1
|
Jin Y, Huang Y, Zhang T, Sun Q, Zhang Y, Zhang P, Wang G, Zhang J, Wu J. Associations of dietary total, heme, non-heme iron intake with diabetes, CVD, and all-cause mortality in men and women with diabetes. Heliyon 2024; 10:e38758. [PMID: 39430450 PMCID: PMC11490858 DOI: 10.1016/j.heliyon.2024.e38758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 09/29/2024] [Accepted: 09/29/2024] [Indexed: 10/22/2024] Open
Abstract
Background Iron metabolism disorders significantly increase the risk of diabetes and its related complications by inducing oxidative stress, inflammation, insulin resistance, and disturbances in glucose and lipid metabolism. However, whether dietary iron intake can influence progression of diabetes remains unclear. The present study aims to investigate the relationship between total iron, heme iron, and non-heme iron intake and diabetes, CVD, and all-cause mortality among men and women with diabetes in the U.S. population. Methods A total of 4416 adults with diabetes(2415 men and 2001 women) from the NHANES 2003-2014 were enrolled. Dietary information was collected by 24-h dietary recall during two nonconsecutive days. Dietary total iron intake was measured based on the dietary survey. Dietary heme iron intake was calculated based on its proportion in dietary total iron intake from each food. non-heme iron is the difference between total iron and heme iron. Diabetes, CVD, and all-cause mortality status were identified as main outcomes. Cox models and RCS analysis were performed to estimate the hazard ratios and 95%CIs. Results For men, the participants with a higher dietary heme iron intake were associated with higher risks of CVD (HRheme iron = 1.61,95%CI:1.03-2.51) and all-cause mortality (HRheme iron = 1.42,95%CI:1.10-1.83). For women, participants in the highest quartile of dietary total/heme/non-heme iron intake had a higher diabetes mortality risk ((HRtotal iron = 2.33,95%CI:1.24-4.38; HRheme iron = 1.87,95%CI:1.00-3.49; HRnon-heme iron = 2.28,95%CI:1.19-4.39), compared to those in the lowest quartile. Additionally, the dose-response curve for the relationship between dietary non-heme iron intake and CVD mortality followed a reverse J-shape in women with diabetes. Conclusions Higher dietary heme iron intake was associated with an increased CVD mortality risk in both men and women with diabetes. Additionally, higher dietary total, heme, and non-heme iron intake was linked to an increased risk of diabetes mortality among women with diabetes. Therefore, women with diabetes should pay more attention on the overconsumption of any type of dietary iron.
Collapse
Affiliation(s)
- Yimin Jin
- Department of General Practice, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yang Huang
- Wu Lian De Memorial Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tongshuai Zhang
- Department of Neurobiology, Harbin Medical University, Harbin, China
- Ministry of Education Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Harbin, China
| | - Qixu Sun
- Department of Digestive System, YANTAI PENGLAI People's Hospital, Yan Tai, China
| | - Yao Zhang
- Department of Neurobiology, Harbin Medical University, Harbin, China
| | - Peiru Zhang
- School of Public Health, Harbin Medical University, Harbin, China
| | - Guangyou Wang
- Department of Neurobiology, Harbin Medical University, Harbin, China
- Ministry of Education Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Harbin, China
| | - Jingyu Zhang
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinrong Wu
- Department of Anaesthesiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
2
|
Li Y, Zhou Q, Zhang K, Meng X. Iron Overload and Abdominal Aortic Aneurysm. Rev Cardiovasc Med 2024; 25:361. [PMID: 39484115 PMCID: PMC11522754 DOI: 10.31083/j.rcm2510361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/15/2024] [Accepted: 04/19/2024] [Indexed: 11/03/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a chronic vascular degenerative disease characterized by progressive segmental dilation of the abdominal aorta. The rupture of an AAA represents a leading cause of death in cardiovascular diseases. Despite numerous experimental and clinical studies examining potential drug targets and therapies, currently there are no pharmaceutical treatment to prevent AAA growth and rupture. Iron is an essential element in almost all living organisms and has important biological functions. Epidemiological studies have indicated that both iron deficiency and overload are associated with adverse clinical outcomes, particularly an increased risk of cardiovascular events. Recent evidence indicates that iron overload is involved in the pathogenesis of abdominal aortic aneurysms. In this review, we provide an overview of the role of iron overload in AAA progression and explore its potential pathological mechanisms. Although the exact molecular mechanisms of iron overload in the development of AAA remain to be elucidated, the inhibition of iron deposition may offer a promising strategy for preventing these aneurysms.
Collapse
Affiliation(s)
- Yunyi Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 250012 Jinan, Shandong, China
| | - Quan Zhou
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 250012 Jinan, Shandong, China
| | - Kai Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 250012 Jinan, Shandong, China
| | - Xiao Meng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 250012 Jinan, Shandong, China
| |
Collapse
|
3
|
Chen W, Zhang H, Shen X, Hong L, Tao H, Xiao J, Nie S, Wei M, Chen M, Zhang C, Yu W. Iron metabolism indexes as predictors of the incidence of cardiac surgery-associated acute kidney surgery. J Cardiothorac Surg 2024; 19:533. [PMID: 39300557 DOI: 10.1186/s13019-024-03080-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 09/15/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is a major complication following cardiac surgery. We explored the clinical utility of iron metabolism indexes for identification of patients at risk for AKI after cardiac surgery. METHODS This prospective observational study included patients who underwent cardiac surgery between March 2023 and June 2023. Iron metabolism indexes were measured upon admission to the intensive care unit. Multivariable logistic regression analyses were performed to explore the relationship between iron metabolism indexes and cardiac surgery-associated AKI (CSA-AKI). Receiver operating characteristic (ROC) curve was used to assess the predictive ability of iron, APACHE II score and the combination of the two indicators. Restricted cubic splines (RCS) was used to further confirm the linear relationship between iron and CSA-AKI. RESULTS Among the 112 recruited patients, 38 (33.9%) were diagnosed with AKI. Multivariable logistic regression analysis indicated that APACHE II score (odds ratio [OR], 1.208; 95% confidence interval [CI], 1.003-1.455, P = 0.036) and iron (OR 1.069; 95% CI 1.009-1.133, P = 0.036) could be used as independent risk factors to predict CSA-AKI. ROC curve analysis showed that iron (area under curve [AUC] = 0.669, 95% CI 0.572-0.757), APACHE II score (AUC = 0.655, 95% CI 0.557-0.744) and iron and APACHE II score combination (AUC = 0.726, 95% CI 0.632-0.807) were predictive indicators for CSA-AKI. RCS further confirmed the linear relationship between iron and CSA-AKI. CONCLUSIONS Elevated iron levels were independently associated with higher risk of CSA-AKI, and there was a linear relationship between iron and CSA-AKI.
Collapse
Affiliation(s)
- Wenxiu Chen
- Department of Intensive Care Unit, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, Jiangsu, China
- Department of Intensive Care Unit, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, China
| | - Hao Zhang
- Department of Nephrology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Xiao Shen
- Department of Intensive Care Unit, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, China
| | - Liang Hong
- Department of Intensive Care Unit, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, China
| | - Hong Tao
- Department of Intensive Care Unit, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, China
| | - Jilai Xiao
- Department of Intensive Care Unit, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, China
| | - Shuai Nie
- Department of Intensive Care Unit, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, China
| | - Meng Wei
- Department of Intensive Care Unit, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, China
| | - Ming Chen
- Department of Intensive Care Unit, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, Jiangsu, China
| | - Cui Zhang
- Department of Intensive Care Unit, Nanjing First Hospital, Nanjing Medical University, No. 68 Changle Road, Nanjing, 210006, Jiangsu, China.
| | - Wenkui Yu
- Department of Intensive Care Unit, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
4
|
Suárez-Ortegón MF, McLachlan S, Fernández-Real JM, Wilson JF, Wild SH. Both low and high body iron stores relate to metabolic syndrome in postmenopausal women: Findings from the VIKING Health Study-Shetland (VIKING I). Eur J Clin Invest 2024:e14312. [PMID: 39239983 DOI: 10.1111/eci.14312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND There are conflicting results among studies on the association between serum ferritin (SF) and metabolic syndrome (MetS), and by groups of sex/menopausal status. To date, there are no studies on British populations. The SF-MetS association might be U/J-shaped. We evaluated whether SF was independently associated with MetS (harmonized definition) in people from Shetland, Scotland. METHODS We analysed cross-sectional data from the Viking Health Study-Shetland (589 premenopausal women [PreMW], 625 postmenopausal women [PostW] and 832 men). Logistic regressions using two approaches, one with the lowest sex and menopausal status-specific ferritin quartile (Q) as the reference and other using the middle two quartiles combined (2-3) as the reference, were conducted to estimate the SF-MetS association. The shape of the association was verified via cubic spline analyses. The associations were adjusted for age, inflammatory and hepatic injury markers, alcohol intake, smoking and BMI. RESULTS Prevalence of MetS was 18.3%. Among PostMW both low and high SF were associated with MetS (fully adjusted odds ratios [95% confidence interval] compared to the middle two quartiles combined were: 1.99 [1.17-3.38] p =.011 for Q1 and 2.10 [1.27-3.49] p =.004 for Q4) This U-shaped pattern was confirmed in the cubic spline analysis in PostMW with a ferritin range of 15-200 ug/L. In men, a positive association between ferritin quartiles with Q1 as the reference, did not remain significant after adjustment for BMI. CONCLUSION Extreme quartiles of iron status were positively associated with MetS in PostMW, while no SF-MetS associations were found in men or PreMW. The ferritin-MetS association pattern differs between populations and U/J-shaped associations may exist.
Collapse
Affiliation(s)
| | | | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain
- Nutrition, Eumetabolism and Health Group, Institut d'Investigació Biomèdica de Girona (IDIBGI-CERCA), Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain
| | - James F Wilson
- Centre for Global Health Research, Usher Institute, Edinburgh, UK
- Centre for Genomic Medicine, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Sarah H Wild
- Usher Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
5
|
von Haehling S, Assmus B, Bekfani T, Dworatzek E, Edelmann F, Hashemi D, Hellenkamp K, Kempf T, Raake P, Schütt KA, Wachter R, Schulze PC, Hasenfuss G, Böhm M, Bauersachs J. Heart failure with preserved ejection fraction: diagnosis, risk assessment, and treatment. Clin Res Cardiol 2024; 113:1287-1305. [PMID: 38602566 PMCID: PMC11371894 DOI: 10.1007/s00392-024-02396-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/02/2024] [Indexed: 04/12/2024]
Abstract
The aetiology of heart failure with preserved ejection fraction (HFpEF) is heterogenous and overlaps with that of several comorbidities like atrial fibrillation, diabetes mellitus, chronic kidney disease, valvular heart disease, iron deficiency, or sarcopenia. The diagnosis of HFpEF involves evaluating cardiac dysfunction through imaging techniques and assessing increased left ventricular filling pressure, which can be measured directly or estimated through various proxies including natriuretic peptides. To better narrow down the differential diagnosis of HFpEF, European and American heart failure guidelines advocate the use of different algorithms including comorbidities that require diagnosis and rigorous treatment during the evaluation process. Therapeutic recommendations differ between guidelines. Whilst sodium glucose transporter 2 inhibitors have a solid evidence base, the recommendations differ with regard to the use of inhibitors of the renin-angiotensin-aldosterone axis. Unless indicated for specific comorbidities, the use of beta-blockers should be discouraged in HFpEF. The aim of this article is to provide an overview of the current state of the art in HFpEF diagnosis, clinical evaluation, and treatment.
Collapse
Affiliation(s)
- Stephan von Haehling
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Strasse 40, 37075, Göttingen, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany.
| | - Birgit Assmus
- Department of Cardiology and Angiology, Universitätsklinikum Gießen und Marburg, Giessen, Germany
| | - Tarek Bekfani
- Department of Cardiology and Angiology, Universitätsklinikum Magdeburg, Magdeburg, Germany
| | - Elke Dworatzek
- Institute of Gender in Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Frank Edelmann
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Campus Virchow-Klinikum, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Djawid Hashemi
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Campus Virchow-Klinikum, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Digital Clinician Scientist Program, Berlin, Germany
| | - Kristian Hellenkamp
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Strasse 40, 37075, Göttingen, Germany
| | - Tibor Kempf
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Philipp Raake
- I. Medical Department, Cardiology, Pneumology, Endocrinology and Intensive Care Medicine, University Hospital Augsburg, University of Augsburg, Augsburg, Germany
| | - Katharina A Schütt
- Department of Internal Medicine I, University Hospital RWTH Aachen, Aachen, Germany
| | - Rolf Wachter
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Strasse 40, 37075, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig, Germany
| | - Paul Christian Schulze
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, FSU, Jena, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Strasse 40, 37075, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | - Michael Böhm
- Kardiologie, Angiologie und Internistische Intensivmedizin, Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, Saarland University, Homburg, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
6
|
Park JA, Yoon JE, Liu X, Chang Y, Maiolino G, Pengo MF, Lin GM, Kwon Y. Cardiovascular Implications of Sleep Disorders Beyond Sleep Apnea. CURRENT SLEEP MEDICINE REPORTS 2024; 10:320-328. [PMID: 39281064 PMCID: PMC11391919 DOI: 10.1007/s40675-024-00302-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2024] [Indexed: 09/18/2024]
Abstract
Purpose of Review Sleep is crucial for human health and life. There is still limited attention to the association between sleep disorders beyond sleep apnea and cardiovascular (CV) health. We investigated the current evidence between non-respiratory sleep disorders and CV health. Recent Findings Current evidence suggests an important association between sleep duration, circadian rhythm, insomnia, disorders of hypersomnolence and CV health. Sleep-related movement disorders exhibit a moderate association with CV health. Further research is needed to explore the effects of each sleep disorder on CV health. Summary Given the close association between non-respiratory sleep disorders and CV health, it is crucial to recognize and address sleep disorders in patients with a high CV risk.
Collapse
Affiliation(s)
- Jung-A Park
- Department of Neurology, Daegu Catholic University Medical Center, Daegu, Korea
| | - Jee-Eun Yoon
- Department of Neurology, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Xiaoyue Liu
- New York University Rory Meyers College of Nursing, New York, NY, USA
| | - Yoonhee Chang
- Staff Physician, Sleep Medicine, Evergreen Health, Kirkland, WA, USA
| | - Giuseppe Maiolino
- Clinica Medica 3, Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Martino F Pengo
- Department of Cardiovascular, Neural and Metabolic Sciences, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Gen-Min Lin
- Department of Medicine, Hualien-Armed Forces General Hospital, Hualien, Taiwan
| | - Younghoon Kwon
- Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
7
|
Yang C, Hu T, Li C, Gong A. Dietary iron intake predicts all-cause and cardiovascular mortality in patients with diabetes. Nutr Diabetes 2024; 14:68. [PMID: 39179569 PMCID: PMC11343882 DOI: 10.1038/s41387-024-00286-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND Limited data exists on the link between dietary iron intake and mortality in diabetes. Our investigation aimed to explore how dietary iron intake correlates with overall and cause-specific mortality in diabetic individuals. METHODS This analysis encompassed 5970 participants with diabetes from the National Health and Nutrition Examination Survey spanning 1999 to 2014. Baseline data were collected through surveys and examinations, with mortality status tracked via National Death Index records until December 31, 2015. Cox proportional hazard models were utilized to calculate hazard ratios (HR) and 95% confidence intervals (CI) for mortality from various causes, including cardiovascular disease (CVD) and cancer. RESULTS The average iron intake among the cohort was 14.1 ± 7.4 mg daily, with an average participant age of 61.3 and 3059 (51.3%) male adults. Over 41,425 person-years of follow-up, 1497 deaths were recorded. Following adjustments for multiple variables, an iron intake between 11.1 and 14.4 mg was associated with the lowest risk of all-cause mortality (HR 0.83 [0.70, 0.99], P < 0.05) compared to the reference group (<8.3 mg). Analysis of dose-response curves revealed an L-shaped pattern in men and a J-shaped pattern in women concerning the relationship between iron intake and all-cause mortality. CONCLUSIONS Our findings suggest a nonlinear association between dietary iron intake and all-cause mortality in individuals with diabetes. Specifically, higher iron intake may increase all-cause mortality risk in men, while potentially exert a protective effect in women.
