1
|
Hou R, Shui W, Xing X, Chen Y, Shen W, Song Q, Wang J. Relationship Between TGF-β1 and Left Ventricular Geometry and Function in Patients With Essential Hypertension. JOURNAL OF CLINICAL ULTRASOUND : JCU 2025. [PMID: 40346927 DOI: 10.1002/jcu.24055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/22/2025] [Accepted: 03/31/2025] [Indexed: 05/12/2025]
Abstract
OBJECTIVE To explore the association of transforming growth factor β1 (TGF-β1) with left ventricular geometry (LVG) and left ventricular function (LVF) in cases having essential hypertension. METHODS This retrospective study of 213 cases of essential hypertension, according to echocardiogram measurements, were split into normal geometry (NG), concentric remodeling (CR), eccentric hypertrophy (EH), and concentric hypertrophy (CH) groups. General clinical data of each patient was analyzed and office blood pressure measurements were performed. Detection of blood biochemistry and serum TGF-β1 content was conducted. The association of TGF-β1 with LVG and LVF parameters was assessed. RESULTS In contrast to the NG and CR groups, the TGF-β1 concentration was higher in the EH and CH groups, and it was most pronounced in the CH group. The TGF-β1 was positively linked to E/e' (r = 0.506, p < 0.001), whereas left ventricular global longitudinal strain (GLS) (r = -0.447, p < 0.001) was negatively correlated. Moreover, serum TGF-β1 levels were independently linked to EH and CH. CONCLUSION TGF-β1 was associated with abnormal LVG and LVF in cases of essential hypertension, indicating that it may induce LVG and LVF abnormalities.
Collapse
Affiliation(s)
- Ran Hou
- Department of Ultrasound, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wen Shui
- Department of Cardiopulmonary Function Examination, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Xueqing Xing
- Department of Ultrasound, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yaodong Chen
- Department of Ultrasound, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wenqian Shen
- Department of Ultrasound, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Qingfei Song
- Department of Ultrasound, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jian Wang
- Department of Ultrasound, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
2
|
Mbadiwe NC, Ezeala-Adikaibe BA, Okwara CC, Orjioke C, Njoku PO, Okoye JU, Ekochin FC, Okereke C, Abonyi MC, Anigbo GE, Unaogu NN, Nwobodo UM. Correlates of Left Ventricular Hypertrophy in a Sub-Urban Low-Income Community in Enugu. A Community-Based Study. Niger J Clin Pract 2024; 27:1464-1472. [PMID: 40033542 DOI: 10.4103/njcp.njcp_362_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 11/21/2024] [Indexed: 03/05/2025]
Abstract
BACKGROUND The prevalence of left ventricular is on the increase, especially in the developing world probably because of high rates of cardiovascular risk factors such as hypertension. Inter-ethnic differences in the prevalence of left ventricular hypertrophy have been demonstrated. AIM The aim of this study was to determine the prevalence of left ventricular hypertrophy in a community and the associated risk factors. METHODS This was a cross-sectional descriptive study carried out in the Enugu State southeast Nigeria. Electrocardiography was conducted using the standard protocols. Left ventricular hypertrophy was diagnosed according to the Sokolow-Lyon criterion. For database management and statistical analyses, we used the SPSS version 26 (IBM Corporation, New York, NY, USA). RESULTS The electrocardiograms of 324 participants were analyzed. Left ventricular hypertrophy was diagnosed in 73 (22.5%), slightly more frequent in males (28.3%) versus 19.1% in females (P = 0.06(. About 18.4% of those with normal systolic blood pressure and 19.6% of those with normal diastolic blood had left ventricular hypertrophy. This increased to 47.4% and 38.1% in those with severe high diastolic and systolic blood pressure measurements, respectively. About 27.5% of the participants with hypertension had left ventricular hypertrophy, whereas 32.9% of newly diagnosed hypertensives had left ventricular hypertrophy. Using Spearman's correlation statistic, left ventricular hypertrophy strongly correlated with systolic hypertension r (P value) =0.12 (0.01); however, the correlation with diastolic hypertension was weak r (P value) =0.1 (0.06). There was a weak positive correlation between gender and left ventricular hypertrophy r (P value) =0.11 (0.06). CONCLUSION The prevalence of left ventricular hypertrophy was 22.5% in a community study in Enugu even among those without hypertension. Public health education on the need for regular cardiovascular checks is advocated even among those with and without hypertension.
Collapse
Affiliation(s)
- N C Mbadiwe
- Department of Medicine, University of Nigeria Teaching Hospital Enugu, Nigeria
| | - B A Ezeala-Adikaibe
- Department of Medicine, University of Nigeria Teaching Hospital Enugu, Nigeria
| | - C C Okwara
- Department of Medicine, University of Nigeria Teaching Hospital Enugu, Nigeria
| | - C Orjioke
- Department of Medicine, Enugu State University Teaching Hospital Enugu, Nigeria
| | - P O Njoku
- Department of Medicine, University of Nigeria Teaching Hospital Enugu, Nigeria
| | - J U Okoye
- Department of Medicine, University of Nigeria Teaching Hospital Enugu, Nigeria
| | - F C Ekochin
- Department of Medicine, Enugu State University Teaching Hospital Enugu, Nigeria
| | - C Okereke
- Department of Medicine, University of Nigeria Teaching Hospital Enugu, Nigeria
| | - M C Abonyi
- Department of Medicine, Enugu State University Teaching Hospital Enugu, Nigeria
| | - G E Anigbo
- Department of Medicine, Enugu State University Teaching Hospital Enugu, Nigeria
| | - N N Unaogu
- Federal Neuropsychiatric Hospital Enugu, Nigeria
| | - U M Nwobodo
- Alex Ekwueme Teaching Hospital Abakiliki, Nigeria
| |
Collapse
|
3
|
Gu L, Wan X, Liu Y, Gong Z, Huang R, Shi Y, Liu H. Mesenchymal stem cells may alleviate angiotensin II-induced myocardial fibrosis and hypertrophy by upregulating SFRS3 expression. Rev Port Cardiol 2024; 43:645-656. [PMID: 38986812 DOI: 10.1016/j.repc.2024.04.010] [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: 10/20/2023] [Revised: 03/29/2024] [Accepted: 04/08/2024] [Indexed: 07/12/2024] Open
Abstract
INTRODUCTION AND OBJECTIVES The development of cardiac fibrosis (CF) and hypertrophy (CH) can lead to heart failure. Mesenchymal stem cells (MSCs) have shown promise in treating cardiac diseases. However, the relationship between MSCs and splicing factor arginine/serine rich-3 (SFRS3) remains unclear. In this study, our objectives are to investigate the effect of MSCs on SFRS3 expression, and their impact on CF and CH. Additionally, we aim to explore the function of the overexpression of SFRS3 in angiotensin II (Ang II)-treated cardiac fibroblasts (CFBs) and cardiac myocytes (CMCs). METHODS Rat cardiac fibroblasts (rCFBs) or rat cardiac myocytes (rCMCs) were co-cultured with rat MSCs (rMSCs). The function of SFRS3 in Ang II-induced rCFBs and rCMCs was studied by overexpressing SFRS3 in these cells, both with and without the presence of rMSCs. We assessed the expression of SFRS3 and evaluated the cell cycle, proliferation and apoptosis of rCFBs and rCMCs. We also measured the levels of interleukin (IL)-β, IL-6 and tumor necrosis factor (TNF)-α and assessed the degree of fibrosis in rCFBs and hypertrophy in rCMCs. RESULTS rMSCs induced SFRS3 expression and promoted cell cycle, proliferation, while reducing apoptosis of Ang II-treated rCFBs and rCMCs. Co-culture of rMSCs with these cells also repressed cytokine production and mitigated the fibrosis of rCFBs, as well as hypertrophy of rCMCs triggered by Ang II. Overexpression of SFRS3 in the rCFBs and rCMCs yielded identical effects to rMSC co-culture. CONCLUSION MSCs may alleviate Ang II-induced cardiac fibrosis and cardiomyocyte hypertrophy by increasing SFRS3 expression in vitro.
Collapse
Affiliation(s)
- Ling Gu
- Department of Critical Care Medicine, Mindong Hospital Affiliated to Fujian Medical University, Fu'an, Fujian, China
| | - Xin Wan
- Department of Critical Care Medicine, Mindong Hospital Affiliated to Fujian Medical University, Fu'an, Fujian, China
| | - Ying Liu
- Department of Critical Care Medicine, Mindong Hospital Affiliated to Fujian Medical University, Fu'an, Fujian, China
| | - Zhenbin Gong
- Department of Critical Care Medicine, Mindong Hospital Affiliated to Fujian Medical University, Fu'an, Fujian, China
| | - Rijin Huang
- Department of Critical Care Medicine, Mindong Hospital Affiliated to Fujian Medical University, Fu'an, Fujian, China
| | - Yundi Shi
- Department of Critical Care Medicine, Mindong Hospital Affiliated to Fujian Medical University, Fu'an, Fujian, China.
| | - Huogen Liu
- Department of Critical Care Medicine, Mindong Hospital Affiliated to Fujian Medical University, Fu'an, Fujian, China.
| |
Collapse
|
4
|
Yao Q, Hu X, Bian T, Zhang Q, Xue Z, Lv Y, Ren S, Chen Y, Zhang D, Chen L. Role of KLF4 and SIAT7A interaction accelerates myocardial hypertrophy induced by Ang II. J Cell Mol Med 2024; 28:e70144. [PMID: 39431583 PMCID: PMC11492152 DOI: 10.1111/jcmm.70144] [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: 01/02/2024] [Revised: 09/05/2024] [Accepted: 10/03/2024] [Indexed: 10/22/2024] Open
Abstract
Sialylation catalysed by sialyltransferase 7A (SIAT7A) plays a role in the development of cardiac hypertrophy. However, the regulatory mechanisms upstream of SIAT7A in this context remain poorly elucidated. Previous study demonstrated that KLF4 activates the SIAT7A gene in ischemic myocardium by binding to its promoter region. Nevertheless, the potential involvement of KLF4 in regulating SIAT7A expression in Ang II-induced hypertrophic cardiomyocytes remains uncertain. This study seeks to deepen the underlying mechanisms of the KLF4 and SIAT7A interaction in the progression of Ang II-induced cardiac hypertrophy. The results showed a concurrent increase in SIAT7A and KLF4 levels in hypertrophic myocardium of essential hypertension patients and in hypertrophic cardiomyocytes stimulated by Ang II. In vitro experiments revealed that reducing KLF4 levels led to a decrease in both SIAT7A synthesis and Sialyl-Tn antigen expression, consequently inhibiting Ang II-induced cardiomyocyte hypertrophy. Intriguingly, reducing SIAT7A levels also resulted in decreased KLF4 expression and suppression cardiomyocyte hypertrophy. Consistent with this, elevating SIAT7A levels increased KLF4 expression and exacerbated cardiomyocyte hypertrophy in both in vivo and in vitro experiments. Additionally, a time-course analysis indicated that KLF4 expression preceded that of SIAT7A. Luciferase reporter assays further confirmed that modulating SIAT7A levels directly influenced the transcriptional activity of KLF4 in cardiomyocytes. In summary, KLF4 expression is upregulated in cardiomyocytes treated with Ang II, which subsequently induces the expression of SIAT7A. The elevated levels of SIAT7A, in turn, enhance the transcription of KLF4. These findings suggest a positive feedback loop between KLF4 and SIAT7A-Sialyl-Tn, ultimately promoting Ang II-induced cardiac hypertrophy.
Collapse
Affiliation(s)
- Qiying Yao
- Department of PhysiologyDalian Medical UniversityDalianLiaoningChina
| | - Xinrui Hu
- Department of PhysiologyDalian Medical UniversityDalianLiaoningChina
| | - Tiantian Bian
- Department of PhysiologyDalian Medical UniversityDalianLiaoningChina
| | - Qing Zhang
- Department of PhysiologyDalian Medical UniversityDalianLiaoningChina
| | - Zhao Xue
- Department of CardiologyThe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Yuesheng Lv
- Institute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Shupeng Ren
- Department of CardiologyThe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Yue Chen
- Department of PhysiologyDalian Medical UniversityDalianLiaoningChina
| | - Dongmei Zhang
- Department of PhysiologyDalian Medical UniversityDalianLiaoningChina
| | - Liang Chen
- Department of CardiologyThe Second Affiliated Hospital of Dalian Medical UniversityDalianChina
| |
Collapse
|
5
|
Soliman Y, Nazir A, Abuelazm M, Seri A, Mahmoud A, Abdelazeem B. The effect of intensive blood pressure lowering on left ventricular hypertrophy in patients with hypertension: a systematic review and meta-analysis of randomized controlled trials. Proc AMIA Symp 2024; 37:700-704. [PMID: 38910799 PMCID: PMC11188804 DOI: 10.1080/08998280.2024.2346409] [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: 12/06/2023] [Accepted: 04/14/2024] [Indexed: 06/25/2024] Open
Abstract
Background We conducted a comprehensive systematic review to examine the efficacy of intensive blood pressure lowering on the risk of left ventricular hypertrophy (LVH). Methods We searched PubMed, Scopus, Web of Science, Cochrane Central, and EMBASE for all relevant randomized controlled trials. The primary outcome was the incidence of left ventricular hypertrophy. We used the risk ratio (RR) and hazard ratio (HR) with a 95% confidence interval as our effect sizes. Results Four studies, comprising 20,747 patients, were included. Intensive blood pressure lowering was linked with a diminished LVH incidence (RR: 0.66, 95% CI [0.56-0.77]). We also found that intensive blood pressure lowering increased the risk of LVH regression in patients with baseline LVH (RR: 1.21, 95% CI [1.11-1.32]). Finally, intensive blood pressure lowering was linked with a reduced risk of cardiovascular disease (HR: 0.71, 95% CI [0.60-0.85]). No significant heterogeneity was seen in either outcome. Conclusion Our study suggests that intensive blood pressure lowering effectively reduces the risk of LVH and cardiovascular disease. An interactive version of our analysis can be accessed here: https://databoard.shinyapps.io/lvh_hypertophy/.
Collapse
Affiliation(s)
| | - Abubakar Nazir
- Faculty of Medicine, King Edward Medical University, Lahore, Pakistan
| | | | - Amith Seri
- Internal Medicine Department, McLaren Health Care, Flint, Michigan, USA
| | | | - Basel Abdelazeem
- Cardiology Department, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
6
|
Du BB, Shi HT, Xiao LL, Li YP, Yao R, Liang C, Tian XX, Yang LL, Kong LY, Du JQ, Zhang ZZ, Zhang YZ, Huang Z. Melanoma differentiation-associated protein 5 prevents cardiac hypertrophy via apoptosis signal-regulating kinase 1-c-Jun N-terminal kinase/p38 signaling. Int J Biol Macromol 2024; 264:130542. [PMID: 38432272 DOI: 10.1016/j.ijbiomac.2024.130542] [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: 11/30/2023] [Revised: 01/28/2024] [Accepted: 02/27/2024] [Indexed: 03/05/2024]
Abstract
Pathological cardiac hypertrophy (CH) is driven by maladaptive changes in myocardial cells in response to pressure overload or other stimuli. CH has been identified as a significant risk factor for the development of various cardiovascular diseases, ultimately resulting in heart failure. Melanoma differentiation-associated protein 5 (MDA5), encoded by interferon-induced with helicase C domain 1 (IFIH1), is a cytoplasmic sensor that primarily functions as a detector of double-stranded ribonucleic acid (dsRNA) viruses in innate immune responses; however, its role in CH pathogenesis remains unclear. Thus, the aim of this study was to examine the relationship between MDA5 and CH using cellular and animal models generated by stimulating neonatal rat cardiomyocytes with phenylephrine and by performing transverse aortic constriction on mice, respectively. MDA5 expression was upregulated in all models. MDA5 deficiency exacerbated myocardial pachynsis, fibrosis, and inflammation in vivo, whereas its overexpression hindered CH development in vitro. In terms of the underlying molecular mechanism, MDA5 inhibited CH development by promoting apoptosis signal-regulating kinase 1 (ASK1) phosphorylation, thereby suppressing c-Jun N-terminal kinase/p38 signaling pathway activation. Rescue experiments using an ASK1 activation inhibitor confirmed that ASK1 phosphorylation was essential for MDA5-mediated cell death. Thus, MDA5 protects against CH and is a potential therapeutic target.
