabetic Complications. J Clin Endocrinol Metab 2018; 103:281-287. [PMID: 29099931 PMCID: PMC5761492 DOI: 10.1210/jc.2017-01593] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 10/27/2017] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Type 2 diabetes mellitus among older women has been associated with increased bone mineral density, but paradoxically with increased fracture risk. Findings among older men have varied, and potential mechanisms have not been fully elucidated. METHODS A retrospective study of male veterans 65 to 99 years of age who received primary care in the Veterans Health Administration from 2000 to 2010, using administrative data from all 146 Veterans Health Administration medical centers linked to Centers for Medicare and Medicaid Services Medicare fee-for-service data. Potential mediating factors of the diabetes-associated risk were evaluated using negative binomial regression models with the outcomes of any clinical fracture and hip fracture. RESULTS Of 2,798,309 Veterans included in the cohort, 900,402 (32.3%) had a diagnosis of diabetes. After adjusting for age, race, ethnicity, body mass index, alcohol and tobacco use, rheumatoid arthritis, and corticosteroid use, the risk of any clinical fracture associated with diabetes was 1.22 (95% confidence interval, 1.21 to 1.23) and that of hip fracture was 1.21 (95% confidence interval, 1.19 to 1.23). Significant mediating factors included peripheral neuropathy, cardiovascular disease, and congestive heart failure, with 45.5% of the diabetes-associated fracture risk explained by these diagnoses. CONCLUSIONS Older male Veterans with diabetes have a 22% increased risk of incident clinical fracture compared with those without. A significant portion of this risk is explained by diabetes-related comorbidities, specifically peripheral neuropathy and congestive heart failure. Identification of these mediating factors suggests possible mechanisms, as well as potential interventions.
Collapse
Affiliation(s)
- Richard H. Lee
- Duke University School of Medicine, Durham, North Carolina 27710
- Durham Veterans Affairs Medical Center, Durham, North Carolina 27705
| | - Richard Sloane
- Duke University School of Medicine, Durham, North Carolina 27710
- Durham Veterans Affairs Medical Center, Durham, North Carolina 27705
| | - Carl Pieper
- Duke University School of Medicine, Durham, North Carolina 27710
| | - Kenneth W. Lyles
- Duke University School of Medicine, Durham, North Carolina 27710
- Durham Veterans Affairs Medical Center, Durham, North Carolina 27705
| | - Robert A. Adler
- Hunter Holmes McGuire Veterans Affairs Medical Center, Richmond, Virginia 23249
- Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298
| | - Courtney Van Houtven
- Duke University School of Medicine, Durham, North Carolina 27710
- Durham Veterans Affairs Medical Center, Durham, North Carolina 27705
| | - Joanne LaFleur
- University of Utah, Salt Lake City, Utah 84112
- Salt Lake City Veterans Affairs Medical Center, Salt Lake City, Utah 84148
| | - Cathleen Colón-Emeric
- Duke University School of Medicine, Durham, North Carolina 27710
- Durham Veterans Affairs Medical Center, Durham, North Carolina 27705
| |
Collapse
|
92
|
Elhoff JJ, Shekerdemian LS. The slow burn: are we neglecting potential long-term cardiovascular sequelae of critical illness in children? THE LANCET CHILD & ADOLESCENT HEALTH 2017; 1:254-256. [PMID: 30169178 DOI: 10.1016/s2352-4642(17)30123-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 10/06/2017] [Indexed: 10/18/2022]
Affiliation(s)
- Justin J Elhoff
- Section of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas 77030, USA
| | - Lara S Shekerdemian
- Section of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas 77030, USA.
| |
Collapse
|
93
|
Cvetinovic N, Isakovic AM, Lainscak M, Dungen HD, Nikolic NM, Loncar G. Procalcitonin in heart failure: hic et nunc. Biomark Med 2017; 11:893-903. [PMID: 28976777 DOI: 10.2217/bmm-2017-0160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Although procalcitonin (PCT) was evaluated for the first time in the setting of heart failure (HF) in 1999, its utility in HF patients is still under examination. Patients with HF have significantly higher plasma PCT concentrations than healthy subjects and PCT levels are associated with severity of HF. It has been confirmed that higher levels of PCT are associated with worse outcomes, such as increased mortality and higher rate of rehospitalization, in HF patients with no evidence of infection. Furthermore, it has been approved that PCT-guided antibiotic treatment in HF patients reduces duration of antibiotic therapy and improves outcomes. This review summarizes current evidence from the published literature of the usefulness and limitations of PCT as a biomarker in HF.