Collapse
Affiliation(s)
- Chenchen Yang
- Department Emergency, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Tingting Hu
- Department Cardiology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Chenglin Li
- Department Cardiothoracic Surgery, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Aifeng Gong
- Department General Practice, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China.
| |
Collapse
|
8
|
Zhang Y, Li B, Cai H, Fu Y, Zheng Y. Associations of iron metabolism and inflammation with all-cause and cardiovascular mortality in a large NHANES community sample: Moderating and mediating effects. Nutr Metab Cardiovasc Dis 2024; 34:1854-1863. [PMID: 38658228 DOI: 10.1016/j.numecd.2024.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/01/2024] [Accepted: 03/21/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND AND AIMS This study aimed to assess the associations between serum iron concentration, C-reactive protein (CRP) concentration and the risk of all-cause mortality and cardiovascular mortality in the general population and to explore potential mediating and moderating effects. METHODS AND RESULTS This study analyzed data from the National Health and Nutrition Examination Survey spanning the years 1999-2010, encompassing 23,634 participants. Cox proportional hazards regression models were employed to investigate the independent associations of serum iron and CRP with all-cause and cardiovascular mortality. Moderation and mediation analyses explored the moderating effect of CRP on the association between the serum iron concentration and all-cause and cardiovascular mortality, and the mediating role of the serum iron concentration in the association between the CRP concentration and all-cause and cardiovascular mortality. After multivariate adjustments in the Cox model, serum iron and CRP levels were independently correlated with both all-cause and cardiovascular mortality risk. Moderation analyses revealed a more pronounced correlation between the serum iron concentration and both all-cause and cardiovascular mortality in participants with higher CRP levels. Mediation analysis indicated that the serum iron concentration partly mediated the impact of CRP on the risk of all-cause mortality (13.79%) and cardiovascular mortality (24.12%). CONCLUSION Serum iron and CRP are independently associated with all-cause and cardiovascular mortality. Moreover, the associations between serum iron concentrations and both all-cause and cardiovascular mortality are more pronounced in individuals with elevated CRP. Serum iron partially mediates the effect of CRP on all-cause and cardiovascular mortality.
Collapse
Affiliation(s)
- Yaoting Zhang
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Bing Li
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun, 130021, China.
| | - He Cai
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Yu Fu
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Yang Zheng
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
9
|
Kremer D, Knobbe TJ, Vinke JSJ, Groothof D, Post A, Annema C, Abrahams AC, van Jaarsveld BC, de Borst MH, Berger SP, Bakker SJL, Eisenga MF. Iron deficiency, anemia, and patient-reported outcomes in kidney transplant recipients. Am J Transplant 2024; 24:1456-1466. [PMID: 38493925 DOI: 10.1016/j.ajt.2024.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 02/29/2024] [Accepted: 03/12/2024] [Indexed: 03/19/2024]
Abstract
Kidney transplant recipients (KTRs) experience more fatigue, anxiety, and depressive symptoms and lower concentration and health-related quality of life (HRQoL) compared with the general population. Anemia is a potential cause that is well-recognized and treated. Iron deficiency, however, is often unrecognized, despite its potential detrimental effects related to and unrelated to anemia. We investigated the interplay of anemia, iron deficiency, and patient-reported outcomes in 814 outpatient KTRs (62% male, age 56 ± 13 years) enrolled in the TransplantLines Biobank and Cohort Study (Groningen, The Netherlands). In total, 28% had iron deficiency (ie, transferrin saturation < 20% and ferritin < 100 μg/L), and 29% had anemia (World Health Organization criteria). In linear regression analyses, iron deficiency, but not anemia, was associated with more fatigue, worse concentration, lower wellbeing, more anxiety, more depressive symptoms, and lower HRQoL, independent of age, sex, estimated glomerular filtration rate, anemia, and other potential confounders. In the fully adjusted logistic regression models, iron deficiency was associated with an estimated 53% higher risk of severe fatigue, a 100% higher risk of major depressive symptoms, and a 51% higher chance of being at risk for sick leave/work disability. Clinical trials are needed to investigate the effect of iron deficiency correction on patient-reported outcomes and HRQoL in KTRs.
Collapse
Affiliation(s)
- Daan Kremer
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Tim J Knobbe
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Joanna Sophia J Vinke
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Dion Groothof
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Adrian Post
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Coby Annema
- Section of Nursing Science, Department of Health Sciences, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Alferso C Abrahams
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Brigit C van Jaarsveld
- Department of Nephrology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Martin H de Borst
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Stefan P Berger
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Stephan J L Bakker
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Michele F Eisenga
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
10
|
Pezacki AT, Gonciarz RL, Okamura T, Matier CD, Torrente L, Cheng K, Miller SG, Ralle M, Ward NP, DeNicola GM, Renslo AR, Chang CJ. A tandem activity-based sensing and labeling strategy reveals antioxidant response element regulation of labile iron pools. Proc Natl Acad Sci U S A 2024; 121:e2401579121. [PMID: 38968123 PMCID: PMC11252945 DOI: 10.1073/pnas.2401579121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/29/2024] [Indexed: 07/07/2024] Open
Abstract
Iron is an essential element for life owing to its ability to participate in a diverse array of oxidation-reduction reactions. However, misregulation of iron-dependent redox cycling can also produce oxidative stress, contributing to cell growth, proliferation, and death pathways underlying aging, cancer, neurodegeneration, and metabolic diseases. Fluorescent probes that selectively monitor loosely bound Fe(II) ions, termed the labile iron pool, are potentially powerful tools for studies of this metal nutrient; however, the dynamic spatiotemporal nature and potent fluorescence quenching capacity of these bioavailable metal stores pose challenges for their detection. Here, we report a tandem activity-based sensing and labeling strategy that enables imaging of labile iron pools in live cells through enhancement in cellular retention. Iron green-1 fluoromethyl (IG1-FM) reacts selectively with Fe(II) using an endoperoxide trigger to release a quinone methide dye for subsequent attachment to proximal biological nucleophiles, providing a permanent fluorescent stain at sites of elevated labile iron. IG1-FM imaging reveals that degradation of the major iron storage protein ferritin through ferritinophagy expands the labile iron pool, while activation of nuclear factor-erythroid 2-related factor 2 (NRF2) antioxidant response elements (AREs) depletes it. We further show that lung cancer cells with heightened NRF2 activation, and thus lower basal labile iron, have reduced viability when treated with an iron chelator. By connecting labile iron pools and NRF2-ARE activity to a druggable metal-dependent vulnerability in cancer, this work provides a starting point for broader investigations into the roles of transition metal and antioxidant signaling pathways in health and disease.
Collapse
Affiliation(s)
- Aidan T. Pezacki
- Department of Chemistry, University of California, Berkeley, CA94720
| | - Ryan L. Gonciarz
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA94158
| | - Toshitaka Okamura
- Department of Chemistry, University of California, Berkeley, CA94720
| | - Carson D. Matier
- Department of Chemistry, University of California, Berkeley, CA94720
| | - Laura Torrente
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL33612
| | - Ke Cheng
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA94158
| | - Sophia G. Miller
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR97239
| | - Martina Ralle
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR97239
| | - Nathan P. Ward
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL33612
| | - Gina M. DeNicola
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL33612
| | - Adam R. Renslo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA94158
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA94158
| | - Christopher J. Chang
- Department of Chemistry, University of California, Berkeley, CA94720
- Department of Molecular and Cell Biology, University of California, Berkeley, CA94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA94720
| |
Collapse
|
11
|
Peri V, Devlin P, Perry L, Richards T, Miles LF. Associations Between Nonanemic Iron Deficiency and Postoperative Outcomes in Cardiac Surgery: A Systematic Review and Meta-Analysis. Anesth Analg 2024; 139:47-57. [PMID: 38241670 DOI: 10.1213/ane.0000000000006822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2024]
Abstract
BACKGROUND Preoperative screening for iron deficiency is a part of patient blood management protocols. This systematic review, meta-analysis, and meta-regression reviews the association between nonanemic iron deficiency and postoperative outcomes in patients undergoing cardiac surgery. We aimed to determine whether preoperative screening for nonanemic iron deficiency should be recommended in patients undergoing cardiac surgery. METHODS Electronic databases MEDLINE (Ovid), Embase (Ovid), and Scopus were searched from inception until December 9, 2022. Studies were considered for inclusion if they (1) used an observational study design; (2) enrolled adult patients undergoing cardiac surgery; and (3) included an iron-deficient and iron-replete group, defined using serum ferritin and/or transferrin saturation. The primary outcome was the length of acute hospital stay. Secondary outcomes included length of intensive care unit stay, requirement for allogeneic red blood cell transfusion, number of red blood cell units transfused, days alive and at home at postoperative days 30 and 90, all-cause postoperative complications, postoperative infection, mortality, and hospital readmission. Meta-regression was performed to assess the effects of study and patient-level factors on the associations between nonanemic iron deficiency and specific outcomes. Individual study quality was assessed using the Risk of Bias in Non-Randomized Studies of Exposures (ROBINS-E) tool. The Grading of Recommendations, Assessment, Development, and Evaluations (GRADE) framework was used to determine the quality of evidence for each outcome. RESULTS Eight studies (2683 patients) were included. No significant association was observed between nonanemic iron deficiency and any of the primary or secondary outcomes except for an increased requirement for allogeneic red blood cell transfusion (odds ratio [OR], 1.39 [95% confidence interval, CI, 1.16-1.68; P < .001; I2 2.8%]). Meta-regression did not identify any potential sources of heterogeneity to explain variation in individual study results. The quality of evidence was rated as "low" or "very low" for all outcomes. CONCLUSIONS Few high-quality studies are available to assess associations between nonanemic iron deficiency and outcomes after cardiac surgery. Acknowledging these limitations, the presence of preoperative nonanemic iron deficiency was not associated with a change in the primary outcome of length of hospital stay, or any patient-centered secondary outcome compared to those without iron deficiency. There was an association with increased requirement for allogeneic red blood cell transfusion, but this did not impact the reported patient-centered outcomes.
Collapse
Affiliation(s)
- Varun Peri
- From the Department of Anaesthesia, Austin Health, Melbourne, Australia
| | - Paula Devlin
- From the Department of Anaesthesia, Austin Health, Melbourne, Australia
| | - Luke Perry
- Department of Critical Care, Melbourne Medical School, The University of Melbourne, Melbourne, Australia
| | - Toby Richards
- Department of Anaesthesiology and Perioperative Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Lachlan F Miles
- From the Department of Anaesthesia, Austin Health, Melbourne, Australia
- Department of Critical Care, Melbourne Medical School, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
12
|
Hirashiki A, Shimizu A, Kamihara T, Kokubo M, Hashimoto K, Ueda I, Murohara T. Prognostic Significance of Serum Uric Acid and Exercise Capacity in Older Adults Hospitalized for Worsening Cardiovascular Disease. J Cardiovasc Dev Dis 2024; 11:165. [PMID: 38921665 PMCID: PMC11203550 DOI: 10.3390/jcdd11060165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/19/2024] [Accepted: 05/20/2024] [Indexed: 06/27/2024] Open
Abstract
Elevated serum uric acid (sUA) is associated with the risk of developing cardiovascular disease (CVD). Here, we examined the prognostic significance of sUA and exercise capacity in 411 Japanese adults (age, ≥65; mean, 81 years) hospitalized for worsening CVD. When the patients were stratified by sUA into three groups (<5.3, 5.4-6.9, >7.0 mg/dL), the high-sUA group had a significantly worse peak VO2 and composite endpoint (rehospitalization due to worsening CVD and all-cause mortality) compared with low- and moderate-sUA groups (p < 0.001). When the patients were stratified by sUA into five groups (sUA < 3.9, 4.0-5.9, 6.0-7.9, 8.0-8.9, and >10.0 mg/dL), the incidence of the composite endpoint was significantly higher in the highest sUA group compared with that in the reference group, but only in women. Univariate Cox regression analysis, but not a multivariate analysis, indicated that sUA was significantly associated with the composite endpoint. Although sUA and exercise capacity may have some degree of prognostic significance in older patients with CVD, this significance may differ between men and women.
Collapse
Affiliation(s)
- Akihiro Hirashiki
- Department of Cardiology, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan; (A.S.); (T.K.); (M.K.)
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan;
| | - Atsuya Shimizu
- Department of Cardiology, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan; (A.S.); (T.K.); (M.K.)
| | - Takahiro Kamihara
- Department of Cardiology, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan; (A.S.); (T.K.); (M.K.)
| | - Manabu Kokubo
- Department of Cardiology, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan; (A.S.); (T.K.); (M.K.)
| | - Kakeru Hashimoto
- Department of Rehabilitation, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan; (K.H.); (I.U.)
| | - Ikue Ueda
- Department of Rehabilitation, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan; (K.H.); (I.U.)
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan;
| |
Collapse
|
13
|
Awad AK, Abdelgalil MS, Gonnah AR, Mouffokes A, Ahmad U, Awad AK, Elbadawy MA, Roberts DH. Intravenous iron for acute and chronic heart failure with reduced ejection fraction (HFrEF) patients with iron deficiency: An updated systematic review and meta-analysis. Clin Med (Lond) 2024; 24:100211. [PMID: 38643833 PMCID: PMC11092397 DOI: 10.1016/j.clinme.2024.100211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 02/23/2024] [Indexed: 04/23/2024]
Abstract
Patients with heart failure (HF) and iron deficiency are at increased risk of adverse clinical outcomes. We searched databases for randomised controlled trials that compared IV iron to placebo, in patients with HF with reduced ejection fraction (HFrEF). A total of 7,813 participants, all having HFrEF with 3,998 receiving IV iron therapy, and 3,815 control recipients were included. There was a significant improvement in Kansas City Cardiomyopathy Questionnaire favouring IV iron with MD 7.39, 95% CI [3.55, 11.22], p = 0.0002. Subgroup analysis, based on acute and chronic HF, has displayed a sustained statistical significance. Additionally, a significant increase in the left ventricular ejection fraction % was observed, with MD 3.76, 95% CI [2.32, 5.21], p < 0.00001. A significant improvement in 6-min walk test was noted, with MD 34.87, 95% CI [20.02, 49.72], p < 0.00001. Furthermore, IV iron showed significant improvement in NYHA class, peak VO2, serum ferritin, and haemoglobin levels. Finally, despite the lack of difference in terms of all-cause hospitalisation and HF-related death, IV iron was associated with a significant reduction in HF-related, any cardiovascular reason hospitalisations, and all-cause death; which supports the need for implementation of IV iron as a standard of care in patients with HF and iron deficiency.