Collapse
Affiliation(s)
- Bin-Bin Du
- Cardiovascular Hospital, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China
| | - Hui-Ting Shi
- Cardiovascular Hospital, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China
| | - Li-Li Xiao
- Cardiovascular Hospital, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China
| | - Ya-Peng Li
- Cardiovascular Hospital, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China
| | - Rui Yao
- Cardiovascular Hospital, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China
| | - Cui Liang
- Cardiovascular Hospital, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China
| | - Xiao-Xu Tian
- Cardiovascular Hospital, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China
| | - Lu-Lu Yang
- Cardiovascular Hospital, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China
| | - Ling-Yao Kong
- Cardiovascular Hospital, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China
| | - Jia-Qi Du
- Cardiovascular Hospital, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China
| | - Zhao-Zhi Zhang
- Cardiovascular Hospital, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China
| | - Yan-Zhou Zhang
- Cardiovascular Hospital, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China.
| | - Zhen Huang
- Cardiovascular Hospital, the First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
7
|
He X, Williams QA, Cantrell AC, Besanson J, Zeng H, Chen JX. TIGAR Deficiency Blunts Angiotensin-II-Induced Cardiac Hypertrophy in Mice. Int J Mol Sci 2024; 25:2433. [PMID: 38397106 PMCID: PMC10889085 DOI: 10.3390/ijms25042433] [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: 01/19/2024] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
Hypertension is the key contributor to pathological cardiac hypertrophy. Growing evidence indicates that glucose metabolism plays an essential role in cardiac hypertrophy. TP53-induced glycolysis and apoptosis regulator (TIGAR) has been shown to regulate glucose metabolism in pressure overload-induced cardiac remodeling. In the present study, we investigated the role of TIGAR in cardiac remodeling during Angiotensin II (Ang-II)-induced hypertension. Wild-type (WT) and TIGAR knockout (KO) mice were infused with Angiotensin-II (Ang-II, 1 µg/kg/min) via mini-pump for four weeks. The blood pressure was similar between the WT and TIGAR KO mice. The Ang-II infusion resulted in a similar reduction of systolic function in both groups, as evidenced by the comparable decrease in LV ejection fraction and fractional shortening. The Ang-II infusion also increased the isovolumic relaxation time and myocardial performance index to the same extent in WT and TIGAR KO mice, suggesting the development of similar diastolic dysfunction. However, the knockout of TIGAR significantly attenuated hypertension-induced cardiac hypertrophy. This was associated with higher levels of fructose 2,6-bisphosphate, PFK-1, and Glut-4 in the TIGAR KO mice. Our present study suggests that TIGAR is involved in the control of glucose metabolism and glucose transporters by Ang-II and that knockout of TIGAR attenuates the development of maladaptive cardiac hypertrophy.
Collapse
Affiliation(s)
| | | | | | | | - Heng Zeng
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; (X.H.); (Q.A.W.); (A.C.C.); (J.B.)
| | - Jian-Xiong Chen
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; (X.H.); (Q.A.W.); (A.C.C.); (J.B.)
| |
Collapse
|
8
|
Ridder A, O'Driscoll J, Khalil A, Thilaganathan B. Routine first-trimester pre-eclampsia screening and maternal left ventricular geometry. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2024; 63:75-80. [PMID: 37448160 DOI: 10.1002/uog.26306] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023]
Abstract
OBJECTIVE Pre-eclampsia (PE) is a pregnancy complication associated with premature cardiovascular disease morbidity and mortality (i.e. before 60 years of age or in the first year postpartum). PE is associated with adverse left ventricular (LV) remodeling in the peri- and postpartum periods, an independent risk factor for cardiovascular disease. This study aimed to compare LV geometry by LV mass (LVM) and LVM index (LVMI) between participants with a high vs low screening risk for preterm PE in the first trimester. METHODS This was a prospective cohort study of singleton pregnancies between 11 + 0 and 13 + 6 weeks' gestation that underwent screening for preterm PE as part of their routine first-trimester ultrasound assessment at a tertiary center in London, UK, from February 2019 until March 2020. Screening for preterm PE was performed using the Fetal Medicine Foundation algorithm. Participants with a screening risk of ≥ 1 in 50 for preterm PE were classified as high risk and those with a screening risk of ≤ 1 in 500 were classified as low risk. All participants underwent two-dimensional and M-mode transthoracic echocardiography. RESULTS A total of 128 participants in the first trimester of pregnancy were included in the analysis, with 57 (44.5%) participants screened as low risk and 71 (55.5%) participants as high risk for PE. The risk groups did not vary in maternal age and gestational age at assessment. Maternal body surface area and body mass index were significantly higher in the high-risk group (all P < 0.05). The high-risk participants were significantly more likely to be Afro-Caribbean, nulliparous and have a family history of hypertensive disease in pregnancy as well as other cardiovascular disease (all P < 0.05). In addition, mean arterial blood pressure (P < 0.001), mean heart rate (P < 0.001), median LVM (130.06 (interquartile range, 113.62-150.50) g vs 97.44 (81.68-114.16) g; P < 0.001) and mean LVMI (72.87 ± 12.2 g/m2 vs 57.54 ± 12.72 g/m2 ; P < 0.001) were significantly higher in the high-risk group. Consequently, those in the high-risk group were more likely to have abnormal LV geometry (37.1% vs 7.0%; P < 0.001). CONCLUSIONS Early echocardiographic assessment in participants at high risk of preterm PE may unmask clinically healthy individuals who are at increased risk for future cardiovascular disease. Adverse cardiac remodeling in the first trimester of pregnancy may be an indicator of decreased cardiovascular reserve and subsequent dysfunctional cardiovascular adaptation in pregnancy. © 2023 The Authors. Ultrasound in Obstetrics & Gynecology published by John Wiley & Sons Ltd on behalf of International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- A Ridder
- Fetal Medicine Unit, St George's University Hospitals NHS Foundation Trust, University of London, London, UK
- Vascular Biology Research Centre, Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| | - J O'Driscoll
- Department of Cardiology, St George's University Hospitals NHS Foundation Trust, University of London, London, UK
- School of Psychology and Life Sciences, Canterbury Christ Church University, Canterbury, UK
| | - A Khalil
- Fetal Medicine Unit, St George's University Hospitals NHS Foundation Trust, University of London, London, UK
- Vascular Biology Research Centre, Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| | - B Thilaganathan
- Fetal Medicine Unit, St George's University Hospitals NHS Foundation Trust, University of London, London, UK
- Vascular Biology Research Centre, Molecular and Clinical Sciences Research Institute, St George's University of London, London, UK
| |
Collapse
|
9
|
Meier AB, Zawada D, De Angelis MT, Martens LD, Santamaria G, Zengerle S, Nowak-Imialek M, Kornherr J, Zhang F, Tian Q, Wolf CM, Kupatt C, Sahara M, Lipp P, Theis FJ, Gagneur J, Goedel A, Laugwitz KL, Dorn T, Moretti A. Epicardioid single-cell genomics uncovers principles of human epicardium biology in heart development and disease. Nat Biotechnol 2023; 41:1787-1800. [PMID: 37012447 PMCID: PMC10713454 DOI: 10.1038/s41587-023-01718-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 02/22/2023] [Indexed: 04/05/2023]
Abstract
The epicardium, the mesothelial envelope of the vertebrate heart, is the source of multiple cardiac cell lineages during embryonic development and provides signals that are essential to myocardial growth and repair. Here we generate self-organizing human pluripotent stem cell-derived epicardioids that display retinoic acid-dependent morphological, molecular and functional patterning of the epicardium and myocardium typical of the left ventricular wall. By combining lineage tracing, single-cell transcriptomics and chromatin accessibility profiling, we describe the specification and differentiation process of different cell lineages in epicardioids and draw comparisons to human fetal development at the transcriptional and morphological levels. We then use epicardioids to investigate the functional cross-talk between cardiac cell types, gaining new insights into the role of IGF2/IGF1R and NRP2 signaling in human cardiogenesis. Finally, we show that epicardioids mimic the multicellular pathogenesis of congenital or stress-induced hypertrophy and fibrotic remodeling. As such, epicardioids offer a unique testing ground of epicardial activity in heart development, disease and regeneration.
Collapse
Affiliation(s)
- Anna B Meier
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Dorota Zawada
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Maria Teresa De Angelis
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
- Department of Experimental and Clinical Medicine, University 'Magna Graecia', Catanzaro, Italy
| | - Laura D Martens
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
- Computational Health Center, Helmholtz Center Munich, Neuherberg, Germany
- Helmholtz Association-Munich School for Data Science (MUDS), Munich, Germany
| | - Gianluca Santamaria
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
- Department of Experimental and Clinical Medicine, University 'Magna Graecia', Catanzaro, Italy
| | - Sophie Zengerle
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Monika Nowak-Imialek
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Jessica Kornherr
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Fangfang Zhang
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Qinghai Tian
- Center for Molecular Signaling (PZMS), Institute for Molecular Cell Biology, Research Center for Molecular Imaging and Screening, Medical Faculty, Saarland University, Homburg, Germany
| | - Cordula M Wolf
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
- Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Christian Kupatt
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Makoto Sahara
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Peter Lipp
- Center for Molecular Signaling (PZMS), Institute for Molecular Cell Biology, Research Center for Molecular Imaging and Screening, Medical Faculty, Saarland University, Homburg, Germany
| | - Fabian J Theis
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
- Computational Health Center, Helmholtz Center Munich, Neuherberg, Germany
| | - Julien Gagneur
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
- Computational Health Center, Helmholtz Center Munich, Neuherberg, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Alexander Goedel
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Karl-Ludwig Laugwitz
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Tatjana Dorn
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Alessandra Moretti
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany.
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany.
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany.
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
10
|
Wen J, Liu G, Liu M, Wang H, Wan Y, Yao Z, Gao N, Sun Y, Zhu L. Transforming growth factor-β and bone morphogenetic protein signaling pathways in pathological cardiac hypertrophy. Cell Cycle 2023; 22:2467-2484. [PMID: 38179789 PMCID: PMC10802212 DOI: 10.1080/15384101.2023.2293595] [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: 06/01/2023] [Revised: 09/22/2023] [Accepted: 10/09/2023] [Indexed: 01/06/2024] Open
Abstract
Pathological cardiac hypertrophy (referred to as cardiac hypertrophy) is a maladaptive response of the heart to a variety of pathological stimuli, and cardiac hypertrophy is an independent risk factor for heart failure and sudden death. Currently, the treatments for cardiac hypertrophy are limited to improving symptoms and have little effect. Elucidation of the developmental process of cardiac hypertrophy at the molecular level and the identification of new targets for the treatment of cardiac hypertrophy are crucial. In this review, we summarize the research on multiple active substances related to the pathogenesis of cardiac hypertrophy and the signaling pathways involved and focus on the role of transforming growth factor-β (TGF-β) and bone morphogenetic protein (BMP) signaling in the development of cardiac hypertrophy and the identification of potential targets for molecular intervention. We aim to identify important signaling molecules with clinical value and hope to help promote the precise treatment of cardiac hypertrophy and thus improve patient outcomes.
Collapse
Affiliation(s)
- Jing Wen
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Guixiang Liu
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Mingjie Liu
- Department of Lung Function, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Huarui Wang
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yunyan Wan
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhouhong Yao
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Nannan Gao
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yuanyuan Sun
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Ling Zhu
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
11
|
Zahedi N, Pourajam S, Zaker E, Kouhpayeh S, Mirbod SM, Tavangar M, Boshtam M, Hatami Kahkesh K, Qian Q, Zhang F, Shariati L, Khanahmad H, Boshtam M. The potential therapeutic impacts of trehalose on cardiovascular diseases as the environmental-influenced disorders: An overview of contemporary findings. ENVIRONMENTAL RESEARCH 2023; 226:115674. [PMID: 36925035 DOI: 10.1016/j.envres.2023.115674] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 06/18/2023]
Abstract
Cardiovascular diseases (CVDs) as environmental-influenced disorders, are a major concern and the leading cause of death worldwide. A range of therapeutic approaches has been proposed, including conventional and novel methods. Natural compounds offer a promising alternative for CVD treatment due to their ability to regulate molecular pathways with minimal adverse effects. Trehalose is natural compound and disaccharide with unique biological functions and cardio-protective properties. The cardio-protective effects of trehalose are generated through its ability to induce autophagy, which is mediated by the transcription factors TFEB and FOXO1. The stimulation of TFEB plays a significant role in regulating autophagy genes and autophagosome formation. Activation of FOXO1 through dephosphorylation of Foxo1 and blocking of p38 mitogen-activated protein kinase (p38 MAPK) also triggers autophagy dramatically. Trehalose has been shown to reduce CVD risk factors, including atherosclerosis, cardiac remodeling after a heart attack, cardiac dysfunction, high blood pressure, and stroke. It also reduces structural abnormalities of mitochondria, cytokine production, vascular inflammation, cardiomyocyte apoptosis, and pyroptosis. This review provides a molecular overview of trehalose's cardioprotective functions, including its mechanisms of autophagy and its potential to improve CVD symptoms based on clinical evidence.
Collapse
Affiliation(s)
- Noushin Zahedi
- Department of Genetics and Molecular biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Samaneh Pourajam
- Department of Internal Medicine, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Erfan Zaker
- Department of Genetics and Molecular biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Medical Genetics, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Shirin Kouhpayeh
- Department of Immunology, Erythron Genetics and Pathobiology Laboratory, Isfahan, Iran
| | - Seyedeh Mahnaz Mirbod
- Department of Cardiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehrsa Tavangar
- Department of Genetics and Molecular biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Kaveh Hatami Kahkesh
- Department of Basic Medical Science, Faculty of Veterinary Medicine, Shahrekord University, Shahrekord, Iran
| | - Qiuping Qian
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, Zhejiang, China
| | - Feng Zhang
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, Zhejiang, China
| | - Laleh Shariati
- Department of Biomaterials, Nanotechnology and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Khanahmad
- Department of Genetics and Molecular biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Maryam Boshtam
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
12
|
Papakonstantinou K, Rorris FP, Schizas N, Antonopoulos C, Samiotis I, Patris V, Geroulakos G, Antoniou GA. Echocardiographic Changes in Cardiac Function After Thoracic Endovascular Aortic Repair: A Systematic Review and Meta-Analysis. Ann Vasc Surg 2023; 90:119-127. [PMID: 36442711 DOI: 10.1016/j.avsg.2022.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/01/2022] [Accepted: 10/11/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Evidence suggests thoracic stent grafts increase the aortic stiffness postimplantation. Our objective was to examine the effect of thoracic aortic stenting on heart function, as demonstrated with echocardiography. METHODS We considered nonrandomized studies examining echocardiographic parameters (left ventricle ejection fraction (LVEF), left ventricle end-diastolic (LVED) and end-systolic diameter (LVESD), posterior wall thickness (LVPWT), interventricular septal thickness (IVST), mass, and mass index) pre and poststent graft implantation in patients with thoracic aortic diseases (aneurysm, dissection, and blunt injury). MEDLINE and CENTRAL were searched (up to March 2021) for eligible studies. The National Institutes of Health Quality Assessment Tool was used for risk of bias assessment. Echocardiographic data pre and postimplantation were compared using the pooled standardized mean difference (SMD) and 95% confidence interval (CI). RESULTS Four studies were included in the meta-analysis. Three of the studies were judged to be "good" quality and one "fair". Nonsignificant differences pre and postimplantation were found for ejection fraction (SMD = -0.53, 95% CI = -1.8 to 0.728, P = 0.406), IVST (SMD = -0.79, 95%, CI = -3.25 to 1.66, P = 0.52), EDD (SMD = -0.10, 95% CI = -0.48 to 0.28, P = 0.60), ESD (SMD = -0.66, 95% CI = -2.35 to 1.02, P = 0.44), and PWT (SMD = -2.20, 95% CI = -5.89 to 1.47, P = 0.24). A trend toward an increase in mass postimplantation was found (SMD = 0.28, 95%, CI = -0.03 to 0.60, P = 0.08), but there was no significant difference in mass index (SMD = 0, 95%, CI = -0.195 to 0.195, P = 1). CONCLUSIONS Thoracic aortic stenting does not appear to significantly impact cardiac physiology as indicated by echocardiographic parameters.
Collapse
Affiliation(s)
| | | | - Nikolaos Schizas
- Department of Cardiovascular and Thoracic Surgery, Evaggelismos General Hospital, Athens, Greece
| | | | - Ilias Samiotis
- Department of Cardiovascular and Thoracic Surgery, Evaggelismos General Hospital, Athens, Greece
| | - Vasileios Patris
- Department of Cardiovascular and Thoracic Surgery, Evaggelismos General Hospital, Athens, Greece
| | - George Geroulakos
- Department of Vascular Surgery, Attikon University Hospital, Athens, Greece
| | - George A Antoniou
- Department of Vascular and Endovascular Surgery, Manchester University NHS Foundation Trust, Manchester, UK; Division of Cardiovascular Sciences, School of Medical Sciences, The University of Manchester, Manchester, UK
| |
Collapse
|
13
|
Esparham A, Shoar S, Kheradmand HR, Ahmadyar S, Dalili A, Rezapanah A, Zandbaf T, Khorgami Z. The Impact of Bariatric Surgery on Cardiac Structure, and Systolic and Diastolic Function in Patients with Obesity: A Systematic Review and Meta-analysis. Obes Surg 2023; 33:345-361. [PMID: 36469205 DOI: 10.1007/s11695-022-06396-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/07/2022]
Abstract
The purpose of this study was to provide pooled data from all studies on the impact of bariatric surgery on cardiac structure, and systolic and diastolic function evaluated by either echocardiography or cardiac magnetic resonance. PubMed, Web of Science, Embase, and Scopus databases were searched. Almost all of cardiac left-side structural indices improved significantly after bariatric surgery. However, right-side structural indices did not change significantly. Left ventricular ejection fraction and most of the diastolic function indices improved significantly after the bariatric surgery. The subgroup analysis showed that the left ventricular mass index decreased more in long-term follow-up (≥ 12 months). In addition, subgroup analysis of studies based on surgery type did not reveal any difference in outcomes between gastric bypass and sleeve gastrectomy groups.
