1
|
Pitt B, Diez J. Possible Role of Gut Microbiota Alterations in Myocardial Fibrosis and Burden of Heart Failure in Hypertensive Heart Disease. Hypertension 2024; 81:1467-1476. [PMID: 38716665 DOI: 10.1161/hypertensionaha.124.23089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
Epidemiological studies have revealed that hypertensive heart disease is a major risk factor for heart failure, and its heart failure burden is growing rapidly. The need to act in the face of this threat requires first an understanding of the multifactorial origin of hypertensive heart disease and second an exploration of new mechanistic pathways involved in myocardial alterations critically involved in cardiac dysfunction and failure (eg, myocardial interstitial fibrosis). Increasing evidence shows that alterations of gut microbiota composition and function (ie, dysbiosis) leading to changes in microbiota-derived metabolites and impairment of the gut barrier and immune functions may be involved in blood pressure elevation and hypertensive organ damage. In this review, we highlight recent advances in the potential contribution of gut microbiota alterations to myocardial interstitial fibrosis in hypertensive heart disease through blood pressure-dependent and blood pressure-independent mechanisms. Achievements in this field should open a new path for more comprehensive treatment of myocardial interstitial fibrosis in hypertensive heart disease and, thus, for the prevention of heart failure.
Collapse
Affiliation(s)
- Bertram Pitt
- Department of Medicine, University of Michigan School of Medicine, Ann Arbor (B.P.)
| | - Javier Diez
- Department of Cardiovascular Diseases, Center for Applied Medical Research and School of Medicine, University of Navarra, Pamplona, Spain (J.D.)
| |
Collapse
|
2
|
Vogt A, Plehn A, Atti C, Nussbaum M, Tongers J, Sedding D, Dutzmann J. Left ventricular structure and function following renal sympathetic denervation in patients with HFpEF: an echocardiographic 9-year long-term follow-up. Front Cardiovasc Med 2024; 11:1408547. [PMID: 38919545 PMCID: PMC11196750 DOI: 10.3389/fcvm.2024.1408547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/23/2024] [Indexed: 06/27/2024] Open
Abstract
Background High blood pressure is a major risk factor for cardiac remodeling and left ventricular hypertrophy, increasing cardiovascular risk and leading to heart failure with preserved ejection fraction (HFpEF). Since renal sympathetic denervation (RDN) reduces blood pressure in the long term, we aimed to investigate the long-term effect of RDN in patients with HFpEF in the present analysis. Methods Patients previously enrolled in a local RDN registry who underwent high-frequency RDN with the use of the Symplicity Flex® renal denervation system between 2011 and 2014 were followed up. The patients were assessed by 24-h ambulatory blood pressure measurement, transthoracic echocardiography, and laboratory tests. We used the echocardiographic and biomarker criteria of the Heart Failure Association (HFA)-PEFF (Pre-test assessment, Echocardiography and Natriuretic Peptide Score, Funkctional testing, and Final aetiology) score to identify patients with HFpEF. Results Echocardiographic assessment was available for 70 patients at a 9-year long-term follow-up. Of these patients, 21 had HFpEF according to the HFA-PEFF score. We found a significant reduction of the HFA-PEFF score from 5.48 ± 0.51 points at baseline to 4.33 ± 1.53 points at the 9-year follow-up (P < 0.01). This decrease was due to a greater reduction in morphological and biomarker subcategories [from 1.95 ± 0.22 to 1.43 ± 0.51 points (P < 0.01) and from 1.52 ± 0.52 to 0.90 ± 0.63 points (P < 0.01), respectively] than in the functional one. Morphologically, there was a reduction in left ventricular hypertrophy and left atrial dilation. Conclusions The present analysis suggests that RDN may lead to a regression of the extent of HFpEF beyond a reduction in blood pressure and thus possibly contribute to an improvement in prognosis. More detailed information will be provided by ongoing randomized sham-controlled trials.
Collapse
Affiliation(s)
- Alexander Vogt
- Department of Internal Medicine III, University Hospital Halle (Saale), Halle (Saale), Germany
| | - Alexander Plehn
- Department of Internal Medicine III, University Hospital Halle (Saale), Halle (Saale), Germany
- Praxisklinik Salzatal, Salzatal, Germany
| | - Carlo Atti
- Department of Internal Medicine, Medical University Graz, Graz, Austria
| | - Michael Nussbaum
- Department of Internal Medicine III, University Hospital Halle (Saale), Halle (Saale), Germany
| | - Jörn Tongers
- Department of Internal Medicine III, University Hospital Halle (Saale), Halle (Saale), Germany
| | - Daniel Sedding
- Department of Internal Medicine III, University Hospital Halle (Saale), Halle (Saale), Germany
| | - Jochen Dutzmann
- Department of Internal Medicine III, University Hospital Halle (Saale), Halle (Saale), Germany
| |
Collapse
|
3
|
Doeblin P, Steinbeis F, Witzenrath M, Hashemi D, Chen W, Weiss KJ, Stawowy P, Kelle S. Half-Dose versus Single-Dose Gadobutrol for Extracellular Volume Measurements in Cardiac Magnetic Resonance. J Cardiovasc Dev Dis 2023; 10:316. [PMID: 37623329 PMCID: PMC10455162 DOI: 10.3390/jcdd10080316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Cardiac magnetic resonance (CMR) imaging with gadolinium-based contrast agents offers unique non-invasive insights into cardiac tissue composition. Myocardial extracellular volume (ECV) has evolved as an objective and robust parameter with broad diagnostic and prognostic implications. For the gadolinium compound gadobutrol, the recommended dose for cardiac imaging, including ECV measurements, is 0.1 mmol/kg (single dose). This dose was optimized for late enhancement imaging, a measure of focal fibrosis. Whether a lower dose is sufficient for ECV measurements is unknown. We aim to evaluate the accuracy of ECV measurements using a half dose of 0.05 mmol/kg gadobutrol compared to the standard single dose of 0.1 mmol/kg. METHODS AND RESULTS From a contemporary trial (NCT04747366, registered 10 February 2021), a total of 25 examinations with available T1 mapping before and after 0.05 and 0.1 mmol/kg gadobutrol were analyzed. ECV values were calculated automatically from pre- and post-contrast T1 relaxation times. T1 and ECV Measurements were performed in the midventricular septum. ECV values after 0.05 and 0.1 mmol/kg gadobutrol were correlated (R2 = 0.920, p < 0.001). ECV values after 0.05 mmol/kg had a bias of +0.9% (95%-CI [0.4; 1.4], p = 0.002) compared to 0.1 mmol/kg gadobutrol, with limits of agreement from -1.5 to 3.3%. CONCLUSIONS CMR with a half dose of 0.05 mmol/kg gadobutrol overestimated ECV by 0.9% compared with a full dose of 0.1 mmol/kg, necessitating adjustment of normal values when using half-dose ECV imaging.
Collapse
Affiliation(s)
- Patrick Doeblin
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, 13353 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10785 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Fridolin Steinbeis
- Department of Infectious Diseases and Respiratory Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Martin Witzenrath
- Department of Infectious Diseases and Respiratory Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- German Center for Lung Research (DZL), 10117 Berlin, Germany
| | - Djawid Hashemi
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, 13353 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10785 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Wensu Chen
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Karl Jakob Weiss
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, 13353 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10785 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Philipp Stawowy
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, 13353 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Sebastian Kelle
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, Augustenburger Platz 1, 13353 Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10785 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
4
|
Pushpakumar S, Singh M, Zheng Y, Akinterinwa OE, Mokshagundam SPL, Sen U, Kalra DK, Tyagi SC. Renal Denervation Helps Preserve the Ejection Fraction by Preserving Endocardial-Endothelial Function during Heart Failure. Int J Mol Sci 2023; 24:ijms24087302. [PMID: 37108465 PMCID: PMC10139195 DOI: 10.3390/ijms24087302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Renal denervation (RDN) protects against hypertension, hypertrophy, and heart failure (HF); however, it is not clear whether RDN preserves ejection fraction (EF) during heart failure (HFpEF). To test this hypothesis, we simulated a chronic congestive cardiopulmonary heart failure (CHF) phenotype by creating an aorta-vena cava fistula (AVF) in the C57BL/6J wild type (WT) mice. Briefly, there are four ways to create an experimental CHF: (1) myocardial infarction (MI), which is basically ligating the coronary artery by instrumenting and injuring the heart; (2) trans-aortic constriction (TAC) method, which mimics the systematic hypertension, but again constricts the aorta on top of the heart and, in fact, exposes the heart; (3) acquired CHF condition, promoted by dietary factors, diabetes, salt, diet, etc., but is multifactorial in nature; and finally, (4) the AVF, which remains the only one wherein AVF is created ~1 cm below the kidneys in which the aorta and vena cava share the common middle-wall. By creating the AVF fistula, the red blood contents enter the vena cava without an injury to the cardiac tissue. This model mimics or simulates the CHF phenotype, for example, during aging wherein with advancing age, the preload volume keeps increasing beyond the level that the aging heart can pump out due to the weakened cardiac myocytes. Furthermore, this procedure also involves the right ventricle to lung to left ventricle flow, thus creating an ideal condition for congestion. The heart in AVF transitions from preserved to reduced EF (i.e., HFpEF to HFrEF). In fact, there are more models of volume overload, such as the pacing-induced and mitral valve regurgitation, but these are also injurious models in nature. Our laboratory is one of the first laboratories to create and study the AVF phenotype in the animals. The RDN was created by treating the cleaned bilateral renal artery. After 6 weeks, blood, heart, and renal samples were analyzed for exosome, cardiac regeneration markers, and the renal cortex proteinases. Cardiac function was analyzed by echocardiogram (ECHO) procedure. The fibrosis was analyzed with a trichrome staining method. The results suggested that there was a robust increase in the exosomes' level in AVF blood, suggesting a compensatory systemic response during AVF-CHF. During AVF, there was no change in the cardiac eNOS, Wnt1, or β-catenin; however, during RDN, there were robust increases in the levels of eNOS, Wnt1, and β-catenin compared to the sham group. As expected in HFpEF, there was perivascular fibrosis, hypertrophy, and pEF. Interestingly, increased levels of eNOS suggested that despite fibrosis, the NO generation was higher and that it most likely contributed to pEF during HF. The RDN intervention revealed an increase in renal cortical caspase 8 and a decrease in caspase 9. Since caspase 8 is protective and caspase 9 is apoptotic, we suggest that RDN protects against the renal stress and apoptosis. It should be noted that others have demonstrated a role of vascular endothelium in preserving the ejection by cell therapy intervention. In the light of foregoing evidence, our findings also suggest that RDN is cardioprotective during HFpEF via preservation of the eNOS and accompanied endocardial-endothelial function.