Collapse
Affiliation(s)
- Ahmed K Awad
- Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | | | - Ahmed R Gonnah
- Imperial College Healthcare NHS Trust, London, United Kingdom.
| | - Adel Mouffokes
- Faculty of Medicine, University of Oran 1 Ahmed Ben Bella, Oran, Algeria
| | | | - Ayman K Awad
- Faculty of Medicine, El-Galala University, Suez, Egypt
| | | | - David Hesketh Roberts
- Lancashire Cardiac Centre, Blackpool, United Kingdom; University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
14
|
Sun Y, Peng W, Lin S, Cui J, Lu J. Iron Metabolic Biomarkers and the Mortality Risk in the General Population: A Nationwide Population-Based Cohort Study. J Endocr Soc 2024; 8:bvae063. [PMID: 38623382 PMCID: PMC11017327 DOI: 10.1210/jendso/bvae063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Indexed: 04/17/2024] Open
Abstract
Context Iron is an essential element in the human body and plays a critical role in many physiological and cellular processes. However, the association between iron status and the risk of all-cause or cause-specific mortality has not been well-investigated. And it is unclear whether the association between iron metabolic biomarkers and the risk of mortality differs between people with and without diabetes mellitus (DM). Objective This work aimed to investigate associations between iron metabolic biomarkers and all-cause and cause-specific mortality risk in the general population, and heterogeneities in the associations among population with and without DM.. Methods A total of 29 166 adults from the National Health and Nutrition Examination Survey (NHANES) III and NHANES 1999 to 2010 were included, with linkage to the National Death Index to December 31, 2019. Cox proportional-hazard models and Fine-Gray subdistribution hazard models were used to estimate associations between iron metabolic biomarkers and outcomes. Results During a median follow-up of 18.83 years, 9378 deaths were observed, including 3420 cardiovascular disease (CVD) deaths and 1969 cancer deaths. A significant linear association between serum ferritin (SF) and all-cause mortality was observed among the overall population and those without DM. J-shaped associations between transferrin saturation (TSAT) and all-cause and CVD mortality were observed among all populations. In the overall population, compared to the first quartile (Q1) group, the adjusted hazard ratio (HR) (95% CI) for all-cause mortality was 1.07 (1.00-1.15), 1.05 (0.98-1.12), 1.13 (1.05-1.21) in Q2, Q3, and Q4 groups for SF, while the HR was 0.94 (0.88-0.99), 0.92 (0.86-0.97), and 0.93 (0.88-0.99) for TSAT. In individuals without DM, the adjusted HR of the Q4 of SF were 1.19 (1.03-1.37) for CVD mortality and 1.25 (1.05-1.48) for cancer mortality. In individuals with DM, the adjusted HRs of the Q4 of TSAT were 0.76 (0.62-0.93) for CVD mortality and 1.47 (1.07-2.03) for cancer mortality. Conclusion Iron metabolism abnormalities increase mortality risk in the general population. The associations of iron status with mortality were significantly different between individuals with and without DM, which indicated tailored strategies for iron homeostasis are needed.
Collapse
Affiliation(s)
- Yuanyuan Sun
- Department of Geriatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, People's Republic of China
| | - Wenyao Peng
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Siqi Lin
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| | - Jingjing Cui
- Department of Geriatric Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, People's Republic of China
| | - Jiapeng Lu
- National Clinical Research Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, People's Republic of China
| |
Collapse
|
15
|
Osei Bonsu E, Addo IY, Boadi C, Boadu EF, Okeke SR. Determinants of iron-rich food deficiency among children under 5 years in sub-Saharan Africa: a comprehensive analysis of Demographic and Health Surveys. BMJ Open 2024; 14:e079856. [PMID: 38458798 DOI: 10.1136/bmjopen-2023-079856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/10/2024] Open
Abstract
BACKGROUND Iron deficiency is a major public health problem that affects the physical and cognitive development of children under 5 years of age (under-5 children) in sub-Saharan Africa (SSA). However, the factors associated with the limited consumption of iron-rich foods in the region are poorly understood. OBJECTIVE This study examined the prevalence and determinants of iron-rich food deficiency among under-5 children in 26 SSA countries. DESIGN This nationally representative quantitative study employed pooled data from Demographic and Health Surveys conducted between 2010 and 2019. METHODS Representative samples comprising 296 850 under-5 children from the various countries were used. Bivariate and multivariate logistic regression models were used to determine the associations between the lack of iron-rich food uptake and various sociodemographic factors. RESULT The overall prevalence of iron-rich food deficiency among the children in the entire sample was 56.75%. The prevalence of iron-rich food deficiency varied widely across the 26 countries, ranging from 42.76% in Congo Democratic Republic to 77.50% in Guinea. Maternal education, particularly primary education (OR 0.62, 95% CI 0.57 to 0.68) and higher education (OR 0.58, 95% CI 0.52 to 0.64), demonstrated a reduced likelihood of iron-rich food deficiency in the sample. Likewise, paternal education, with both primary education (OR 0.69, 95% CI 0.63 to 0.75) and higher education (OR 0.66, 95% CI 0.60 to 0.73) showed decreased odds of iron-rich food deficiency. Postnatal visits contributed significantly to reducing the odds of iron-rich food deficiency (OR 0.90, 95% CI 0.83 to 0.95), along with antenatal visits, which also had a positive impact (OR 0.84, 95% CI 0.74 to 0.95). Finally, residents in rural areas showed slightly higher odds of iron-rich food deficiency (OR 1.12, 95% CI 1.10 to 1.28). CONCLUSION Based on the findings, interventions targeting iron-food deficiency in the SSA region should take into strong consideration the key determinants highlighted in this study.
Collapse
Affiliation(s)
- Emmanuel Osei Bonsu
- Department of Epidemiology and Biostatistics, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | | | - Caleb Boadi
- Department of Operations and Management Information Systems, University of Ghana, Legon, Ghana
| | | | | |
Collapse
|
16
|
Kotit S. Benefits of intravenous iron supplementation in heart failure. Glob Cardiol Sci Pract 2024; 2024:e202410. [PMID: 38746071 PMCID: PMC11090186 DOI: 10.21542/gcsp.2024.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/14/2024] [Indexed: 05/16/2024] Open
Abstract
Introduction: Iron deficiency (ID) is one of the most frequent comorbidities in patients with heart failure (HF) and is estimated to be present in up to 80% of acute patients regardless of their ejection fraction. Randomized controlled trials have shown that supplementary intravenous iron results in improved clinical outcomes; however, the current understanding of the effects of intravenous iron on morbidity and mortality remains limited. Study and results: The meta-analysis pooled individual participant data from three randomized placebo-controlled trials of ferric carboxymaltose (FCM) in adult patients (n = 4,501) with heart failure and iron deficiency (CONFIRM-HF, AFFIRM-AHF, and HEART-FID). FCM therapy significantly reduced the co-primary composite endpoint of total cardiovascular hospitalizations and cardiovascular death, with a rate ratio (RR 0.86; 95% CI 0.75 to 0.98; p = 0.029). FCM therapy was associated with a 17% relative rate reduction in total cardiovascular hospitalizations (RR 0.83; 95% CI 0.73 to 0.96; p = 0.009) and a 16% relative rate reduction in total heart failure hospitalizations (RR 0.84; 95% CI 0.71 to 0.98; p = 0.025). Lessons learned: The meta-analysis shows that in iron-deficient patients with heart failure and reduced or mildly reduced left ventricular ejection fraction, intravenous ferric carboxymaltose (FCM) is associated with a reduced risk of total cardiovascular hospitalization and cardiovascular mortality. These findings indicate that intravenous FCM should be considered in iron-deficient patients with heart failure and reduced or mildly reduced ejection fractions.
Collapse
|
17
|
Wang T, Chen X, Wang K, Ju J, Yu X, Yu W, Liu C, Wang Y. Cardiac regeneration: Pre-existing cardiomyocyte as the hub of novel signaling pathway. Genes Dis 2024; 11:747-759. [PMID: 37692487 PMCID: PMC10491875 DOI: 10.1016/j.gendis.2023.01.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 01/22/2023] [Accepted: 01/30/2023] [Indexed: 09/12/2023] Open
Abstract
In the mammalian heart, cardiomyocytes are forced to withdraw from the cell cycle shortly after birth, limiting the ability of the heart to regenerate and repair. The development of multimodal regulation of cardiac proliferation has verified that pre-existing cardiomyocyte proliferation is an essential driver of cardiac renewal. With the continuous development of genetic lineage tracking technology, it has been revealed that cell cycle activity produces polyploid cardiomyocytes during the embryonic, juvenile, and adult stages of cardiogenesis, but newly formed mononucleated diploid cardiomyocytes also elevated sporadically during myocardial infarction. It implied that adult cardiomyocytes have a weak regenerative capacity under the condition of ischemia injury, which offers hope for the clinical treatment of myocardial infarction. However, the regeneration frequency and source of cardiomyocytes are still low, and the mechanism of regulating cardiomyocyte proliferation remains further explained. It is noteworthy to explore what force triggers endogenous cardiomyocyte proliferation and heart regeneration. Here, we focused on summarizing the recent research progress of emerging endogenous key modulators and crosstalk with other signaling pathways and furnished valuable insights into the internal mechanism of heart regeneration. In addition, myocardial transcription factors, non-coding RNAs, cyclins, and cell cycle-dependent kinases are involved in the multimodal regulation of pre-existing cardiomyocyte proliferation. Ultimately, awakening the myocardial proliferation endogenous modulator and regeneration pathways may be the final battlefield for the regenerative therapy of cardiovascular diseases.
Collapse
Affiliation(s)
- Tao Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266023, China
| | - Xinzhe Chen
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266023, China
| | - Kai Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266023, China
| | - Jie Ju
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266023, China
| | - Xue Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266023, China
| | - Wanpeng Yu
- College of Medicine, Qingdao University, Qingdao, Shandong 266023, China
| | - Cuiyun Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266023, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, Shandong 266023, China
| |
Collapse
|
18
|
Sarate N, Sonawane R, Pai V, Karatela S, Mulkalwar A. Iron Deficiency: A Silent Threat in Patients With Heart Failure With Reduced Ejection Fraction. Cureus 2024; 16:e53542. [PMID: 38445122 PMCID: PMC10912968 DOI: 10.7759/cureus.53542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2024] [Indexed: 03/07/2024] Open
Abstract
Background Iron deficiency is a prevalent and clinically significant comorbidity in patients with heart failure with reduced ejection fraction (HFrEF). Despite its high prevalence, its impact on clinical outcomes, mortality, and various physiological parameters remains a subject of ongoing investigation. The findings of this study are anticipated to contribute valuable insights into the management and prognosis of patients with HFrEF, potentially informing future interventions and improving patient outcomes. This study aimed to assess the clinical profile of iron deficiency and its implications on morbidity and mortality in patients with HFrEF. Methodology A prospective cohort study was conducted at King Edward Memorial Hospital, India, involving 371 patients with HFrEF. Participants underwent comprehensive clinical and laboratory assessments, evaluating iron deficiency with signs, symptoms, comorbidities, dyspnea, elevated jugular venous pressure (JVP), past medical history, and various hematological and biochemical parameters. Results Overall, 50% of HFrEF participants were iron deficient (n = 185), of whom 80% (n = 148) had anemia against 43% (n = 81) anemics in iron repletes (n = 186). Of the 185 iron-deficient patients, 44 (11.86%) had absolute iron deficiency and 141 (38%) had functional iron deficiency. Iron deficiency significantly correlated with increased mortality in HFrEF patients (χ2 (1, N = 371) = 3.88, p = 0.048). A large positive correlation was observed between absolute iron deficiency and dyspnea severity (r2 = 0.949, p = 0.026). Statistically significant differences were found in hemoglobin (anemia), serum iron, serum ferritin, total iron-binding capacity, and transferrin saturation between iron-deficient and iron-replete patients (p < 0.05). However, no statistically significant difference in left ventricular ejection fraction between iron-deficient and replete patients was noted. Conclusions Iron deficiency emerges as more than a mere comorbidity in heart failure, becoming a prognostic factor with multifaceted outcomes. Its impact extends beyond cardiovascular consequences, encompassing adverse manifestations such as anemia, ascites, edema, dyspnea, elevated JVP, and a heightened risk of mortality. This intricate interplay positions iron deficiency as a critical determinant, significantly influencing the clinical trajectory and outcomes for patients with HFrEF.
Collapse
Affiliation(s)
- Nitin Sarate
- Department of Medicine, Seth Gordhandas Sunderdas Medical College and King Edward Memorial Hospital, Mumbai, IND
| | - Rahul Sonawane
- Department of Medicine, Seth Gordhandas Sunderdas Medical College and King Edward Memorial Hospital, Mumbai, IND
| | - Vinayak Pai
- Department of Medicine, Seth Gordhandas Sunderdas Medical College and King Edward Memorial Hospital, Mumbai, IND
| | - Shifa Karatela
- Department of Medicine, Medical College Baroda and Sir Sayajirao General Hospital, Vadodara, IND
| | - Alhad Mulkalwar
- Department of Pharmacology, Dr. D. Y. Patil Medical College, Hospital & Research Centre, Pune, IND
| |
Collapse
|
19
|
Yang YS, Wang FN, Zhang YP, Yang F, Xue JJ. Novel Bis-pyrazoline Fluorescent Probe for Cu 2+ and Fe 3+ Detection and Application in Cell Imaging. J Fluoresc 2024; 34:159-167. [PMID: 37166610 DOI: 10.1007/s10895-023-03213-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/16/2023] [Indexed: 05/12/2023]
Abstract
A fluorescent probe Y((1,1'-([1,1'-biphenyl]-4,4'-diylbis(3-(2-hydroxyphenyl)-4,5-dihydro-1H-pyrazole-5,1-diyl)) bis(ethan-1-one))) was designed and synthesized, which could be used to Cu2+ and Fe3+ sensors. Through the study of optical properties, the probe Y shows good selectivity and sensitivity to Cu2+ and Fe3+ in aqueous tetrahydrofuran solution [10.0 mM HEPES, pH 7.4, THF-H2O = 9:1(v/v)] with has excellent anti-interference performance, and its detection limits were 0.931 uΜ for Cu2+ and 0.401uΜ for Fe3+. The coordination mechanism of probe Y with Cu2+ and Fe3+ was speculated and verified at DFT level and HRNM. By Hela cytotoxicity and imaging tests, probe Y not only has good biocompatibility, but also can be used for sensing Cu2+ in cells.
Collapse
Affiliation(s)
- Yun-Shang Yang
- School of Petrochemical Engineering & Key Laboratory of Low Carbon Energy and Chemical Engineering of Gansu, Lanzhou University of Technology, Lanzhou, 730050, China.
| | - Fu-Nian Wang
- School of Petrochemical Engineering & Key Laboratory of Low Carbon Energy and Chemical Engineering of Gansu, Lanzhou University of Technology, Lanzhou, 730050, China
| | - Ying-Peng Zhang
- School of Petrochemical Engineering & Key Laboratory of Low Carbon Energy and Chemical Engineering of Gansu, Lanzhou University of Technology, Lanzhou, 730050, China.
| | - Feng Yang
- School of Petrochemical Engineering & Key Laboratory of Low Carbon Energy and Chemical Engineering of Gansu, Lanzhou University of Technology, Lanzhou, 730050, China
| | - Ji-Jun Xue
- State Key Laboratory of Applied Organic Chemistry & College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| |
Collapse
|
20
|
Cheng X, Caruso C, Lam WA, Graham MD. Marginated aberrant red blood cells induce pathologic vascular stress fluctuations in a computational model of hematologic disorders. SCIENCE ADVANCES 2023; 9:eadj6423. [PMID: 38019922 PMCID: PMC10686556 DOI: 10.1126/sciadv.adj6423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023]
Abstract
Red blood cell (RBC) disorders such as sickle cell disease affect billions worldwide. While much attention focuses on altered properties of aberrant RBCs and corresponding hemodynamic changes, RBC disorders are also associated with vascular dysfunction, whose origin remains unclear and which provoke severe consequences including stroke. Little research has explored whether biophysical alterations of RBCs affect vascular function. We use a detailed computational model of blood that enables characterization of cell distributions and vascular stresses in blood disorders and compare simulation results with experimental observations. Aberrant RBCs, with their smaller size and higher stiffness, concentrate near vessel walls (marginate) because of contrasts in physical properties relative to normal cells. In a curved channel exemplifying the geometric complexity of the microcirculation, these cells distribute heterogeneously, indicating the importance of geometry. Marginated cells generate large transient stress fluctuations on vessel walls, indicating a mechanism for the observed vascular inflammation.