Collapse
Affiliation(s)
- Ali Esparham
- Student Research Committee, College of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeed Shoar
- Department of Clinical Research, ScientificWriting Corp, Houston, TX, USA
| | - Hamid Reza Kheradmand
- Student Research Committee, College of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soheil Ahmadyar
- Student Research Committee, College of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amin Dalili
- Department of Surgery, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Surgical Oncology Research Center, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Rezapanah
- Department of Surgery, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Surgical Oncology Research Center, Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Tooraj Zandbaf
- Department of Surgery, School of Medicine, Islamic Azad University, Mashhad, Iran
| | - Zhamak Khorgami
- Department of Surgery, University of Oklahoma College of Community Medicine, Tulsa, OK, USA.
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
14
|
Wang H, Zhang B, Wu WC, Zhu ZH, Wang H. Change of Left Ventricular Geometric Pattern in Patients with Preserved Ejection Fraction Undergoing Coronary Artery Bypass Grafting. J Cardiovasc Transl Res 2022; 15:1444-1454. [PMID: 35543834 DOI: 10.1007/s12265-022-10249-6] [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: 12/06/2021] [Accepted: 03/31/2022] [Indexed: 12/16/2022]
Abstract
Left ventricular (LV) remodeling and geometric patterns are associated with variations in prognosis. Two hundred twenty-eight patients who underwent selective isolated coronary artery bypass grafting (CABG) were included, divided into normal geometry, concentric remodeling, concentric hypertrophy, and eccentric hypertrophy at baseline. More than half participants with normal geometry at baseline remained in that category, and similar ratio of concentric remodeling reverted to normal geometry on follow-up. The concentric hypertrophy at baseline tended to progress to eccentric geometry rather than normal geometry, while changes from eccentric to concentric hypertrophy was uncommon. iLVEDD had a significant association with an increased risk of developing an abnormal geometric pattern from a normal or concentric remodeling pattern, and iLVESD and LAScd involved in the regression from an abnormal geometric pattern. Thus, dynamic changes in LV geometric pattern are common on 1-year follow-up after CABG and LA strain has an incremental role for early detection in this process.
Collapse
Affiliation(s)
- Han Wang
- Department of Echocardiography, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Beilishi Road, XiCheng District, Beijing, 100037, People's Republic of China
| | - Bing Zhang
- Department of Echocardiography, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Beilishi Road, XiCheng District, Beijing, 100037, People's Republic of China
| | - Wei-Chun Wu
- Department of Echocardiography, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Beilishi Road, XiCheng District, Beijing, 100037, People's Republic of China
| | - Zhen-Hui Zhu
- Department of Echocardiography, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Beilishi Road, XiCheng District, Beijing, 100037, People's Republic of China
| | - Hao Wang
- Department of Echocardiography, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.167 Beilishi Road, XiCheng District, Beijing, 100037, People's Republic of China.
| |
Collapse
|
15
|
Kis M, Dogan Y, Yildirim A, Güzel T, Bekar L, Akhan O, Dogdus M, Harbalıoğlu H, Karabulut D, Soydan E, Zoghi M, Ergene O. Evaluation of demographic, clinical, and aetiological data of patients admitted to cardiology clinics and diagnosed with left ventricular hypertrophy in Turkish population (LVH-TR). Acta Cardiol 2022; 77:836-845. [PMID: 36222672 DOI: 10.1080/00015385.2022.2119670] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Left ventricular hypertrophy (LVH) is potentially modifiable cardiovascular risk factor often overlooked in clinical practice. For this reason, we planned to LVH-TR (Left Ventricular Hypertrophy in Turkish Population) trial to determine the aetiological causes and demographic characteristics of LVH patients. METHODS Our study was a multicentre, national, observational study and included 886 patients who applied to the cardiology clinics in 22 centres between February 2020 and August 2021. In the initial evaluation, the Fabry disease (FD) and cardiac amyloidosis (CA) algorithm was followed in patients whose definitive etiologic cause(s) could not be identified. RESULTS The most common aetiological causes of LVH in our study were hypertension with a rate of 56.6%, heart valve disease with 8.2%, and hypertrophic cardiomyopathy with 7.5%. Athlete's heart was detected in eight patients, LV non-compaction was detected in four patients. The rate of LVH of unknown cause was 18.8%. FD was suspected in 143 patients, and CA was suspected in 16 patients. There were 43 (4.85%) patients with low α-galactosidase A enzyme levels. GLA gene mutation analysis was positive in 1.58% of all patients, and these patients were diagnosed with FD, and 15 (1.69%) patients were diagnosed with CA by endomyocardial biopsy method. CONCLUSION In the aetiology of LVH, the rate of LVH of unknown cause was high. FD and CA should be considered primarily in this patient group. Early diagnosis of the disease by following the schemes leading to FD and CA was essential in starting treatment before the progression of the disease.
Collapse
Affiliation(s)
- Mehmet Kis
- Department of Cardiology, Dokuz Eylul University, Izmir, Turkey
| | - Yasemin Dogan
- Department of Cardiology, Kayseri City Hospital, Kayseri, Turkey
| | - Abdullah Yildirim
- Department of Cardiology, Adana City Training and Research Hospital, Adana, Turkey
| | - Tuncay Güzel
- Department of Cardiology, Akhisar State Hospital, Manisa, Turkey
| | - Lutfu Bekar
- Department of Cardiology, Hitit University Faculty of Medicine, Corum, Turkey
| | - Onur Akhan
- Department of Cardiology, Bilecik State Hospital, Bilecik, Turkey
| | - Mustafa Dogdus
- Department of Cardiology, Usak University Training and Research Hospital, Usak, Turkey
| | - Hazar Harbalıoğlu
- Department of Cardiology, Düzce Atatürk State Hospital, Duzce, Turkey
| | - Dilay Karabulut
- Department of Cardiology, Bakırkoy Dr. Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Elton Soydan
- Department of Cardiology, EGE University, Izmir, Turkey
| | - Mehdi Zoghi
- Department of Cardiology, EGE University, Izmir, Turkey
| | - Oktay Ergene
- Department of Cardiology, Dokuz Eylul University, Izmir, Turkey
| | | |
Collapse
|
16
|
Liu CM, Hsieh ME, Hu YF, Wei TY, Wu IC, Chen PF, Lin YJ, Higa S, Yagi N, Chen SA, Tseng VS. Artificial Intelligence-Enabled Model for Early Detection of Left Ventricular Hypertrophy and Mortality Prediction in Young to Middle-Aged Adults. Circ Cardiovasc Qual Outcomes 2022; 15:e008360. [PMID: 35959675 DOI: 10.1161/circoutcomes.121.008360] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Concealed left ventricular hypertrophy (LVH) is a prevalent condition that is correlated with a substantial risk of cardiovascular events and mortality, especially in young to middle-aged adults. Early identification of LVH is warranted. In this work, we aimed to develop an artificial intelligence (AI)-enabled model for early detection and risk stratification of LVH using 12-lead ECGs. METHODS By deep learning techniques on the ECG recordings from 28 745 patients (20-60 years old), the AI model was developed to detect verified LVH from transthoracic echocardiography and evaluated on an independent cohort. Two hundred twenty-five patients from Japan were externally validated. Cardiologists' diagnosis of LVH was based on conventional ECG criteria. The area under the curve (AUC), sensitivity, and specificity were applied to evaluate the model performance. A Cox regression model estimated the independent effects of AI-predicted LVH on cardiovascular or all-cause death. RESULTS The AUC of the AI model in diagnosing LVH was 0.89 (sensitivity: 90.3%, specificity: 69.3%), which was significantly better than that of the cardiologists' diagnosis (AUC, 0.64). In the second independent cohort, the diagnostic performance of the AI model was consistent (AUC, 0.86; sensitivity: 85.4%, specificity: 67.0%). After a follow-up of 6 years, AI-predicted LVH was independently associated with higher cardiovascular or all-cause mortality (hazard ratio, 1.91 [1.04-3.49] and 1.54 [1.20-1.97], respectively). The predictive power of the AI model for mortality was consistently valid among patients of different ages, sexes, and comorbidities, including hypertension, diabetes, stroke, heart failure, and myocardial infarction. Last, we also validated the model in the international independent cohort from Japan (AUC, 0.83). CONCLUSIONS The AI model improved the detection of LVH and mortality prediction in the young to middle-aged population and represented an attractive tool for risk stratification. Early identification by the AI model gives every chance for timely treatment to reverse adverse outcomes.
Collapse
Affiliation(s)
- Chih-Min Liu
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taiwan (C.-M.L., Y.-F.H., Y.-J.L., S.-A.C.).,Institute of Clinical Medicine and Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan (C.-M.L., Y.-F.H., Y.-J.L., S.-A.C.)
| | - Ming-En Hsieh
- Institute of Data Science and Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan (M.-E.H., T.-Y.W., V.S.T.)
| | - Yu-Feng Hu
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taiwan (C.-M.L., Y.-F.H., Y.-J.L., S.-A.C.).,Institute of Clinical Medicine and Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan (C.-M.L., Y.-F.H., Y.-J.L., S.-A.C.).,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (Y.-F.H.)
| | - Tzu-Yin Wei
- Institute of Data Science and Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan (M.-E.H., T.-Y.W., V.S.T.)
| | - I-Chien Wu
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli, Taiwan (I.-C.W., P.-F.C.)
| | - Pei-Fen Chen
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli, Taiwan (I.-C.W., P.-F.C.)
| | - Yenn-Jiang Lin
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taiwan (C.-M.L., Y.-F.H., Y.-J.L., S.-A.C.).,Institute of Clinical Medicine and Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan (C.-M.L., Y.-F.H., Y.-J.L., S.-A.C.)
| | - Satoshi Higa
- Cardiac Electrophysiology and Pacing Laboratory, Division of Cardiovascular Medicine, Makiminato Central Hospital, Okinawa, Japan (S.H.)
| | - Nobumori Yagi
- Division of Cardiovascular Medicine, Nakagami Hospital, Okinawa, Japan (N.Y.)
| | - Shih-Ann Chen
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taiwan (C.-M.L., Y.-F.H., Y.-J.L., S.-A.C.).,Institute of Clinical Medicine and Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan (C.-M.L., Y.-F.H., Y.-J.L., S.-A.C.).,Cardiovascular Center, Taichung Veterans General Hospital, Taiwan (S.-A.C.).,National Chung Hsing University, Taichung, Taiwan (S.-A.C.)
| | - Vincent S Tseng
- Institute of Data Science and Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan (M.-E.H., T.-Y.W., V.S.T.).,Department of Computer Science, National Yang Ming Chiao Tung University, Hsinchu, Taiwan (V.S.T.)
| |
Collapse
|
17
|
Bariatric Surgery Improves Heart Geometry and Plasticity. Obes Surg 2022; 32:1-6. [PMID: 35501635 DOI: 10.1007/s11695-022-06016-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 03/10/2022] [Accepted: 03/16/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Obesity is commonly associated with increased sympathetic tone, changes in heart geometry, and mortality. The aforementioned translates into a higher and potentially modifiable mortality risk for this specific population. OBJECTIVES The aim of the study was to analyze the extent of changes in the heart ventricular structure following rapid weight loss after bariatric surgery. SETTING Academic, university-affiliated hospital. METHODS We retrospectively reviewed all the patients that underwent bariatric surgery at our institution between 2010 and 2015. Data analyzed included demographics, BMI, and associated medical problems. Preoperative and postoperative echography readings were compared looking at the heart geometry, cardiac volumes, and wall thickness. RESULTS Fifty-one patients who had bariatric surgery and had echocardiography before and after the surgery were identified. There were 33 females (64.7%). The mean age was 63.4 ± 12.0 years with an average BMI of 40.3 ± 6.3. The mean follow-up was 1.2 years after the procedure. At 1 year follow-up 25 patients (49%, p = 0.01) showed normal left ventricular geometry. The left ventricular mass (229 ± 82.1 vs 193.2 ± 42.5, p<0.01) and the left ventricular end diastolic volume (129.4 ± 53 vs 96.4 ± 36.5, p = 0.01) showed a significant modification following the procedure. There was a significant improvement in the interventricular septal thickness (p = 0.01) and relative wall thickness (p < 0.01) following surgery. CONCLUSION The patients with obesity present a significant cardiac remodeling from concentric remodeling to normal geometry after bariatric surgery. The decrease in BMI has a direct effect on improvement of the left ventricular structure. Further studies must be carried out to define the damage of obesity to diastolic function.
Collapse
|
18
|
Jakicic JM, Rogers RJ, Lang W, Gibbs BB, Yuan N, Fridman Y, Schelbert EB. Impact of weight loss with diet or diet plus physical activity on cardiac magnetic resonance imaging and cardiovascular disease risk factors: Heart Health Study randomized trial. Obesity (Silver Spring) 2022; 30:1039-1056. [PMID: 35470972 PMCID: PMC9813917 DOI: 10.1002/oby.23412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/31/2022] [Accepted: 02/22/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVE The primary aim of this study was to examine the change in left ventricular mass (LVM) in adults with overweight or obesity in response to a behavioral weight-loss intervention, with variable physical activity (PA) prescriptions. METHODS A total of 383 adults were randomized to a 12-month intervention of diet modification (DIET), DIET plus 150 min/wk of PA (DIET+MODPA), or DIET plus prescription of 250 min/wk of PA (DIET+HIGHPA). LVM was measured with cardiac magnetic resonance imaging. RESULTS Twelve-month weight loss was -10.2% (95% CI: -11.7% to -8.8%) in the DIET group, -11.0% (95% CI: -12.4% to -9.5%) in the DIET+MODPA group, and -10.3% (95% CI: -11.8% to -8.9%) in the DIET+HIGHPA group. LVM decreased at 12 months in the DIET group (-2.9 g [95% CI: -5.2 to -0.7]; p = 0.0114), with no change observed in the DIET+MODPA group (-0.8 g [95% CI: -3.0 to 1.5]; p = 0.4979) or the DIET+HIGHPA group (-1.1 g [95% CI: -3.3 to 1.1]; p = 0.3299). CONCLUSIONS Weight loss through dietary modification resulted in reduced LVM, whereas, when combined with at least 150 min/wk of prescribed moderate-to-vigorous PA, LVM was preserved. These may both be favorable adaptations to weight loss and PA in adults with overweight or obesity that warrant further investigation to understand the clinical implications of these changes on cardiovascular disease risk.
Collapse
Affiliation(s)
- John M Jakicic
- AdventHealth, Translational Research Institute, Orlando, Florida, USA
| | - Renee J Rogers
- AdventHealth, Translational Research Institute, Orlando, Florida, USA
| | - Wei Lang
- Center on Aging and Mobility, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Bethany B Gibbs
- Department of Health and Human Development, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Nalingna Yuan
- Department of Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yaron Fridman
- Asheville Cardiology Associates, Mission Health, Asheville, North Carolina, USA
| | - Erik B Schelbert
- School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Minneapolis Heart Institute East, Saint Paul, Minnesota, USA
| |
Collapse
|
19
|
Sobhani S, Raji S, Aghaee A, Pirzadeh P, Ebrahimi Miandehi E, Shafiei S, Akbari M, Eslami S. Body mass index, lipid profile, and hypertension contribute to prolonged QRS complex. Clin Nutr ESPEN 2022; 50:231-237. [DOI: 10.1016/j.clnesp.2022.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/24/2022] [Accepted: 05/17/2022] [Indexed: 10/18/2022]
|
20
|
Aluja D, Delgado-Tomás S, Ruiz-Meana M, Barrabés JA, Inserte J. Calpains as Potential Therapeutic Targets for Myocardial Hypertrophy. Int J Mol Sci 2022; 23:ijms23084103. [PMID: 35456920 PMCID: PMC9032729 DOI: 10.3390/ijms23084103] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 03/26/2022] [Accepted: 04/06/2022] [Indexed: 11/25/2022] Open
Abstract
Despite advances in its treatment, heart failure remains a major cause of morbidity and mortality, evidencing an urgent need for novel mechanism-based targets and strategies. Myocardial hypertrophy, caused by a wide variety of chronic stress stimuli, represents an independent risk factor for the development of heart failure, and its prevention constitutes a clinical objective. Recent studies performed in preclinical animal models support the contribution of the Ca2+-dependent cysteine proteases calpains in regulating the hypertrophic process and highlight the feasibility of their long-term inhibition as a pharmacological strategy. In this review, we discuss the existing evidence implicating calpains in the development of cardiac hypertrophy, as well as the latest advances in unraveling the underlying mechanisms. Finally, we provide an updated overview of calpain inhibitors that have been explored in preclinical models of cardiac hypertrophy and the progress made in developing new compounds that may serve for testing the efficacy of calpain inhibition in the treatment of pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- David Aluja
- Cardiovascular Diseases Research Group, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (D.A.); (S.D.-T.); (M.R.-M.); (J.A.B.)
| | - Sara Delgado-Tomás
- Cardiovascular Diseases Research Group, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (D.A.); (S.D.-T.); (M.R.-M.); (J.A.B.)
| | - Marisol Ruiz-Meana
- Cardiovascular Diseases Research Group, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (D.A.); (S.D.-T.); (M.R.-M.); (J.A.B.)