Collapse
Affiliation(s)
- Sathnur Pushpakumar
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Mahavir Singh
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Yuting Zheng
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Oluwaseun E Akinterinwa
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Sri Prakash L Mokshagundam
- Division of Endocrinology, Metabolism and Diabetes and Robley Rex VA Medical Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Utpal Sen
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Dinesh K Kalra
- Division of Cardiovascular Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Suresh C Tyagi
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
5
|
Nordmeyer S, Kraus M, Ziehm M, Kirchner M, Schafstedde M, Kelm M, Niquet S, Stephen MM, Baczko I, Knosalla C, Schapranow MP, Dittmar G, Gotthardt M, Falcke M, Regitz-Zagrosek V, Kuehne T, Mertins P. Disease- and sex-specific differences in patients with heart valve disease: a proteome study. Life Sci Alliance 2023; 6:e202201411. [PMID: 36627164 PMCID: PMC9834574 DOI: 10.26508/lsa.202201411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Pressure overload in patients with aortic valve stenosis and volume overload in mitral valve regurgitation trigger specific forms of cardiac remodeling; however, little is known about similarities and differences in myocardial proteome regulation. We performed proteome profiling of 75 human left ventricular myocardial biopsies (aortic stenosis = 41, mitral regurgitation = 17, and controls = 17) using high-resolution tandem mass spectrometry next to clinical and hemodynamic parameter acquisition. In patients of both disease groups, proteins related to ECM and cytoskeleton were more abundant, whereas those related to energy metabolism and proteostasis were less abundant compared with controls. In addition, disease group-specific and sex-specific differences have been observed. Male patients with aortic stenosis showed more proteins related to fibrosis and less to energy metabolism, whereas female patients showed strong reduction in proteostasis-related proteins. Clinical imaging was in line with proteomic findings, showing elevation of fibrosis in both patient groups and sex differences. Disease- and sex-specific proteomic profiles provide insight into cardiac remodeling in patients with heart valve disease and might help improve the understanding of molecular mechanisms and the development of individualized treatment strategies.
Collapse
Affiliation(s)
- Sarah Nordmeyer
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Institute for Cardiovascular Computer-Assisted Medicine, Berlin, Germany
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Department of Congenital Heart Disease - Pediatric Cardiology, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Milena Kraus
- Hasso Plattner Institute for Digital Engineering, Digital Health Center, University of Potsdam, Potsdam, Germany
| | - Matthias Ziehm
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Proteomics Platform, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marieluise Kirchner
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Proteomics Platform, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marie Schafstedde
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Institute for Cardiovascular Computer-Assisted Medicine, Berlin, Germany
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Department of Congenital Heart Disease - Pediatric Cardiology, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marcus Kelm
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Institute for Cardiovascular Computer-Assisted Medicine, Berlin, Germany
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Department of Congenital Heart Disease - Pediatric Cardiology, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sylvia Niquet
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Proteomics Platform, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Mariet Mathew Stephen
- Hasso Plattner Institute for Digital Engineering, Digital Health Center, University of Potsdam, Potsdam, Germany
| | - Istvan Baczko
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary
| | - Christoph Knosalla
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart, Department of Cardiothoracic and Vascular Surgery, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Matthieu-P Schapranow
- Hasso Plattner Institute for Digital Engineering, Digital Health Center, University of Potsdam, Potsdam, Germany
| | - Gunnar Dittmar
- Proteomics of Cellular Signaling, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Michael Gotthardt
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Neuromuscular and Cardiovascular Cell Biology, Berlin, Germany
| | - Martin Falcke
- Max Delbrück Center for Molecular Medicine, Mathematical Cell Physiology, Berlin, Germany
| | - Vera Regitz-Zagrosek
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Cardiology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Titus Kuehne
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Institute for Cardiovascular Computer-Assisted Medicine, Berlin, Germany
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Department of Congenital Heart Disease - Pediatric Cardiology, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Philipp Mertins
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Proteomics Platform, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
6
|
Abstract
Hypertensive heart disease (HHD) is currently the second leading cause of heart failure. The prevalence of HHD and its associated risk of heart failure have increased despite substantial improvement in arterial hypertension treatment and control in the recent decades. Therefore, the prevention of heart failure in patients with HHD represents an unmet medical need, due to its clinical, economic, and social impact. In this conceptual framework, we call to action because the time has come for diagnosis and treatment of patients with HHD not to be limited to assessment of morphological and functional left ventricular changes, blood pressure control, and left ventricular hypertrophy regression. We propose a further perspective incorporating also the detection and reversal of the histological changes that develop in the hypertensive heart and that lead to the structural remodeling of the myocardium. In particular, we focus on the diagnosis and treatment of myocardial interstitial fibrosis, likely the lesion most critically involved in the transition from subclinical HHD to clinically overt heart failure. In this context, it is worth considering whether the use of imaging and circulating biomarkers for the noninvasive diagnosis of myocardial interstitial fibrosis should be incorporated in the medical study of hypertensive patients, especially of those with HHD.
Collapse
Affiliation(s)
- Javier Díez
- Center for Applied Medical Research (CIMA), and School of Medicine, Universidad de Navarra, and Center for Network Biomedical Research of Cardiovascular Diseases (CIBERCV), Carlos III Institute of Health, Madrid, Spain (J.D.)
| | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, TX, and Department of Medicine, University of Mississippi School of Medicine, Jackson (J.B.)
| |
Collapse
|
7
|
Cardiac Magnetic Resonance in Hypertensive Heart Disease: Time for a New Chapter. Diagnostics (Basel) 2022; 13:diagnostics13010137. [PMID: 36611429 PMCID: PMC9818319 DOI: 10.3390/diagnostics13010137] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
Hypertension is one of the most important cardiovascular risk factors, associated with significant morbidity and mortality. Chronic high blood pressure leads to various structural and functional changes in the myocardium. Different sophisticated imaging methods are developed to properly estimate the severity of the disease and to prevent possible complications. Cardiac magnetic resonance can provide a comprehensive assessment of patients with hypertensive heart disease, including accurate and reproducible measurement of left and right ventricle volumes and function, tissue characterization, and scar quantification. It is important in the proper evaluation of different left ventricle hypertrophy patterns to estimate the presence and severity of myocardial fibrosis, as well as to give more information about the benefits of different therapeutic modalities. Hypertensive heart disease often manifests as a subclinical condition, giving exceptional value to cardiac magnetic resonance as an imaging modality capable to detect subtle changes. In this article, we are giving a comprehensive review of all the possibilities of cardiac magnetic resonance in patients with hypertensive heart disease.
Collapse
|
8
|
Bertacchini F, Agabiti Rosei C, Buso G, Cappellini S, Stassaldi D, Aggiusti C, Salvetti M, Paini A, De Ciuceis C, Muiesan ML. Subclinical HMOD in Hypertension: Left Ventricular Diastolic Dysfunction. High Blood Press Cardiovasc Prev 2022; 29:585-593. [PMID: 36352335 PMCID: PMC9708770 DOI: 10.1007/s40292-022-00548-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 10/25/2022] [Indexed: 06/16/2023] Open
Abstract
Arterial hypertension represents an important risk factor for the development of cardiac, vascular and renal events, predisposing to heart failure, acute coronary syndromes, peripheral artery disease, stroke, and chronic renal disease. Arterial hypertension leads to the development of subclinical hypertension mediated organ damage (HMOD) which has prognostic relevance and may influence the choice of treatment options. Alterations of cardiac structure and function represent the more widely assessed form of HMOD. This manuscript will focus on the diagnostic opportunities, prognostic significance and treatment of diastolic dysfunction alterations.
Collapse
Affiliation(s)
- Fabio Bertacchini
- Department of Clinical and Experimental Sciences, 2a Medicina-ASST Spedali Civili Brescia, University of Brescia, 25100, Brescia, Italy
| | - Claudia Agabiti Rosei
- Department of Clinical and Experimental Sciences, 2a Medicina-ASST Spedali Civili Brescia, University of Brescia, 25100, Brescia, Italy
| | - Giacomo Buso
- Department of Clinical and Experimental Sciences, 2a Medicina-ASST Spedali Civili Brescia, University of Brescia, 25100, Brescia, Italy
| | - Sara Cappellini
- Department of Clinical and Experimental Sciences, 2a Medicina-ASST Spedali Civili Brescia, University of Brescia, 25100, Brescia, Italy
| | - Deborah Stassaldi
- Department of Clinical and Experimental Sciences, 2a Medicina-ASST Spedali Civili Brescia, University of Brescia, 25100, Brescia, Italy
| | - Carlo Aggiusti
- Department of Clinical and Experimental Sciences, 2a Medicina-ASST Spedali Civili Brescia, University of Brescia, 25100, Brescia, Italy
| | - Massimo Salvetti
- Department of Clinical and Experimental Sciences, 2a Medicina-ASST Spedali Civili Brescia, University of Brescia, 25100, Brescia, Italy
| | - Anna Paini
- Department of Clinical and Experimental Sciences, 2a Medicina-ASST Spedali Civili Brescia, University of Brescia, 25100, Brescia, Italy
| | - Carolina De Ciuceis
- Department of Clinical and Experimental Sciences, 2a Medicina-ASST Spedali Civili Brescia, University of Brescia, 25100, Brescia, Italy
| | - Maria Lorenza Muiesan
- Department of Clinical and Experimental Sciences, 2a Medicina-ASST Spedali Civili Brescia, University of Brescia, 25100, Brescia, Italy.
| |
Collapse
|
9
|
Cavus E, Schneider JN, Bei der Kellen R, di Carluccio E, Ziegler A, Tahir E, Bohnen S, Avanesov M, Radunski UK, Chevalier C, Jahnke C, Ojeda F, Kirchhof P, Blankenberg S, Adam G, Lund GK, Muellerleile K. Impact of Sex and Cardiovascular Risk Factors on Myocardial T1, Extracellular Volume Fraction, and T2 at 3 Tesla: Results From the Population-Based, Hamburg City Health Study. Circ Cardiovasc Imaging 2022; 15:e014158. [PMID: 36126126 DOI: 10.1161/circimaging.122.014158] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Reliable reference intervals are crucial for clinical application of myocardial T1 and T2 mapping cardiovascular magnetic resonance imaging. This study evaluated the impact of sex and cardiovascular risk factors on myocardial T1, extracellular volume fraction (ECV), and T2 at 3T in the population-based HCHS (Hamburg City Health Study). METHODS The final study sample consisted of 1576 consecutive HCHS participants between 46 and 78 years without prevalent heart disease, including 1020 (67.3%) participants with hypertension and 110 (7.5%) with diabetes. T1 and T2 mapping were performed on a 3T scanner using 5b(3b)3b modified Look-Locker inversion recovery and T2 prepared, fast-low-angle shot sequence, respectively. Stepwise regression analyses were performed to identify variables with an independent impact on T1, ECV, and T2. Reference intervals were defined as the interval between the 2.5% and 97.5% quantiles. RESULTS Sex was the major independent influencing factor of myocardial native T1, ECV, and T2. Female patients had significantly higher upper limits of reference intervals for native T1 (1112-1261 versus 1079-1241 ms), ECV (23%-33% versus 22%-32%), and T2 (36-46 versus 35-45 ms) compared with male patients (all P<0.001). Cardiovascular risk factors, such as diabetes and hypertension, did not systematically affect native T1. There was an independent association of T2 by hypertension and, to a lesser degree, by left ventricular mass, heart rate (all P<0.001), and body mass index (P=0.001). CONCLUSIONS Sex needs to be considered as the major, independent influencing factor for clinical application of myocardial T1, ECV, and T2 measurements. Consequently, sex-specific reference intervals should be used in clinical routine. Our findings suggest that there is no need for specific reference intervals for myocardial T1 and ECV measurements in individuals with cardiovascular risk factors. However, hypertension should be considered as an additional factor for clinical application of T2 measurements. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: NCT03934957.