Collapse
Affiliation(s)
- Xiaopo Cheng
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Christina Caruso
- Aflac Cancer and Blood Disorders Center of Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - Wilbur A. Lam
- Aflac Cancer and Blood Disorders Center of Children’s Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30307, USA
- Wallace H. Coulter Department of Biomedical Engineering. Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Michael D. Graham
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
21
|
Ariya M, Sharafi M, Afrashteh S. Association between latent profile of dietary intake and cardiovascular diseases (CVDs): Results from Fasa Adults Cohort Study (FACS). Sci Rep 2023; 13:17749. [PMID: 37853042 PMCID: PMC10584898 DOI: 10.1038/s41598-023-44766-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 10/12/2023] [Indexed: 10/20/2023] Open
Abstract
Cardiovascular diseases (CVDs) have been among the most significant non-communicable diseases. Dietary risks account for the most cause of CVDs mortalities. Evaluating overall dietary patterns (through the Latent profile of dietary intake) can provide a more accurate prediction regarding the prevalence of CVDs. The present cross-sectional study aimed to investigate the relationship between the latent profile of dietary intake and CVDs prevalence. The population of the Fasa Adults Cohort Study was employed to gather the data (n = 8319). A modified FFQ was employed to assess eating behaviors. Minerals, as well as the energy intake and total fiber, were measured using Nutritionist IV software (version 7.0). To estimate the prevalence of CVDs, accurate records of patients' histories were made. Individuals were clustered according to their dietary intake using latent profile analysis. The mean age was 48.75 ± 9.59 years, and 53.28% (4430) were women. 63.9% of participants with low Socioeconomic Status (SES) were in the low-intake profile (P < 0.001), and high SES increases the odds of being in the high-intake profile (ORhigh/low = 2.87, 95% CI 2.55-3.24). The low-intake group had the lowest amount of physical activity (Met) (P < 0.001). The result of multivariate logistic regression revealed that categorized in the low-intake group significantly increased the development of CVDs (OR = 1.32, 95% CI 1.07-1.63, P = 0.010). The mean micronutrients and total fiber, in individuals with a low intake profile, were significantly lower than other groups (P < 0.001). Overall, we estimated that a low intake of all food groups increases the odds of developing CVDs significantly.
Collapse
Affiliation(s)
- Mohammad Ariya
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Nutrition, Fasa University of Medical Sciences, Fasa, Iran
| | - Mehdi Sharafi
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran.
- Social Determinants in Health Promotion Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Sima Afrashteh
- Department of Biostatistics and Epidemiology, Faculty of Health and Nutrition, Bushehr University of Medical Sciences, Bushehr, Iran
| |
Collapse
|
22
|
Yera HO, Khan A, Akinlade OM, Champsi A, Glouzon VNJ, Spencer C. Improving the Outcome of Patients With Heart Failure: Assessment of Iron Deficiency and Intravenous Iron Replacement. Cureus 2023; 15:e47027. [PMID: 37965394 PMCID: PMC10642709 DOI: 10.7759/cureus.47027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2023] [Indexed: 11/16/2023] Open
Abstract
Background Iron deficiency (ID) has been shown to be a significant co-morbidity in patients with heart failure (HF), independent of their anaemia status. Correction of ID has been shown to improve quality of life, recurrent heart failure hospitalizations and morbidity. A quality improvement project was designed to improve the assessment and treatment of iron deficiency in HF patients in our tertiary care centre. Methods and results An initial baseline dataset was collected, followed by two cycles of interventions to help improve the care of HF patients admitted to our hospital over a two-month period. The Plan-Do-Study-Act (PDSA) cycle approach was applied, with the first intervention involving raising awareness of the importance and need to assess the iron status of HF patients through education provided to doctors, nurses and patients. Furthermore, information leaflets were produced and disseminated across the medical wards and through social media forums. The post-intervention datasets were collected and compared to the baseline outcomes. Baseline data showed that only four (20%) of heart failure patients had their iron status checked. Following the interventions, screening for ID increased to 80% (16), of which 85% (11) of those who identified as iron deficient received intravenous iron replacement. Conclusion The project was successful in improving the practice of screening for iron deficiency and intravenous replacement of iron in patients with HF.
Collapse
Affiliation(s)
- Hassan O Yera
- Internal Medicine, The Shrewsbury and Telford Hospital NHS Trust, Telford, GBR
| | - Ahsan Khan
- Cardiology, Heart and Lung Centre, New Cross Hospital, Wolverhampton, GBR
| | | | - Asgher Champsi
- Cardiology, Heart and Lung Centre, New Cross Hospital, Wolverhampton, GBR
| | - Van Nam J Glouzon
- Internal Medicine, Heart and Lung Centre, New Cross Hospital, Wolverhampton, GBR
| | - Charles Spencer
- Cardiology, Heart and Lung Centre, New Cross Hospital, Wolverhampton, GBR
| |
Collapse
|
23
|
Skalny AV, Korobeinikova TV, Zabroda NN, Chang JS, Chao JCJ, Aschner M, Paoliello MMB, Burtseva TI, Tinkov AA. Interactive Effects of Obesity and Hypertension on Patterns of Hair Essential Trace Element and Mineral Content in Adult Women. Biol Trace Elem Res 2023; 201:4677-4687. [PMID: 36648598 DOI: 10.1007/s12011-023-03561-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023]
Abstract
The objective of the present study was to evaluate potential similar patterns and interactive effects of obesity and hypertension on hair essential trace element and mineral content in adult women. In this cross-sectional study, a total of 607 adult women divided into controls (n = 101), groups with obesity without hypertension (n = 199), hypertension without obesity (n = 143), and both obesity and hypertension (n = 164) were included in the study. Assessment of hair mineral and trace element levels was performed by inductively-coupled plasma mass-spectrometry. Hair Ca, Mg, Co, and Mn levels in women with obesity, hypertension, and both diseases were significantly lower, compared to controls. Hair Mg levels in women with obesity and hypertension were significantly lower, whereas hair Na and K were found to be higher when compared to other groups. Hair Fe and V content in obese patients was lower than in other groups. Obesity was associated with lower hair Cu levels, whereas patients with hypertension had higher hair Cu content. Hypertension was also associated with higher hair Cr and Se content irrespective of body weight. Hair Zn levels in obese women with and without hypertension were significantly lower than those in healthy controls and normal-weight women with hypertension. In multiple regression models hair Mg was considered as a significant negative predictor of both systolic and diastolic blood pressure values. The observed alterations in hair trace element and mineral content provide an additional link between obesity and hypertension, although further detailed studies are required.
Collapse
Affiliation(s)
- Anatoly V Skalny
- Center for Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia
- Yaroslavl State University, Yaroslavl, 150003, Russia
| | - Tatiana V Korobeinikova
- Center for Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia
| | - Nadezhda N Zabroda
- Center for Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia
| | - Jung-Su Chang
- Taipei Medical University, Taipei, 11031, Taiwan
- Nutrition Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan
| | - Jane C-J Chao
- Taipei Medical University, Taipei, 11031, Taiwan
- Nutrition Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Monica M B Paoliello
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | | | - Alexey A Tinkov
- Center for Bioelementology and Human Ecology, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia.
- Yaroslavl State University, Yaroslavl, 150003, Russia.
| |
Collapse
|
24
|
Dai Y, Ignatyeva N, Xu H, Wali R, Toischer K, Brandenburg S, Lenz C, Pronto J, Fakuade FE, Sossalla S, Zeisberg EM, Janshoff A, Kutschka I, Voigt N, Urlaub H, Rasmussen TB, Mogensen J, Lehnart SE, Hasenfuss G, Ebert A. An Alternative Mechanism of Subcellular Iron Uptake Deficiency in Cardiomyocytes. Circ Res 2023; 133:e19-e46. [PMID: 37313752 DOI: 10.1161/circresaha.122.321157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 05/26/2023] [Indexed: 06/15/2023]
Abstract
BACKGROUND Systemic defects in intestinal iron absorption, circulation, and retention cause iron deficiency in 50% of patients with heart failure. Defective subcellular iron uptake mechanisms that are independent of systemic absorption are incompletely understood. The main intracellular route for iron uptake in cardiomyocytes is clathrin-mediated endocytosis. METHODS We investigated subcellular iron uptake mechanisms in patient-derived and CRISPR/Cas-edited induced pluripotent stem cell-derived cardiomyocytes as well as patient-derived heart tissue. We used an integrated platform of DIA-MA (mass spectrometry data-independent acquisition)-based proteomics and signaling pathway interrogation. We employed a genetic induced pluripotent stem cell model of 2 inherited mutations (TnT [troponin T]-R141W and TPM1 [tropomyosin 1]-L185F) that lead to dilated cardiomyopathy (DCM), a frequent cause of heart failure, to study the underlying molecular dysfunctions of DCM mutations. RESULTS We identified a druggable molecular pathomechanism of impaired subcellular iron deficiency that is independent of systemic iron metabolism. Clathrin-mediated endocytosis defects as well as impaired endosome distribution and cargo transfer were identified as a basis for subcellular iron deficiency in DCM-induced pluripotent stem cell-derived cardiomyocytes. The clathrin-mediated endocytosis defects were also confirmed in the hearts of patients with DCM with end-stage heart failure. Correction of the TPM1-L185F mutation in DCM patient-derived induced pluripotent stem cells, treatment with a peptide, Rho activator II, or iron supplementation rescued the molecular disease pathway and recovered contractility. Phenocopying the effects of the TPM1-L185F mutation into WT induced pluripotent stem cell-derived cardiomyocytes could be ameliorated by iron supplementation. CONCLUSIONS Our findings suggest that impaired endocytosis and cargo transport resulting in subcellular iron deficiency could be a relevant pathomechanism for patients with DCM carrying inherited mutations. Insight into this molecular mechanism may contribute to the development of treatment strategies and risk management in heart failure.
Collapse
Affiliation(s)
- Yuanyuan Dai
- Heart Research Center Goettingen, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University of Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., S.S., E.M.Z., S.E.L., G.H., A.E.)
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
| | - Nadezda Ignatyeva
- Heart Research Center Goettingen, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University of Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., S.S., E.M.Z., S.E.L., G.H., A.E.)
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
| | - Hang Xu
- Heart Research Center Goettingen, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University of Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., S.S., E.M.Z., S.E.L., G.H., A.E.)
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
| | - Ruheen Wali
- Heart Research Center Goettingen, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University of Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., S.S., E.M.Z., S.E.L., G.H., A.E.)
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
| | - Karl Toischer
- Heart Research Center Goettingen, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University of Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., S.S., E.M.Z., S.E.L., G.H., A.E.)
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
- Heart Center, Clinic for Cardiology and Pneumology, University Medical Center Goettingen (K.T., S.B., S.S., G.H.), University of Goettingen, Germany
| | - Sören Brandenburg
- Heart Research Center Goettingen, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University of Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., S.S., E.M.Z., S.E.L., G.H., A.E.)
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
- Heart Center, Clinic for Cardiology and Pneumology, University Medical Center Goettingen (K.T., S.B., S.S., G.H.), University of Goettingen, Germany
| | - Christof Lenz
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
- Department of Clinical Chemistry, University Medical Center Goettingen, (C.L., H.U.), University of Goettingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC; C.L., F.E.F., N.V., S.E.L.), University of Goettingen, Germany
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Goettingen (C.L., H.U.)
| | - Julius Pronto
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, (J.P., F.E.F., N.V.), University of Goettingen, Germany
| | - Funsho E Fakuade
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, (J.P., F.E.F., N.V.), University of Goettingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC; C.L., F.E.F., N.V., S.E.L.), University of Goettingen, Germany
| | - Samuel Sossalla
- Heart Research Center Goettingen, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University of Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., S.S., E.M.Z., S.E.L., G.H., A.E.)
- Heart Center, Clinic for Cardiology and Pneumology, University Medical Center Goettingen (K.T., S.B., S.S., G.H.), University of Goettingen, Germany
- Department for Internal Medicine II, University Medical Center Regensburg (S.S.)
| | - Elisabeth M Zeisberg
- Heart Research Center Goettingen, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University of Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., S.S., E.M.Z., S.E.L., G.H., A.E.)
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
| | - Andreas Janshoff
- Institute for Physical Chemistry (A.J.), University of Goettingen, Germany
| | - Ingo Kutschka
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
- Department of Thoracic and Cardiovascular Surgery, University Medical Center Göttingen (I.K.)
| | - Niels Voigt
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, (J.P., F.E.F., N.V.), University of Goettingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC; C.L., F.E.F., N.V., S.E.L.), University of Goettingen, Germany
| | - Henning Urlaub
- Department of Clinical Chemistry, University Medical Center Goettingen, (C.L., H.U.), University of Goettingen, Germany
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Goettingen (C.L., H.U.)
| | | | - Jens Mogensen
- Department of Cardiology, Aalborg University Hospital, Denmark (J.M.)
| | - Stephan E Lehnart
- Heart Research Center Goettingen, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University of Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., S.S., E.M.Z., S.E.L., G.H., A.E.)
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC; C.L., F.E.F., N.V., S.E.L.), University of Goettingen, Germany
| | - Gerd Hasenfuss
- Heart Research Center Goettingen, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University of Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., S.S., E.M.Z., S.E.L., G.H., A.E.)
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
- Heart Center, Clinic for Cardiology and Pneumology, University Medical Center Goettingen (K.T., S.B., S.S., G.H.), University of Goettingen, Germany
| | - Antje Ebert
- Heart Research Center Goettingen, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University of Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., S.S., E.M.Z., S.E.L., G.H., A.E.)
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
| |
Collapse
|
25
|
Vukadinović D, Abdin A, Emrich I, Schulze PC, von Haehling S, Böhm M. Efficacy and safety of intravenous iron repletion in patients with heart failure: a systematic review and meta-analysis. Clin Res Cardiol 2023; 112:954-966. [PMID: 37074386 PMCID: PMC10116902 DOI: 10.1007/s00392-023-02207-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 03/24/2023] [Indexed: 04/20/2023]
Abstract
INTRODUCTION AFFIRM-AHF and IRONMAN demonstrated lower rates of the combined endpoint recurrent heart failure (HF) hospitalizations and cardiovascular death (CVD) using intravenous (IV) ferric carboxymaltose (FCM) and ferric derisomaltose (FDI), respectively in patients with HF and iron deficiency (ID) utilizing prespecified COVID-19 analyses. MATERIAL AND METHODS We meta-analyzed efficacy, between trial heterogeneity and data robustness for the primary endpoint and CVD in AFFIRM-AHF and IRONMAN. As sensitivity analysis, we analyzed data from all eligible exploratory trials investigating FCM/FDI in HF. RESULTS FCM/FDI reduced the primary endpoint (RR = 0.81, 95% CI 0.69-0.95, p = 0.01, I2 = 0%), with the number needed to treat (NNT) being 7. Power was 73% and findings were robust with fragility index (FI) of 94 and fragility quotient (FQ) of 0.041. Effects of FCM/FDI were neutral concerning CVD (OR = 0.88, 95% CI 0.71-1.09, p = 0.24, I2 = 0%). Power was 21% while findings were fragile with reverse FI of 14 and reversed FQ of 0.006. The sensitivity analysis from all eligible trials (n = 3258) confirmed positive effects of FCM/FDI on the primary endpoint (RR = 0.77, 95% CI 0.66-0.90, p = 0.0008, I2 = 0%), with NNT being 6. Power was 91% while findings were robust (FI of 147 and FQ of 0.045). Effect on CVD was neutral (RR = 0.87, 95% CI 0.71-1.07, p = 0.18, I2 = 0%). Power was 10% while findings were fragile (reverse FI of 7 and reverse FQ of 0.002). Rate of infections (OR = 0.85, 95% CI 0.71-1.02, p = 0.09, I2 = 0%), vascular disorder (OR = 0.84, 95% CI 0.57-1.25, p = 0.34, I2 = 0%) and general or injection-site related disorders (OR = 1.39, 95% CI 0.88-1.29, p = 0.16, I2 = 30%) were comparable between groups. There was no relevant heterogeneity (I2 > 50%) between the trials for any of the analyzed outcomes. CONCLUSIONS Use of FCM/FDI is safe and reduces the composite of recurrent HF hospitalizations and CVD, while effects on CVD alone are based on available level of data indeterminate. Findings concerning composite outcomes exhibit a high level of robustness without heterogeneity between trials with FCM and FDI.