Collapse
Affiliation(s)
- Natasa Cvetinovic
- Department of Cardiology, Zvezdara University Medical Center, Belgrade, Serbia
| | | | - Mitja Lainscak
- Department of Internal Medicine, General Hospital Murska Sobota, Slovenia.,Faculty of Medicine, University of Ljubljana, Slovenia
| | - Hans-Durk Dungen
- Department of Cardiology, Campus Virchow, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Natasa Markovic Nikolic
- Department of Cardiology, Zvezdara University Medical Center, Belgrade, Serbia.,School of Medicine, University of Belgrade, Serbia
| | - Goran Loncar
- Department of Cardiology, Zvezdara University Medical Center, Belgrade, Serbia.,School of Medicine, University of Belgrade, Serbia
| |
Collapse
|
94
|
Lichtenauer M, Jirak P, Wernly B, Paar V, Rohm I, Jung C, Schernthaner C, Kraus J, Motloch LJ, Yilmaz A, Hoppe UC, Christian Schulze P, Kretzschmar D, Pistulli R. A comparative analysis of novel cardiovascular biomarkers in patients with chronic heart failure. Eur J Intern Med 2017; 44:31-38. [PMID: 28579310 DOI: 10.1016/j.ejim.2017.05.027] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/19/2017] [Accepted: 05/25/2017] [Indexed: 12/26/2022]
Abstract
BACKGROUND Heart failure (HF) with reduced ejection fraction remains a major therapeutic challenge. The aim of this study was to investigate the role of novel cardiovascular biomarkers, i.e. soluble suppression of tumorigenicity (sST2), growth-differentiation factor-15 (GDF-15), soluble urokinase plasminogen activator receptor (suPAR) and heart-type fatty acid binding protein (H-FABP) in patients with ischaemic (ICM) or dilative cardiomyopathy (DCM). MATERIALS AND METHODS A total of 200 patients were enrolled in this study: 65 were diagnosed with DCM and 59 patients suffering from ICM were included. 76 patients without coronary artery disease or signs of heart failure were included as controls. Plasma samples of all patients were analyzed by use of ELISA. RESULTS Levels of sST2, suPAR and H-FABP were significantly higher in ICM and DCM patients compared to the control group (p<0.0001). However, there were no significant differences between ICM and DCM in biomarker levels. Ejection fraction correlated inversely with cardiac biomarkers (sST2 p<0.0001, GDF-15 p=0.0394, suPAR p=0.0029, H-FABP p<0.0001). Similarly, CRP levels also showed a positive correlation with cardiac biomarkers. Renal insufficiency (p<0.0001) and diabetes (sST2 p=0.0021, GDF-15 p=0.0055, suPAR p=0.0339, H-FABP p=0.0010) were significantly associated with a rise in cardiac biomarkers. CONCLUSION Novel cardiovascular biomarkers such as ST2, GDF-15, uPAR and H-FABP could offer a great potential for more precise diagnostic in ICM and DCM patients. H-FABP was the most promising marker in our study, followed by sST2, uPAR and GDF-15. Additional prospective studies will be necessary to further evaluate the potential clinical benefits in routine treatment of HF.
Collapse
Affiliation(s)
- Michael Lichtenauer
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Austria.
| | - Peter Jirak
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Austria
| | - Bernhard Wernly
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Austria
| | - Vera Paar
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Austria
| | - Ilonka Rohm
- Universitätsherzzentrum Thüringen, Clinic of Internal Medicine I, Department of Cardiology, Friedrich Schiller University Jena, Germany
| | - Christian Jung
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Duesseldorf, Germany
| | - Christiana Schernthaner
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Austria
| | - Johannes Kraus
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Austria
| | - Lukas J Motloch
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Austria
| | - Atilla Yilmaz
- Clinic of Internal Medicine II, Elisabeth Klinikum Schmalkalden, Germany
| | - Uta C Hoppe
- Clinic of Internal Medicine II, Department of Cardiology, Paracelsus Medical University of Salzburg, Austria
| | - P Christian Schulze
- Universitätsherzzentrum Thüringen, Clinic of Internal Medicine I, Department of Cardiology, Friedrich Schiller University Jena, Germany
| | - Daniel Kretzschmar
- Universitätsherzzentrum Thüringen, Clinic of Internal Medicine I, Department of Cardiology, Friedrich Schiller University Jena, Germany
| | - Rudin Pistulli