Collapse
Affiliation(s)
- Ersin Cavus
- Department of Cardiology, University Heart and Vascular Center Hamburg Eppendorf, Germany (E.C., J.N.S., R.B.d.K., E.d.C., A.Z., S. Bohnen, U.K.R., C.C., C.J., F.O., P.K., S. Blankenberg, K.M.).,Deutsches Zentrum für Herz-Kreislauf-Forschung e.V. (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany (E.C., P.K., S. Blankenberg, K.M.)
| | - Jan N Schneider
- Department of Cardiology, University Heart and Vascular Center Hamburg Eppendorf, Germany (E.C., J.N.S., R.B.d.K., E.d.C., A.Z., S. Bohnen, U.K.R., C.C., C.J., F.O., P.K., S. Blankenberg, K.M.)
| | - Ramona Bei der Kellen
- Department of Cardiology, University Heart and Vascular Center Hamburg Eppendorf, Germany (E.C., J.N.S., R.B.d.K., E.d.C., A.Z., S. Bohnen, U.K.R., C.C., C.J., F.O., P.K., S. Blankenberg, K.M.)
| | - Eleonora di Carluccio
- Department of Cardiology, University Heart and Vascular Center Hamburg Eppendorf, Germany (E.C., J.N.S., R.B.d.K., E.d.C., A.Z., S. Bohnen, U.K.R., C.C., C.J., F.O., P.K., S. Blankenberg, K.M.).,Cardio-Care, Medizincampus Davos, Switzerland (E.d.C., A.Z.)
| | - Andreas Ziegler
- Department of Cardiology, University Heart and Vascular Center Hamburg Eppendorf, Germany (E.C., J.N.S., R.B.d.K., E.d.C., A.Z., S. Bohnen, U.K.R., C.C., C.J., F.O., P.K., S. Blankenberg, K.M.).,Cardio-Care, Medizincampus Davos, Switzerland (E.d.C., A.Z.).,School of Mathematics, Statistics and Computer Science, University of KwaZulu-Natal, Pietermaritzburg, South Africa (A.Z.)
| | - Enver Tahir
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Hospital Hamburg Eppendorf, Germany (E.T., M.A., G.A., G.K.L.)
| | - Sebastian Bohnen
- Department of Cardiology, University Heart and Vascular Center Hamburg Eppendorf, Germany (E.C., J.N.S., R.B.d.K., E.d.C., A.Z., S. Bohnen, U.K.R., C.C., C.J., F.O., P.K., S. Blankenberg, K.M.)
| | - Maxim Avanesov
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Hospital Hamburg Eppendorf, Germany (E.T., M.A., G.A., G.K.L.)
| | - Ulf K Radunski
- Department of Cardiology, University Heart and Vascular Center Hamburg Eppendorf, Germany (E.C., J.N.S., R.B.d.K., E.d.C., A.Z., S. Bohnen, U.K.R., C.C., C.J., F.O., P.K., S. Blankenberg, K.M.)
| | - Celeste Chevalier
- Department of Cardiology, University Heart and Vascular Center Hamburg Eppendorf, Germany (E.C., J.N.S., R.B.d.K., E.d.C., A.Z., S. Bohnen, U.K.R., C.C., C.J., F.O., P.K., S. Blankenberg, K.M.)
| | - Charlotte Jahnke
- Department of Cardiology, University Heart and Vascular Center Hamburg Eppendorf, Germany (E.C., J.N.S., R.B.d.K., E.d.C., A.Z., S. Bohnen, U.K.R., C.C., C.J., F.O., P.K., S. Blankenberg, K.M.)
| | - Francisco Ojeda
- Department of Cardiology, University Heart and Vascular Center Hamburg Eppendorf, Germany (E.C., J.N.S., R.B.d.K., E.d.C., A.Z., S. Bohnen, U.K.R., C.C., C.J., F.O., P.K., S. Blankenberg, K.M.)
| | - Paulus Kirchhof
- Department of Cardiology, University Heart and Vascular Center Hamburg Eppendorf, Germany (E.C., J.N.S., R.B.d.K., E.d.C., A.Z., S. Bohnen, U.K.R., C.C., C.J., F.O., P.K., S. Blankenberg, K.M.).,Deutsches Zentrum für Herz-Kreislauf-Forschung e.V. (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany (E.C., P.K., S. Blankenberg, K.M.)
| | - Stefan Blankenberg
- Department of Cardiology, University Heart and Vascular Center Hamburg Eppendorf, Germany (E.C., J.N.S., R.B.d.K., E.d.C., A.Z., S. Bohnen, U.K.R., C.C., C.J., F.O., P.K., S. Blankenberg, K.M.).,Deutsches Zentrum für Herz-Kreislauf-Forschung e.V. (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany (E.C., P.K., S. Blankenberg, K.M.)
| | - Gerhard Adam
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Hospital Hamburg Eppendorf, Germany (E.T., M.A., G.A., G.K.L.)
| | - Gunnar K Lund
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Hospital Hamburg Eppendorf, Germany (E.T., M.A., G.A., G.K.L.)
| | - Kai Muellerleile
- Department of Cardiology, University Heart and Vascular Center Hamburg Eppendorf, Germany (E.C., J.N.S., R.B.d.K., E.d.C., A.Z., S. Bohnen, U.K.R., C.C., C.J., F.O., P.K., S. Blankenberg, K.M.).,Deutsches Zentrum für Herz-Kreislauf-Forschung e.V. (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany (E.C., P.K., S. Blankenberg, K.M.)
| |
Collapse
|
10
|
Zhang SJ, Chang D, Jin JY, Wang YL, Wang L, Wang YC, Wang Z, Ju S. Myocardial Extracellular Volume Fraction Measured by Cardiac Magnetic Resonance Imaging Negatively Correlates With Cardiomyocyte Breadth in a Healthy Porcine Model. Front Cardiovasc Med 2022; 9:791963. [PMID: 35369328 PMCID: PMC8968101 DOI: 10.3389/fcvm.2022.791963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 02/11/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundThe extracellular volume fraction (ECV) derived from cardiac magnetic resonance imaging (MRI) is extensively used to evaluate myocardial fibrosis. However, due to the limited histological verification in healthy individuals, it remains unclear whether the size of cardiomyocytes may play a potential role in the physiological changes of ECV. The aim of this study was to examine the association between cardiomyocyte size and myocardial ECV by using a healthy porcine model.MethodsSixteen domestic healthy pigs were anesthetized and underwent cardiac MRI with mechanical controlled breathing. Intravenous contrast medium was introduced at a dose of 0.2–0.25 mmol/kg. The interventricular septum ECV was calculated using an established MRI procedure, which was based on the pre- and post-contrast T1 values of the heart and individual blood hematocrit. The cardiomyocyte breadth (CmyB) in cross section was measured by hematoxylin and eosin staining to reflect the cardiomyocyte size.ResultsData were successfully acquired from 14 pigs. The CmyB was obtained from the myocardial tissues corresponding to the region of interest on cardiac MRI. The mean ± SD of the ECV was 0.253 ± 0.043, and the mean ± SD of the CmyB was 10.02 ± 0.84 μm. The ECV exhibited a negative correlation with the CmyB (r = −0.729, p = 0.003).ConclusionThe myocardial ECV detected by cardiac MRI is negatively correlated with the CmyB in healthy pigs, demonstrating that the size of cardiomyocytes is potentially associated with the ECV under physiological conditions.
Collapse
Affiliation(s)
- Shi-Jun Zhang
- Department of Radiology, Zhongda Hospital, Jiangsu Key Laboratory of Molecular and Functional Imaging, Medical School of Southeast University, Nanjing, China
| | - Di Chang
- Department of Radiology, Zhongda Hospital, Jiangsu Key Laboratory of Molecular and Functional Imaging, Medical School of Southeast University, Nanjing, China
| | - Ji-Yang Jin
- Department of Radiology, Zhongda Hospital, Jiangsu Key Laboratory of Molecular and Functional Imaging, Medical School of Southeast University, Nanjing, China
| | - Ya-Ling Wang
- Department of Radiology, Zhongda Hospital, Jiangsu Key Laboratory of Molecular and Functional Imaging, Medical School of Southeast University, Nanjing, China
| | - Lin Wang
- Department of Radiology, Zhongda Hospital, Jiangsu Key Laboratory of Molecular and Functional Imaging, Medical School of Southeast University, Nanjing, China
| | - Yuan-Cheng Wang
- Department of Radiology, Zhongda Hospital, Jiangsu Key Laboratory of Molecular and Functional Imaging, Medical School of Southeast University, Nanjing, China
| | - Zhen Wang
- Department of Anesthesiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Shenghong Ju
- Department of Radiology, Zhongda Hospital, Jiangsu Key Laboratory of Molecular and Functional Imaging, Medical School of Southeast University, Nanjing, China
- *Correspondence: Shenghong Ju,
| |
Collapse
|
11
|
Bazoukis G, Thomopoulos C, Tse G, Vassiliou VS, Liu T, Dimitriadis K, Tatakis F, Konstantinou K, Doumas M, Tsioufis K. Impact of renal sympathetic denervation on cardiac magnetic resonance-derived cardiac indices in hypertensive patients - A meta-analysis. J Cardiol 2021; 78:314-321. [PMID: 34088560 DOI: 10.1016/j.jjcc.2021.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/20/2021] [Accepted: 04/09/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Renal sympathetic denervation (RDN) is a safe device-based option for the treatment of hypertension although current guidelines do not recommend its use in routine clinical practice. In this meta-analysis, we investigated the effects of RDN in cardiac magnetic resonance (CMR)-derived cardiac indices. METHODS This meta-analysis was performed in accordance with the PRISMA statement. A comprehensive systematic search of MEDLINE database and Cochrane library through to January 2021 was performed. The inclusion criteria were studies that enrolled patients undergoing RDN in whom CMR data were provided for left ventricular end-diastolic volume indexed to body surface area (BSA) (LVEDVI), left ventricular end-systolic volume indexed (LVESVI), left ventricular mass indexed (LVMI), and left ventricular ejection fraction (LVEF) pre and post RDN. A random effects model was used for the analyses. RESULTS Our search strategy revealed 9 studies that were finally included in the meta-analysis (n=300 patients, mean age: 60 years old, males: 59%). Compared to control group, RDN patients showed significantly lower values in the attained volumes (LVEDVI: -6.70 ml/m2, p=0.01; LVESVI: -3.63 ml/m2, p=0.006). Moreover, RDN group achieved a statistically significant higher attained LVEF (3.49%, p=0.01). A non-significant difference was found in the attained LVMI between RDN and control groups (-2.59 g/m2, p=0.39). Compared to pre-RDN values, RDN reduces significantly the LVMI, the LVEDVI, and the LVESVI while a non-significant change of LVEF was found. CONCLUSIONS In conclusion, the current study demonstrates the potential beneficial role of RDN in CMR-derived cardiac indices that reflect adverse remodeling. However, large, randomized studies are needed to elucidate the role of RDN in cardiac remodeling in hypertension, heart failure, and other clinical settings.