Collapse
Affiliation(s)
- Davor Vukadinović
- Klinik Für Innere Medizin III, Kardiologie, Angiologie Und Internistische Intensivmedizin, Universitätsklinikum Des Saarlandes, Universität Des Saarlandes, Kirrberger Strasse, 66421, Homburg/Saar, Germany.
| | - Amr Abdin
- Klinik Für Innere Medizin III, Kardiologie, Angiologie Und Internistische Intensivmedizin, Universitätsklinikum Des Saarlandes, Universität Des Saarlandes, Kirrberger Strasse, 66421, Homburg/Saar, Germany
| | - Insa Emrich
- Klinik Für Innere Medizin III, Kardiologie, Angiologie Und Internistische Intensivmedizin, Universitätsklinikum Des Saarlandes, Universität Des Saarlandes, Kirrberger Strasse, 66421, Homburg/Saar, Germany
| | - P Christian Schulze
- Department of Internal Medicine I, Division of Cardiology, Angiology and Intensive Medical Care, Friedrich-Schiller-University, University Hospital Jena, Jena, Germany
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, Georg-August-University, University Medical Centre, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site, Göttingen, Germany
| | - Michael Böhm
- Klinik Für Innere Medizin III, Kardiologie, Angiologie Und Internistische Intensivmedizin, Universitätsklinikum Des Saarlandes, Universität Des Saarlandes, Kirrberger Strasse, 66421, Homburg/Saar, Germany
| |
Collapse
|
26
|
Liu F, Liu Y, Xu S, Wang Q, Xu F, Liu Y. Mendelian randomization study reveals a causal relationship between serum iron status and coronary heart disease and related cardiovascular diseases. Front Cardiovasc Med 2023; 10:1152201. [PMID: 37383700 PMCID: PMC10294586 DOI: 10.3389/fcvm.2023.1152201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/29/2023] [Indexed: 06/30/2023] Open
Abstract
Background Growing observational studies have shown that abnormal systemic iron status is associated with Coronary heart disease (CHD). However, these results from observational studies was not entirely consistent.It remains unclear whether this relationship represents causality.It is necessary to explore the causal relationship between iron status and CHD and related cardiovascular diseases (CVD). Objective We aimed to investigate the potential casual relationship between serum iron status and CHD and related CVD using a two-sample Mendelian randomization (MR) approach. Methods Genetic statistics for single nucleotide polymorphisms (SNPs) between four iron status parameters were identified in a large-scale genome-wide association study (GWAS) conducted by the Iron Status Genetics organization. Three independent single nucleotide polymorphisms (SNPs) (rs1800562, rs1799945, and rs855791) aligned with four iron status biomarkers were used as instrumental variables. CHD and related CVD genetic statistics We used publicly available summary-level GWAS data. Five different MR methods random effects inverse variance weighting (IVW), MR Egger, weighted median, weighted mode, and Wald ratio were used to explore the causal relationship between serum iron status and CHD and related CVD. Results In the MR analysis, we found that the causal effect of serum iron (OR = 0.995, 95% CI = 0.992-0.998, p = 0.002) was negatively associated with the odds of coronary atherosclerosis (AS). Transferrin saturation (TS) (OR = 0.885, 95% CI = 0.797-0.982, p = 0.02) was negatively associated with the odds of Myocardial infarction (MI). Conclusion This MR analysis provides evidence for a causal relationship between whole-body iron status and CHD development. Our study suggests that a high iron status may be associated with a reduced risk of developing CHD.
Collapse
Affiliation(s)
- Fenglan Liu
- The Second Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yanfei Liu
- The Second Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shihan Xu
- The Second Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qing Wang
- The Second Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengqin Xu
- The Second Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yue Liu
- National Clinical Research Center for TCM Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
27
|
Hamed M, Elseidy SA, Ahmed A, Thakker R, Mansoor H, Khalili H, Mohsen A, Mamas MA, Banerjee S, Kumbhani DJ, Elgendy IY, Elbadawi A. Intravenous iron therapy among patients with heart failure and iron deficiency: An updated meta-analysis of randomized controlled trials. Heliyon 2023; 9:e17245. [PMID: 37383191 PMCID: PMC10293724 DOI: 10.1016/j.heliyon.2023.e17245] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/10/2023] [Accepted: 06/12/2023] [Indexed: 06/30/2023] Open
Abstract
Background Randomized clinical trials (RCTs) evaluating the role of intravenous (IV) iron administration in patients with heart failure (HF) and iron deficiency (ID) have yielded inconsistent results. Methods Electronic search of MEDLINE, EMBASE and OVID databases was performed until November 2022 for RCTs that evaluated the role of IV iron administration in patients with HF and ID. The main study outcomes were the composite of HF hospitalization or cardiovascular mortality, and individual outcome of HF hospitalization. Summary estimates were evaluated using random effects model. Results The final analysis included 12 RCTs with 3,492 patients (1,831 patients in the IV iron group and 1,661 patients in the control group). The mean follow-up was 8.3 months. IV iron was associated with a lower incidence in the composite of HF hospitalization or cardiovascular mortality (31.9% vs. 45.3%; relative risk [RR] 0.72; 95% confidence interval [CI] 0.59-0.88) and individual outcome of HF hospitalization (28.4% vs. 42.2; RR 0.69; 95% CI 0.57-0.85). There was no significant difference between both groups in cardiovascular mortality (RR 0.88; 95% CI 0.75-1.04) and all-cause mortality (RR 0.95; 95% CI 0.83-1.09). IV iron was associated with lower New York Heart Association class and higher left ventricular ejection fraction (LVEF). Meta-regression analyses showed no effect modification for the main outcomes based on age, hemoglobin level, ferritin level or LVEF. Conclusion Among patients with HF and ID, IV iron administration was associated with reduction in the composite of HF hospitalization or cardiovascular mortality and driven by a reduction in HF hospitalization.
Collapse
Affiliation(s)
- Mohamed Hamed
- Department of Internal Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Sheref A. Elseidy
- Department of Internal Medicine, Rochester General Hospital, Rochester, NY, USA
| | - Asmaa Ahmed
- Department of Internal Medicine, Rochester General Hospital, Rochester, NY, USA
| | - Ravi Thakker
- Division of Cardiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Hend Mansoor
- Department of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Houman Khalili
- Division of Cardiology, Memorial Healthcare System, Hollywood, FL, USA
| | - Amr Mohsen
- Division of Cardiology, Loma Linda University, Loma Linda, CA, USA
| | - Mamas A. Mamas
- Keele Cardiovascular Research Group, Keele University, Keele, UK
| | - Subhash Banerjee
- Division of Cardiology, Baylor University Medical Center, Dallas, TX, USA
| | - Dharam J. Kumbhani
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Islam Y. Elgendy
- Division of Cardiovascular Medicine, Gill Heart Institute, University of Kentucky, Lexington, KY, USA
| | - Ayman Elbadawi
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
28
|
Xi X, Wu Q, Wang X, Sun X, Yu G, Jiang L, Wu H, Zhang L. The association between iron metabolism with the change of blood pressure and risk of hypertension: A large cross-sectional study. J Trace Elem Med Biol 2023; 79:127193. [PMID: 37269648 DOI: 10.1016/j.jtemb.2023.127193] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 05/01/2023] [Accepted: 05/02/2023] [Indexed: 06/05/2023]
Abstract
BACKGROUND The relationship between iron metabolism and variations in blood pressure and hypertension risk is still not clear. This study aimed to determine whether iron metabolism is associated with changes in blood pressure and hypertension prevalence in the general United States population. METHODS The National Health and Nutrition Examination Survey (NAHNES) database contains data on 116876 Americans from 1999 to 2020 years. Data from the NHANES database were used to examine the relationships between iron metabolism (serum iron [SI], serum ferritin [SF], and soluble transferrin receptor [sTfR]) and changes in blood pressure and hypertension prevalence. Generalized linear models and restricted cubic spline (RCS) plot curves were used to estimate the relationship between iron metabolism and hypertension. Further, generalized additive models with smooth functions were used to identify the relationship between iron metabolism and blood pressure. Finally, a stratified subgroup analysis was performed. RESULTS A total of 6710 participants were included in our analysis. The RCS plot showed a linear relationship between SI, as well as sTfR, and hypertension prevalence. SF and hypertension prevalence were associated in a J-shape. In addition, the relationship between SI and systolic blood pressure (SBP) and diastolic blood pressure (DBP) decreased initially and then increased. A correlation between SF, SBP, and DBP first decreased, then increased, and finally decreased. A positive linear correlation existed between sTfR and SBP, but it increased and then decreased with DBP. CONCLUSION The correlation between SF and hypertension prevalence displayed a J-curve. In contrast, the correlation between SI, as well as sTfR, and hypertension risk was negative and positive, respectively.
Collapse
Affiliation(s)
- Xiaolong Xi
- Department of Critical Care Medicine, Shaoxing People's Hospital, No.568 Zhong Xing Road, Shaoxing, Zhejiang, 312000, China
| | - Qiong Wu
- Department of Cardiology, Kunshan Hospital of Traditional Chinese Medicine, Suzhou, Jiangsu, 215300, China
| | - Xiaotong Wang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Xuedong Sun
- Department of Critical Care Medicine, Shaoxing People's Hospital, No.568 Zhong Xing Road, Shaoxing, Zhejiang, 312000, China
| | - Guofeng Yu
- Department of Critical Care Medicine, Shaoxing People's Hospital, No.568 Zhong Xing Road, Shaoxing, Zhejiang, 312000, China
| | - Lixian Jiang
- Department of Cardiology, Wuxi No.2 People's Hospital, Wuxi Clinical College of Nanjing Medical University, Wuxi, Jiangsu, 214000, China
| | - Hanzhi Wu
- Department of Cardiology, Wuxi No.2 People's Hospital, Wuxi Clinical College of Nanjing Medical University, Wuxi, Jiangsu, 214000, China
| | - Lizhu Zhang
- Department of Cardiology, Jiangnan University Medical Center, No.68 Zhongshan Road, Wuxi, Jiangsu, 214001, China.
| |
Collapse
|
29
|
Campodonico J, Carulli E, Doni F, Russo GL, Junod D, Gaudenzi Asinelli M, Bonomi A, De Martino F, Vignati C, Pezzuto B, Agostoni P. Is red distribution width a valid tool to predict impaired iron transport in heart failure? Front Cardiovasc Med 2023; 10:1133233. [PMID: 37113703 PMCID: PMC10126241 DOI: 10.3389/fcvm.2023.1133233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 03/08/2023] [Indexed: 04/29/2023] Open
Abstract
Background Impaired iron transport (IIT) is a form of iron deficiency (ID) defined as transferrin saturation (TSAT) < 20% irrespective of serum ferritin levels. It is frequently observed in heart failure (HF) where it negatively affects prognosis irrespective of anaemia. Objectives In this retrospective study we searched for a surrogate biomarker of IIT. Methods We tested the predictive power of red distribution width (RDW), mean corpuscular volume (MCV) and mean corpuscular haemoglobin concentration (MCHC) to detect IIT in 797 non-anaemic HF patients. Results At ROC analysis, RDW provided the best AUC (0.6928). An RDW cut-off value of 14.2% identified patients with IIT, with positive and negative predictive values of 48 and 80%, respectively. Comparison between the true and false negative groups showed that estimated glomerular filtration rate (eGFR) was significantly higher (p = 0.0092) in the true negative vs. false negative group. Therefore, we divided the study population according to eGFR value: 109 patients with eGFR ≥ 90 ml/min/1.73 m2, 318 patients with eGFR 60-89 ml/min/1.73 m2, 308 patients with eGFR 30-59 ml/min/1.73 m2 and 62 patients with eGFR < 30 ml/min/1.73 m2. In the first group, positive and negative predictive values were 48 and 81% respectively, 51 and 85% in the second group, 48 and 73% in the third group and 43 and 67% in the fourth group. Conclusion RDW may be seen as a reliable marker to exclude IIT in non-anaemic HF patients with eGFR ≥60 ml/min/1.73 m2.
Collapse
Affiliation(s)
- Jeness Campodonico
- Centro Cardiologico Monzino, IRCCS, Milano, Italy
- Department of Clinical Science and Community Health, Cardiovascular Section, University of Milan, Milan, Italy
| | - Ermes Carulli
- Department of Clinical Science and Community Health, Cardiovascular Section, University of Milan, Milan, Italy
| | - Francesco Doni
- Department of Clinical Science and Community Health, Cardiovascular Section, University of Milan, Milan, Italy
| | - Gerardo Lo Russo
- Department of Clinical Science and Community Health, Cardiovascular Section, University of Milan, Milan, Italy
| | - Daniele Junod
- Department of Clinical Science and Community Health, Cardiovascular Section, University of Milan, Milan, Italy
| | | | - Alice Bonomi
- Centro Cardiologico Monzino, IRCCS, Milano, Italy
| | | | | | | | - Piergiuseppe Agostoni
- Centro Cardiologico Monzino, IRCCS, Milano, Italy
- Department of Clinical Science and Community Health, Cardiovascular Section, University of Milan, Milan, Italy
| |
Collapse
|
30
|
Lanser L, Plaikner M, Schroll A, Burkert FR, Seiwald S, Fauser J, Petzer V, Bellmann-Weiler R, Fritsche G, Tancevski I, Duftner C, Pircher A, Seeber A, Zoller H, Kremser C, Henninger B, Weiss G. Tissue iron distribution in patients with anemia of inflammation: Results of a pilot study. Am J Hematol 2023; 98:890-899. [PMID: 36880875 DOI: 10.1002/ajh.26909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/20/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023]
Abstract
Anemia of inflammation (AI) is frequently present in subjects with inflammatory disorders, primarily caused by inflammation-driven iron retention in macrophages. So far, only limited data on qualitative and quantitative estimates of tissue iron retention in AI patients exist. We performed a prospective cohort study analyzing splenic, hepatic, pancreatic, and cardiac iron content with MRI-based R2*-relaxometry in AI patients, including subjects with concomitant true iron deficiency (AI+IDA) hospitalized between 05/2020-01/2022. Control groups were individuals without inflammation. Spleen R2* values in AI patients with ferritin ≤200 μg/L (AI+IDA) were comparable with those found in controls. In AI patients with ferritin >200 μg/L, spleen (47.6 s-1 vs. 19.3 s-1 , p < .001) and pancreatic R2* values (32.5 s-1 vs. 24.9 s-1 , p = .011) were significantly higher compared with controls, while liver and heart R2*-values did not differ. Higher spleen R2* values were associated with higher ferritin, hepcidin, CRP, and IL-6 concentrations. Spleen R2* values normalized in AI patients after recovery (23.6 s-1 vs. 47.6 s-1 , p = .008), while no changes were found in patients with baseline AI+IDA. This is the first study investigating tissue iron distribution in patients with inflammatory anemia and AI with concomitant true iron deficiency. The results support the findings in animal models demonstrating iron retention in macrophages, which are primarily accumulating in the spleen under inflammatory conditions. MRI-related iron measurement may help to better characterize actual iron needs and to define better biomarker thresholds in the diagnosis of true ID in patients with AI. It may qualify as a useful diagnostic method to estimate the need for iron supplementation and to guide therapy.