- Universitätsherzzentrum Thüringen, Clinic of Internal Medicine I, Department of Cardiology, Friedrich Schiller University Jena, Germany
| |
Collapse
|
95
|
Xuan W, Huang W, Wang R, Chen C, Chen Y, Wang Y, Tan X. Elevated circulating IL-32 presents a poor prognostic outcome in patients with heart failure after myocardial infarction. Int J Cardiol 2017; 243:367-373. [PMID: 28747035 DOI: 10.1016/j.ijcard.2017.03.065] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/20/2017] [Accepted: 03/15/2017] [Indexed: 02/05/2023]
Abstract
BACKGROUND Interleukin-32 (IL-32) is a newly discovered proinflammatory cytokine. However, there are limited data regarding IL-32 as a biomarker for heart failure (HF). In this study, we assessed the prognostic value of IL-32 in patients with chronic HF after myocardial infarction (MI). METHODS AND RESULTS Over a period of 1.8years, we prospectively enrolled 100 patients with chronic HF after MI. IL-32, NT-proBNP, Matrix metallopeptidase 9 (MMP-9), procollagen type I (PI) and type III (PIII) were measured at baseline. Study endpoint was adverse cardiac events. High IL-32 levels were associated with numerous factors that are related to deteriorate cardiac function and cardiac fibrosis. Strong expression of IL-32 was detected in human cardiomyocytes from HF tissue. ROC curve revealed the area under the curve of IL-32 for predicting negative outcome of HF was 0.72 (95% CI: 0.60-0.83, P<0.01). Kaplan-Meier statistics showed that the risk of adverse cardiac event was 5.75 fold (hazard ratio 5.75, 95% CI 1.53-21.58, P=0.009), which increased in the highest quartile (>296pg/mL). Cox regression analysis revealed IL-32 was an independent predictor for cardiac events (hazard ratio 2.78, 95% CI 1.02-7.57, P=0.046). Recombinant IL-32 significantly exacerbated infarct size in a mouse model of MI. IL-32 upregulated expression of MMP-9, PIII and transforming growth factor beta in rat fibroblasts. CONCLUSION IL-32 might be a novel predictor of adverse cardiac event in patients with HF after MI. The pro-fibrotic effect of IL-32 may contribute to adverse cardiac remodeling and progression to HF.
Collapse
Affiliation(s)
- Wanling Xuan
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Weixing Huang
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Ruijie Wang
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Chang Chen
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yequn Chen
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yan Wang
- Department of Radiology, University of California San Francisco, San Francisco, CA, United States
| | - Xuerui Tan
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China.
| |
Collapse
|
96
|
Radley G, Pieper IL, Thornton CA. The effect of ventricular assist device-associated biomaterials on human blood leukocytes. J Biomed Mater Res B Appl Biomater 2017; 106:1730-1738. [PMID: 28888071 DOI: 10.1002/jbm.b.33981] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 08/01/2017] [Accepted: 08/16/2017] [Indexed: 12/29/2022]
Abstract
Ventricular assist devices (VADs) are an effective bridging or destination therapy for patients with advanced stage heart failure. These devices remain susceptible to adverse events including infection, bleeding, and thrombus; events linked to the foreign body response. Therefore, the biocompatibility of all biomaterials used is crucial to the success of medical devices. Biomaterials common in VADs-DLC: diamond-like carbon coated stainless steel; Sap: single-crystal sapphire; SiN: silicon nitride; Ti: titanium alloy; and ZTA: zirconia-toughened alumina-were tested for their biocompatibility through incubation with whole human blood for 2 h with mild agitation. Blood was then removed and used for: complete cell counts; leukocyte activation and death, and the production of key inflammatory cytokines. All were compared to time 0 and an un-exposed 2 h sample. Monocyte numbers were lower after exposure to DLC, SiN, and ZTA and monocytes showed evidence of activation with DLC, Sap, and SiN. Neutrophils and lymphocytes were unaffected. This approach allows comprehensive analysis of the potential blood damaging effects of biomaterials. Monocyte activation by DLC, Sap, ZTA, and SiN warrants further investigation linking effects on this cell type to unfavorable inflammatory/thrombogenic responses to VADs and other blood handling devices. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 1730-1738, 2018.