Collapse
Affiliation(s)
- George Bazoukis
- First Cardiology Clinic, Medical School, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece.
| | | | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Vassilios S Vassiliou
- Norfolk and Norwich University Hospital, Norwich, UK; Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Kyriakos Dimitriadis
- First Cardiology Clinic, Medical School, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Fotios Tatakis
- First Cardiology Clinic, Medical School, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Konstantinos Konstantinou
- First Cardiology Clinic, Medical School, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Michael Doumas
- 2nd Propedeutic Department of Internal Medicine, Aristotle University, Thessaloniki, Greece
| | - Konstantinos Tsioufis
- First Cardiology Clinic, Medical School, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| |
Collapse
|
12
|
Nordmeyer S, Lee CB, Goubergrits L, Knosalla C, Berger F, Falk V, Ghorbani N, Hireche-Chikaoui H, Zhu M, Kelle S, Kuehne T, Kelm M. Circulatory efficiency in patients with severe aortic valve stenosis before and after aortic valve replacement. J Cardiovasc Magn Reson 2021; 23:15. [PMID: 33641670 PMCID: PMC7919094 DOI: 10.1186/s12968-020-00686-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 10/29/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Circulatory efficiency reflects the ratio between total left ventricular work and the work required for maintaining cardiovascular circulation. The effect of severe aortic valve stenosis (AS) and aortic valve replacement (AVR) on left ventricular/circulatory mechanical power and efficiency is not yet fully understood. We aimed to quantify left ventricular (LV) efficiency in patients with severe AS before and after surgical AVR. METHODS Circulatory efficiency was computed from cardiovascular magnetic resonance (CMR) imaging derived volumetric data, echocardiographic and clinical data in patients with severe AS (n = 41) before and 4 months after AVR and in age and sex-matched healthy subjects (n = 10). RESULTS In patients with AS circulatory efficiency was significantly decreased compared to healthy subjects (9 ± 3% vs 12 ± 2%; p = 0.004). There were significant negative correlations between circulatory efficiency and LV myocardial mass (r = - 0.591, p < 0.001), myocardial fibrosis volume (r = - 0.427, p = 0.015), end systolic volume (r = - 0.609, p < 0.001) and NT-proBNP (r = - 0.444, p = 0.009) and significant positive correlation between circulatory efficiency and LV ejection fraction (r = 0.704, p < 0.001). After AVR, circulatory efficiency increased significantly in the total cohort (9 ± 3 vs 13 ± 5%; p < 0.001). However, in 10/41 (24%) patients, circulatory efficiency remained below 10% after AVR and, thus, did not restore to normal values. These patients also showed less reduction in myocardial fibrosis volume compared to patients with restored circulatory efficiency after AVR. CONCLUSION In our cohort, circulatory efficiency is reduced in patients with severe AS. In 76% of cases, AVR leads to normalization of circulatory efficiency. However, in 24% of patients, circulatory efficiency remained below normal values even after successful AVR. In these patients also less regression of myocardial fibrosis volume was seen. Trial Registration clinicaltrials.gov NCT03172338, June 1, 2017, retrospectively registered.
Collapse
Affiliation(s)
- S Nordmeyer
- Department of Congenital Heart Disease, German Heart Centre Berlin, Berlin, Germany.
- Institute for Imaging Science and Computational Modelling in Cardiovascular Medicine, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - C B Lee
- Institute for Imaging Science and Computational Modelling in Cardiovascular Medicine, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - L Goubergrits
- Institute for Imaging Science and Computational Modelling in Cardiovascular Medicine, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - C Knosalla
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, German Heart Centre Berlin, Berlin, Germany
| | - F Berger
- Department of Congenital Heart Disease, German Heart Centre Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - V Falk
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, German Heart Centre Berlin, Berlin, Germany
| | - N Ghorbani
- Institute for Imaging Science and Computational Modelling in Cardiovascular Medicine, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - H Hireche-Chikaoui
- Department of Internal Medicine and Cardiology, German Heart Centre Berlin, Berlin, Germany
| | - M Zhu
- Institute for Imaging Science and Computational Modelling in Cardiovascular Medicine, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - S Kelle
- Department of Internal Medicine and Cardiology, German Heart Centre Berlin, Berlin, Germany
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - T Kuehne
- Department of Congenital Heart Disease, German Heart Centre Berlin, Berlin, Germany
- Institute for Imaging Science and Computational Modelling in Cardiovascular Medicine, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - M Kelm
- Department of Congenital Heart Disease, German Heart Centre Berlin, Berlin, Germany
- Institute for Imaging Science and Computational Modelling in Cardiovascular Medicine, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
13
|
Nasser SB, Doeblin P, Doltra A, Schnackenburg B, Wassilew K, Berger A, Gebker R, Bigvava T, Hennig F, Pieske B, Kelle S. Cardiac Myxomas Show Elevated Native T1, T2 Relaxation Time and ECV on Parametric CMR. Front Cardiovasc Med 2020; 7:602137. [PMID: 33330663 PMCID: PMC7710854 DOI: 10.3389/fcvm.2020.602137] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/27/2020] [Indexed: 12/15/2022] Open
Abstract
Introduction: While cardiac tumors are rare, their identification and differentiation has wide clinical implications. Recent cardiac magnetic resonance (CMR) parametric mapping techniques allow for quantitative tissue characterization. Our aim was to examine the range of values encountered in cardiac myxomas in correlation to histological measurements. Methods and Results: Nine patients with histologically proven cardiac myxomas were included. CMR (1.5 Tesla, Philips) including parametric mapping was performed in all patients pre-operatively. All data are reported as mean ± standard deviation. Compared to myocardium, cardiac myxomas demonstrated higher native T1 relaxation times (1,554 ± 192 ms vs. 1,017 ± 58 ms, p < 0.001), ECV (46.9 ± 13.0% vs. 27.1 ± 2.6%, p = 0.001), and T2 relaxation times (209 ± 120 ms vs. 52 ± 3 ms, p = 0.008). Areas with LGE showed higher ECV than areas without (54.3 ± 17.8% vs. 32.7 ± 18.6%, p = 0.042), with differences in native T1 relaxation times (1,644 ± 217 ms vs. 1,482 ± 351 ms, p = 0.291) and T2 relaxation times (356 ± 236 ms vs. 129 ± 68 ms, p = 0.155) not reaching statistical significance. Conclusions: Parametric CMR showed elevated native T1 and T2 relaxation times and ECV values in cardiac myxomas compared to normal myocardium, reflecting an increased interstitial space and fluid content. This might help in the differentiation of cardiac myxomas from other tumor entities.
Collapse
Affiliation(s)
- Sarah B Nasser
- Department of Cardiology, Dar Al Fouad Hospital, Cairo, Egypt
| | - Patrick Doeblin
- Department of Internal Medicine/Cardiology, Deutsches Herzzentrum Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Adelina Doltra
- Department of Internal Medicine/Cardiology, Deutsches Herzzentrum Berlin, Berlin, Germany.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Cardiovascular Institute, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | | | - Katharina Wassilew
- Department of Pathology, Rigshospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Alexander Berger
- Department of Internal Medicine/Cardiology, Deutsches Herzzentrum Berlin, Berlin, Germany
| | - Rolf Gebker
- Department of Internal Medicine/Cardiology, Deutsches Herzzentrum Berlin, Berlin, Germany
| | | | - Felix Hennig
- Department of Cardiothoracic Surgery, Deutsches Herzzentrum Berlin, Berlin, Germany
| | - Burkert Pieske
- Department of Internal Medicine/Cardiology, Deutsches Herzzentrum Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.,Department of Cardiology, Charité University Medicine Berlin, Berlin, Germany
| | - Sebastian Kelle
- Department of Internal Medicine/Cardiology, Deutsches Herzzentrum Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.,Department of Cardiology, Charité University Medicine Berlin, Berlin, Germany
| |
Collapse
|
14
|
Estimation of total collagen volume: a T1 mapping versus histological comparison study in healthy Landrace pigs. Int J Cardiovasc Imaging 2020; 36:1761-1769. [PMID: 32409978 PMCID: PMC7438377 DOI: 10.1007/s10554-020-01881-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/10/2020] [Indexed: 12/16/2022]
Abstract
Right ventricular biopsy represents the gold standard for the assessment of myocardial fibrosis and collagen content. This invasive technique, however, is accompanied by perioperative complications and poor reproducibility. Extracellular volume (ECV) measured through cardiovascular magnetic resonance (CMR) has emerged as a valid surrogate method to assess fibrosis non-invasively. Nonetheless, ECV provides an overestimation of collagen concentration since it also considers interstitial space. Our study aims to investigate the feasibility of estimating total collagen volume (TCV) through CMR by comparing it with the TCV measured at histology. Seven healthy Landrace pigs were acutely instrumented closed-chest and transported to the MRI facility for measurements. For each protocol, CMR imaging at 3T was acquired. MEDIS software was used to analyze T1 mapping and ECV for both the left ventricular myocardium (LVmyo) and left ventricular septum (LVseptum). ECV was then used to estimate TCVCMR at LVmyo and LVseptum following previously published formulas. Tissues were prepared following an established protocol and stained with picrosirius red to analyze the TCVhisto in LVmyo and LVseptum. TCV measured at LVmyo and LVseptum with both histology (8 ± 5 ml and 7 ± 3 ml, respectively) and T1-Mapping (9 ± 5 ml and 8 ± 6 ml, respectively) did not show any regional differences. TCVhisto and TCVCMR showed a good level of data agreement by Bland–Altman analysis. Estimation of TCV through CMR may be a promising way to non-invasively assess myocardial collagen content and may be useful to track disease progression or treatment response.
Collapse
|
15
|
Kiuchi MG, Ho JK, Nolde JM, Gavidia LML, Carnagarin R, Matthews VB, Schlaich MP. Sympathetic Activation in Hypertensive Chronic Kidney Disease - A Stimulus for Cardiac Arrhythmias and Sudden Cardiac Death? Front Physiol 2020; 10:1546. [PMID: 32009970 PMCID: PMC6974800 DOI: 10.3389/fphys.2019.01546] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 12/09/2019] [Indexed: 12/11/2022] Open
Abstract
Studies have revealed a robust and independent correlation between chronic kidney disease (CKD) and cardiovascular (CV) events, including death, heart failure, and myocardial infarction. Recent clinical trials extend this range of adverse CV events, including malignant ventricular arrhythmias and sudden cardiac death (SCD). Moreover, other studies point out that cardiac structural and electrophysiological changes are a common occurrence in this population. These processes are likely contributors to the heightened hazard of arrhythmias in CKD population and may be useful indicators to detect patients who are at a higher SCD risk. Sympathetic overactivity is associated with increased CV risk, specifically in the population with CKD, and it is a central feature of the hypertensive state, occurring early in its clinical course. Sympathetic hyperactivity is already evident at the earliest clinical stage of CKD and is directly related to the progression of renal failure, being most pronounced in those with end-stage renal disease. Sympathetic efferent and afferent neural activity in kidney failure is a crucial facilitator for the perpetuation and evolvement of the disease. Here, we will revisit the role of the feedback loop of the sympathetic neural cycle in the context of CKD and how it may aggravate several of the risk factors responsible for causing SCD. Targeting the overactive sympathetic nervous system therapeutically, either pharmacologically or with newly available device-based approaches, may prove to be a pivotal intervention to curb the substantial burden of cardiac arrhythmias and SCD in the high-risk population of patients with CKD.