Collapse
Affiliation(s)
- Lukas Lanser
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Michaela Plaikner
- Department of Radiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Andrea Schroll
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Stefanie Seiwald
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Josia Fauser
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Austria
| | - Verena Petzer
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Austria
| | - Rosa Bellmann-Weiler
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Gernot Fritsche
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Ivan Tancevski
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Christina Duftner
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas Pircher
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas Seeber
- Department of Internal Medicine V, Medical University of Innsbruck, Innsbruck, Austria
| | - Heinz Zoller
- Department of Internal Medicine I and Christian Doppler Laboratory on Iron and Phosphate Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christian Kremser
- Department of Radiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Benjamin Henninger
- Department of Radiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria.,Christian Doppler Laboratory for Iron Metabolism of Anemia Research, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
31
|
Jiang T, Huang J, Ran G, Song Q, Wang C. A colorimetric and fluorometric dual-mode carbon dots probe derived from phenanthroline precursor for the selective detection of Fe 2+ and Fe 3. ANAL SCI 2023; 39:325-333. [PMID: 36539607 DOI: 10.1007/s44211-022-00236-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022]
Abstract
Iron's metabolism is heavily involved in the regulation of redox balance for cell functions, however, the simultaneous monitoring of Fe2+/3+ concentration is still a great challenge due to their transitional nature in biological systems. A novel type of carbon dots (CDs) was synthesized by solvothermal treatment with 5-amino-1,10-phenanthroline (Aphen) and salicylic acid as precursors, and the resulting targeted CDs (T-CDs) were used to simultaneously detect Fe2+ and Fe3+. Comprehensive experimental characterizations revealed that the strong binding affinity of Aphen moiety to Fe2+ leads to the formation of rigid T-CDs aggregates, which causes a substantial enhancement of fluorescence intensity, whereas Fe3+ could cause the fluorescence quenching of T-CDs due to the oxidation-reduction induced electron transfer. These different fluorescence responses allow T-CDs to sensitively differentiate Fe2+ from Fe3+, and give the limit of detection (LOD) of 1.78 and 2.78 μM for Fe2+ and Fe3+, respectively. Furthermore, the Aphen dominated structure endows the T-CDs with a colorimetric response to Fe2+ with a LOD of 0.13 μM, which is very different from Fe3+. Thus, the dynamic changes of Fe2+ and Fe3+ in solution can be accurately monitored by T-CDs within the total iron concentration of 50 μM, which is probably the most sensitive dual-mode probe reported so far. In addition, this probe is successfully applied to detect the Fe2+/3+ concentration in cells, demonstrating a huge application potential in the sensing of the dynamic equilibrium of these important transition metals during the cell metabolism or stimulated process. The dynamic changes of Fe2+ and Fe3+ in solution can be accurately monitored by carbon dots based on the colorimetric and fluorometric dual-mode.
Collapse
Affiliation(s)
- Tao Jiang
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, International Joint Research Center for Photoresponsive Molecules and Materials, School of Chemical and Material Engineering, Jiangnan University, Wuxi, 214122, China
| | - Jianfeng Huang
- Department of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China
| | - Guoxia Ran
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, International Joint Research Center for Photoresponsive Molecules and Materials, School of Chemical and Material Engineering, Jiangnan University, Wuxi, 214122, China
| | - Qijun Song
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, International Joint Research Center for Photoresponsive Molecules and Materials, School of Chemical and Material Engineering, Jiangnan University, Wuxi, 214122, China
| | - Chan Wang
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, International Joint Research Center for Photoresponsive Molecules and Materials, School of Chemical and Material Engineering, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
32
|
Wang W, Wang Y, Xi H, Song Z, Zhang W, Xie L, Ma D, Qin N, Wang H. Extension Peptide of Plant Ferritin from Setaria italica Presents a Novel Fold. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:934-943. [PMID: 36576327 DOI: 10.1021/acs.jafc.2c07595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The extension peptide (EP) is the most distinctive feature of mature plant ferritin. Some EPs have exhibited serine-like protease activity, which is associated with iron uptake and release. EP forms a helix and a long loop, followed by a conserved core helical bundle. However, whether the EP adopts a stable or uniform folding pattern in all plants remains unclear. To clarify this, we investigated the crystal structure of ferritin-1 from Setaria italica (SiFer1), a type of monocotyledon. In our structure of SiFer1, the EP is different from other EPs in other solved structures of plant ferritins and consisted of a pair of β-sheets, a shorter helix, and two loops, which masks two hydrophobic pockets on the outer surface of every subunit. Furthermore, sequence analysis and structure comparison suggest that the EPs in ferritins from monocotyledons may adopt a novel fold pattern, and the conformations of EPs in ferritins are alterable among different plant species. Furthermore, additional eight iron atoms were first founded binding in the fourfold channels, demonstrating the vital function of fourfold channels in iron diffusion. In all, our structure provides new clues for understanding plant ferritins and the functions of the EP.
Collapse
Affiliation(s)
- Wenming Wang
- Institute of Molecular Science, Shanxi University, Taiyuan 030006, China
| | - Yuan Wang
- Institute of Molecular Science, Shanxi University, Taiyuan 030006, China
| | - Hongfang Xi
- Institute of Molecular Science, Shanxi University, Taiyuan 030006, China
| | - Zidan Song
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Wenlong Zhang
- College of Chinese Medicine and Food Engineering, Experimental Management Center, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Leilei Xie
- College of Chinese Medicine and Food Engineering, Experimental Management Center, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Danyang Ma
- Institute of Molecular Science, Shanxi University, Taiyuan 030006, China
| | - Nan Qin
- College of Chinese Medicine and Food Engineering, Experimental Management Center, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Hongfei Wang
- Institute of Molecular Science, Shanxi University, Taiyuan 030006, China
| |
Collapse
|
33
|
Li J, Bi Z, Wang L, Xia Y, Xie Y, Liu Y. Recent Advances in Strategies for Imaging Detection and Intervention of Cellular Senescence. Chembiochem 2023; 24:e202200364. [PMID: 36163425 DOI: 10.1002/cbic.202200364] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/14/2022] [Indexed: 01/10/2023]
Abstract
Cellular senescence is a stable cell cycle arrest state that can be triggered by a wide range of intrinsic or extrinsic stresses. Increased burden of senescent cells in various tissues is thought to contribute to aging and age-related diseases. Thus, the detection and interventions of senescent cells are critical for longevity and treatment of disease. However, the highly heterogeneous feature of senescence makes it challenging for precise detection and selective clearance of senescent cells in different age-related diseases. To address this issue, considerable efforts have been devoted to developing senescence-targeting molecular theranostic strategies, based on the potential biomarkers of cellular senescence. Herein, we review recent advances in the field of anti-senescence research and highlight the specific visualization and elimination of senescent cells. Additionally, the challenges in this emerging field are outlined.
Collapse
Affiliation(s)
- Jili Li
- Molecular Science and Biomedicine Laboratory (MBL) State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering Aptamer Engineering Center of Hunan Province Hunan University, Changsha, Hunan, 410082, P. R. China
| | - Zhengyan Bi
- Molecular Science and Biomedicine Laboratory (MBL) State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering Aptamer Engineering Center of Hunan Province Hunan University, Changsha, Hunan, 410082, P. R. China
| | - Linlin Wang
- Molecular Science and Biomedicine Laboratory (MBL) State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering Aptamer Engineering Center of Hunan Province Hunan University, Changsha, Hunan, 410082, P. R. China
| | - Yinghao Xia
- Molecular Science and Biomedicine Laboratory (MBL) State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering Aptamer Engineering Center of Hunan Province Hunan University, Changsha, Hunan, 410082, P. R. China
| | - Yuqi Xie
- Molecular Science and Biomedicine Laboratory (MBL) State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering Aptamer Engineering Center of Hunan Province Hunan University, Changsha, Hunan, 410082, P. R. China
| | - Yanlan Liu
- Molecular Science and Biomedicine Laboratory (MBL) State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering Aptamer Engineering Center of Hunan Province Hunan University, Changsha, Hunan, 410082, P. R. China
| |
Collapse
|
34
|
Wang L, Cai J, Qiao T, Li K. Ironing out macrophages in atherosclerosis. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1-10. [PMID: 36647723 PMCID: PMC10157607 DOI: 10.3724/abbs.2022196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
<p indent="0mm">The most common cause of death worldwide is atherosclerosis and related cardiovascular disorders. Macrophages are important players in the pathogenesis of atherosclerosis and perform critical functions in iron homeostasis due to recycling iron by phagocytosis of senescent red blood cells and regulating iron availability in the tissue microenvironment. With the growth of research on the "iron hypothesis" of atherosclerosis, macrophage iron has gradually become a hotspot in the refined iron hypothesis. Macrophages with the M1, M2, M(Hb), Mox, and other phenotypes have been defined with different iron-handling capabilities related to the immune function and immunometabolism of macrophages, which influence the progression of atherosclerosis. In this review, we focus on macrophage iron and its effects on the development of atherosclerosis. We also cover the contradictory discoveries and propose a possible explanation. Finally, pharmaceutical modulation of macrophage iron is discussed as a promising target for atherosclerosis therapy.</p>.
Collapse
Affiliation(s)
- Lei Wang
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Jing Cai
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Tong Qiao
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Kuanyu Li
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing 210093, China
| |
Collapse
|
35
|
Santos HO, May TL, Bueno AA. Eating more sardines instead of fish oil supplementation: Beyond omega-3 polyunsaturated fatty acids, a matrix of nutrients with cardiovascular benefits. Front Nutr 2023; 10:1107475. [PMID: 37143475 PMCID: PMC10153001 DOI: 10.3389/fnut.2023.1107475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 03/27/2023] [Indexed: 05/06/2023] Open
Abstract
Omega-3 polyunsaturated fatty acids (n-3 PUFA) play a significant role in the prevention and management of cardiometabolic diseases associated with a mild chronic pro-inflammatory background, including type 2 diabetes, hypertension, hypertriglyceridaemia, and fatty liver disease. The effects of n-3 PUFA supplements specifically, remain controversial regarding reducing risks of cardiovascular events. n-3 PUFA supplements come at a cost for the consumer and can result in polypharmacy for patients on pharmacotherapy. Sardines are a well-known, inexpensive source of n-3 PUFA and their consumption could reduce the need for n-3 PUFA supplementation. Moreover, sardines contain other cardioprotective nutrients, although further insights are crucial to translate a recommendation for sardine consumption into clinical practice. The present review discusses the matrix of nutrients contained in sardines which confer health benefits for cardiometabolism, beyond n-3 PUFA. Sardines contain calcium, potassium, magnesium, zinc, iron, taurine, arginine and other nutrients which together modulate mild inflammation and exacerbated oxidative stress observed in cardiovascular disease and in haemodynamic dysfunction. In a common serving of sardines, calcium, potassium, and magnesium are the minerals at higher amounts to elicit clinical benefits, whilst other nutrients are present in lower but valuable amounts. A pragmatic approach towards the consumption of such nutrients in the clinical scenario should be adopted to consider the dose-response relationship effects on physiological interactions. As most recommendations currently available are based on an indirect rationale of the physiological actions of the nutrients found in sardines, randomised clinical trials are warranted to expand the evidence on the benefits of sardine consumption.
Collapse
Affiliation(s)
- Heitor O. Santos
- School of Medicine, Federal University of Uberlandia (UFU), Uberlandia, Minas Gerais, Brazil
- *Correspondence: Heitor O. Santos,
| | - Theresa L. May
- School of Science and the Environment, University of Worcester, Worcester, United Kingdom
| | - Allain A. Bueno
- School of Science and the Environment, University of Worcester, Worcester, United Kingdom
| |
Collapse
|
36
|
Qin M, Shao B, Lin L, Zhang ZQ, Sheng ZG, Qin L, Shao J, Zhu BZ. Molecular mechanism of the unusual biphasic effects of the natural compound hinokitiol on iron-induced cellular DNA damage. Free Radic Biol Med 2023; 194:163-171. [PMID: 36476568 DOI: 10.1016/j.freeradbiomed.2022.11.042] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/19/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
Hinokitiol is a natural monoterpene compound found in the heartwood of cupressaceous plants that have anticancer and anti-inflammatory properties. However, few studies have focused on its effect on iron-mediated cellular DNA damage. Here we show that hinokitiol exhibited unusual biphasic effects on iron-induced DNA damage in a molar ratio (hinokitiol/iron) dependent manner in HeLa cells. Under low ratios (<3:1), hinokitiol markedly enhanced DNA damage induced by Fe(II) or Fe(II)-H2O2; However, when the ratios increased over 3:1, the DNA damage was progressively inhibited. We found that the total cytoplasmic and nuclear iron concentration increased as the ratios of hinokitiol/iron increased. However, the cellular level of labile iron pool (LIP) only increased at ratios lower than 3, and the ROS generation is consistent with LIP change. Hinokitiol was found to interact with iron to form lipophilic hinokitiol-iron complexes with different stoichiometry and redox-activity by complementary applications of various analytical methods. Taken together, we propose that the enhancement of iron-induced cellular DNA damage by hinokitiol at low ratios (<3:1) was due to formation of lipophilic and redox-active iron complexes which facilitated cellular iron uptake and •OH production, while the inhibition at ratios higher than 3 was due to formation of redox-inactive iron complexes. These new findings will help us to design more effective drugs for the prevention and treatment of a series of iron-related diseases via regulating the two critical physicochemical factors (lipophilicity and redox activity of iron complexes) by simple natural compounds with iron-chelating properties.
Collapse
Affiliation(s)
- Miao Qin
- School of Public Health, Weifang Medical University, Weifang, Shandong, 261053, China; School of Public Health, Jining Medical University, Jining, Shandong, 272013, China; State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Bo Shao
- School of Public Health, Weifang Medical University, Weifang, Shandong, 261053, China; School of Public Health, Jining Medical University, Jining, Shandong, 272013, China; State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China.
| | - Li Lin
- School of Public Health, Weifang Medical University, Weifang, Shandong, 261053, China; School of Public Health, Jining Medical University, Jining, Shandong, 272013, China
| | - Zhao-Qiang Zhang
- School of Public Health, Jining Medical University, Jining, Shandong, 272013, China
| | - Zhi-Guo Sheng
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; School of Resources and Environment, The University of Chinese Academy of Sciences, Beijing, China
| | - Li Qin
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jie Shao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; School of Resources and Environment, The University of Chinese Academy of Sciences, Beijing, China
| | - Ben-Zhan Zhu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; School of Resources and Environment, The University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
37
|
Wong SG, Vorakunthada Y, Lee-Iannotti J, Johnson KG. Sleep-related motor disorders. HANDBOOK OF CLINICAL NEUROLOGY 2023; 195:383-397. [PMID: 37562879 DOI: 10.1016/b978-0-323-98818-6.00012-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Sleep-related motor disorders include non-rapid-eye movement (NREM) sleep parasomnias, rapid-eye movement (REM), sleep parasomnias including REM sleep behavior disorder (RBD), isolated motor phenomena in sleep, and periodic limb movement disorder. Restless legs syndrome (RLS) occurs while awake but is closely related to sleep and has a circadian pattern. The pontine sublaterodorsal tegmental nucleus has an important role in aligning motor control with sleep states, and dysfunction in this region can explain motor activities including cataplexy and loss of REM atonia seen in REM sleep behavior disorder. This chapter begins with a review of motor control in sleep. The rest of the chapter summarizes the clinical presentation, epidemiology, differential and treatment of NREM, REM, and isolated sleep-related motor disorders as well as restless legs syndrome.