Collapse
Affiliation(s)
- Gemma Radley
- Institute of Life Science, Swansea University Medical School, Swansea, Wales, UK.,Calon Cardio - Technology Ltd., Institute of Life Science, Swansea, Wales, UK
| | - Ina Laura Pieper
- Institute of Life Science, Swansea University Medical School, Swansea, Wales, UK.,Calon Cardio - Technology Ltd., Institute of Life Science, Swansea, Wales, UK
| | - Catherine A Thornton
- Institute of Life Science, Swansea University Medical School, Swansea, Wales, UK
| |
Collapse
|
97
|
Pasqualetti G, Seghieri M, Santini E, Rossi C, Vitolo E, Giannini L, Malatesta MG, Calsolaro V, Monzani F, Solini A. P2X 7 Receptor and APOE Polymorphisms and Survival from Heart Failure: A Prospective Study in Frail Patients in a Geriatric Unit. Aging Dis 2017; 8:434-441. [PMID: 28840058 PMCID: PMC5524806 DOI: 10.14336/ad.2016.1202] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 12/02/2016] [Indexed: 11/29/2022] Open
Abstract
Heart failure (HF) is one of the most frequent cause of hospitalization in elderly and often coexists with concurrent geriatric syndromes, like cognitive disturbances; various pathophysiological mechanisms are shared by HF and cognitive decline, notably a substrate of low-grade inflammation. We investigated whether SNPs in the purinergic receptor (P2X7R) and apolipoprotein (APO) E genes, both involved in a series of inflammatory responses, are associated to HF or cognitive impairment and are able to predict post-discharge mortality in the elderly. We prospectively analyzed 198 patients (age 85 ± 8 years, predominantly females) admitted to a Geriatric unit for acute HF, whose diagnosis was based on clinical signs, brain natriuretic peptide (BNP) values and ecocardiography in uncertain diagnosis (BNP values between 100 and 400 pg/mL); cognitive performance was assesed by Short Portable Mental Status Questionnaire (SPMSQ). In all the participants, SNPs rs208294 and rs3751143 for P2X7R gene and rs429558 and rs7412 for APOE gene were assessed. Information on all-cause mortality was adjudicated by medical records review 36 months after discharge. We found no relationship between P2X7R and APOE polymorphisms and 36-month post-discharge mortality; a better outcome for overall survival was observed in patients with BNP values below the median (281 pg/mL) (p=0.002) persisting after adjustment for renal function and age, and in those with cognitive impairment (p<0.001). Patients harboring APOE-ε4 genotype showed higher BNP concentrations than noncarriers (1289.9 ± 226.9 vs 580.5 ± 90.2 pg/mL respectively,p=0.004), whereas none of the studied SNPs were associated to impairment in cognitive performance. In conclusion, neither P2X7R or APOE genotype seem to predict long-term mortality in elderly patients. Interestingly, APOE-ε4 genotype was associated to higher BNP values, suggesting a putative interaction between genetic and biochemical markers in identifying people at risk for HF.
Collapse
Affiliation(s)
- Giuseppe Pasqualetti
- Department of Clinical and Experimental Medicine University of Pisa, Pisa, Italy
| | - Marta Seghieri
- Department of Clinical and Experimental Medicine University of Pisa, Pisa, Italy
| | - Eleonora Santini
- Department of Clinical and Experimental Medicine University of Pisa, Pisa, Italy
| | - Chiara Rossi
- Department of Clinical and Experimental Medicine University of Pisa, Pisa, Italy
| | - Edoardo Vitolo
- Department of Clinical and Experimental Medicine University of Pisa, Pisa, Italy
| | - Livia Giannini
- Department of Clinical and Experimental Medicine University of Pisa, Pisa, Italy
| | | | - Valeria Calsolaro
- Department of Clinical and Experimental Medicine University of Pisa, Pisa, Italy
| | - Fabio Monzani
- Department of Clinical and Experimental Medicine University of Pisa, Pisa, Italy
| | - Anna Solini
- Department of Clinical and Experimental Medicine University of Pisa, Pisa, Italy
| |
Collapse
|
98
|
Tabrez S, Jabir NR, Firoz CK, Hindawi S, Shakil S, Damanhouri GA, Zaidi SK. Estimation of Interleukin-1β Promoter (−31 C/T and −511 T/C) Polymorphisms and Its Level in Coronary Artery Disease Patients. J Cell Biochem 2017; 118:2977-2982. [DOI: 10.1002/jcb.25958] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 02/27/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Shams Tabrez
- King Fahd Medical Research Center; King Abdulaziz University; Jeddah Saudi Arabia
| | - Nasimudeen R. Jabir
- King Fahd Medical Research Center; King Abdulaziz University; Jeddah Saudi Arabia
| | - Chelapram K. Firoz
- King Fahd Medical Research Center; King Abdulaziz University; Jeddah Saudi Arabia
| | - Salwa Hindawi
- Faculty of Medicine; Department of Hematology; King Abdulaziz University Hospital; Jeddah Saudi Arabia