Collapse
Affiliation(s)
- Márcio Galindo Kiuchi
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit/Medical Research Foundation, The University of Western Australia, Perth, WA, Australia
| | - Jan K Ho
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit/Medical Research Foundation, The University of Western Australia, Perth, WA, Australia
| | - Janis Marc Nolde
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit/Medical Research Foundation, The University of Western Australia, Perth, WA, Australia
| | - Leslie Marisol Lugo Gavidia
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit/Medical Research Foundation, The University of Western Australia, Perth, WA, Australia
| | - Revathy Carnagarin
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit/Medical Research Foundation, The University of Western Australia, Perth, WA, Australia
| | - Vance B Matthews
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit/Medical Research Foundation, The University of Western Australia, Perth, WA, Australia
| | - Markus P Schlaich
- Dobney Hypertension Centre, School of Medicine - Royal Perth Hospital Unit/Medical Research Foundation, The University of Western Australia, Perth, WA, Australia.,Departments of Cardiology and Nephrology, Royal Perth Hospital, Perth, WA, Australia.,Neurovascular Hypertension & Kidney Disease Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| |
Collapse
|
16
|
Effect of renal denervation procedure on left ventricular mass, myocardial strain and diastolic function by CMR on a 12-month follow-up. Jpn J Radiol 2019; 37:642-650. [DOI: 10.1007/s11604-019-00854-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 07/04/2019] [Indexed: 12/12/2022]
|
17
|
Kiuchi MG, Nolde JM, Villacorta H, Carnagarin R, Chan JJSY, Lugo-Gavidia LM, Ho JK, Matthews VB, Dwivedi G, Schlaich MP. New Approaches in the Management of Sudden Cardiac Death in Patients with Heart Failure-Targeting the Sympathetic Nervous System. Int J Mol Sci 2019; 20:E2430. [PMID: 31100908 PMCID: PMC6567277 DOI: 10.3390/ijms20102430] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/10/2019] [Accepted: 05/12/2019] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases (CVDs) have been considered the most predominant cause of death and one of the most critical public health issues worldwide. In the past two decades, cardiovascular (CV) mortality has declined in high-income countries owing to preventive measures that resulted in the reduced burden of coronary artery disease (CAD) and heart failure (HF). In spite of these promising results, CVDs are responsible for ~17 million deaths per year globally with ~25% of these attributable to sudden cardiac death (SCD). Pre-clinical data demonstrated that renal denervation (RDN) decreases sympathetic activation as evaluated by decreased renal catecholamine concentrations. RDN is successful in reducing ventricular arrhythmias (VAs) triggering and its outcome was not found inferior to metoprolol in rat myocardial infarction model. Registry clinical data also suggest an advantageous effect of RDN to prevent VAs in HF patients and electrical storm. An in-depth investigation of how RDN, a minimally invasive and safe method, reduces the burden of HF is urgently needed. Myocardial systolic dysfunction is correlated to neuro-hormonal overactivity as a compensatory mechanism to keep cardiac output in the face of declining cardiac function. Sympathetic nervous system (SNS) overactivity is supported by a rise in plasma noradrenaline (NA) and adrenaline levels, raised central sympathetic outflow, and increased organ-specific spillover of NA into plasma. Cardiac NA spillover in untreated HF individuals can reach ~50-fold higher levels compared to those of healthy individuals under maximal exercise conditions. Increased sympathetic outflow to the renal vascular bed can contribute to the anomalies of renal function commonly associated with HF and feed into a vicious cycle of elevated BP, the progression of renal disease and worsening HF. Increased sympathetic activity, amongst other factors, contribute to the progress of cardiac arrhythmias, which can lead to SCD due to sustained ventricular tachycardia. Targeted therapies to avoid these detrimental consequences comprise antiarrhythmic drugs, surgical resection, endocardial catheter ablation and use of the implantable electronic cardiac devices. Analogous NA agents have been reported for single photon-emission-computed-tomography (SPECT) scans usage, specially the 123I-metaiodobenzylguanidine (123I-MIBG). Currently, HF prognosis assessment has been improved by this tool. Nevertheless, this radiotracer is costly, which makes the use of this diagnostic method limited. Comparatively, positron-emission-tomography (PET) overshadows SPECT imaging, because of its increased spatial definition and broader reckonable methodologies. Numerous ANS radiotracers have been created for cardiac PET imaging. However, so far, [11C]-meta-hydroxyephedrine (HED) has been the most significant PET radiotracer used in the clinical scenario. Growing data has shown the usefulness of [11C]-HED in important clinical situations, such as predicting lethal arrhythmias, SCD, and all-cause of mortality in reduced ejection fraction HF patients. In this article, we discussed the role and relevance of novel tools targeting the SNS, such as the [11C]-HED PET cardiac imaging and RDN to manage patients under of SCD risk.
Collapse
Affiliation(s)
- Márcio Galindo Kiuchi
- Dobney Hypertension Cenre, School of Medicine-Royal Perth Hospital Unit, Faculty of Medicine, Dentistry & Health Sciences, The University of Western Australia Level 3, MRF Building, Rear 50 Murray St, Perth 6000, MDBP: M570, Australia.
| | - Janis Marc Nolde
- Dobney Hypertension Cenre, School of Medicine-Royal Perth Hospital Unit, Faculty of Medicine, Dentistry & Health Sciences, The University of Western Australia Level 3, MRF Building, Rear 50 Murray St, Perth 6000, MDBP: M570, Australia.
| | - Humberto Villacorta
- Cardiology Division, Department of Medicine, Universidade Federal Fluminense, Niterói, Rio de Janeiro 24033-900, Brazil.
| | - Revathy Carnagarin
- Dobney Hypertension Cenre, School of Medicine-Royal Perth Hospital Unit, Faculty of Medicine, Dentistry & Health Sciences, The University of Western Australia Level 3, MRF Building, Rear 50 Murray St, Perth 6000, MDBP: M570, Australia.
| | - Justine Joy Su-Yin Chan
- Dobney Hypertension Cenre, School of Medicine-Royal Perth Hospital Unit, Faculty of Medicine, Dentistry & Health Sciences, The University of Western Australia Level 3, MRF Building, Rear 50 Murray St, Perth 6000, MDBP: M570, Australia.
| | - Leslie Marisol Lugo-Gavidia
- Dobney Hypertension Cenre, School of Medicine-Royal Perth Hospital Unit, Faculty of Medicine, Dentistry & Health Sciences, The University of Western Australia Level 3, MRF Building, Rear 50 Murray St, Perth 6000, MDBP: M570, Australia.
| | - Jan K Ho
- Dobney Hypertension Cenre, School of Medicine-Royal Perth Hospital Unit, Faculty of Medicine, Dentistry & Health Sciences, The University of Western Australia Level 3, MRF Building, Rear 50 Murray St, Perth 6000, MDBP: M570, Australia.
| | - Vance B Matthews
- Dobney Hypertension Cenre, School of Medicine-Royal Perth Hospital Unit, Faculty of Medicine, Dentistry & Health Sciences, The University of Western Australia Level 3, MRF Building, Rear 50 Murray St, Perth 6000, MDBP: M570, Australia.
| | - Girish Dwivedi
- Harry Perkins Institute of Medical Research and Fiona Stanley Hospital, The University of Western Australia, Perth 6150, Australia.
| | - Markus P Schlaich
- Dobney Hypertension Cenre, School of Medicine-Royal Perth Hospital Unit, Faculty of Medicine, Dentistry & Health Sciences, The University of Western Australia Level 3, MRF Building, Rear 50 Murray St, Perth 6000, MDBP: M570, Australia.
- Departments of Cardiology and Nephrology, Royal Perth Hospital, Perth 6000, Australia.
- Neurovascular Hypertension & Kidney Disease Laboratory, Baker Heart and Diabetes Institute, Melbourne 3004, Australia.
| |
Collapse
|
18
|
Zheng H, Katsurada K, Liu X, Knuepfer MM, Patel KP. Specific Afferent Renal Denervation Prevents Reduction in Neuronal Nitric Oxide Synthase Within the Paraventricular Nucleus in Rats With Chronic Heart Failure. Hypertension 2019; 72:667-675. [PMID: 30012866 DOI: 10.1161/hypertensionaha.118.11071] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Renal denervation (RDN) has been shown to restore endogenous neuronal nitric oxide synthase (nNOS) in the paraventricular nucleus (PVN) and reduce sympathetic drive during chronic heart failure (CHF). The purpose of the present study was to assess the contribution of afferent renal nerves to the nNOS-mediated sympathetic outflow within the PVN in rats with CHF. CHF was induced in rats by ligation of the left coronary artery. Four weeks after surgery, selective afferent RDN (A-RDN) was performed by bilateral perivascular application of capsaicin on the renal arteries. Seven days after intervention, nNOS protein expression, nNOS immunostaining signaling, and diaphorase-positive stained cells were significantly decreased in the PVN of CHF rats, changes that were reversed by A-RDN. A-RDN reduced basal lumbar sympathetic nerve activity in rats with CHF (8.5%±0.5% versus 17.0%±1.2% of max). Microinjection of nNOS inhibitor L-NMMA (L-NG-monomethyl arginine citrate) into the PVN produced a blunted increase in lumbar sympathetic nerve activity in rats with CHF. This response was significantly improved after A-RDN (Δ lumbar sympathetic nerve activity: 25.7%±2.4% versus 11.2%±0.9%). Resting afferent renal nerves activity was substantially increased in CHF compared with sham rats (56.3%±2.4% versus 33.0%±4.7%). These results suggest that intact afferent renal nerves contribute to the reduction of nNOS in the PVN. A-RDN restores nNOS and thus attenuates the sympathoexcitation. Also, resting afferent renal nerves activity is elevated in CHF rats, which may highlight a crucial neural mechanism arising from the kidney in the maintenance of enhanced sympathetic drive in CHF.
Collapse
Affiliation(s)
- Hong Zheng
- From the Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion (H.Z., X.L.)
| | - Kenichi Katsurada
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (K.K., K.P.P.)
| | - Xuefei Liu
- From the Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion (H.Z., X.L.)
| | - Mark M Knuepfer
- Department of Pharmacology and Physiology, St. Louis University School of Medicine, MO (M.M.K.)
| | - Kaushik P Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (K.K., K.P.P.)
| |
Collapse
|
19
|
Heck SL, Gulati G, Hoffmann P, von Knobelsdorff-Brenkenhoff F, Storås TH, Ree AH, Gravdehaug B, Røsjø H, Steine K, Geisler J, Schulz-Menger J, Omland T. Effect of candesartan and metoprolol on myocardial tissue composition during anthracycline treatment: the PRADA trial. Eur Heart J Cardiovasc Imaging 2019; 19:544-552. [PMID: 29106497 DOI: 10.1093/ehjci/jex159] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 05/24/2017] [Indexed: 02/04/2023] Open
Abstract
Aims Anthracycline treatment may cause myocyte loss and expansion of the myocardial extracellular volume (ECV) fraction by oedema and fibrosis. We tested the hypotheses that adjuvant treatment for early breast cancer with the anthracycline epirubicin is dose dependently associated with increased ECV fraction and total ECV, as well as reduced total myocardial cellular volume, and that these changes could be prevented by concomitant angiotensin or beta-adrenergic blockade. Methods and results PRevention of cArdiac Dysfunction during Adjuvant breast cancer therapy (PRADA) was a 2 × 2 factorial, placebo-controlled, double-blinded trial of candesartan and metoprolol. Sixty-nine women had valid ECV measurements. ECV fraction, total ECV, and total cellular volume were measured by cardiovascular magnetic resonance before and at the completion of anthracycline therapy. ECV fraction increased from 27.5 ± 2.7% to 28.6 ± 2.9% (P = 0.002). A cumulative doxorubicin equivalent dose of 268 mg/m2 was associated with greater increase in ECV fraction than doses <268 mg/m2 (mean change 3.4% [95% confidence interval (CI) 1.2, 5.5] vs. 0.7% [95% CI 0.0, 1.5], P = 0.006), as well as greater increase in total ECV (1.9 mL [95% CI 0.4, 3.5] vs. 0.1 mL [95% CI -0.6, 0.8], P = 0.04). In patients receiving candesartan, total cellular volume decreased (-3.5 mL [95% CI - 4.7, -2.2], P < 0.001) while in patients not receiving candesartan, it remained unchanged (P = 0.45; between group difference P = 0.003). Conclusions Anthracycline therapy is associated with dose-dependent increase in ECV fraction and total ECV. Concomitant treatment with candesartan reduces left ventricular total cellular volume.