- Centro de Investigación en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - José A. Barrabés
- Cardiovascular Diseases Research Group, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (D.A.); (S.D.-T.); (M.R.-M.); (J.A.B.)
- Centro de Investigación en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Javier Inserte
- Cardiovascular Diseases Research Group, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (D.A.); (S.D.-T.); (M.R.-M.); (J.A.B.)
- Centro de Investigación en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-934894038
| |
Collapse
|
21
|
Kruizinga MD, Houdijk EC, van der Kaay DC, van Berkel Y, Filippini L, Stuurman FE, Cohen AF, Driessen GJ, Kruizinga MD. Objective Home-Monitoring of Physical Activity, Cardiovascular Parameters, and Sleep in Pediatric Obesity. Digit Biomark 2022; 6:19-29. [DOI: 10.1159/000522185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 12/20/2021] [Indexed: 11/19/2022] Open
Abstract
<b><i>Introduction:</i></b> Clinical research and treatment of childhood obesity is challenging, and objective biomarkers obtained in a home-setting are needed. The aim of this study was to determine the potential of novel digital endpoints gathered by a home-monitoring platform in pediatric obesity. <b><i>Methods:</i></b> In this prospective observational study, 28 children with obesity aged 6–16 years were included and monitored for 28 days. Patients wore a smartwatch, which measured physical activity (PA), heart rate (HR), and sleep. Furthermore, daily blood pressure (BP) measurements were performed. Data from 128 healthy children were utilized for comparison. Differences between patients and controls were assessed via linear mixed effect models. <b><i>Results:</i></b> Data from 28 patients (average age 11.6 years, 46% male, average body mass index 30.9) and 128 controls (average age 11.1 years, 46% male, average body mass index 18.0) were analyzed. Patients were recruited between November 2018 and February 2020. For patients, the median compliance for the measurements ranged from 55% to 100% and the highest median compliance was observed for the smartwatch-related measurements (81–100%). Patients had a lower daily PA level (4,597 steps vs. 6,081 steps, 95% confidence interval [CI] 862–2,108) and peak PA level (1,115 steps vs. 1,392 steps, 95% CI 136–417), a higher nighttime HR (81 bpm vs. 71 bpm, 95% CI 6.3–12.3) and daytime HR (98 bpm vs. 88 bpm, 95% CI 7.6–12.6), a higher systolic BP (115 mm Hg vs. 104 mm Hg, 95% CI 8.1–14.5) and diastolic BP (76 mm Hg vs. 65 mm Hg, 95% CI 8.7–12.7), and a shorter sleep duration (difference 0.5 h, 95% CI 0.2–0.7) compared to controls. <b><i>Conclusion:</i></b> Remote monitoring via wearables in pediatric obesity has the potential to objectively measure the disease burden in the home-setting. The novel endpoints demonstrate significant differences in PA level, HR, BP, and sleep duration between patients and controls. Future studies are needed to determine the capacity of the novel digital endpoints to detect effect of interventions.
Collapse
|
22
|
Monda E, Palmiero G, Lioncino M, Rubino M, Cirillo A, Fusco A, Caiazza M, Verrillo F, Diana G, Mauriello A, Iavarone M, Losi MA, De Rimini ML, Dellegrottaglie S, D’Andrea A, Bossone E, Pacileo G, Limongelli G. Multimodality Imaging in Cardiomyopathies with Hypertrophic Phenotypes. J Clin Med 2022; 11:868. [PMID: 35160323 PMCID: PMC8836956 DOI: 10.3390/jcm11030868] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/29/2022] [Accepted: 02/04/2022] [Indexed: 12/16/2022] Open
Abstract
Multimodality imaging is a comprehensive strategy to investigate left ventricular hypertrophy (LVH), providing morphologic, functional, and often clinical information to clinicians. Hypertrophic cardiomyopathy (HCM) is defined by an increased LV wall thickness not only explainable by abnormal loading conditions. In the context of HCM, multimodality imaging, by different imaging techniques, such as echocardiography, cardiac magnetic resonance, cardiac computer tomography, and cardiac nuclear imaging, provides essential information for diagnosis, sudden cardiac death stratification, and management. Furthermore, it is essential to uncover the specific cause of HCM, such as Fabry disease and cardiac amyloidosis, which can benefit of specific treatments. This review aims to elucidate the current role of multimodality imaging in adult patients with HCM.
Collapse
Affiliation(s)
- Emanuele Monda
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, AORN Ospedali dei Colli-Monaldi Hospital, 80131 Naples, Italy; (E.M.); (G.P.); (M.L.); (M.R.); (A.C.); (A.F.); (M.C.); (F.V.); (G.D.); (A.M.); (M.I.); (G.P.)
| | - Giuseppe Palmiero
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, AORN Ospedali dei Colli-Monaldi Hospital, 80131 Naples, Italy; (E.M.); (G.P.); (M.L.); (M.R.); (A.C.); (A.F.); (M.C.); (F.V.); (G.D.); (A.M.); (M.I.); (G.P.)
| | - Michele Lioncino
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, AORN Ospedali dei Colli-Monaldi Hospital, 80131 Naples, Italy; (E.M.); (G.P.); (M.L.); (M.R.); (A.C.); (A.F.); (M.C.); (F.V.); (G.D.); (A.M.); (M.I.); (G.P.)
| | - Marta Rubino
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, AORN Ospedali dei Colli-Monaldi Hospital, 80131 Naples, Italy; (E.M.); (G.P.); (M.L.); (M.R.); (A.C.); (A.F.); (M.C.); (F.V.); (G.D.); (A.M.); (M.I.); (G.P.)
| | - Annapaola Cirillo
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, AORN Ospedali dei Colli-Monaldi Hospital, 80131 Naples, Italy; (E.M.); (G.P.); (M.L.); (M.R.); (A.C.); (A.F.); (M.C.); (F.V.); (G.D.); (A.M.); (M.I.); (G.P.)
| | - Adelaide Fusco
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, AORN Ospedali dei Colli-Monaldi Hospital, 80131 Naples, Italy; (E.M.); (G.P.); (M.L.); (M.R.); (A.C.); (A.F.); (M.C.); (F.V.); (G.D.); (A.M.); (M.I.); (G.P.)
| | - Martina Caiazza
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, AORN Ospedali dei Colli-Monaldi Hospital, 80131 Naples, Italy; (E.M.); (G.P.); (M.L.); (M.R.); (A.C.); (A.F.); (M.C.); (F.V.); (G.D.); (A.M.); (M.I.); (G.P.)
| | - Federica Verrillo
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, AORN Ospedali dei Colli-Monaldi Hospital, 80131 Naples, Italy; (E.M.); (G.P.); (M.L.); (M.R.); (A.C.); (A.F.); (M.C.); (F.V.); (G.D.); (A.M.); (M.I.); (G.P.)
| | - Gaetano Diana
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, AORN Ospedali dei Colli-Monaldi Hospital, 80131 Naples, Italy; (E.M.); (G.P.); (M.L.); (M.R.); (A.C.); (A.F.); (M.C.); (F.V.); (G.D.); (A.M.); (M.I.); (G.P.)
| | - Alfredo Mauriello
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, AORN Ospedali dei Colli-Monaldi Hospital, 80131 Naples, Italy; (E.M.); (G.P.); (M.L.); (M.R.); (A.C.); (A.F.); (M.C.); (F.V.); (G.D.); (A.M.); (M.I.); (G.P.)
| | - Michele Iavarone
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, AORN Ospedali dei Colli-Monaldi Hospital, 80131 Naples, Italy; (E.M.); (G.P.); (M.L.); (M.R.); (A.C.); (A.F.); (M.C.); (F.V.); (G.D.); (A.M.); (M.I.); (G.P.)
| | - Maria Angela Losi
- Department of Advanced Biomedical Sciences, Federico II University of Naples, 80138 Naples, Italy;
| | - Maria Luisa De Rimini
- Department of Nuclear Medicine, AORN Ospedali dei Colli-Monaldi Hospital, 80131 Naples, Italy;
| | - Santo Dellegrottaglie
- Cardiovascular MRI Laboratory, Division of Cardiology, Ospedale Medico-Chirurgico Accreditato Villa dei Fiori, 80011 Acerra, Italy;
| | - Antonello D’Andrea
- Unit of Cardiology and Intensive Coronary Care, “Umberto I” Hospital, 84014 Nocera Inferiore, Italy;
| | - Eduardo Bossone
- Department of Cardiology, Cardarelli Hospital, 80131 Naples, Italy;
| | - Giuseppe Pacileo
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, AORN Ospedali dei Colli-Monaldi Hospital, 80131 Naples, Italy; (E.M.); (G.P.); (M.L.); (M.R.); (A.C.); (A.F.); (M.C.); (F.V.); (G.D.); (A.M.); (M.I.); (G.P.)
| | - Giuseppe Limongelli
- Inherited and Rare Cardiovascular Diseases, Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, AORN Ospedali dei Colli-Monaldi Hospital, 80131 Naples, Italy; (E.M.); (G.P.); (M.L.); (M.R.); (A.C.); (A.F.); (M.C.); (F.V.); (G.D.); (A.M.); (M.I.); (G.P.)
| |
Collapse
|
23
|
Shibata R, Shinohara K, Ikeda S, Iyonaga T, Matsuura T, Kashihara S, Ito K, Kishi T, Hirooka Y, Tsutsui H. Transient receptor potential vanilloid 1-expressing cardiac afferent nerves may contribute to cardiac hypertrophy in accompany with an increased expression of brain-derived neurotrophic factor within nucleus tractus solitarius in a pressure overload model. Clin Exp Hypertens 2022; 44:249-257. [DOI: 10.1080/10641963.2022.2029470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Risa Shibata
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Keisuke Shinohara
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Center for Clinical and Translational Research, Kyushu University Hospital, Fukuoka, Japan
| | - Shota Ikeda
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeshi Iyonaga
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Taku Matsuura
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Soichiro Kashihara
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koji Ito
- Ito Heart Clinic, Fukuoka, Japan
| | - Takuya Kishi
- Graduate School of Medicine (Cardiology), International University of Health and Welfare, Fukuoka, Japan
| | - Yoshitaka Hirooka
- Department of Medical Technology and Sciences, International University of Health and Welfare, Fukuoka, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
24
|
Isa Tafreshi R, Radgoodarzi M, Arjmandi Rafsanjani K, Soheilipour F. Subclinical Left Ventricular Dysfunction in Children and Adolescence With Thalassemia Intermedia. Front Pediatr 2022; 10:774528. [PMID: 35783313 PMCID: PMC9249082 DOI: 10.3389/fped.2022.774528] [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: 09/12/2021] [Accepted: 05/24/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Cardiac complications are important causes of morbidity in patients with thalassemia intermedia (TI). We aimed to assess left ventricular (LV) function, using new tissue Doppler imaging (TDI) indices, in order to diagnose early ventricular impairment in asymptomatic children and adolescence with the TI. MATERIALS AND METHODS We investigated possible differences in echocardiographic systolic and diastolic parameters between a population of 28 asymptomatic patients (mean age, 13.6 ± 5.7 years) and 35 age-matched healthy control members. All of them underwent 2-D, pulsed Doppler, and tissue Doppler echocardiographic studies for the assessment of the LV mass, Trans-mitral velocities, mitral annular systolic and diastolic velocities, myocardial performance index (MPI), and myocardial acceleration during isovolumic contraction (IVA). The cardiac iron load was estimated by magnetic resonance imaging T2*. RESULTS Left ventricular hypertrophy (LVH) was found in 13 (46.4%) patients. We found significantly reduced TDI-derived peak systolic myocardial velocity (s') in patients, whereas no significant difference was identified between the patients and control group members when the IVA was compared. The ratio of peak mitral inflow velocity to annular early diastolic velocity (E/e') of the mitral valve as an index of the diastolic function was significantly higher in patients (9 ± 1 vs. 6 ± 1, p < 0.05). Choosing a TDI-derived MPI > 0.33 as a cutoff point, the global LV dysfunction was detected with a sensitivity of 78% and a specificity of 80%. The patients with LVH significantly exhibited higher values of TDI-MPI and lower values of s' velocity and IVA when compared against the subjects with normal LV mass. CONCLUSION Subtle LV systolic and diastolic dysfunction develops early in young patients with the TI who have normal cardiac iron concentration. Moreover, LV remodeling as a main cardiac adaptive response plays a principal role in developing myocardial impairment.
Collapse
Affiliation(s)
- Roya Isa Tafreshi
- Department of Pediatric Cardiology, Ali Asghar Children's Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Radgoodarzi
- Department of Pediatrics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Kadijeh Arjmandi Rafsanjani
- Department of Pediatric Hematology and Oncology, Ali Asghar Children's Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fahimeh Soheilipour
- Department of Pediatric Endocrinology, Minimally Invasive Surgery Research Center, Aliasghar Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Odajima S, Tanaka H, Fujimoto W, Kuroda K, Yamashita S, Imanishi J, Iwasaki M, Todoroki T, Okuda M, Hayashi T, Konishi A, Shinohara M, Toh R, Hirata KI. Efficacy of Renin-angiotensin-aldosterone-system inhibitors for heart failure with preserved ejection fraction and left ventricular hypertrophy -from the KUNIUMI Registry Acute Cohort. J Cardiol 2021; 79:703-710. [PMID: 34924235 DOI: 10.1016/j.jjcc.2021.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 10/28/2021] [Accepted: 11/14/2021] [Indexed: 01/17/2023]
Abstract
BACKGROUND Heterogeneity of heart failure (HF) with preserved ejection fraction (HFpEF) would contribute to the difficulty in identifying effective treatments, and interest in the phenogrouping of HFpEF as a potential means for predicting patients who respond to cardioprotective drugs has been increasing. METHODS We studied 468 first-hospitalized HFpEF patients among 1971 acute-hospitalized HF patients from KUNIUMI Registry Acute Cohort. The primary endpoint was defined as HF-rehospitalization and cardiovascular death over a median follow-up period of 508 days. RESULTS In HFpEF patients with left ventricular hypertrophy (LVH), patients prescribed renin-angiotensin-aldosterone-system (RAAS) inhibitors had similar outcomes compared to those without (HR, 0.77; 95% CI 0.51-1.16; p = 0.21), and the outcome was also similar between patients with and without RAAS inhibitors' prescription in HFpEF patients without LVH. Moreover, in HFpEF patients with LVH and mild-moderate chronic kidney disease (CKD), which was determined as an estimated glomerular filtration rate (eGFR) of 30-60 mL/min/1.73 m2, patients prescribed RAAS inhibitors had significantly favorable outcomes compared to those without (HR 0.39; 95% CI 0.19-0.80; p = 0.01). In HFpEF patients with LVH and severe CKD, which was defined as eGFR <30 mL/min/1.73 m2, the outcome was similar between patients with and without RAAS inhibitor prescription. Multivariable Cox regression analysis showed that the prescription of RAAS inhibitors was the only independent predictor of outcome in HFpEF patients with LVH and mild-moderate CKD (HR 0.49; 95% CI 0.25-0.94; p = 0.03). CONCLUSIONS Our findings showed the importance of HFpEF phenogrouping for identifying effective pharmacological treatments.