| | - Shazi Shakil
- Center of Excellence in Genomic Medicine Research; King Abdulaziz University; Jeddah Saudi Arabia
- Faculty of Applied Medical Sciences; Department of Medical Laboratory Technology; King Abdulaziz University; Jeddah Saudi Arabia
| | - Ghazi A. Damanhouri
- King Fahd Medical Research Center; King Abdulaziz University; Jeddah Saudi Arabia
| | - Syed Kashif Zaidi
- Center of Excellence in Genomic Medicine Research; King Abdulaziz University; Jeddah Saudi Arabia
| |
Collapse
|
99
|
Abstract
Excessive feeding is associated with an increase in the incidence of chronic metabolic diseases, such as obesity, insulin resistance, and type 2 diabetes. Metabolic disturbance induces chronic low-grade inflammation in metabolically-important organs, such as the liver and adipose tissue. Many of the inflammatory signalling pathways are directly triggered by nutrients. The pro-inflammatory mediators in adipocytes and macrophages infiltrating adipose tissue promote both local and systemic pro-inflammatory status. Metabolic cardiomyopathy is a chronic metabolic disease characterized by structural and functional alterations and interstitial fibrosis without coronary artery disease or hypertension. In the early stage of metabolic cardiomyopathy, metabolic disturbance is not accompanied by substantial changes in myocardial structure and cardiac function. However, metabolic disturbance induces subcellular low-grade inflammation in the heart, and in turn, subcellular component abnormalities, such as oxidative stress, mitochondrial dysfunction, endoplasmic reticulum stress, and impaired calcium handling, leading to impaired myocardial relaxation. In the advanced stage, the vicious cycle of subcellular component abnormalities, inflammatory cell infiltration, and neurohumoral activation induces cardiomyocyte injury and death, and cardiac fibrosis, resulting in impairment of both diastolic and systolic functions. This review discusses some recent advances in understanding involvement of inflammation in metabolic cardiomyopathy.
Collapse
|
100
|
Fujita SI, Tanaka S, Maeda D, Morita H, Fujisaka T, Takeda Y, Ito T, Ishizaka N. Serum Soluble Urokinase-Type Plasminogen Activator Receptor Is Associated with Low Left Ventricular Ejection Fraction and Elevated Plasma Brain-Type Natriuretic Peptide Level. PLoS One 2017; 12:e0170546. [PMID: 28135310 PMCID: PMC5279735 DOI: 10.1371/journal.pone.0170546] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 01/06/2017] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Recent studies have suggested that soluble urokinase plasminogen activator receptor (suPAR), a biomarker of subclinical levels of inflammation, is significantly correlated with cardiovascular events. PURPOSE We investigated the association between suPAR and left ventricular ejection fraction (LVEF), left ventricular mass index (LVMI), and plasma B-type natriuretic peptide (BNP) among cardiac inpatients. METHODS AND RESULTS In total, 242 patients (mean age 71.3 ± 9.8 years; 70 women) admitted to the cardiology department were enrolled in the study. suPAR was significantly correlated with LVEF (R = -0.24, P<0.001), LVMI (R = 0.16, P = 0.014) and BNP (R = 0.46, P<0.001). In logistic regression analysis, the highest suPAR tertile (> 3236 pg/mL) was associated with low LVEF (< 50%) and elevated BNP (> 300 pg/mL) with an odds ratio of 3.84 (95% confidence interval [CI], 1.22-12.1) and 5.36 (95% CI, 1.32-21.8), respectively, after adjusting for age, sex, log-transformed estimated glomerular filtration rate (log(eGFR)), C-reactive protein, and diuretic use. The association between suPAR and LVMI was not statistically significant. In multivariate receiver operating characteristic analysis, addition of log(suPAR) to the combination of age, sex, log(eGFR) and CRP incrementally improved the prediction of low LVEF (area under the curve [AUC], 0.827 to 0.852, P = 0.046) and BNP ≥ 300 pg/mL (AUC, 0.869 to 0.906; P = 0.029). CONCLUSIONS suPAR was associated with low LVEF and elevated BNP, but not with left ventricular hypertrophy, independent of CRP, renal function, and diuretic use among cardiac inpatients who were not undergoing chronic hemodialysis.
Collapse
Affiliation(s)
- Shu-ichi Fujita
- Department of Cardiology, Osaka Medical College, Osaka, Japan
| | - Suguru Tanaka
- Department of Cardiology, Osaka Medical College, Osaka, Japan
| | - Daichi Maeda
- Department of Cardiology, Osaka Medical College, Osaka, Japan
| | - Hideaki Morita
- Department of Cardiology, Osaka Medical College, Osaka, Japan
| | | | | | - Takahide Ito
- Department of Cardiology, Osaka Medical College, Osaka, Japan
| | - Nobukazu Ishizaka
- Department of Cardiology, Osaka Medical College, Osaka, Japan
- * E-mail:
| |
Collapse
|