Collapse
Affiliation(s)
- Siri Lagethon Heck
- Department of Cardiology, Division of Medicine, Akershus University Hospital, Sykehusveien 25, 1478 Lørenskog, Norway.,Center for Heart Failure Research, University of Oslo, Campus AHUS, Sykehusveien 25, 1474 Nordbyhagen, Norway.,Department of Radiology, Akershus University Hospital, Sykehusveien 25, 1478 Lørenskog, Norway
| | - Geeta Gulati
- Department of Cardiology, Division of Medicine, Akershus University Hospital, Sykehusveien 25, 1478 Lørenskog, Norway.,Center for Heart Failure Research, University of Oslo, Campus AHUS, Sykehusveien 25, 1474 Nordbyhagen, Norway
| | - Pavel Hoffmann
- Section for Interventional Cardiology, Department of Cardiology, Oslo University Hospital, Ullevål, Kirkeveien 166, 0450 Oslo, Norway
| | - Florian von Knobelsdorff-Brenkenhoff
- Working Group Cardiovascular Magnetic Resonance, Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrueck Center for Molecular Medicine; and HELIOS Klinikum Berlin Buch, Department of Cardiology and Nephrology, Lindenberger Weg 80, 13125 Berlin, Germany.,Clinic Agatharied, Dept. of Cardiology, Ludwig-Maximilians-University of Munich, Norbert-Kerkel-Platz, 83734 Hausham, Germany
| | - Tryggve Holck Storås
- Department for Diagnostic Physics, KRN, Oslo University Hospital, Postboks 4950 Nydalen, 0424 OSLO, Norway
| | - Anne Hansen Ree
- Department of Oncology, Division of Medicine, Akershus University Hospital, Sykehusveien 25, 1478 Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, Campus AHUS, Sykehusveien 25, 1474 Nordbyhagen, Norway
| | - Berit Gravdehaug
- Department of Breast and Endocrine Surgery, Division of Surgery, Akershus University Hospital, Campus AHUS, Sykehusveien 25, 1478 Nordbyhagen, Norway; Lørenskog, Norway
| | - Helge Røsjø
- Department of Cardiology, Division of Medicine, Akershus University Hospital, Sykehusveien 25, 1478 Lørenskog, Norway.,Center for Heart Failure Research, University of Oslo, Campus AHUS, Sykehusveien 25, 1474 Nordbyhagen, Norway
| | - Kjetil Steine
- Department of Cardiology, Division of Medicine, Akershus University Hospital, Sykehusveien 25, 1478 Lørenskog, Norway.,Center for Heart Failure Research, University of Oslo, Campus AHUS, Sykehusveien 25, 1474 Nordbyhagen, Norway
| | - Jürgen Geisler
- Department of Oncology, Division of Medicine, Akershus University Hospital, Sykehusveien 25, 1478 Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, Campus AHUS, Sykehusveien 25, 1474 Nordbyhagen, Norway
| | - Jeanette Schulz-Menger
- Working Group Cardiovascular Magnetic Resonance, Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max-Delbrueck Center for Molecular Medicine; and HELIOS Klinikum Berlin Buch, Department of Cardiology and Nephrology, Lindenberger Weg 80, 13125 Berlin, Germany
| | - Torbjørn Omland
- Department of Cardiology, Division of Medicine, Akershus University Hospital, Sykehusveien 25, 1478 Lørenskog, Norway.,Center for Heart Failure Research, University of Oslo, Campus AHUS, Sykehusveien 25, 1474 Nordbyhagen, Norway
| |
Collapse
|
20
|
Liu X, Yan D, Guo SW. Sensory nerve-derived neuropeptides accelerate the development and fibrogenesis of endometriosis. Hum Reprod 2019; 34:452-468. [PMID: 30689856 DOI: 10.1093/humrep/dey392] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 12/04/2018] [Accepted: 12/14/2018] [Indexed: 12/16/2022] Open
Affiliation(s)
- Xishi Liu
- Shanghai OB/GYN Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China
| | - Dingmin Yan
- Shanghai OB/GYN Hospital, Fudan University, Shanghai, China
| | - Sun-Wei Guo
- Shanghai OB/GYN Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China
| |
Collapse
|
21
|
|
22
|
Warchol-Celinska E, Prejbisz A, Kadziela J, Florczak E, Januszewicz M, Michalowska I, Dobrowolski P, Kabat M, Sliwinski P, Klisiewicz A, Topor-Madry R, Narkiewicz K, Somers VK, Sobotka PA, Witkowski A, Januszewicz A. Renal Denervation in Resistant Hypertension and Obstructive Sleep Apnea. Hypertension 2018; 72:381-390. [PMID: 29941516 DOI: 10.1161/hypertensionaha.118.11180] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 04/18/2018] [Accepted: 04/29/2018] [Indexed: 12/31/2022]
Affiliation(s)
| | | | - Jacek Kadziela
- Department of Interventional Cardiology and Angiology (J.K., A.W.)
| | - Elzbieta Florczak
- From the Department of Hypertension (E.W.-C., A.P., E.F., M.K., A.J.)
| | - Magdalena Januszewicz
- Institute of Cardiology, Warsaw, Poland; Second Department of Radiology, Medical University of Warsaw, Poland (M.J.)
| | | | | | - Marek Kabat
- From the Department of Hypertension (E.W.-C., A.P., E.F., M.K., A.J.)
| | - Pawel Sliwinski
- Second Department of Respiratory Medicine, Institute of Tuberculosis and Lung Diseases, Warsaw, Poland (P.S.)
| | - Anna Klisiewicz
- Department of Interventional Cardiology and Angiology (J.K., A.W.)
- Department of Congenital Heart Diseases (P.D., A.K.)
| | - Roman Topor-Madry
- Institute of Public Health, Jagiellonian University Medical College, Cracow, Poland (R.T.-M.)
| | - Krzysztof Narkiewicz
- Department of Hypertension and Diabetology, Medical University of Gdansk, Poland (K.N.)
| | - Virend K. Somers
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (V.K.S.)
| | - Paul A. Sobotka
- Department of Cardiovascular Diseases, Ohio State University, Columbus (P.A.S.)
- Rox Medical, Inc, San Clemente, CA (P.A.S.)
| | | | | |
Collapse
|
23
|
Abstract
Purpose of Review The etiology of hypertension, a critical public health issue affecting one in three US adults, involves the integration of the actions of multiple organ systems, including the renal sympathetic nerves. The renal sympathetic nerves, which are comprised of both afferent (sensory input) and efferent (sympathetic outflow) arms, have emerged as a major potential therapeutic target to treat hypertension and disease states exhibiting excess renal sympathetic activity. Recent Findings This review highlights recent advances in both clinical and basic science that have provided new insight into the distribution, function, and reinnervation of the renal sympathetic nerves, with a focus on the renal afferent nerves, in hypertension and hypertension-evoked disease states including salt-sensitive hypertension, obesity-induced hypertension, and chronic kidney disease. Summary Increased understanding of the differential role of the renal afferent versus efferent nerves in the pathophysiology of hypertension has the potential to identify novel targets and refine therapeutic interventions designed to treat hypertension.
Collapse
|
24
|
Pinkham MI, Loftus MT, Amirapu S, Guild SJ, Quill G, Woodward WR, Habecker BA, Barrett CJ. Renal denervation in male rats with heart failure improves ventricular sympathetic nerve innervation and function. Am J Physiol Regul Integr Comp Physiol 2017; 312:R368-R379. [PMID: 28052866 DOI: 10.1152/ajpregu.00313.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 12/14/2016] [Accepted: 01/02/2017] [Indexed: 01/19/2023]
Abstract
Heart failure is characterized by the loss of sympathetic innervation to the ventricles, contributing to impaired cardiac function and arrhythmogenesis. We hypothesized that renal denervation (RDx) would reverse this loss. Male Wistar rats underwent myocardial infarction (MI) or sham surgery and progressed into heart failure for 4 wk before receiving bilateral RDx or sham RDx. After additional 3 wk, left ventricular (LV) function was assessed, and ventricular sympathetic nerve fiber density was determined via histology. Post-MI heart failure rats displayed significant reductions in ventricular sympathetic innervation and tissue norepinephrine content (nerve fiber density in the LV of MI+sham RDx hearts was 0.31 ± 0.05% vs. 1.00 ± 0.10% in sham MI+sham RDx group, P < 0.05), and RDx significantly increased ventricular sympathetic innervation (0.76 ± 0.14%, P < 0.05) and tissue norepinephrine content. MI was associated with an increase in fibrosis of the noninfarcted ventricular myocardium, which was attenuated by RDx. RDx improved LV ejection fraction and end-systolic and -diastolic areas when compared with pre-RDx levels. This is the first study to show an interaction between renal nerve activity and cardiac sympathetic nerve innervation in heart failure. Our findings show denervating the renal nerves improves cardiac sympathetic innervation and function in the post-MI failing heart.
Collapse
Affiliation(s)
| | - Michael T Loftus
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Satya Amirapu
- Department of Anatomy and Radiology, University of Auckland, Auckland, New Zealand
| | - Sarah-Jane Guild
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Gina Quill
- Department of Medicine, University of Auckland, Auckland, New Zealand; and
| | - William R Woodward
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon
| | - Beth A Habecker
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon
| | - Carolyn J Barrett
- Department of Physiology, University of Auckland, Auckland, New Zealand
| |
Collapse
|
25
|
Fengler K, Rommel KP, Okon T, Schuler G, Lurz P. Renal sympathetic denervation in therapy resistant hypertension - pathophysiological aspects and predictors for treatment success. World J Cardiol 2016; 8:436-46. [PMID: 27621771 PMCID: PMC4997524 DOI: 10.4330/wjc.v8.i8.436] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 06/21/2016] [Accepted: 07/14/2016] [Indexed: 02/06/2023] Open
Abstract
Many forms of human hypertension are associated with an increased systemic sympathetic activity. Especially the renal sympathetic nervous system has been found to play a prominent role in this context. Therefore, catheter-interventional renal sympathetic denervation (RDN) has been established as a treatment for patients suffering from therapy resistant hypertension in the past decade. The initial enthusiasm for this treatment was markedly dampened by the results of the Symplicity-HTN-3 trial, although the transferability of the results into clinical practice to date appears to be questionable. In contrast to the extensive use of RDN in treating hypertensive patients within or without clinical trial settings over the past years, its effects on the complex pathophysiological mechanisms underlying therapy resistant hypertension are only partly understood and are part of ongoing research. Effects of RDN have been described on many levels in human trials: From altered systemic sympathetic activity across cardiac and metabolic alterations down to changes in renal function. Most of these changes could sustainably change long-term morbidity and mortality of the treated patients, even if blood pressure remains unchanged. Furthermore, a number of promising predictors for a successful treatment with RDN have been identified recently and further trials are ongoing. This will certainly help to improve the preselection of potential candidates for RDN and thereby optimize treatment outcomes. This review summarizes important pathophysiologic effects of renal denervation and illustrates the currently known predictors for therapy success.