Collapse
Affiliation(s)
- Susumu Odajima
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hidekazu Tanaka
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Wataru Fujimoto
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan; Department of Cardiology, Hyogo Prefectural Awaji Medical Center, Sumoto, Japan
| | - Koji Kuroda
- Department of Cardiology, Hyogo Prefectural Awaji Medical Center, Sumoto, Japan
| | - Soichiro Yamashita
- Department of Cardiology, Hyogo Prefectural Awaji Medical Center, Sumoto, Japan
| | - Junichi Imanishi
- Department of Cardiology, Hyogo Prefectural Awaji Medical Center, Sumoto, Japan
| | - Masamichi Iwasaki
- Department of Cardiology, Hyogo Prefectural Awaji Medical Center, Sumoto, Japan
| | - Takashi Todoroki
- Department of Cardiology, Hyogo Prefectural Awaji Medical Center, Sumoto, Japan
| | - Masanori Okuda
- Department of Cardiology, Hyogo Prefectural Awaji Medical Center, Sumoto, Japan
| | - Takatoshi Hayashi
- Department of Cardiology, Hyogo Prefectural Awaji Medical Center, Sumoto, Japan
| | - Akihide Konishi
- Clinical & Translational Research Center, Kobe University Hospital, Kobe, Japan
| | - Masakazu Shinohara
- Division of Epidemiology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ryuji Toh
- Division of Evidence-based Laboratory Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ken-Ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan; Division of Evidence-based Laboratory Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
26
|
MicroRNAs: From Junk RNA to Life Regulators and Their Role in Cardiovascular Disease. CARDIOGENETICS 2021. [DOI: 10.3390/cardiogenetics11040023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
MicroRNAs (miRNAs) are single-stranded small non-coding RNA (18–25 nucleotides) that until a few years ago were considered junk RNA. In the last twenty years, they have acquired more importance thanks to the understanding of their influence on gene expression and their role as negative regulators at post-transcriptional level, influencing the stability of messenger RNA (mRNA). Approximately 5% of the genome encodes miRNAs which are responsible for regulating numerous signaling pathways, cellular processes and cell-to-cell communication. In the cardiovascular system, miRNAs control the functions of various cells, such as cardiomyocytes, endothelial cells, smooth muscle cells and fibroblasts, playing a role in physiological and pathological processes and seeming also related to variations in contractility and hereditary cardiomyopathies. They provide a new perspective on the pathophysiology of disorders such as hypertrophy, fibrosis, arrhythmia, inflammation and atherosclerosis. MiRNAs are differentially expressed in diseased tissue and can be released into the circulation and then detected. MiRNAs have become interesting for the development of new diagnostic and therapeutic tools for various diseases, including heart disease. In this review, the concept of miRNAs and their role in cardiomyopathies will be introduced, focusing on their potential as therapeutic and diagnostic targets (as biomarkers).
Collapse
|
27
|
Mohan M, Dihoum A, Mordi IR, Choy AM, Rena G, Lang CC. Left Ventricular Hypertrophy in Diabetic Cardiomyopathy: A Target for Intervention. Front Cardiovasc Med 2021; 8:746382. [PMID: 34660744 PMCID: PMC8513785 DOI: 10.3389/fcvm.2021.746382] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/02/2021] [Indexed: 12/12/2022] Open
Abstract
Heart failure is an important manifestation of diabetic heart disease. Before the development of symptomatic heart failure, as much as 50% of patients with type 2 diabetes mellitus (T2DM) develop asymptomatic left ventricular dysfunction including left ventricular hypertrophy (LVH). Left ventricular hypertrophy (LVH) is highly prevalent in patients with T2DM and is a strong predictor of adverse cardiovascular outcomes including heart failure. Importantly regression of LVH with antihypertensive treatment especially renin angiotensin system blockers reduces cardiovascular morbidity and mortality. However, this approach is only partially effective since LVH persists in 20% of patients with hypertension who attain target blood pressure, implicating the role of other potential mechanisms in the development of LVH. Moreover, the pathophysiology of LVH in T2DM remains unclear and is not fully explained by the hyperglycemia-associated cellular alterations. There is a growing body of evidence that supports the role of inflammation, oxidative stress, AMP-activated kinase (AMPK) and insulin resistance in mediating the development of LVH. The recognition of asymptomatic LVH may offer an opportune target for intervention with cardio-protective therapy in these at-risk patients. In this article, we provide a review of some of the key clinical studies that evaluated the effects of allopurinol, SGLT2 inhibitor and metformin in regressing LVH in patients with and without T2DM.
Collapse
Affiliation(s)
- Mohapradeep Mohan
- Division of Mental Health and Wellbeing, Warwick Medical School, University of Warwick, Coventry, United Kingdom.,Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Adel Dihoum
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Ify R Mordi
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Anna-Maria Choy
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Graham Rena
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Chim C Lang
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom.,UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
28
|
Gul R, Alsalman N, Bazighifan A, Alfadda AA. Comparative beneficial effects of nebivolol and nebivolol/valsartan combination against mitochondrial dysfunction in angiotensin II-induced pathology in H9c2 cardiomyoblasts. J Pharm Pharmacol 2021; 73:1520-1529. [PMID: 34453839 DOI: 10.1093/jpp/rgab124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 08/03/2021] [Indexed: 11/14/2022]
Abstract
OBJECTIVES Considering the complementary nature of signalling mechanisms and the therapeutic effects of nebivolol, a β1-adrenoreceptor antagonist, and valsartan, an angiotensin receptor blocker (ARB), here we aimed to investigate whether nebivolol/valsartan combination would complement the cardioprotective effects of nebivolol on angiotensin II (ANG II)-induced pathology in H9c2 cardiomyoblasts. METHODS H9c2 cardiomyoblasts were used to investigate the protective effects of nebivolol and nebivolol and valsartan combination against ANG II-induced pathology. Reactive oxygen species (ROS) generation was determined by 2',7'-dichlorofluorescein diacetate (DCFDA) and MitoSOX Red staining. Real-time PCR and immunoblotting were employed to quantify the changes in mRNA and protein expression levels, respectively. KEY FINDINGS Our data revealed that pretreatment with nebivolol and nebivolol/valsartan combination significantly reduced ANG II-induced oxidative stress and mTORC1 signalling. Concurrently, ANG II-induced activation of inflammatory cytokines and fetal gene expressions were significantly suppressed by nebivolol and nebivolol/valsartan combination. Pretreatment with nebivolol and nebivolol/valsartan combination alleviated ANG II-induced impairment of mitochondrial biogenesis by restoring the gene expression levels of PGC-1α, TFAM, NRF-1 and SIRT3. Our data further show that nebivolol and nebivolol/valsartan combination mediated up-regulation in mitochondrial biogenesis is accompanied by decrease in ANG II-stimulated mitochondrial ROS generation as well as increase in expression of mitochondrial fusion genes MFN2 and OPA1, indicative of improved mitochondrial dynamics. SUMMARY These findings suggest that both nebivolol and nebivolol/valsartan combination exert protective effects on ANG II-induced mitochondrial dysfunction by alleviating its biogenesis and dynamics. Moreover, addition of valsartan to nebivolol do not produce any additive effects compared with nebivolol alone on ANG II-induced cardiac pathology.
Collapse
Affiliation(s)
- Rukhsana Gul
- Obesity Research Center, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Nouf Alsalman
- Obesity Research Center, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Arwa Bazighifan
- Obesity Research Center, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Assim A Alfadda
- Obesity Research Center, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia.,Department of Medicine, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia.,Strategic Center for Diabetes Research, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
29
|
Wen J, Liu L, Li J, He Y. A review of nardosinone for pharmacological activities. Eur J Pharmacol 2021; 908:174343. [PMID: 34265296 DOI: 10.1016/j.ejphar.2021.174343] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/16/2021] [Accepted: 07/11/2021] [Indexed: 11/20/2022]
Abstract
Nardostachys jatamansi is a natural medicinal plant that is widely used in Asia for the treatment of various neurological and cardiac diseases, and nardosinone is the main active ingredient of N. jatamansi, which has the potential to treat a variety of diseases. Herein, we summarize the reported chemical structure, pharmacokinetics and pharmacological potential of nardosinone, and point out areas for further research. We obtained studies that were related to the chemical structure and pharmacological activities of nardosinone from several databases. Previous studies have shown that nardosinone has anti-inflammatory effects, anti-hypertrophic effect in cardiomyocytes, enhances activity of the nerve growth factor and promotes neural stem cells to proliferate and differentiate. However, the molecular mechanism of how nardosinone promotes proliferation and differentiation of neural stem cells, and its role in resisting cardiomyocyte hypertrophy remains unclear and needs to be further studied. Overall, nardosinone has the potential to treat bacterial infections, periodontitis, cardiac diseases, neurodegenerative diseases and cancer. However, the gaps found in the literature is the lack of more comprehensive information regarding the pharmacokinetics and toxicology of nardosinone.
Collapse
Affiliation(s)
- Jiawei Wen
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Linqiu Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Junjun Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Yang He
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China.
| |
Collapse
|
30
|
Wang H, Zhao M, Magnussen CG, Xi B. Change in waist circumference over 2 years and the odds of left ventricular hypertrophy among Chinese children. Nutr Metab Cardiovasc Dis 2021; 31:2484-2489. [PMID: 34088584 DOI: 10.1016/j.numecd.2021.04.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/18/2021] [Accepted: 04/30/2021] [Indexed: 11/15/2022]
Abstract
BACKGROUND AND AIMS High waist circumference (WC) is associated with left ventricular mass index (LVMI) in childhood. However, no studies have assessed the association between WC change and left ventricular hypertrophy (LVH) in childhood. This study aimed to investigate the association between change in WC status over 2 years on LVH among Chinese children. METHODS AND RESULTS Data were from a population-based prospective cohort study in China. Children without LVH at baseline (n = 1067) were assigned to four WC status change groups (persistent normal WC, WC loss, WC gain, and persistent abdominal obesity). Over a 2-year follow-up, 103 (out of 1067) children had LVH. LVMI levels were the highest among the persistent abdominal obesity group (31.5 ± 3.8 g/m 2.7), lower in the WC gain group (31.0 ± 3.6 g/m 2.7) and the WC loss group (29.8 ± 3.7 g/m 2.7), and lowest in the persistent normal WC group (29.1 ± 3.7 g/m 2.7). Compared with children in the persistent normal WC group, the odds of LVH was highest in the persistent abdominal obesity group [odds ratio (OR) = 3.57, 95% confidence interval (CI): 2.18-5.83], followed by the WC gain group (OR = 2.85, 95% CI: 1.50-5.41). In contrast, the odds of LVH was not increased in the WC loss group (OR = 0.93, 95% CI: 0.21-4.07). CONCLUSION Although these findings highlight the importance of maintaining normal WC in childhood to reduce the odds of developing LVH, our data suggest the increased odds associated with abdominal obesity can be reversed by WC loss.
Collapse
Affiliation(s)
- Huan Wang
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Min Zhao
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Costan G Magnussen
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia; Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland; Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Bo Xi
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| |
Collapse
|
31
|
Katz DH, Tahir UA, Ngo D, Benson MD, Gao Y, Shi X, Nayor M, Keyes MJ, Larson MG, Hall ME, Correa A, Sinha S, Shen D, Herzig M, Yang Q, Robbins JM, Chen ZZ, Cruz DE, Peterson B, Vasan RS, Wang TJ, Wilson JG, Gerszten RE. Multiomic Profiling in Black and White Populations Reveals Novel Candidate Pathways in Left Ventricular Hypertrophy and Incident Heart Failure Specific to Black Adults. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2021; 14:e003191. [PMID: 34019435 PMCID: PMC8497179 DOI: 10.1161/circgen.120.003191] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 05/05/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND Increased left ventricular (LV) mass is associated with adverse cardiovascular events including heart failure (HF). Both increased LV mass and HF disproportionately affect Black individuals. To understand the underlying mechanisms, we undertook a proteomic screen in a Black cohort and compared the findings to results from a White cohort. METHODS We measured 1305 plasma proteins using the SomaScan platform in 1772 Black participants (mean age, 56 years; 62% women) in JHS (Jackson Heart Study) with LV mass assessed by 2-dimensional echocardiography. Incident HF was assessed in 1600 participants. We then compared protein associations in JHS to those observed in White participants from FHS (Framingham Heart Study; mean age, 54 years; 56% women). RESULTS In JHS, there were 110 proteins associated with LV mass and 13 proteins associated with incident HF hospitalization with false discovery rate <5% after multivariable adjustment. Several proteins showed expected associations with both LV mass and HF, including NT-proBNP (N-terminal pro-B-type natriuretic peptide; β=0.04; P=2×10-8; hazard ratio, 1.48; P=0.0001). The strongest association with LV mass was novel: LKHA4 (leukotriene-A4 hydrolase; β=0.05; P=5×10-15). This association was confirmed on an alternate proteomics platform and further supported by related metabolomic data. Fractalkine/CX3CL1 (C-X3-C Motif Chemokine Ligand 1) showed a novel association with incident HF (hazard ratio, 1.32; P=0.0002). While established biomarkers such as cystatin C and NT-proBNP showed consistent associations in Black and White individuals, LKHA4 and fractalkine were significantly different between the two groups. CONCLUSIONS We identified several novel biological pathways specific to Black adults hypothesized to contribute to the pathophysiologic cascade of LV hypertrophy and incident HF including LKHA4 and fractalkine.
Collapse
Affiliation(s)
- Daniel H. Katz
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Usman A. Tahir
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Debby Ngo
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Mark D. Benson
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Yan Gao
- Univ of Mississippi Medical Center, Jackson, MS
| | - Xu Shi
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Matthew Nayor
- Cardiology Division, Department of Medicine, Massachusetts General Hospital
| | - Michelle J. Keyes
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
- Framingham Heart Study, Framingham
| | | | | | | | - Sumita Sinha
- Whitehead Institute for Biomedical Research, Cambridge
| | - Dongxiao Shen
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Matthew Herzig
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Jeremy M. Robbins
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Zsu-Zsu Chen
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Daniel E. Cruz
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Bennet Peterson
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | | | - Thomas J. Wang
- Department of Medicine, UT Southwestern Medical Center, Dallas, TX
| | - James G. Wilson
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Robert E. Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
- Broad Institute of Harvard and MIT, Cambridge, MA
| |
Collapse
|
32
|
Complementary value of ECG and echocardiographic left ventricular hypertrophy for prediction of adverse outcomes in the general population. J Hypertens 2021; 39:548-555. [PMID: 33543885 DOI: 10.1097/hjh.0000000000002652] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To investigate whether ECG left ventricular hypertrophy (ECG-LVH) has prognostic value independent of echocardiography LVH (Echo-LVH). METHODS Participants (N = 9744, mean age, 53.81 ± 10.49 years and 45.5% male) from the Northeast China Rural Cardiovascular Health Study were included. Associations between Echo-LVH (sex-specific left ventricular mass normalized to BSA) and ECG-LVH (diagnosed using the Cornell-voltage duration product) and adverse outcomes were evaluated using Cox regression. The value of ECG-LVH for predicting adverse events was evaluated by reclassification and discrimination analyses. RESULTS Median follow-up was 4.65 years; 563 participants developed incident stroke or coronary heart disease (CHD) and 402 died. Compared with participants without either condition, those with both Echo-LVH and ECG-LVH had a significantly increased risk of incident stroke or CHD (hazard ratio, 2.42; 95% confidence interval, 1.82-3.22) and mortality (2.58; 1.85-3.60). ECG-LVH remained an independent risk factors for both outcomes when ECG-LVH and Echo-LVH were included in the model as separate variables [incident stroke or CHD (1.43; 1.14-1.79); mortality (1.41; 1.08-1.84)]. Reclassification and discrimination analyses indicated ECG-LVH addition could improve the conventional model for predicting adverse outcomes within 4 years. These relationships persisted after excluding participants with cardiovascular disease history or taking antihypertension drugs or upon applying other ECG-LVH and Echo-LVH diagnostic criteria. CONCLUSION Our study provides strong evidence that ECG-LVH is associated with adverse outcomes, independent of Echo-LVH. Clinically, ECG-LVH could be considered as a consequential factor, especially in those with Echo-LVH. These findings have potential clinical relevance for risk stratification.
Collapse
|
33
|
Masjoan Juncos JX, Shakil S, Bradley WE, Wei CC, Zafar I, Powell P, Mariappan N, Louch WE, Ford DA, Ahmad A, Dell'Italia LJ, Ahmad S. Chronic cardiac structural damage, diastolic and systolic dysfunction following acute myocardial injury due to bromine exposure in rats. Arch Toxicol 2021; 95:179-193. [PMID: 32979061 PMCID: PMC7855670 DOI: 10.1007/s00204-020-02919-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/17/2020] [Indexed: 12/16/2022]
Abstract
Accidental bromine spills are common and its large industrial stores risk potential terrorist attacks. The mechanisms of bromine toxicity and effective therapeutic strategies are unknown. Our studies demonstrate that inhaled bromine causes deleterious cardiac manifestations. In this manuscript we describe mechanisms of delayed cardiac effects in the survivors of a single bromine exposure. Rats were exposed to bromine (600 ppm for 45 min) and the survivors were sacrificed at 14 or 28 days. Echocardiography, hemodynamic analysis, histology, transmission electron microscopy (TEM) and biochemical analysis of cardiac tissue were performed to assess functional, structural and molecular effects. Increases in right ventricular (RV) and left ventricular (LV) end-diastolic pressure and LV end-diastolic wall stress with increased LV fibrosis were observed. TEM images demonstrated myofibrillar loss, cytoskeletal breakdown and mitochondrial damage at both time points. Increases in cardiac troponin I (cTnI) and N-terminal pro brain natriuretic peptide (NT-proBNP) reflected myofibrillar damage and increased LV wall stress. LV shortening decreased as a function of increasing LV end-systolic wall stress and was accompanied by increased sarcoendoplasmic reticulum calcium ATPase (SERCA) inactivation and a striking dephosphorylation of phospholamban. NADPH oxidase 2 and protein phosphatase 1 were also increased. Increased circulating eosinophils and myocardial 4-hydroxynonenal content suggested increased oxidative stress as a key contributing factor to these effects. Thus, a continuous oxidative stress-induced chronic myocardial damage along with phospholamban dephosphorylation are critical for bromine-induced chronic cardiac dysfunction. These findings in our preclinical model will educate clinicians and public health personnel and provide important endpoints to evaluate therapies.