Collapse
Affiliation(s)
- Stephanie G Wong
- Department of Medicine, University of Arizona College of Medicine, Phoenix, AZ, United States
| | - Yuttiwat Vorakunthada
- Department of Medicine, University of Arizona College of Medicine, Phoenix, AZ, United States
| | - Joyce Lee-Iannotti
- Department of Medicine, University of Arizona College of Medicine, Phoenix, AZ, United States
| | - Karin G Johnson
- Department of Neurology, University of Massachusetts Chan School of Medicine-Baystate, Springfield, MA, United States; Institute for Healthcare Delivery and Population Science, University of Massachusetts Chan School of Medicine-Baystate, Springfield, MA, United States.
| |
Collapse
|
38
|
Bhardwaj B, Kolte D, Zhao Y, Alu MC, Zahr F, Passeri JJ, Inglessis I, Vlahakes GJ, Garcia S, Cohen DJ, Makkar RR, Kodali S, Thourani VH, Kapadia S, Palacios IF, Leon MB, Smith CR, Mack MJ, Elmariah S. Severity of and Recovery From Anemia After Transcatheter Aortic Valve Replacement: An Analysis of the PARTNER Trials and Registries. JOURNAL OF THE SOCIETY FOR CARDIOVASCULAR ANGIOGRAPHY & INTERVENTIONS 2023; 2:100531. [PMID: 39132543 PMCID: PMC11307816 DOI: 10.1016/j.jscai.2022.100531] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 08/13/2024]
Abstract
Background Anemia is associated with increased mortality in patients undergoing transcatheter aortic valve replacement (TAVR); however, data on the effect of the severity of and recovery from anemia on clinical outcomes are limited. This study examined the impact of the severity of and recovery from anemia after TAVR. Methods Patients with symptomatic, severe aortic stenosis across all surgical risk groups from the Placement of Aortic Transcatheter Valves (PARTNER) I, II, and III trials and registries who underwent TAVR were analyzed. Baseline anemia was defined as mild (hemoglobin [Hb] level ≥11.0 g/dL and <13.0 g/dL for men and ≥11.0 g/dL and <12.0 g/dL for women) and moderate-to-severe anemia (Hb level <11.0 g/dL). Recovery from anemia was defined as an increase of ≥1 g/dL in the Hb level. Patients with missing Hb information and major bleeding within 30 days were excluded. The association of the severity of and recovery from anemia with clinical outcomes was analyzed using multivariable Cox proportional hazards regression models. The primary outcome was 1-year all-cause mortality. Results The Kaplan-Meier estimate for 1-year all-cause mortality was 5.4%, 8.2%, and 14.5% in patients with no, mild, and moderate-to-severe anemia, respectively (P < .001). Recovery from anemia at 30 days occurred in 8.4% (229/2730) of all patients. Compared with those without baseline or 30-day anemia, patients with recovery from anemia had similar 1-year mortality (hazard ratio, 1.02; CI, 0.50-2.08; P = .96), whereas those without recovery from anemia had higher 1-year mortality (hazard ratio, 1.82; CI, 1.17-2.85; P = .009). Conclusions In patients undergoing TAVR, moderate-to-severe anemia is independently associated with increased 1-year mortality, and recovery from anemia after TAVR is associated with favorable outcomes. Further efforts are needed to determine whether preprocedural correction of anemia improves post-TAVR outcomes.
Collapse
Affiliation(s)
- Bhaskar Bhardwaj
- Division of Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Dhaval Kolte
- Cardiology Division, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts
| | | | - Maria C. Alu
- Cardiovascular Research Foundation, New York, New York
| | - Firas Zahr
- Division of Cardiology, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Jonathan J. Passeri
- Cardiology Division, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts
| | - Ignacio Inglessis
- Cardiology Division, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts
| | - Gus J. Vlahakes
- Cardiology Division, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts
| | - Santiago Garcia
- Minneapolis Heart Institute Foundation, Minneapolis, Minnesota
| | - David J. Cohen
- Cardiovascular Research Foundation, New York, New York
- Division of Cardiovascular Medicine, St Francis Hospital, Roslyn, New York
| | - Raj R. Makkar
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Susheel Kodali
- Cardiovascular Research Foundation, New York, New York
- Division of Cardiology, Columbia University Irving Medical Center/NewYork-Presbyterian Hospital, New York, New York
| | - Vinod H. Thourani
- Department of Cardiovascular Surgery, Piedmont Heart Institute, Atlanta, Georgia
| | - Samir Kapadia
- Department of Cardiovascular Medicine, Heart, Vascular & Thoracic Institute, Cleveland Clinic, Cleveland, Ohio
| | - Igor F. Palacios
- Cardiology Division, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts
| | - Martin B. Leon
- Cardiovascular Research Foundation, New York, New York
- Division of Cardiology, Columbia University Irving Medical Center/NewYork-Presbyterian Hospital, New York, New York
| | - Craig R. Smith
- Cardiovascular Research Foundation, New York, New York
- Division of Cardiology, Columbia University Irving Medical Center/NewYork-Presbyterian Hospital, New York, New York
| | - Michael J. Mack
- Department of Cardiothoracic Surgery, Baylor University Medical Center, Dallas, Texas
| | - Sammy Elmariah
- Cardiology Division, Department of Medicine, University of California San Francisco, San Francisco, California
| |
Collapse
|
39
|
Savarese G, von Haehling S, Butler J, Cleland JGF, Ponikowski P, Anker SD. Iron deficiency and cardiovascular disease. Eur Heart J 2023; 44:14-27. [PMID: 36282723 PMCID: PMC9805408 DOI: 10.1093/eurheartj/ehac569] [Citation(s) in RCA: 74] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 08/11/2022] [Accepted: 09/27/2022] [Indexed: 02/07/2023] Open
Abstract
Iron deficiency (ID) is common in patients with cardiovascular disease. Up to 60% of patients with coronary artery disease, and an even higher proportion of those with heart failure (HF) or pulmonary hypertension have ID; the evidence for cerebrovascular disease, aortic stenosis and atrial fibrillation is less robust. The prevalence of ID increases with the severity of cardiac and renal dysfunction and is probably more common amongst women. Insufficient dietary iron, reduced iron absorption due to increases in hepcidin secondary to the low-grade inflammation associated with atherosclerosis and congestion or reduced gastric acidity, and increased blood loss due to anti-thrombotic therapy or gastro-intestinal or renal disease may all cause ID. For older people in the general population and patients with HF with reduced ejection fraction (HFrEF), both anaemia and ID are associated with a poor prognosis; each may confer independent risk. There is growing evidence that ID is an important therapeutic target for patients with HFrEF, even if they do not have anaemia. Whether this is also true for other HF phenotypes or patients with cardiovascular disease in general is currently unknown. Randomized trials showed that intravenous ferric carboxymaltose improved symptoms, health-related quality of life and exercise capacity and reduced hospitalizations for worsening HF in patients with HFrEF and mildly reduced ejection fraction (<50%). Since ID is easy to treat and is effective for patients with HFrEF, such patients should be investigated for possible ID. This recommendation may extend to other populations in the light of evidence from future trials.
Collapse
Affiliation(s)
- Gianluigi Savarese
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Heart and Vascular Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| | - Javed Butler
- Department of Medicine, University of Mississippi School of Medicine, Jackson, MS, USA
- Baylor Scott and White Research Institute, Dallas TX, USA
| | - John G F Cleland
- Robertson Centre for Biostatistics and Clinical Trials, Institute of Health & Wellebing, University of Glasgow, Glasgow, UK
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Piotr Ponikowski
- Department of Heart Diseases, Wroclaw Medical University, Wrocław, Poland
- Centre for Heart Diseases, University Hospital, Wroclaw, Poland
| | - Stefan D Anker
- Department of Cardiology (CVK) and Berlin Institute of Health Centre for Regenerative Therapies, German Centre for Cardiovascular Research (DZHK) partner site Berlin; Charité Universitätsmedizin Berlin, Germany
| |
Collapse
|
40
|
Zhang Y, Xue N, Jia W, Chen X, Chen X, Li H, Wang B, Guo Y, Chen J, Tian H. Associations between serum soluble transferrin receptor and the prevalence of cancers. Front Oncol 2022; 12:1039930. [PMID: 36568176 PMCID: PMC9773974 DOI: 10.3389/fonc.2022.1039930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022] Open
Abstract
Background As increasing experimental evidence suggests that iron metabolism play crucial roles in cancer and non-cancer conditions, there is a lack of data on serum soluble transferrin receptor (sTfR), a promising marker representing unmet cellular iron demands, between cancer risk from epidemiological studies. Here, we aimed to evaluate the predictive value of sTfR and cancer prevalence. Materials and methods We analyzed on 5,480 adult participants from 2015 to 2018 National Health and Nutrition Examination Survey (NHANES). Spearman correlation analysis was performed to investigate the correlations between sTfR and other characteristics. To identify the associations between sTfR and the prevalence of cancers, stratified multivariable logistic regression models, subgroup and sensitivity analyses were also performed. Results In tertile analyses, participants in the highest level of sTfR were significantly associated with increased prevalence of total cancers [odds ratio (OR) = 1.53, 95% confidence interval (CI): 1.15-2.02] as compared with those at the lowest tertile. Each unit increment in ln-transformed sTfR concentration was shown to be associated with 39% increased risks of total cancers. Similar associations were found in males rather than females. Further subgroup and sensitivity analyses indicated that, in continuous and tertile analyses, sTfR was more closely associated with male- and female-specific cancers of prostate and testis (2.35: 1.03-5.40; 2.03: 1.00-4.09; respectively), and breast, cervix, ovary and uterus (1.92: 1.11-3.35; 1.66: 1.02-2.69; respectively). Conclusions Our findings suggested that elevated level of sTfR was associated with the prevalence of cancers, especially in sex-specific cancers. In order to better determine them, further research in humans will be required.
Collapse
Affiliation(s)
- Yuzhuo Zhang
- The 8th Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, China,Foshan Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Nianci Xue
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenyu Jia
- The 8th Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, China,Foshan Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Xikang Chen
- The 8th Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, China,Foshan Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Xuezhang Chen
- The 8th Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, China,Foshan Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Hongliang Li
- The 8th Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, China,Foshan Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Bin Wang
- The 8th Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, China,Foshan Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Yi Guo
- The 8th Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, China,Foshan Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Ju Chen
- The 8th Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, China,Foshan Hospital of Traditional Chinese Medicine, Guangzhou, China,*Correspondence: Huaqin Tian, ; Ju Chen,
| | - Huaqin Tian
- The 8th Clinical Medical College, Guangzhou University of Chinese Medicine, Foshan, China,Foshan Hospital of Traditional Chinese Medicine, Guangzhou, China,*Correspondence: Huaqin Tian, ; Ju Chen,
| |
Collapse
|
41
|
Lupu M, Tudor D, Filip A. Iron metabolism and cardiovascular disease: Basic to translational purviews and therapeutical approach. Rev Port Cardiol 2022; 41:1037-1046. [PMID: 36228833 DOI: 10.1016/j.repc.2021.09.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 08/09/2021] [Accepted: 09/13/2021] [Indexed: 11/15/2022] Open
Abstract
Iron interactions with the cardiovascular system were proposed about half a century ago, yet a clear-cut understanding of this micronutrient and its intricacies with acute and chronic events is still lacking. In chronic heart failure, patients with decreased iron stores appear to benefit from intravenous administration of metallic formulations, whereas acute diseases (e.g., myocardial infarction, stroke) are barely studied in randomized controlled trials in humans. However, proof-of-concept studies have indicated that the dual redox characteristics of iron could be involved in atherosclerosis, necrosis, and ferroptosis. To this end, we sought to review the currently available body of literature pertaining to these temporal profiles of heart diseases, as well as the pathophysiologic mechanism by which iron enacts, underlining key points related to treatment options.
Collapse
Affiliation(s)
- Mihai Lupu
- Iuliu Hatieganu University of Medicine and Pharmacy, Department of Physiology, Cluj-Napoca, Romania.
| | - Diana Tudor
- Iuliu Hatieganu University of Medicine and Pharmacy, Department of Physiology, Cluj-Napoca, Romania
| | - Adriana Filip
- Iuliu Hatieganu University of Medicine and Pharmacy, Department of Physiology, Cluj-Napoca, Romania
| |
Collapse
|
42
|
Wang T, Cheng J, Wang Y. Genetic support of a causal relationship between iron status and atrial fibrillation: a Mendelian randomization study. GENES & NUTRITION 2022; 17:8. [PMID: 35637428 PMCID: PMC9153204 DOI: 10.1186/s12263-022-00708-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 03/31/2022] [Indexed: 11/10/2022]
Abstract
Abstract
Background
Atrial fibrillation is the most common arrhythmia disease. Animal and observational studies have found a link between iron status and atrial fibrillation. However, the causal relationship between iron status and AF remains unclear. The purpose of this investigation was to use Mendelian randomization (MR) analysis, which has been widely applied to estimate the causal effect, to reveal whether systemic iron status was causally related to atrial fibrillation.
Methods
Single nucleotide polymorphisms (SNPs) strongly associated (P < 5 × 10−8) with four biomarkers of systemic iron status were obtained from a genome-wide association study involving 48,972 subjects conducted by the Genetics of Iron Status consortium. Summary-level data for the genetic associations with atrial fibrillation were acquired from the AFGen (Atrial Fibrillation Genetics) consortium study (including 65,446 atrial fibrillation cases and 522,744 controls). We used a two-sample MR analysis to obtain a causal estimate and further verified credibility through sensitivity analysis.
Results
Genetically instrumented serum iron [OR 1.09; 95% confidence interval (CI) 1.02–1.16; p = 0.01], ferritin [OR 1.16; 95% CI 1.02–1.33; p = 0.02], and transferrin saturation [OR 1.05; 95% CI 1.01–1.11; p = 0.01] had positive effects on atrial fibrillation. Genetically instrumented transferrin levels [OR 0.90; 95% CI 0.86–0.97; p = 0.006] were inversely correlated with atrial fibrillation.
Conclusion
In conclusion, our results strongly elucidated a causal link between genetically determined higher iron status and increased risk of atrial fibrillation. This provided new ideas for the clinical prevention and treatment of atrial fibrillation.
Collapse
|
43
|
Hof L, Old O, Steinbicker A, Meybohm P, Choorapoikayil S, Zacharowski K. Iron deficiency in cardiac surgical patients. ACTA ANAESTHESIOLOGICA BELGICA 2022. [DOI: 10.56126/73.4.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Iron is an essential element and involved in a variety of metabolic processes including oxygen transport, cellular energy production, energy metabolism of heart muscles, brain function, cell growth and cell differentiation. Preoperative anaemia is an independent risk factor for poor outcome. Recently, iron deficiency was considered only in the context of anaemia. However, negative consequences of iron deficiency in the absence of anaemia have been described for patients undergoing cardiac surgery. To date, the benefit of intravenous iron supplementation in these patients has been controversially debated. In this review, we discuss the latest progress in studies of intravenous iron supplementation in iron deficient cardiac surgical patients.
Collapse
|
44
|
Association between iron status and incident coronary artery disease: a population based-cohort study. Sci Rep 2022; 12:17490. [PMID: 36261681 PMCID: PMC9581887 DOI: 10.1038/s41598-022-22275-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 10/12/2022] [Indexed: 01/12/2023] Open
Abstract
Disorders of iron metabolism has been implicated in cardiovascular disease. However, the association of serum iron stores and coronary artery disease (CAD) remains inconsistent. Here, we investigated the associations of serum iron metabolism with the incidence of CAD, the severity of coronary artery stenosis, metabolic biomarkers, and the risk of major adverse cardiovascular event (MACE). A total of 643 CAD patients and 643 healthy controls were enrolled to assess the associations of serum iron status with the presence of CAD, the severity of CAD, and the risk of MACE. Serum iron metabolism and other metabolic markers were measured in all subjects. All statistical analyses were analyzed using SPSS22.0 software and STATA statistical package. Serum level of iron metabolism markers, including serum iron, unsaturated transferrin iron binding capacity (UIBC), Total iron binding capacity (TIBC) levels, in CAD groups was significantly lower than the control group (P < 0.001). UIBC and TIBC were negatively correlated with ferritin in both sexes. Each unit increase of serum iron and TIBC were found to have a protective role for CAD in women (iron: OR 0.794, 95% CI (0.647-0.973), TIBC: OR 0.891, 95% CI (0.795-0.999), P < 0.05). However, high ferritin level was significant associated the CAD incident in both sexes (OR 1.029, 95% CI (1.002-1.058) in men, OR 1.013, 95% CI (1.0-1.025) in women, P < 0.05). Serum iron metabolism markers exhibited no significant association with the severity of CAD. Increased serum level of iron and TIBC levels were found to have a protective role for CAD in women, but not in men. Elevated serum ferritin is independently and positively associated with CAD in men and women.