Collapse
Affiliation(s)
- Karl Fengler
- Karl Fengler, Karl Philipp Rommel, Thomas Okon, Gerhard Schuler, Philipp Lurz, Department of Internal Medicine/Cardiology, University of Leipzig - Heart Center, 04289 Leipzig, Germany
| | - Karl Philipp Rommel
- Karl Fengler, Karl Philipp Rommel, Thomas Okon, Gerhard Schuler, Philipp Lurz, Department of Internal Medicine/Cardiology, University of Leipzig - Heart Center, 04289 Leipzig, Germany
| | - Thomas Okon
- Karl Fengler, Karl Philipp Rommel, Thomas Okon, Gerhard Schuler, Philipp Lurz, Department of Internal Medicine/Cardiology, University of Leipzig - Heart Center, 04289 Leipzig, Germany
| | - Gerhard Schuler
- Karl Fengler, Karl Philipp Rommel, Thomas Okon, Gerhard Schuler, Philipp Lurz, Department of Internal Medicine/Cardiology, University of Leipzig - Heart Center, 04289 Leipzig, Germany
| | - Philipp Lurz
- Karl Fengler, Karl Philipp Rommel, Thomas Okon, Gerhard Schuler, Philipp Lurz, Department of Internal Medicine/Cardiology, University of Leipzig - Heart Center, 04289 Leipzig, Germany
| |
Collapse
|
26
|
Wei Y, Xu J, Zhou G, Chen S, Ouyang P, Liu S. Renal Denervation Suppresses the Inducibility of Atrial Fibrillation in a Rabbit Model for Atrial Fibrosis. PLoS One 2016; 11:e0160634. [PMID: 27529427 PMCID: PMC4986963 DOI: 10.1371/journal.pone.0160634] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 07/23/2016] [Indexed: 11/18/2022] Open
Abstract
Renal denervation (RD) was reported to reduce the susceptibility of atrial fibrillation (AF), but the underlying mechanism has not been well understood. This study was performed to investigate the effect of RD on the inducibility of AF in a rabbit model for atrial fibrosis and to explore the potential mechanisms. Thirty-five rabbits were randomly assigned into sham-operated group (n = 12), abdominal aortic constriction (AAC) group (n = 12) and AAC with RD (AAC-RD) group (n = 11). The incidence of AF induced by burst pacing in atriums was determined. Blood was collected to measure the levels of rennin, angiotensin II and aldosterone. Atrial samples were preserved to evaluate protein and gene expression of collagen, connective tissue growth factor (CTGF) and transforming growth factor-β1 (TGF-β1). Our data suggested cardiac structure remodeling and atrial fibrosis were successfully induced by AAC. Compared with the AAC group, the AAC-RD rabbits had smaller ascending aortic diameter and left ventricular end-systolic diameter. For burst pacing at the left atrium (LA), AF was induced in two of the 12 rabbits in the sham-operated group, 10 of the 12 rabbits in the AAC group, and 2 of the 11 rabbits in the AAC-RD group, with great difference among the three groups (P = 0.001). The percentage of LA burst stimulations with induced AF achieved 47.2% in the AAC group, which was higher than those in both the AAC-RD (12.1%) and the Sham-operated (5.6%) groups. Significantly increasing intercellular space in the AAC group (P<0.001) compared with the sham-operated rabbits. RD clearly decreased the volume fraction of collagen in LA and right atrium compared with that of the AAC group (P< 0.01). AAC-induced elevation of collagen I, CTGF and TGF-β1 was suppressed by RD. In conclusion, RD suppressed the inducibility of AF in a rabbit model for pressure associated atrial fibrosis, potentially by modulating renin-angiotensin-aldosterone system and decreasing pro-fibrotic factors.
Collapse
Affiliation(s)
- Yong Wei
- Department of cardiology, Shanghai Songjiang Central Hospital, Shanghai, 201600, China
- Department of cardiology, Shanghai First People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200800, China
| | - Juan Xu
- Department of cardiology, Shanghai First People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200800, China
| | - Genqing Zhou
- Department of cardiology, Shanghai First People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200800, China
| | - Songwen Chen
- Department of cardiology, Shanghai First People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200800, China
| | - Ping Ouyang
- Department of cardiology, Shanghai Songjiang Central Hospital, Shanghai, 201600, China
| | - Shaowen Liu
- Department of cardiology, Shanghai First People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200800, China
- * E-mail:
| |
Collapse
|
27
|
Fu L, Xu Y, Tu L, Huang H, Zhang Y, Chen Y, Tao L, Shen X. Oxymatrine inhibits aldosterone-induced rat cardiac fibroblast proliferation and differentiation by attenuating smad-2,-3 and-4 expression: an in vitro study. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 16:241. [PMID: 27457615 PMCID: PMC4960670 DOI: 10.1186/s12906-016-1231-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 07/19/2016] [Indexed: 01/07/2023]
Abstract
BACKGROUND We previously demonstrated oxymatrine, an alkaloid from the Chinese medicine radix Sophorae flavescentis, ameliorates hemodynamic disturbances and cardiac fibrosis; however, the underlying mechanisms are unclear. Here, we investigated the effect and mechanism of action of oxymatrine on aldosterone-induced cardiac fibroblast to myofibroblast differentiation in vitro. METHODS Cardiac fibroblasts were isolated purified from neonatal Sprague Dawley rats. The optimal concentration of aldosterone to stimulate cardiac fibroblast proliferation was determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Cardiac fibroblasts were pretreated with 7.57 × 10(-4) mol/L or 3.78 × 10(-4) mol/L oxymatrine or without oxymatrine for 2 h, and then coincubated with 1 × 10(-8) mol/L aldosterone for 48 h. The MTT assay and Masson staining were used to detect the cardiac fibroblast proliferation and myofibroblast differentiation. The secretion of type I and III collagen was measured by commercial ELISA kits, and the hydroxyproline content was determined by the colorimetric assay. Western blotting assayed the Smad-2, Smad-3, and Smad-4 protein expression in cardiac fibroblasts. RESULTS The present results confirmed that aldosterone induced cardiac fibroblast to myofibroblast proliferation and differentiation. The MTT assay and Masson staining indicated oxymatrine significantly inhibited aldosterone-induced cardiac fibroblast proliferation and myofibroblast differentiation. Oxymatrine significantly inhibited aldosterone-induced secretion of type I and III collagen, as indicated by commercial ELISA kits, and aldosterone-induced increase in hydroxyproline content, as indicated by a colorimetric assay. Western blotting revealed oxymatrine attenuated aldosterone-induced Smad-2, Smad-3, and Smad-4 expression in cardiac fibroblasts. CONCLUSION Oxymatrine can inhibit cardiac fibroblast proliferation and differentiation into myofibroblasts via a mechanism linked to attenuation of the Smad signaling pathway.
Collapse
Affiliation(s)
- Lingyun Fu
- Department of Pharmacology of Materia Medica, Guizhou Medical University, Huaxi University town, Guian New District, Guizhou, 550025, China
- The Key Laboratory of Optimal Utilization of Natural Medicinal Resources, Guizhou Medical University, Huaxi University town, Guian new district, Guizhou, 550025, China
| | - Yini Xu
- The Key Laboratory of Optimal Utilization of Natural Medicinal Resources, Guizhou Medical University, Huaxi University town, Guian new district, Guizhou, 550025, China
| | - Ling Tu
- Department of Pharmacology of Materia Medica, Guizhou Medical University, Huaxi University town, Guian New District, Guizhou, 550025, China
- The Key Laboratory of Optimal Utilization of Natural Medicinal Resources, Guizhou Medical University, Huaxi University town, Guian new district, Guizhou, 550025, China
| | - Haifeng Huang
- Department of Pharmacology of Materia Medica, Guizhou Medical University, Huaxi University town, Guian New District, Guizhou, 550025, China
- The Key Laboratory of Optimal Utilization of Natural Medicinal Resources, Guizhou Medical University, Huaxi University town, Guian new district, Guizhou, 550025, China
| | - Yanyan Zhang
- The Key Laboratory of Optimal Utilization of Natural Medicinal Resources, Guizhou Medical University, Huaxi University town, Guian new district, Guizhou, 550025, China
| | - Yan Chen
- Department of Pharmacology of Materia Medica, Guizhou Medical University, Huaxi University town, Guian New District, Guizhou, 550025, China
- The Key Laboratory of Optimal Utilization of Natural Medicinal Resources, Guizhou Medical University, Huaxi University town, Guian new district, Guizhou, 550025, China
| | - Ling Tao
- The Key Laboratory of Optimal Utilization of Natural Medicinal Resources, Guizhou Medical University, Huaxi University town, Guian new district, Guizhou, 550025, China
| | - Xiangchun Shen
- Department of Pharmacology of Materia Medica, Guizhou Medical University, Huaxi University town, Guian New District, Guizhou, 550025, China.
- The Key Laboratory of Optimal Utilization of Natural Medicinal Resources, Guizhou Medical University, Huaxi University town, Guian new district, Guizhou, 550025, China.
| |
Collapse
|
28
|
Effects of Renal Denervation on Renal Artery Function in Humans: Preliminary Study. PLoS One 2016; 11:e0150662. [PMID: 27003912 PMCID: PMC4803336 DOI: 10.1371/journal.pone.0150662] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 02/16/2016] [Indexed: 11/24/2022] Open
Abstract
Aim To study the effects of RD on renal artery wall function non-invasively using magnetic resonance. Methods and Results 32 patients undergoing RD were included. A 3.0 Tesla magnetic resonance of the renal arteries was performed before RD and after 6-month. We quantified the vessel sharpness of both renal arteries using a quantitative analysis tool (Soap-Bubble®). In 17 patients we assessed the maximal and minimal cross-sectional area of both arteries, peak velocity, mean flow, and renal artery distensibility. In a subset of patients wall shear stress was assessed with computational flow dynamics. Neither renal artery sharpness nor renal artery distensibility differed significantly. A significant increase in minimal and maximal areas (by 25.3%, p = 0.008, and 24.6%, p = 0.007, respectively), peak velocity (by 16.9%, p = 0.021), and mean flow (by 22.4%, p = 0.007) was observed after RD. Wall shear stress significantly decreased (by 25%, p = 0.029). These effects were observed in blood pressure responders and non-responders. Conclusions RD is not associated with adverse effects at renal artery level, and leads to an increase in cross-sectional areas, velocity and flow and a decrease in wall shear stress.