Collapse
MESH Headings
- Animals
- Bromine
- Calcium-Binding Proteins/metabolism
- Cardiomegaly/chemically induced
- Cardiomegaly/metabolism
- Cardiomegaly/pathology
- Cardiomegaly/physiopathology
- Cardiotoxicity
- Diastole
- Disease Models, Animal
- Fibrosis
- Male
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/ultrastructure
- Myocardium/metabolism
- Myocardium/ultrastructure
- NADPH Oxidase 2/metabolism
- Natriuretic Peptide, Brain/metabolism
- Oxidative Stress/drug effects
- Peptide Fragments/metabolism
- Phosphorylation
- Protein Phosphatase 1/metabolism
- Rats, Sprague-Dawley
- Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
- Systole
- Time Factors
- Troponin I/metabolism
- Ventricular Dysfunction, Left/chemically induced
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/pathology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Right/chemically induced
- Ventricular Dysfunction, Right/metabolism
- Ventricular Dysfunction, Right/pathology
- Ventricular Dysfunction, Right/physiopathology
- Ventricular Function, Left
- Ventricular Function, Right
- Ventricular Remodeling
- Rats
Collapse
Affiliation(s)
- Juan Xavier Masjoan Juncos
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, #322 BMRII, 901 19th St. South, Birmingham, AL, 35294, USA
| | - Shazia Shakil
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, #322 BMRII, 901 19th St. South, Birmingham, AL, 35294, USA
| | - Wayne E Bradley
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Veterans Affairs Medical Center, Birmingham, AL, USA
| | - Chih-Chang Wei
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Veterans Affairs Medical Center, Birmingham, AL, USA
| | - Iram Zafar
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, #322 BMRII, 901 19th St. South, Birmingham, AL, 35294, USA
| | - Pamela Powell
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Veterans Affairs Medical Center, Birmingham, AL, USA
| | - Nithya Mariappan
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, #322 BMRII, 901 19th St. South, Birmingham, AL, 35294, USA
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- Center for Heart Failure Research, KG Jebsen Cardiac Research Center, University of Oslo, Oslo, Norway
| | - David A Ford
- Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University, St. Louis, MO, USA
| | - Aftab Ahmad
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, #322 BMRII, 901 19th St. South, Birmingham, AL, 35294, USA
| | - Louis J Dell'Italia
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Veterans Affairs Medical Center, Birmingham, AL, USA
| | - Shama Ahmad
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, #322 BMRII, 901 19th St. South, Birmingham, AL, 35294, USA.
| |
Collapse
|
34
|
Corrêa JWDN, Prado CM, Riul ME, Araújo AV, Rossi MA, Bendhack LM. Reversion of cardiovascular remodelling in renovascular hypertensive 2K-1C rats by renin-angiotensin system inhibitors. Clin Exp Pharmacol Physiol 2020; 47:1965-1977. [PMID: 32688435 DOI: 10.1111/1440-1681.13384] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 12/30/2022]
Abstract
OBJECTIVES Evaluate whether the RAS dual blockade would induce additional beneficial effects on cardiovascular remodelling when compared to monotherapy in renal hypertensive two kidneys-one clip (2K-1C) rats. METHODS Hypertensive 2K-1C and normotensive (2K) rats were treated for 14 days with submaximal doses of losartan (LOS), enalapril (ENA), losartan plus enalapril (LOS + ENA) or vehicle (water). Blood pressure and some parameters of cardiovascular remodelling were evaluated. RESULTS Systolic blood pressure (SBP) was higher in 2K-1C (209 ± 3 mm Hg, P < .05) than in 2K (113 ± 1 mm Hg) rats. There was an additional effect in 2K-1C treated with LOS + ENA (153 ± 9 mm Hg) on lowering SBP when compared to LOS (184 ± 12 mm Hg) or ENA (177 ± 9 mm Hg). None of the treatments had effect on SBP in 2K rats. In 2K-1C, cardiomyocyte hypertrophy was reduced by all treatments, although the cardiac hypertrophy indexes remained unchanged. 2K-1C aortas presented medial thickening that was partially reduced by the treatments. Intimal hyperplasia observed in 2K-1C (15.56 ± 0.89 µm vs 8.24 ± 0.80 µm) was reversed by ENA (9.52 ± 0.45 µm) or LOS + ENA (8.17 ± 0.53 µm). Collagen deposition was increased in 2K-1C hearts (1.77 ± 0.16 vs 1.28 ± 0.09) and aortas (8.1 ± 0.6 vs 5.2 ± 0.2). Treatment with LOS reduced (1.12 ± 0.14%) and ENA (0.81 ± 0.11%) or LOS + ENA (0.86 ± 0.11%) additionally diminished collagen only in 2K-1C hearts. CONCLUSIONS Submaximal doses of ACEi and/or ARB have inhibitory actions on cardiac remodelling and vascular hypertrophy not entirely dependent on their effects on blood pressure normalization in renovascular hypertensive rats. Combined therapy produced additional reduction in blood pressure than monotherapy despite a similar inhibition on cardiovascular remodelling.
Collapse
Affiliation(s)
| | - Cibele Maria Prado
- Department of Pathology, School of Medicine of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Maria Elena Riul
- Department of Pathology, School of Medicine of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Alice Valença Araújo
- Nucleus of Public Health, Academic Center of Vitória (CAV), Federal University of Pernambuco (UFPE), Vitória de Santo Antão, Brazil
| | - Marcos Antonio Rossi
- Department of Pathology, School of Medicine of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Lusiane Maria Bendhack
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, Brazil
| |
Collapse
|
35
|
Şahin AA, Yildirim C, Dogan Z, Demir AR, Panc C, Yalcin AA, Kalkan AK, Celik O. Evaluation of early electrocardiographic changes after successful percutaneous stent implantation to isolated coarctation of aorta. J Electrocardiol 2020; 63:124-128. [PMID: 33189064 DOI: 10.1016/j.jelectrocard.2020.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/07/2020] [Accepted: 10/24/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND Coarctation of aorta (CoA) is a congenital obstructive lesion characterized by narrowing of the aorta in which concludes as increase in afterload. Percutaneous stent implantation to CoA is a treatment of choice in older children and adults. Pathology related to CoA mainly caused by increased afterload and left ventricular hypertrophy. Electrocardiographic (ECG) findings are also related to left ventricular hypertrophy (LVH). Evidence shows that, in variety of diseases, the correction of the pathology might normalize ECG findings and ventricular dysfunction related to increase in afterload. Therefore the aim of this study was to compare the pre- and postprocedural ECG findings of the patients who underwent percutaneous intervention for isolated CoA. METHODS After exclusion criterion was applied, 30 patients were included into study, retrospectively. ECG records before the procedure and 3 months after the procedure of the patients were evaluated. The parameters related to LVH, ventricular and atrial conduction were evaluated and compared between pre- and post-procedural ECG records. RESULTS The findings showed that parameters of atrial conduction including P wave maximum duration (p < 0.001) and p wave dispersion (p < 0.001) were significantly decreased after stent implantation. Additionally, ventricular repolarization parameters including QT duration (p = 0.039), Tpe interval (p < 0.001), Tpe / QT (p = 0.038) and Tpe / QTc (p = 0.003) were significantly decreased after stent implantation. Sokolow-Lyon criteria (p < 0.003) and voltage in selected leads were significantly decreased after intervention. CONCLUSION Percutaneous intervention to CoA might regress LVH parameters in ECG and improve atrial and ventricular repolarization in ECG, which might lead to decreased event of atrial and ventricular arrhythmias in patients with isolated CoA.
Collapse
Affiliation(s)
- Ahmet Anıl Şahin
- Department Cardiology, University of Health Sciences, Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey; Department of Cardiology, Halic University, School of Medicine, Istanbul, Turkey.
| | - Ceren Yildirim
- Department Cardiology, University of Health Sciences, Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Zekeriya Dogan
- Department of Cardiology, Marmara University, School of Medicine, Istanbul, Turkey
| | - Ali Rıza Demir
- Department Cardiology, University of Health Sciences, Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Cafer Panc
- Department Cardiology, University of Health Sciences, Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Ahmet Arif Yalcin
- Department Cardiology, University of Health Sciences, Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Ali Kemal Kalkan
- Department Cardiology, University of Health Sciences, Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Omer Celik
- Department Cardiology, University of Health Sciences, Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
36
|
Mohan M, Al-Talabany S, McKinnie A, Mordi IR, Singh JSS, Gandy SJ, Baig F, Hussain MS, Bhalraam U, Khan F, Choy AM, Matthew S, Houston JG, Struthers AD, George J, Lang CC. A randomized controlled trial of metformin on left ventricular hypertrophy in patients with coronary artery disease without diabetes: the MET-REMODEL trial. Eur Heart J 2020; 40:3409-3417. [PMID: 30993313 PMCID: PMC6823615 DOI: 10.1093/eurheartj/ehz203] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/01/2019] [Accepted: 04/02/2019] [Indexed: 12/25/2022] Open
Abstract
Aim We tested the hypothesis that metformin may regress left ventricular hypertrophy (LVH) in patients who have coronary artery disease (CAD), with insulin resistance (IR) and/or pre-diabetes. Methods and results We randomly assigned 68 patients (mean age 65 ± 8 years) without diabetes who have CAD with IR and/or pre-diabetes to receive either metformin XL (2000 mg daily dose) or placebo for 12 months. Primary endpoint was change in left ventricular mass indexed to height1.7 (LVMI), assessed by magnetic resonance imaging. In the modified intention-to-treat analysis (n = 63), metformin treatment significantly reduced LVMI compared with placebo group (absolute mean difference −1.37 (95% confidence interval: −2.63 to −0.12, P = 0.033). Metformin also significantly reduced other secondary study endpoints such as: LVM (P = 0.032), body weight (P = 0.001), subcutaneous adipose tissue (P = 0.024), office systolic blood pressure (BP, P = 0.022) and concentration of thiobarbituric acid reactive substances, a biomarker for oxidative stress (P = 0.04). The glycated haemoglobin A1C concentration and fasting IR index did not differ between study groups at the end of the study. Conclusion Metformin treatment significantly reduced LVMI, LVM, office systolic BP, body weight, and oxidative stress. Although LVH is a good surrogate marker of cardiovascular (CV) outcome, conclusive evidence for the cardio-protective role of metformin is required from large CV outcomes trials. ![]()
Collapse
Affiliation(s)
- Mohapradeep Mohan
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - Shaween Al-Talabany
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - Angela McKinnie
- NHS Tayside Clinical Radiology, Ninewells Hospital & Medical School, Dundee, DD1 9SY, UK
| | - Ify R Mordi
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - Jagdeep S S Singh
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - Stephen J Gandy
- Department of Medical Physics, NHS Tayside, Ninewells Hospital & Medical School, Dundee, DD1 9SY, UK
| | - Fatima Baig
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - Muhammad S Hussain
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - U Bhalraam
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - Faisel Khan
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - Anna-Maria Choy
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - Shona Matthew
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - John Graeme Houston
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - Allan D Struthers
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - Jacob George
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - Chim C Lang
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK
| |
Collapse
|
37
|
Djordjevic DB, Tasic IS, Kostic ST, Stamenkovic BN, Lovic MB, Djordjevic ND, Koracevic GP, Lovic DB. Electrocardiographic criteria which have the best prognostic significance in hypertensive patients with echocardiographic hypertrophy of left ventricle: 15-year prospective study. Clin Cardiol 2020; 43:1017-1023. [PMID: 32492247 PMCID: PMC7462184 DOI: 10.1002/clc.23402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/15/2020] [Accepted: 05/26/2020] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Electrocardiography is the first-choice technique for detecting left ventricular hypertrophy in patients with arterial hypertension. It is necessary to know the probable outcome for every patient during the treatment, with the aim of improving cardiovascular event prevention. HYPOTHESIS Certain electrocardiographic criteria for left ventricular hypertrophy may predict outcomes of patients with left ventricular hypertrophy during a 15-year follow-up. METHODS Fifteen-year prospective study of 83 consecutive patients (53 male and 30 female; mean age 55.3 ± 8.1) with echocardiographic left ventricular hypertrophy (left ventricular mass index 170.3 ± 31.6 g/m2 ). Electrocardiographic left ventricular hypertrophy was determined by means of Gubner-Ungerleider voltage, Lewis voltage, voltage of R wave in aVL lead, Lyon-Sokolow voltage, Cornell voltage and Cornell product, voltage RV6 and RV5 ratio, Romhilt-Estes score, Framingham criterion and Perugia criterion. RESULTS One or more composite events were registered in 32 (38.5%) patients during 15-year follow-up. Positive Lyon-Sokolow score (17.6% vs. 47.3%; P < 0.05), Lewis voltage (9.8% vs. 21.9%; P < 0.05), Cornell voltage (15.7% vs. 37.5%; P < 0.05), and Cornell product (9.8% vs. 34.4%; P < 0.01) were more frequent in a group of patients with composite events. Odd ratio for Cornell product was 4.819 (95% CI 1.486-15.627). CONCLUSION Patients with echocardiographic left ventricular hypertrophy who had positive Lewis voltage, Lyon-Sokolow voltage, Cornell voltage, and Cornell product showed worse 15-year outcome. The strongest predictor of cardiovascular events was positive result of Cornell product.
Collapse
Affiliation(s)
| | - Ivan S. Tasic
- University of NisMedical FacultyNisSerbia
- Institute Niska BanjaNisSerbia
| | | | | | | | | | | | - Dragan B. Lovic
- Singidunum University School of MedicineClinic for Internal Diseases IntermedicaNisSerbia
- Veterans Affairs Medical CenterWashingtonDistrict of ColumbiaUSA
| |
Collapse
|
38
|
Han BG, Lee JY, Kim MR, Shin H, Kim JS, Yang JW, Kim JY. Fluid overload is a determinant for cardiac structural and functional impairments in type 2 diabetes mellitus and chronic kidney disease stage 5 not undergoing dialysis. PLoS One 2020; 15:e0235640. [PMID: 32730268 PMCID: PMC7392282 DOI: 10.1371/journal.pone.0235640] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 06/19/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Fluid overload is common in patients with diabetes and chronic kidney disease (DM and CKD; DMCKD) and can lead to structural and functional cardiac abnormalities including left ventricular hypertrophy (LVH) and left ventricular diastolic dysfunction (LVDD). Fluid overload represents a crucial step in the pathophysiological pathways to chronic heart failure in patients with end-stage renal disease. We evaluated the impact of fluid overload on cardiac alterations in patients with diabetes and non-dialysis-dependent CKD stage 5 (DMCKD5-ND) without intrinsic heart disease. METHODS Bioimpedance spectroscopy, echocardiography, and N-terminal prohormone of B-type natriuretic peptide (NT-proBNP) measurement were performed in 135 consecutive patients on the same day. Patients were divided into groups by tertiles of overhydration/extracellular water (OH/ECW) per bioimpedance spectroscopy. RESULTS Fluid balance markers including OH/ECW and NT-proBNP were significantly higher in the LVDD+LVH group. OH/ECW and its exacerbation were positively associated with the ratio between early mitral inflow and annular early diastolic velocities (E/e' ratio) and left ventricular mass index (LVMI). The prevalence of LVH progressively increased across increasing tertiles of OH/ECW. In multiple regression analyses, OH/ECW as a continuous and categorical variable was independently associated with the E/e' ratio and LVMI after adjustment for multiple confounding factors. CONCLUSIONS Fluid overload was independently associated with LVDD and LVH in patients with DMCKD5-ND. Our study suggests that structural and functional cardiac abnormalities and volume status should be evaluated simultaneously in patients with early-stage DMCKD rather than only DMCKD5-ND, in addition to intensive blood pressure and glycemic control, regardless of evident cardiovascular disease.