Collapse
|
45
|
Xu C, Li W, Li T, Yuan J, Pang X, Liu T, Liang B, Cheng L, Sun X, Dong S. Iron metabolism-related genes reveal predictive value of acute coronary syndrome. Front Pharmacol 2022; 13:1040845. [PMID: 36330096 PMCID: PMC9622999 DOI: 10.3389/fphar.2022.1040845] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/04/2022] [Indexed: 11/25/2022] Open
Abstract
Iron deficiency has detrimental effects in patients with acute coronary syndrome (ACS), which is a common nutritional disorder and inflammation-related disease affects up to one-third people worldwide. However, the specific role of iron metabolism in ACS progression is opaque. In this study, we construct an iron metabolism-related genes (IMRGs) based molecular signature of ACS and to identify novel iron metabolism gene markers for early stage of ACS. The IMRGs were mainly collected from Molecular Signatures Database (mSigDB) and two relevant studies. Two blood transcriptome datasets GSE61144 and GSE60993 were used for constructing the prediction model of ACS. After differential analysis, 22 IMRGs were differentially expressed and defined as DEIGs in the training set. Then, the 22 DEIGs were trained by the Elastic Net to build the prediction model. Five genes, PADI4, HLA-DQA1, LCN2, CD7, and VNN1, were determined using multiple Elastic Net calculations and retained to obtain the optimal performance. Finally, the generated model iron metabolism-related gene signature (imSig) was assessed by the validation set GSE60993 using a series of evaluation measurements. Compared with other machine learning methods, the performance of imSig using Elastic Net was superior in the validation set. Elastic Net consistently scores the higher than Lasso and Logistic regression in the validation set in terms of ROC, PRC, Sensitivity, and Specificity. The prediction model based on iron metabolism-related genes may assist in ACS early diagnosis.
Collapse
Affiliation(s)
- Cong Xu
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
| | - Wanyang Li
- School of Mathematics, South China University of Technology, Guangzhou, China
| | - Tangzhiming Li
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
| | - Jie Yuan
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
| | - Xinli Pang
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
| | - Tao Liu
- International Digital Economy Academy, Shenzhen, China
| | - Benhui Liang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, China
| | - Lixin Cheng
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
- *Correspondence: Lixin Cheng, ; Xin Sun, ; Shaohong Dong,
| | - Xin Sun
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
- *Correspondence: Lixin Cheng, ; Xin Sun, ; Shaohong Dong,
| | - Shaohong Dong
- Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
- *Correspondence: Lixin Cheng, ; Xin Sun, ; Shaohong Dong,
| |
Collapse
|
46
|
Jenkins R, Vamos EP, Mason KE, Daras K, Taylor-Robinson D, Bambra C, Millett C, Laverty AA. Local area public sector spending and nutritional anaemia hospital admissions in England: a longitudinal ecological study. BMJ Open 2022; 12:e059739. [PMID: 36175095 PMCID: PMC9528630 DOI: 10.1136/bmjopen-2021-059739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION Reductions in local government spending may have impacts on diets and health which increase the risk of hospital admissions for nutritional anaemias. Mechanisms include potential impacts of changes to local authority (LA) services (eg, housing services) on personal resources and food access, availability and provision. We therefore investigated the association between changes in LA spending and nutritional anaemia-related hospital admissions. Specifically, we address whether greater cuts to LA spending were linked to increased hospital admissions for nutritional anaemias. DESIGN Longitudinal analysis of LA panel data using Poisson fixed effects regression models. SETTING 312 LAs in England (2005-2018). MAIN EXPOSURE Total LA service expenditure per capita per year. MAIN OUTCOME Principal and total nutritional anaemia hospital admissions, for all ages and stratified by age (0-14, 15-64, 65+ years). RESULTS LA service expenditure increased by 9% between 2005 and 2009 then decreased by 20% between 2010 and 2018. Total nutritional anaemia hospital admissions increased between 2005 and 2018 from 173 to 633 admissions per 100 000 population. A £100 higher LA service spending was associated with a 1.9% decrease in total nutritional anaemia hospital admissions (adjusted incidence rate ratio (aIRR): 0.98, 95% CI: 0.96 to 0.99). When stratified by age, this was seen only in adults. A £100 higher LA service spending was associated with a 2.6% decrease in total nutritional anaemia hospital admissions in the most deprived LAs (aIRR: 0.97, 95% CI: 0.95 to 1.0). CONCLUSION Increased LA spending was associated with reduced hospital admissions for nutritional anaemia. Austerity-related reductions had the opposite effect, increasing admissions, with greater impacts in more deprived areas. This adds further evidence to the potential negative impacts of austerity policies on health and health inequalities. Among other impacts, re-investing in LA services may prevent hospital admissions associated with nutritional anaemias.
Collapse
Affiliation(s)
- Rosemary Jenkins
- Public Health Policy Evaluation Unit, School of Public Health, Imperial College London, Charing Cross Campus; The Reynolds Building; St Dunstan's Road, London W6 8RP, UK
| | - Eszter P Vamos
- Public Health Policy Evaluation Unit, School of Public Health, Imperial College London, Charing Cross Campus; The Reynolds Building; St Dunstan's Road, London W6 8RP, UK
| | - Kate E Mason
- Department of Public Health, Policy and Systems, Institute of Population Health, University of Liverpool; Waterhouse Building Block F, 2nd Floor, Liverpool L69 3BX, UK
| | - Konstantinos Daras
- Department of Public Health, Policy and Systems, Institute of Population Health, University of Liverpool; Waterhouse Building Block F, 2nd Floor, Liverpool L69 3BX, UK
| | - David Taylor-Robinson
- Department of Public Health, Policy and Systems, Institute of Population Health, University of Liverpool; Waterhouse Building Block F, 2nd Floor, Liverpool L69 3BX, UK
| | - Clare Bambra
- Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, NE1 4LE, UK
| | - Christopher Millett
- Public Health Policy Evaluation Unit, School of Public Health, Imperial College London, Charing Cross Campus; The Reynolds Building; St Dunstan's Road, London W6 8RP, UK
| | - Anthony A Laverty
- Public Health Policy Evaluation Unit, School of Public Health, Imperial College London, Charing Cross Campus; The Reynolds Building; St Dunstan's Road, London W6 8RP, UK
| |
Collapse
|
47
|
Role of Iron-Related Oxidative Stress and Mitochondrial Dysfunction in Cardiovascular Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5124553. [PMID: 36120592 PMCID: PMC9473912 DOI: 10.1155/2022/5124553] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/28/2022] [Accepted: 08/09/2022] [Indexed: 11/18/2022]
Abstract
Iron is indispensable in numerous biologic processes, but abnormal iron regulation and accumulation is related to pathological processes in cardiovascular diseases. However, the underlying mechanisms still need to be further explored. Iron plays a key role in metal-catalyzed oxidative reactions that generate reactive oxygen species (ROS), which can cause oxidative stress. As the center for oxygen and iron utilization, mitochondria are vulnerable to damage from iron-induced oxidative stress and participate in processes involved in iron-related damage in cardiovascular disease, although the mechanism remains unclear. In this review, the pathological roles of iron-related oxidative stress in cardiovascular diseases are summarized, and the potential effects and mechanisms of mitochondrial iron homeostasis and dysfunction in these diseases are especially highlighted.
Collapse
|
48
|
de Carvalho WC, de Sousa TL, de Jesus ACL, Souza LA, Dos Santos LC, da Silva SVS, Santos DCMB, de Freitas Santos Júnior A, Korn MDGA. Effect of losartan potassium, metformin hydrochloride, and simvastatin on in vitro bioaccessibility of Cu, Fe, Mn, and Zn in oat flour from Brazil. J Trace Elem Med Biol 2022; 73:127032. [PMID: 35797925 DOI: 10.1016/j.jtemb.2022.127032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 06/23/2022] [Accepted: 06/29/2022] [Indexed: 10/17/2022]
Abstract
BACKGROUND The simultaneous administration of drugs with food can compromise the bioaccessibility and absorption of nutrients. The objective of this study was to evaluate the influence of the use of losartan potassium (LP), metformin hydrochloride (MH), and simvastatin (S) on the in vitro bioaccessibility of micronutrients (Cu, Fe, Mn, and Zn) in oat flour from Bahia, Brazil. METHODS The experimental procedure consisted of the in vitro extraction of the bioaccessible fraction of Cu, Fe, Mn, and Zn in oat flour-with and without LP (50 mg), MH (500 mg), and S (20 mg)-using the unified bioaccessibility method (UBM), simulating the conditions of the gastrointestinal tract. For decomposition of the samples (oat flour and residue), a digester block with a closed system was used. To determine the total content (flour and residual fraction) and bioaccessible micronutrients, inductively coupled plasma optical emission spectrometry (ICP OES) was used. RESULTS The bioaccessible contents (µg g-1) without the addition of drugs were: Cu 5.86 ± 0.21, Fe 32.80 ± 1.32, Mn 87.90 ± 1.90, and Zn 30.33 ± 2.05, with bioaccessibility ranging from 31.5 % for Fe to 99 % for Mn. The in vitro extraction method was validated by mass balance with recovery values from 89.78 % for Cu to 101.94 % for Mn. The range of bioaccessible contents (µg g-1) were: Cu (<4.14), Fe (32.10 ± 0.20-54.10 ± 2.03), Mn (81.40 ± 0.93-93.22 ± 0.80), and Zn (<10.80-29.11 ± 2.20). The estimation of the bioaccessibility of Cu, Mn, and Zn in oat flour were compromised in the presence of LP, MH, and S (p < 0.05). CONCLUSION Chemical interactions can occur between drugs and micronutrients. Taken together, our results highlight that LP, MH, and S can interfere with the bioaccessibility of Cu, Fe, Mn, and Zn in oat flour in patients who use these drugs, suggesting its rational use in further investigations.
Collapse
Affiliation(s)
- Wellington Correia de Carvalho
- Chemistry Institute, Universidade Federal da Bahia, Salvador, Bahia 40170-115, Brazil; Instituto Federal de Educação, Ciência e Tecnologia da Bahia (IFBA), Campus Porto Seguro, Porto Seguro, Bahia 45810-000, Brazil
| | - Thaís Luz de Sousa
- Chemistry Institute, Universidade Federal da Bahia, Salvador, Bahia 40170-115, Brazil
| | | | - Laís Araújo Souza
- Chemistry Institute, Universidade Federal da Bahia, Salvador, Bahia 40170-115, Brazil
| | | | | | | | | | | |
Collapse
|
49
|
Severino P, D’Amato A, Prosperi S, Myftari V, Labbro Francia A, Önkaya M, Notari C, Papisca I, Canuti ES, Yarden Revivo M, Birtolo LI, Celli P, Galardo G, Maestrini V, d’Ettorre G, Mancone M, Fedele F. The Mutual Relationship among Cardiovascular Diseases and COVID-19: Focus on Micronutrients Imbalance. Nutrients 2022; 14:3439. [PMID: 36014944 PMCID: PMC9416353 DOI: 10.3390/nu14163439] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 12/16/2022] Open
Abstract
Micronutrients are ions and vitamins humbly required by the human body. They play a main role in several physiological mechanisms and their imbalance is strongly associated with potentially-fatal complications. Micronutrient imbalance is associated with many cardiovascular diseases, such as arrythmias, heart failure, and ischemic heart disease. It has been also observed in coronavirus disease 2019 (COVID-19), particularly in most severe patients. The relationship between cardiovascular diseases and COVID-19 is mutual: the latter triggers cardiovascular disease onset and worsening while patients with previous cardiovascular disease may develop a more severe form of COVID-19. In addition to the well-known pathophysiological mechanisms binding COVID-19 and cardiovascular diseases together, increasing importance is being given to the impact of micronutrient alterations, often present during COVID-19 and able to affect the balance responsible for a good functioning of the cardiovascular system. In particular, hypokalemia, hypomagnesemia, hyponatremia, and hypocalcemia are strongly associated with worse outcome, while vitamin A and D deficiency are associated with thromboembolic events in COVID-19. Thus, considering how frequent the cardiovascular involvement is in patients with COVID-19, and how it majorly affects their prognosis, this manuscript provides a comprehensive review on the role of micronutrient imbalance in the interconnection between COVID-19 and cardiovascular diseases.
Collapse
Affiliation(s)
- Paolo Severino
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Andrea D’Amato
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Silvia Prosperi
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Vincenzo Myftari
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Aurora Labbro Francia
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Merve Önkaya
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Claudia Notari
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Ilaria Papisca
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Elena Sofia Canuti
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Mia Yarden Revivo
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Lucia Ilaria Birtolo
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Paola Celli
- Anesthesiology and Intensive Care Unit, Sapienza University of Rome, Policlinico Umberto I, 00161 Rome, Italy
| | - Gioacchino Galardo
- Medical Emergency Unit, Sapienza University of Rome, Policlinico Umberto I, 00185 Rome, Italy
| | - Viviana Maestrini
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Gabriella d’Ettorre
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| | - Massimo Mancone
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Francesco Fedele
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| |
Collapse
|
50
|
Li Y, Luo W, Liang B. Circulating trace elements status in COVID-19 disease: A meta-analysis. Front Nutr 2022; 9:982032. [PMID: 36034929 PMCID: PMC9411985 DOI: 10.3389/fnut.2022.982032] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 07/27/2022] [Indexed: 12/18/2022] Open
Abstract
Trace elements are a group of essential metals or metalloids, which are necessary for life, and present in minute amounts. Despite substantial researches highlighting the importance of trace elements in Coronavirus disease 2019 (COVID-19) diseases, a thorough evaluation of the levels of circulating trace elements is lacking. Therefore, we conducted a systematic review and meta-analysis to evaluate the trace element status (Zn, Fe, Cu, Mg, and Se) in COVID-19 disease. We also assessed the relationship between circulating trace elements and COVID-19 disease severity and survival status during follow-up. We searched comprehensively MEDLINE, Web of Science, CNKI, and WangFang databases without language restriction, between November 1, 2019 and April 1, 2022. The search identified 1,566 preliminary references. A total of 49 studies met the eligibility criteria and were included in the review, and 42 studies were included in the final meta-analysis. Meta-analysis showed that COVID-19 patients had significantly lower circulating Zn (SMD: -0.83, 95% CI: -1.19 to -0.46, P < 0.001), Fe (SMD: -1.56, 95% CI: -2.90 to -0.21, P = 0.023), and Se (SMD: -0.75, 95% CI: -0.94 to -0.56, P < 0.001) levels than healthy controls, and circulating Zn (SMD: -0.47, 95% CI: -0.75 to -0.18, P = 0.002), Fe (SMD: -0.45, 95% CI: -0.79 to -0.12, P = 0.008), and Se (SMD: -0.27, 95% CI: -0.49 to -0.04, P = 0.020) levels were associated with the presence of severity status in COVID-19 patients. Moreover, circulating Fe levels in non-survivors were significantly lower than survivors in COVID-19 (SMD: -0.28, 95% CI: -0.44 to -0.12, P = 0.001). However, there was no significant difference in Cu and Mg levels between COVID-19 patients and controls, severity and non-severity status, and survivors and non-survivors (all P > 0.05). Taken together, COVID-19 patients displayed lower circulating levels of Zn, Fe, and Se, and their levels were associated with severity status. Moreover, circulating Fe levels may provide part of the explanation for the unfavorable survival status. Therefore, we presumed optimistically that supplements of trace elements might provide an adjutant treatment in the early stages of COVID-19. Systematic review registration [https://www.crd.york.ac.uk/prospero], identifier [CRD42022348599].
Collapse
Affiliation(s)
- Yunhui Li
- Clinical Laboratory, PLA North Military Command Region General Hospital, Shenyang, China
| | - Weihe Luo
- Department of Medical Engineering, PLA North Military Command Region General Hospital, Shenyang, China
| | - Bin Liang
- Department of Bioinformatics, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, China
| |
Collapse
|