Collapse
|
29
|
Impact of Renal Sympathetic Denervation on Left Ventricular Structure and Function at 1-Year Follow-Up. PLoS One 2016; 11:e0149855. [PMID: 26934735 PMCID: PMC4774915 DOI: 10.1371/journal.pone.0149855] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 02/05/2016] [Indexed: 01/19/2023] Open
Abstract
Background Catheter-based sympathetic renal denervation (RDN) is a recent therapeutic option for patients with resistant hypertension. However, the impact of RDN in left ventricular (LV) mass and function is not completely established. Our aim was to evaluate the effects of RDN on LV structure and function (systolic and diastolic) in patients with resistant hypertension (HTN). Methods and Results From a single centre prospective registry including 65 consecutive patients with resistant HTN submitted to RDN between July-2011 and April-2015, 31 patients with baseline and 1-year follow-up echocardiogram were included in this analysis. Mean age was 65±7 years, 48% were males, 71% had type 2 diabetes. Most had hypertension lasting for more than 10 years (90%), and were being treated with a median number of 6 anti-hypertensive drugs, including 74% on spironolactone. At 1-year, there was a significant decrease both on office SBP (176±24 to 149±13mmHg, p<0.001) and DBP (90±14 to 79±11mmHg, p<0.001), and also in 24h ABPM SBP (150±20 to 132±14mmhg, p<0.001) and DBP (83±10 to 74±9mmHg, p<0.001). There was also a significant decrease in LV mass from 152±32 to 136±34g/m2 (p<0.001), an increase in LV end diastolic volume (93±18 to 111±27 mL, p = 0.004), an increase in LV ejection fraction (65±9 to 68±9%, p = 0.001) and mitral valve E deceleration time (225±49 to 247±51ms, p = 0.015) at 1-year follow up. There were no significant changes in left atrium volume index or in the distribution of patients among the different left ventricle geometric patterns and diastolic function subgroups. Conclusions In this single centre registry of patients with resistant hypertension, renal denervation was associated with significant reduction in both office and ABPM blood pressure and a significant decrease in left ventricle mass evaluated by transthoracic echocardiogram at 1 year follow-up.
Collapse
|
30
|
Schelbert EB, Messroghli DR. State of the Art: Clinical Applications of Cardiac T1 Mapping. Radiology 2016; 278:658-76. [DOI: 10.1148/radiol.2016141802] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
31
|
The effects of renal denervation on resistant hypertension patients: a meta-analysis. Blood Press Monit 2016; 21:206-14. [PMID: 26901340 DOI: 10.1097/mbp.0000000000000177] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
We carried out this meta-analysis to assess the effects of renal denervation (RDN) on resistant hypertension patients. According to the collaborative review group search strategy, we searched MEDLINE (1996 to 2015.10); EBCO (1996 to 2015.10) and CNKI. A meta-analysis was carried out using RevMan 5.0. We identified 11 reports that fulfilled the inclusion criteria for our review. Controlled trials reporting systolic blood pressure (SBP), diastolic blood pressure in RDN, and control groups at the 6-month follow-up in patients with resistant hypertension were systematically reviewed. Pooled analysis of all 11 included studies showed significant reductions in SBP (weighted mean difference -13.9 mmHg, 95% confidence interval -21.17 to -6.63, P=0.00025, I=93%) and diastolic blood pressure (weighted mean difference -4.41 mmHg, 95% confidence interval -6.95 to -1.88, P=0.004, I=90%) compared with the control group at the 6-month follow-up. Six controlled trials reported specific values of ambulatory SBP that showed no significant difference between two groups. It has also been found that RDN has benefits in protecting cardiac and renal function compared with the control group without increasing adverse events. In conclusion, this meta-analysis shows that RDN is superior to the control group in lowering office blood pressure rather than ambulatory SBP, and might have other potential benefits to protect heart and renal function.
Collapse
|
32
|
Reductions of left ventricular mass and atrial size following renal denervation: a meta-analysis. Clin Res Cardiol 2016; 105:648-656. [PMID: 26838292 DOI: 10.1007/s00392-016-0964-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 01/19/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND Renal denervation (RDN), a novel therapy for resistant hypertension, has been shown to have an effect on cardiac remodeling in several small studies. We aimed to pool currently available data to assess the effects of RDN on left ventricular hypertrophy (LVH) and left atrial (LA) enlargement. METHODS AND RESULTS Two investigators independently searched PubMed, EMBASE and Cochrane Library Central Register of Controlled Trials database for studies reporting change in left ventricular mass index (LVMI) or LA size before and after RDN. Twelve publications met our pre-defined inclusion criteria. Echocardiographic data showed that RDN markedly reduced both LVMI [weighted mean difference (WMD) = -15.77 g/m(2); 95 % confidence interval (CI) -22.51 to -9.02 g/m(2)] and LA diameter [WMD = -2.48 mm; 95 % CI -4.12 to -0.83 mm] after 6 months. Data from cardiac magnetic resonance also showed a significant reduction in LVMI [WMD = -5.43 g/m(2), 95 % CI -10.01 to -0.35 g/m(2)) at 6 months. Changes in LVH and LA size at 12 months were more pronounced than those at 6 months. Meta-regression analysis failed to demonstrate a significant relationship between RDN-induced LVMI reduction and BP lowering at 6 months. CONCLUSIONS RDN led to significant regressions of both LVH and LA enlargement at 6 months, which were sustained at least up to 12 months.
Collapse
|
33
|
Long-term effects of multielectrode renal denervation on cardiac adaptations in resistant hypertensive patients with left ventricular hypertrophy. J Hum Hypertens 2016; 30:714-719. [DOI: 10.1038/jhh.2015.127] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Revised: 11/25/2015] [Accepted: 12/04/2015] [Indexed: 11/09/2022]
|
34
|
Chronic kidney disease and risk factors responsible for sudden cardiac death: a whiff of hope? Kidney Res Clin Pract 2015; 35:3-9. [PMID: 27069851 PMCID: PMC4811986 DOI: 10.1016/j.krcp.2015.11.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 11/11/2015] [Accepted: 11/18/2015] [Indexed: 02/07/2023] Open
Abstract
Several studies have shown a strong independent association between chronic kidney disease (CKD) and cardiovascular events, including death, heart failure, and myocardial infarction. Recent clinical trials extend this range of adverse cardiovascular events, also including ventricular arrhythmias and sudden cardiac death. Furthermore, other studies suggest structural remodeling of the heart and electrophysiological alterations in this population. These processes may explain the increased risk of arrhythmia in kidney disease and help to identify patients who are at increased risk of sudden cardiac death. Sympathetic hyperactivity is well known to increase cardiovascular risk in CKD patients and is a hallmark of essential hypertensive state that occurs early in the clinical course of the disease. In CKD, the sympathetic hyperactivity seems to be expressed at the earliest clinical stage of the disease, showing a direct relationship with the severity of the condition of renal failure, being more pronounced in the terminal stage of CKD. The sympathetic efferent and afferent neural activity in kidney failure is a key mediator for the maintenance and progression of the disease. The aim of this review was to show that the feedback loop of this cycle, due to adrenergic hyperactivity, also aggravates many of the risk factors responsible for causing sudden cardiac death and may be a potential target modifiable by percutaneous renal sympathetic denervation. If it is feasible and effective in end-stage renal disease, little is known.
Collapse
|
35
|
Doltra A, Nasser SB, Messroghli D, Gebker R, Schnackenburg B, Pieske B, Kelle S. T1 Mapping for the Study of Cardiac Hypertrophy. CURRENT CARDIOVASCULAR IMAGING REPORTS 2015. [DOI: 10.1007/s12410-015-9362-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
36
|
Affiliation(s)
- Paulette Wehner
- Joan C Edwards School of Medicine, Marshall University, Huntington, WV (P.W., J.I.S.)
| | - Joseph I. Shapiro
- Joan C Edwards School of Medicine, Marshall University, Huntington, WV (P.W., J.I.S.)
| |
Collapse
|
37
|
Chin CWL, Pawade TA, Newby DE, Dweck MR. Risk Stratification in Patients With Aortic Stenosis Using Novel Imaging Approaches. Circ Cardiovasc Imaging 2015. [PMID: 26198161 DOI: 10.1161/circimaging.115.003421] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Calvin W L Chin
- From the BHF/Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (C.W.L.C., T.A.P., D.E.N., M.R.D.); and Department of Cardiovascular Medicine, National Heart Center Singapore, Singapore (C.W.L.C.).
| | - Tania A Pawade
- From the BHF/Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (C.W.L.C., T.A.P., D.E.N., M.R.D.); and Department of Cardiovascular Medicine, National Heart Center Singapore, Singapore (C.W.L.C.)
| | - David E Newby
- From the BHF/Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (C.W.L.C., T.A.P., D.E.N., M.R.D.); and Department of Cardiovascular Medicine, National Heart Center Singapore, Singapore (C.W.L.C.)
| | - Marc R Dweck
- From the BHF/Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (C.W.L.C., T.A.P., D.E.N., M.R.D.); and Department of Cardiovascular Medicine, National Heart Center Singapore, Singapore (C.W.L.C.)
| |
Collapse
|
38
|
Booth LC, May CN, Yao ST. The role of the renal afferent and efferent nerve fibers in heart failure. Front Physiol 2015; 6:270. [PMID: 26483699 PMCID: PMC4589650 DOI: 10.3389/fphys.2015.00270] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/14/2015] [Indexed: 01/14/2023] Open
Abstract
Renal nerves contain afferent, sensory and efferent, sympathetic nerve fibers. In heart failure (HF) there is an increase in renal sympathetic nerve activity (RSNA), which can lead to renal vasoconstriction, increased renin release and sodium retention. These changes are thought to contribute to renal dysfunction, which is predictive of poor outcome in patients with HF. In contrast, the role of the renal afferent nerves remains largely unexplored in HF. This is somewhat surprising as there are multiple triggers in HF that have the potential to increase afferent nerve activity, including increased venous pressure and reduced kidney perfusion. Some of the few studies investigating renal afferents in HF have suggested that at least the sympatho-inhibitory reno-renal reflex is blunted. In experimentally induced HF, renal denervation, both surgical and catheter-based, has been associated with some improvements in renal and cardiac function. It remains unknown whether the effects are due to removal of the efferent renal nerve fibers or afferent renal nerve fibers, or a combination of both. Here, we review the effects of HF on renal efferent and afferent nerve function and critically assess the latest evidence supporting renal denervation as a potential treatment in HF.
Collapse
Affiliation(s)
- Lindsea C Booth
- Florey Institute of Neuroscience and Mental Health, University of Melbourne Melbourne, VIC, Australia
| | - Clive N May
- Florey Institute of Neuroscience and Mental Health, University of Melbourne Melbourne, VIC, Australia
| | - Song T Yao
- Florey Institute of Neuroscience and Mental Health, University of Melbourne Melbourne, VIC, Australia
| |
Collapse
|
39
|
Briasoulis A, Bakris GL. A clinician's perspective of the role of renal sympathetic nerves in hypertension. Front Physiol 2015; 6:75. [PMID: 25859218 PMCID: PMC4373248 DOI: 10.3389/fphys.2015.00075] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 02/23/2015] [Indexed: 01/29/2023] Open
Abstract
The renal sympathetic nerves have significant contribution to the control of different aspects of kidney function. Early animal studies of renal denervation in a large number of different models of hypertension showed that that RDN improved BP control. Recently, data from prospective cohorts and randomized studies showed that renal denervation therapy (RDN) is a safe procedure but is associated with only modest reduction of ambulatory blood pressure (BP) in patients on intensive medical therapy. The main goal of this article is to review the results of preclinical and clinical studies on the contribution of the renal sympathetic nervous system to hypertension and the therapeutic applications of catheter-based renal denervation.
Collapse
Affiliation(s)
- Alexandros Briasoulis
- American Society of Hypertension Comprehensive Hypertension Center, Department of Medicine, The University of Chicago Medicine Chicago, IL, USA
| | - George L Bakris
- American Society of Hypertension Comprehensive Hypertension Center, Department of Medicine, The University of Chicago Medicine Chicago, IL, USA
| |
Collapse
|