Collapse
Affiliation(s)
- Byoung-Geun Han
- Department of Nephrology, Yonsei University Wonju College of Medicine, Wonju, Kang-won, Korea
| | - Jun Young Lee
- Department of Nephrology, Yonsei University Wonju College of Medicine, Wonju, Kang-won, Korea
| | - Mi Ryung Kim
- Department of Nephrology, Yonsei University Wonju College of Medicine, Wonju, Kang-won, Korea
| | - Hanwul Shin
- Department of Nephrology, Yonsei University Wonju College of Medicine, Wonju, Kang-won, Korea
| | - Jae-Seok Kim
- Department of Nephrology, Yonsei University Wonju College of Medicine, Wonju, Kang-won, Korea
| | - Jae-Won Yang
- Department of Nephrology, Yonsei University Wonju College of Medicine, Wonju, Kang-won, Korea
| | - Jong Yeon Kim
- Department of Neurosurgery, Yonsei University Wonju College of Medicine, Wonju, Kang-won, Korea
- * E-mail:
| |
Collapse
|
39
|
Promoting role of circ-Jarid2/miR-129-5p/Celf1 axis in cardiac hypertrophy. Gene Ther 2020; 28:718-728. [PMID: 32632266 DOI: 10.1038/s41434-020-0165-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 05/20/2020] [Accepted: 06/02/2020] [Indexed: 11/09/2022]
Abstract
Cardiac hypertrophy is imposed much pressure on heart and threatening our live. Previous study suggested that dysregulation of Celf1 is largely connecting to neonatal cardiac dysfunction. Hence, we aimed to explore the precise function and probable regulatory mechanism upstream of Celf1in cardiac hypertrophy. Here, Ang-II treatment was implemented to stimulate hypertrophic phenotypes inH9C2 and MCM cells. Immunofluorescence assay was conducted to measure the surface area of cardiomyocytes. And qRT-PCR assay was conducted to investigate gene expression. Moreover, western blot assay was conducted to probe the protein levels. Results uncovered that Celf1 expression was increased dependent on elevated Ang-II concentration, and that inhibited Celf1 could relieve the Ang-II-caused cardiac hypertrophy. Significantly, Celf1was found to be targeted by miR-129-5p but then released via the sponging role of circ-Jarid2. Furthermore, circ-Jarid2 was found to promote cardiac hypertrophy, whereas miR-129-5p played suppressing parts in hypertrophic cardiomyocytes. Moreover, we verified circ-Jarid2 contributed to cardiac hypertrophy via miR-129-5p/Celf1 axis both in vitro and in vivo. In conclusion, circ-Jarid2/miR-129-5p/Celf1 axis aggravates cardiac hypertrophy, which provides new ideas for developing treatment strategies for patients with cardiac hypertrophy.
Collapse
|
40
|
Siasos G, Bletsa E, Stampouloglou PK, Oikonomou E, Tsigkou V, Paschou SA, Vlasis K, Marinos G, Vavuranakis M, Stefanadis C, Tousoulis D. MicroRNAs in cardiovascular disease. Hellenic J Cardiol 2020; 61:165-173. [PMID: 32305497 DOI: 10.1016/j.hjc.2020.03.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/08/2020] [Accepted: 03/19/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease (CVD) remains the predominant cause of human morbidity and mortality in developed countries. Currently, microRNAs have been investigated in many diseases as well-promising biomarkers for diagnosis, prognosis, and disease monitoring. Plenty studies have been designed so as to elucidate the properties of microRNAs in the classification and risk stratification of patients with CVD and also to evaluate their potentials in individualized management and guide treatment decisions. Therefore, in this review article, we aimed to present the most recent data concerning the role of microRNAs as potential novel biomarkers for cardiovascular disease.
Collapse
Affiliation(s)
- Gerasimos Siasos
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece; Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Evanthia Bletsa
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Panagiota K Stampouloglou
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Evangelos Oikonomou
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Vasiliki Tsigkou
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Stavroula A Paschou
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Konstantinos Vlasis
- Department of Anatomy, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgios Marinos
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Manolis Vavuranakis
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Christodoulos Stefanadis
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Dimitris Tousoulis
- Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| |
Collapse
|
41
|
Prognostic utility of electrocardiograms in patients with hypertension older than 65 years. The PAFRES study. Rev Clin Esp 2020. [DOI: 10.1016/j.rceng.2019.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
42
|
Association of Urine Albumin/Creatinine Ratio below 30 mg/g and Left Ventricular Hypertrophy in Patients with Type 2 Diabetes. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5240153. [PMID: 32076606 PMCID: PMC6996706 DOI: 10.1155/2020/5240153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/18/2019] [Accepted: 12/23/2019] [Indexed: 01/19/2023]
Abstract
Several studies show that even a level of urine albumin/creatinine ratio (UACR) within the normal range (below 30 mg/g) increases the risk of cardiovascular diseases. We speculate that mildly increased UACR is related to left ventricular hypertrophy (LVH) in patients with type 2 diabetes mellitus (T2DM). In this retrospective study, 317 patients with diabetes with normal UACR, of whom 62 had LVH, were included. The associations between UACR and laboratory indicators, as well as LVH, were examined using multivariate linear regression and logistic regression, respectively. The diagnostic efficiency and the optimal cutoff point of UACR for LVH were evaluated using the area under the receiver operating characteristic curve (AUC) and Youden index. Our results showed that patients with LVH had significantly higher UACR than those without LVH (P < 0.001). The prevalence of LVH presented an upward trend with the elevation of UACR. UACR was independently and positively associated with hemoglobin A1c (P < 0.001). UACR can differentiate LVH (AUC = 0.682, 95% CI (0.602–0.760), P < 0.001). The optimal cutoff point determined with the Youden index was UACR = 10.2 mg/g. When categorized by this cutoff point, the odds ratio (OR) for LVH in patients in the higher UACR group (10.2–30 mg/g) was 3.104 (95% CI: 1.557–6.188, P=0.001) compared with patients in the lower UACR group (<10.2 mg/g). When UACR was analyzed as a continuous variable, every double of increased UACR, the OR for LVH was 1.511 (95% CI: 1.047–2.180, P=0.028). Overall, UACR below 30 mg/g is associated with LVH in patients with T2DM. The optimal cutoff value of UACR for identifying LVH in diabetes is 10 mg/g.
Collapse
|
43
|
Saraiva RM. Do We Need to Know the Left Ventricular Geometry Patterns of the Brazilian Population? Arq Bras Cardiol 2020; 114:66-67. [PMID: 32049172 PMCID: PMC7025312 DOI: 10.36660/abc.20190838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Roberto M Saraiva
- Fundação Oswaldo Cruz - Instituto Nacional de Infectologia Evandro Chagas, Rio de Janeiro, RJ - Brazil
| |
Collapse
|
44
|
Kawel-Boehm N, Kronmal R, Eng J, Folsom A, Burke G, Carr JJ, Shea S, Lima JAC, Bluemke DA. Left Ventricular Mass at MRI and Long-term Risk of Cardiovascular Events: The Multi-Ethnic Study of Atherosclerosis (MESA). Radiology 2019; 293:107-114. [PMID: 31453766 PMCID: PMC6776886 DOI: 10.1148/radiol.2019182871] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 07/02/2019] [Accepted: 07/12/2019] [Indexed: 01/19/2023]
Abstract
Background Few data exist on the long-term risk prediction of elevated left ventricular (LV) mass quantified by MRI for cardiovascular (CV) events in a contemporary, ethnically diverse cohort. Purpose To assess the long-term impact of elevated LV mass on CV events in a prospective cohort study of a multiethnic population in relationship to risk factors and coronary artery calcium (CAC) score. Materials and Methods The Multi-Ethnic Study of Atherosclerosis, or MESA (ClinicalTrials.gov: NCT00005487), is an ongoing prospective multicenter population-based study in the United States. A total of 6814 participants (age range, 45-84 years) free of clinical CV disease at baseline were enrolled between 2000 and 2002. In 4988 participants (2613 [52.4%] women; mean age, 62 years ± 10.1 [standard deviation]) followed over 15 years for CV events, LV mass was derived from cardiac MRI at baseline enrollment by using semiautomated software at a central core laboratory. Cox proportional hazard models, Kaplan-Meier curves, and z scores were applied to assess the impact of LV hypertrophy. Results A total of 290 participants had hard coronary heart disease (CHD) events (207 myocardial infarctions [MIs], 95 CHD deaths), 57 had other CV disease-related deaths, and 215 had heart failure (HF). LV hypertrophy was an independent predictor of hard CHD events (hazard ratio [HR]: 2.7; 95% confidence interval [CI]: 1.9, 3.8), MI (HR: 2.8; 95% CI: 1.8, 4.0), CHD death (HR: 4.3; 95% CI: 2.5, 7.3), other CV death (HR: 7.5; 95% CI: 4.2, 13.5), and HF (HR: 5.4; 95% CI: 3.8, 7.5) (P < .001 for all end points). LV hypertrophy was a stronger predictor than CAC for CHD death, other CV death, and HF (z scores: 5.4 vs 3.4, 6.8 vs 2.4, and 9.7 vs 3.2 for LV hypertrophy vs CAC, respectively). Kaplan-Meier analysis demonstrated an increased risk of CV events in participants with LV hypertrophy, particularly after 5 years. Conclusion Elevated left ventricular mass was strongly associated with hard coronary heart disease events, other cardiovascular death, and heart failure over 15 years of follow-up, independent of traditional risk factors and coronary artery calcium score. © RSNA, 2019 See also the editorial by Hanneman in this issue.
Collapse
Affiliation(s)
- Nadine Kawel-Boehm
- From the Department of Radiology, Kantonsspital Graubuenden,
Loestrasse 170, 7000 Chur, Switzerland (N.K.); Collaborative Health Studies
Coordinating Center, University of Washington, Seattle, Wash (R.K.); Department
of Radiology and Radiological Science (J.E.) and Division of Cardiology
(J.A.C.L.), Johns Hopkins University, Baltimore, Md; Division of Epidemiology
and Community Health, University of Minnesota, Minneapolis, Minn (A.F.);
Department of Public Health Sciences, Wake Forest University, Winston-Salem, NC
(G.B.); Department of Radiology, Vanderbilt University Medical Center,
Nashville, Tenn (J.J.C.); Department of Medicine, Columbia University, New York,
NY (S.S.); and Department of Radiology, University of Wisconsin School of
Medicine and Public Health, Madison, Wis (D.A.B.)
| | - Richard Kronmal
- From the Department of Radiology, Kantonsspital Graubuenden,
Loestrasse 170, 7000 Chur, Switzerland (N.K.); Collaborative Health Studies
Coordinating Center, University of Washington, Seattle, Wash (R.K.); Department
of Radiology and Radiological Science (J.E.) and Division of Cardiology
(J.A.C.L.), Johns Hopkins University, Baltimore, Md; Division of Epidemiology
and Community Health, University of Minnesota, Minneapolis, Minn (A.F.);
Department of Public Health Sciences, Wake Forest University, Winston-Salem, NC
(G.B.); Department of Radiology, Vanderbilt University Medical Center,
Nashville, Tenn (J.J.C.); Department of Medicine, Columbia University, New York,
NY (S.S.); and Department of Radiology, University of Wisconsin School of
Medicine and Public Health, Madison, Wis (D.A.B.)
| | - John Eng
- From the Department of Radiology, Kantonsspital Graubuenden,
Loestrasse 170, 7000 Chur, Switzerland (N.K.); Collaborative Health Studies
Coordinating Center, University of Washington, Seattle, Wash (R.K.); Department
of Radiology and Radiological Science (J.E.) and Division of Cardiology
(J.A.C.L.), Johns Hopkins University, Baltimore, Md; Division of Epidemiology
and Community Health, University of Minnesota, Minneapolis, Minn (A.F.);
Department of Public Health Sciences, Wake Forest University, Winston-Salem, NC
(G.B.); Department of Radiology, Vanderbilt University Medical Center,
Nashville, Tenn (J.J.C.); Department of Medicine, Columbia University, New York,
NY (S.S.); and Department of Radiology, University of Wisconsin School of
Medicine and Public Health, Madison, Wis (D.A.B.)
| | - Aaron Folsom
- From the Department of Radiology, Kantonsspital Graubuenden,
Loestrasse 170, 7000 Chur, Switzerland (N.K.); Collaborative Health Studies
Coordinating Center, University of Washington, Seattle, Wash (R.K.); Department
of Radiology and Radiological Science (J.E.) and Division of Cardiology
(J.A.C.L.), Johns Hopkins University, Baltimore, Md; Division of Epidemiology
and Community Health, University of Minnesota, Minneapolis, Minn (A.F.);
Department of Public Health Sciences, Wake Forest University, Winston-Salem, NC
(G.B.); Department of Radiology, Vanderbilt University Medical Center,
Nashville, Tenn (J.J.C.); Department of Medicine, Columbia University, New York,
NY (S.S.); and Department of Radiology, University of Wisconsin School of
Medicine and Public Health, Madison, Wis (D.A.B.)
| | - Gregory Burke
- From the Department of Radiology, Kantonsspital Graubuenden,
Loestrasse 170, 7000 Chur, Switzerland (N.K.); Collaborative Health Studies
Coordinating Center, University of Washington, Seattle, Wash (R.K.); Department
of Radiology and Radiological Science (J.E.) and Division of Cardiology
(J.A.C.L.), Johns Hopkins University, Baltimore, Md; Division of Epidemiology
and Community Health, University of Minnesota, Minneapolis, Minn (A.F.);
Department of Public Health Sciences, Wake Forest University, Winston-Salem, NC
(G.B.); Department of Radiology, Vanderbilt University Medical Center,
Nashville, Tenn (J.J.C.); Department of Medicine, Columbia University, New York,
NY (S.S.); and Department of Radiology, University of Wisconsin School of
Medicine and Public Health, Madison, Wis (D.A.B.)
| | - J. Jeffrey Carr
- From the Department of Radiology, Kantonsspital Graubuenden,
Loestrasse 170, 7000 Chur, Switzerland (N.K.); Collaborative Health Studies
Coordinating Center, University of Washington, Seattle, Wash (R.K.); Department
of Radiology and Radiological Science (J.E.) and Division of Cardiology
(J.A.C.L.), Johns Hopkins University, Baltimore, Md; Division of Epidemiology
and Community Health, University of Minnesota, Minneapolis, Minn (A.F.);
Department of Public Health Sciences, Wake Forest University, Winston-Salem, NC
(G.B.); Department of Radiology, Vanderbilt University Medical Center,
Nashville, Tenn (J.J.C.); Department of Medicine, Columbia University, New York,
NY (S.S.); and Department of Radiology, University of Wisconsin School of
Medicine and Public Health, Madison, Wis (D.A.B.)
| | - Steven Shea
- From the Department of Radiology, Kantonsspital Graubuenden,
Loestrasse 170, 7000 Chur, Switzerland (N.K.); Collaborative Health Studies
Coordinating Center, University of Washington, Seattle, Wash (R.K.); Department
of Radiology and Radiological Science (J.E.) and Division of Cardiology
(J.A.C.L.), Johns Hopkins University, Baltimore, Md; Division of Epidemiology
and Community Health, University of Minnesota, Minneapolis, Minn (A.F.);
Department of Public Health Sciences, Wake Forest University, Winston-Salem, NC
(G.B.); Department of Radiology, Vanderbilt University Medical Center,
Nashville, Tenn (J.J.C.); Department of Medicine, Columbia University, New York,
NY (S.S.); and Department of Radiology, University of Wisconsin School of
Medicine and Public Health, Madison, Wis (D.A.B.)
| | - João A. C. Lima
- From the Department of Radiology, Kantonsspital Graubuenden,
Loestrasse 170, 7000 Chur, Switzerland (N.K.); Collaborative Health Studies
Coordinating Center, University of Washington, Seattle, Wash (R.K.); Department
of Radiology and Radiological Science (J.E.) and Division of Cardiology
(J.A.C.L.), Johns Hopkins University, Baltimore, Md; Division of Epidemiology
and Community Health, University of Minnesota, Minneapolis, Minn (A.F.);
Department of Public Health Sciences, Wake Forest University, Winston-Salem, NC
(G.B.); Department of Radiology, Vanderbilt University Medical Center,
Nashville, Tenn (J.J.C.); Department of Medicine, Columbia University, New York,
NY (S.S.); and Department of Radiology, University of Wisconsin School of
Medicine and Public Health, Madison, Wis (D.A.B.)
| | - David A. Bluemke
- From the Department of Radiology, Kantonsspital Graubuenden,
Loestrasse 170, 7000 Chur, Switzerland (N.K.); Collaborative Health Studies
Coordinating Center, University of Washington, Seattle, Wash (R.K.); Department
of Radiology and Radiological Science (J.E.) and Division of Cardiology
(J.A.C.L.), Johns Hopkins University, Baltimore, Md; Division of Epidemiology
and Community Health, University of Minnesota, Minneapolis, Minn (A.F.);
Department of Public Health Sciences, Wake Forest University, Winston-Salem, NC
(G.B.); Department of Radiology, Vanderbilt University Medical Center,
Nashville, Tenn (J.J.C.); Department of Medicine, Columbia University, New York,
NY (S.S.); and Department of Radiology, University of Wisconsin School of
Medicine and Public Health, Madison, Wis (D.A.B.)
| |
Collapse
|
45
|
The microRNA in ventricular remodeling: the miR-30 family. Biosci Rep 2019; 39:BSR20190788. [PMID: 31320543 PMCID: PMC6680373 DOI: 10.1042/bsr20190788] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/07/2019] [Accepted: 07/16/2019] [Indexed: 12/14/2022] Open
Abstract
Ventricular remodeling (VR) is a complex pathological process of cardiomyocyte apoptosis, cardiac hypertrophy, and myocardial fibrosis, which is often caused by various cardiovascular diseases (CVDs) such as hypertension, acute myocardial infarction, heart failure (HF), etc. It is also an independent risk factor for a variety of CVDs, which will eventually to damage the heart function, promote cardiovascular events, and lead to an increase in mortality. MicroRNAs (miRNAs) can participate in a variety of CVDs through post-transcriptional regulation of target gene proteins. Among them, microRNA-30 (miR-30) is one of the most abundant miRNAs in the heart. In recent years, the study found that the miR-30 family can participate in VR through a variety of mechanisms, including autophagy, apoptosis, oxidative stress, and inflammation. VR is commonly found in ischemic heart disease (IHD), hypertensive heart disease (HHD), diabetic cardiomyopathy (DCM), antineoplastic drug cardiotoxicity (CTX), and other CVDs. Therefore, we will review the relevant mechanisms of the miR-30 in VR induced by various diseases.
Collapse
|
46
|
Kattih B, Elling LS, Weiss C, Bea M, Zwadlo C, Bavendiek U, Bauersachs J, Heineke J. Anti-androgenic therapy with finasteride in patients with chronic heart failure - a retrospective propensity score based analysis. Sci Rep 2019; 9:10139. [PMID: 31300720 PMCID: PMC6626053 DOI: 10.1038/s41598-019-46640-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 07/01/2019] [Indexed: 12/20/2022] Open
Abstract
Sex hormones influence the prevalence and the outcome of heart diseases. The conversion of testosterone to its more active metabolite dihydrotestosterone drives cardiac growth and dysfunction, while inhibition of this step by the anti-androgenic drug finasteride counteracts these pathological processes in preclinical models. In this retrospective, observational study, we aim to investigate whether finasteride, which is in clinical use mainly for prostate disease, might ameliorate cardiac hypertrophy and heart failure in patients. Retrospective chart review of 1041 medical cases with heart failure between 1995 and 2015 was conducted. Stratification was performed by concomitant prostate treatment status (tamsulosin versus finasteride). A propensity score analysis yielded a total of 328 matched medical cases without residual differences in the baseline patient characteristics. In this propensity score matched samples, anti-androgenic therapy with finasteride was associated with significantly reduced left ventricular hypertrophy (interventricular septal thickness 13.3 ± 2.4 mm control vs. 12.6 ± 2.1 mm finasteride group (p = 0.029); estimated average treatment effects on the treated: −0.7 mm, 95% CI mean difference −1.3 to −0.1). In this retrospective analysis anti-androgenic therapy with finasteride for prostate disease was associated with attenuated cardiac hypertrophy in patients with heart failure. Therefore, our data encourage further analysis of this approach in larger heart failure patient cohorts.
Collapse
Affiliation(s)
- Badder Kattih
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Street 1, 30625, Hannover, Germany. .,Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim of Heidelberg University, University Medical Centre Mannheim, Ludolf-Krehl Street 7-11, 68167, Mannheim, Germany.
| | - Lukas Simon Elling
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Street 1, 30625, Hannover, Germany
| | - Christel Weiss
- Department for Medical Statistics and Biomathematics, Medical Faculty Mannheim of Heidelberg University, University Medical Centre Mannheim, Ludolf-Krehl Street 9-13, 68167, Mannheim, Germany
| | - Marieke Bea
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Street 1, 30625, Hannover, Germany
| | - Carolin Zwadlo
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Street 1, 30625, Hannover, Germany
| | - Udo Bavendiek
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Street 1, 30625, Hannover, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Street 1, 30625, Hannover, Germany
| | - Joerg Heineke
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Street 1, 30625, Hannover, Germany. .,Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim of Heidelberg University, University Medical Centre Mannheim, Ludolf-Krehl Street 7-11, 68167, Mannheim, Germany.
| |
Collapse
|
47
|
Sun D, Li C, Liu J, Wang Z, Liu Y, Luo C, Chen Y, Wen S. Expression Profile of microRNAs in Hypertrophic Cardiomyopathy and Effects of microRNA-20 in Inducing Cardiomyocyte Hypertrophy Through Regulating Gene MFN2. DNA Cell Biol 2019; 38:796-807. [PMID: 31295012 DOI: 10.1089/dna.2019.4731] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Myocardial hypertrophy is an important cause of heart failure and sudden death. Studies have shown that Mitofusin-2 (MFN2) is downregulated in myocardial hypertrophy, but the upstream regulation mechanism underlying its downexpression in cardiomyocytes is still unclear. This study aims to identify the expression profile of microRNAs (miRNAs) in hypertrophic cardiomyopathy (HCM) and explore the function of miRNA-20 in inducing cardiomyocyte hypertrophy through regulating MFN2. Through miRNA + mRNA microarray analysis, 1451 miRNAs were identified, 367 miRNAs expressed differently between groups. Meanwhile, a number of 24,718 mRNAs were identified, among which 5850 mRNAs were upregulated and 3005 mRNAs were downregulated in HCM group compared with the control group. Expression of hsa-miRNA-20a-5p was 2.26 times higher in the HCM group compared with the control group and 7 target gene prediction programs predicted MFN2 as a target of miRNA-20. In vitro model of hypertrophic cardiomyocytes displayed high expression level of miRNA-20, atrial natriuretic peptide (ANP) mRNA, and protein, accompanying low expression level of Mfn2 mRNA and protein, which meant miRNA-20 played a role in cardiomyocyte hypertrophy and might interact with MFN2 to function. Thereafter, overexpression of miRNA-20 led to cell hypertrophy accompanied with lowly expressed Mfn2 mRNA and protein. When transfected with miRNA-20 inhibitors, the expression of miRNA-20 and ANP gene was attenuated and MFN2 was the other way around. The cell surface area of Ang II group and mimic group was significantly larger compared with the control group, and in the inhibitor+Ang II group, the area was significantly decreased compared with the Ang II group. Dual-luciferase assays showed that miRNA-20 bound to 3' untranslated region of MFN2 and inhibited its expression. In conclusion, hypertrophic myocardium and normal myocardium have different miRNA expression profiles and the effect of miRNA-20 reducing the expression of MFN2 plays a role in promoting cardiomyocyte hypertrophy.
Collapse
Affiliation(s)
- Dongdong Sun
- 1Department of Hypertension Research, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,2Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China.,3Beijing Laboratory for Cardiovascular Precision Medicine (PXM2017_014226_000037), Beijing, China
| | - Chuang Li
- 1Department of Hypertension Research, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,2Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China.,3Beijing Laboratory for Cardiovascular Precision Medicine (PXM2017_014226_000037), Beijing, China
| | - Jielin Liu
- 1Department of Hypertension Research, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,2Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China.,3Beijing Laboratory for Cardiovascular Precision Medicine (PXM2017_014226_000037), Beijing, China
| | - Zuoguang Wang
- 1Department of Hypertension Research, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,2Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China.,3Beijing Laboratory for Cardiovascular Precision Medicine (PXM2017_014226_000037), Beijing, China
| | - Ya Liu
- 1Department of Hypertension Research, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,2Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China.,3Beijing Laboratory for Cardiovascular Precision Medicine (PXM2017_014226_000037), Beijing, China
| | - Chen Luo
- 1Department of Hypertension Research, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,2Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China.,3Beijing Laboratory for Cardiovascular Precision Medicine (PXM2017_014226_000037), Beijing, China
| | - Yanyu Chen
- 1Department of Hypertension Research, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,2Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China.,3Beijing Laboratory for Cardiovascular Precision Medicine (PXM2017_014226_000037), Beijing, China
| | - Shaojun Wen
- 1Department of Hypertension Research, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,2Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China.,3Beijing Laboratory for Cardiovascular Precision Medicine (PXM2017_014226_000037), Beijing, China
| |
Collapse
|
48
|
Guedes Ramallo P, Morillas Blasco P, Gómez Martínez MJ, Núñez Martínez L, Romero Valero A, Peris Castelló F, Rodríguez Santiago FM, Vicente Ibarra N, Quintanilla Tello MA, Castilla Cabanes E, Fácila Rubio L, Pallarés-Carratalá V. Prognostic utility of electrocardiograms in patients with hypertension older than 65 years. The PAFRES study. Rev Clin Esp 2019; 220:100-108. [PMID: 31272678 DOI: 10.1016/j.rce.2019.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/04/2019] [Indexed: 10/26/2022]
Abstract
OBJECTIVE Left ventricular hypertrophy is the most common marker of target organ damage in arterial hypertension. Electrocardiograms are typically performed to identify left ventricular hypertrophy. The aim of this study was to analyse the prognostic utility of other electrocardiographic abnormalities in patients with arterial hypertension, beyond ventricular hypertrophy. MATERIALS AND METHODS The study included 1003 patients older than 65years with arterial hypertension. We recorded risk factors, previous cardiovascular history and medical treatment and analysed various electrocardiographic abnormalities including the Sokolow-Lyon index, the Cornell index, ventricular overload and branch blocks. The study conducted a 2-year follow-up, recording the major cardiovascular events (mortality, myocardial infarction, stroke and hospitalisation for heart failure). RESULTS The study population's mean age was 72.9±5.8years, 47.5% of whom were men. During the follow-up, 13.9% of the patients experienced a major cardiovascular event. These patients were older, more often smokers and engaged in less physical exercise, without presenting differences in the antihypertensive therapy or blood pressure control. The ventricular overload pattern (HR: 1.93; 95%CI: 1.160-3.196; P=.011) and the complete left bundle branch block (HR: 2.27; 95%CI: 1.040-4.956; P=.040) behaved as independent electrocardiographic predictors of major cardiovascular events; however, left ventricular hypertrophy using the Sokolow and/or Cornell index did not behave as such. CONCLUSIONS For patients with hypertension, the presence in the baseline electrocardiogram of complete left bundle branch block or a pattern of ventricular overload identifies a population at increased cardiovascular risk.
Collapse
Affiliation(s)
- P Guedes Ramallo
- Servicio de Cardiología, Hospital General Universitario de Elche, Elche, Alicante, España.
| | - P Morillas Blasco
- Servicio de Cardiología, Hospital General Universitario de Elche, Elche, Alicante, España
| | - M J Gómez Martínez
- Servicio de Cardiología, Hospital General Universitario de Elche, Elche, Alicante, España
| | - L Núñez Martínez
- Servicio de Cardiología, Hospital General Universitario de Elche, Elche, Alicante, España
| | - A Romero Valero
- Servicio de Cardiología, Hospital General Universitario de Elche, Elche, Alicante, España
| | - F Peris Castelló
- Servicio de Cardiología, Hospital General Universitario de Elche, Elche, Alicante, España
| | - F M Rodríguez Santiago
- Servicio de Cardiología, Hospital General Universitario de Elche, Elche, Alicante, España
| | - N Vicente Ibarra
- Servicio de Cardiología, Hospital General Universitario de Elche, Elche, Alicante, España
| | - M A Quintanilla Tello
- Servicio de Cardiología, Hospital General Universitario de Elche, Elche, Alicante, España
| | - E Castilla Cabanes
- Servicio de Cardiología, Hospital General Universitario de Elche, Elche, Alicante, España
| | - L Fácila Rubio
- Servicio de Cardiología, Hospital General de Valencia, Valencia, España
| | - V Pallarés-Carratalá
- Unidad de Vigilancia de la Salud, Unión de Mutuas. Departamento de Medicina, Universitat Jaume I, Castellón, España
| |
Collapse
|
49
|
Left ventricular mass in offspring of diabetic mothers: at 5–7 years old. Int J Diabetes Dev Ctries 2019. [DOI: 10.1007/s13410-018-0625-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
50
|
Prasad GVR, Yan AT, Nash MM, Kim SJ, Wald R, Wald R, Lok C, Gunaratnam L, Karur GR, Kirpalani A, Connelly PW. Determinants of Left Ventricular Characteristics Assessed by Cardiac Magnetic Resonance Imaging and Cardiovascular Biomarkers Related to Kidney Transplantation. Can J Kidney Health Dis 2018; 5:2054358118809974. [PMID: 30542623 PMCID: PMC6236646 DOI: 10.1177/2054358118809974] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/10/2018] [Indexed: 01/03/2023] Open
Abstract
Background: Cardiac magnetic resonance (CMR) imaging accurately and precisely measures
left ventricular (LV) mass and function. Identifying mechanisms by which LV
mass change and functional improvement occur in some end-stage kidney
disease (ESKD) patients may help to appropriately target kidney transplant
(KT) recipients for further investigation and intervention. The
concentration of serum adiponectin, a cardiovascular biomarker, increases in
cardiac failure, its production being enhanced by B-type natriuretic peptide
(BNP), and both serum adiponectin and BNP concentrations decline
posttransplantation. Objective: We tested the hypothesis that kidney transplantation alters LV
characteristics that relate to serum adiponectin concentrations. Design: Prospective and observational cohort study. Setting: The study was performed at 3 adult kidney transplant and dialysis centers in
Ontario, Canada. Patients: A total of 82 KT candidate subjects were recruited (39 to the KT group and 43
to the dialysis group). Predialysis patients were excluded. Measurements: Subjects underwent CMR with a 1.5-tesla whole-body magnetic resonance scanner
using a phased-array cardiac coil and retrospective vectorographic gating.
LV mass, LV ejection fraction (LVEF), LV end-systolic volume (LVESV), and LV
end-diastolic volume (LVEDV) were measured by CMR pre-KT and again 12 months
post-KT (N = 39), or 12 months later if still receiving dialysis (N = 43).
LV mass, LVESV, and LVEDV were indexed for height (m2.7) to
calculate left ventricular mass index (LVMI), left ventricular end-systolic
volume index (LVESVI), and left ventricular end-diastolic volume index
(LVEDVI), respectively. Serum total adiponectin and N-terminal proBNP
(NT-proBNP) concentrations were measured at baseline, 3 months, and 12
months. Methods: We performed a prospective 1:1 observational study comparing KT candidates
with ESKD either receiving a living donor organ (KT group) or waiting for a
deceased donor organ (dialysis group). Results: Left ventricular mass index change was −1.98 ± 5.5 and −0.36 ± 5.7
g/m2.7 for KT versus dialysis subjects (P =
.44). Left ventricular mass change was associated with systolic blood
pressure (SBP) (P = .0008) and average LV mass
(P = .0001). Left ventricular ejection fraction did not
improve (2.9 ± 6.6 vs 0.7 ± 4.9 %, P = .09), while LVESVI
and LVEDVI decreased more post-KT than with continued dialysis (−3.36 ± 5.6
vs −0.22 ± 4.4 mL/m2.7, P < .01 and −4.9 ±
8.5 vs −0.3 ± 9.2 mL/m2.7, P = .02). Both
adiponectin (−7.1 ± 11.3 vs −0.11 ± 7.9 µg/mL, P <
.0001) and NT-proBNP (−3811 ± 8130 vs 1665 ± 20013 pg/mL, P
< .0001) declined post-KT. Post-KT adiponectin correlated with NT-proBNP
(P = .001), but not estimated glomerular filtration
rate (eGFR) (P = .13). Change in adiponectin did not
correlate with change in LVEF in the KT group (Spearman ρ = 0.16,
P = .31) or dialysis group (Spearman ρ = 0.19,
P = .21). Limitations: Few biomarkers of cardiac function were measured to fully contextualize their
role during changing kidney function. Limited intrapatient biomarker
sampling and CMR measurements precluded constructing dose-response curves of
biomarkers to LV mass and function. The CMR timing in relation to dialysis
was not standardized. Conclusions: The LVESVI and LVEDVI but not LVMI or LVEF improve post-KT. LVMI and LVEF
change is independent of renal function and adiponectin. As adiponectin
correlates with NT-proBNP post-KT, improved renal function through KT
restores the normal heart-endocrine axis.
Collapse
Affiliation(s)
- G V Ramesh Prasad
- Division of Nephrology, St. Michael's Hospital, University of Toronto, ON, Canada
| | - Andrew T Yan
- Division of Cardiology, St. Michael's Hospital, University of Toronto, ON, Canada
| | - Michelle M Nash
- Renal Transplant Program, St. Michael's Hospital, Toronto, ON, Canada
| | - S Joseph Kim
- Division of Nephrology, Toronto General Hospital, University of Toronto, ON, Canada
| | - Ron Wald
- Division of Nephrology, St. Michael's Hospital, University of Toronto, ON, Canada
| | - Rachel Wald
- Division of Cardiology, Toronto General Hospital, University of Toronto, ON, Canada
| | - Charmaine Lok
- Division of Nephrology, Toronto General Hospital, University of Toronto, ON, Canada
| | - Lakshman Gunaratnam
- Division of Nephrology, London Health Sciences Centre, Western University, ON, Canada
| | - Gauri R Karur
- Division of Cardiology, St. Michael's Hospital, University of Toronto, ON, Canada
| | - Anish Kirpalani
- Department of Medical Imaging, St. Michael's Hospital, University of Toronto, ON, Canada
| | - Philip W Connelly
- Division of Endocrinology and Metabolism, St. Michael's Hospital, University of Toronto, ON, Canada
| |
Collapse
|