51
|
Huang YJ, Ferrari MW, Lin S, Wang ZH. Recent advances on the Role of Gut Microbiota in the Development of Heart Failure by Mediating Immune Metabolism. Curr Probl Cardiol 2024; 49:102128. [PMID: 37802162 DOI: 10.1016/j.cpcardiol.2023.102128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 09/30/2023] [Indexed: 10/08/2023]
Abstract
The association between gut microbiota and the development of heart failure has become a research hotspot in recent years and the impact of gut microbiota on heart failure has attracted growing interest. From 2006 to 2021, the global research on gut microbiota and heart failure has gradually expanded, indicating a developed and promising research field. There were 40 countries, 196 institutions, and 257 authors involved in the publication on the relationship between gut microbiota and heart failure, respectively. In patients with heart failure, inadequate visceral perfusion leads to ischemia and intestinal edema, which compromise the gut barrier. This subsequently results in the translocation of bacteria and bacterial metabolites into the circulatory system and causes local and systemic inflammatory responses. The gastrointestinal tract contains the largest number of immune cells in the human body and gut microbiota play important roles in the immune system by promoting immune tolerance to symbiotic bacteria. Studies have shown that probiotics can act on gut microorganisms, thereby increasing choline metabolism and reducing plasma TMA and TMAO concentrations, thus inhibiting the development of heart failure. Meanwhile, probiotics induce the production of inflammatory suppressors to maintain gut immune stability and inhibit the progression of heart failure by reducing ventricular remodeling. Here, we review the current understanding of gut microbiota-driven immune dysfunction in experimental and clinical heart failure, as well as the therapeutic interventions that could be used to address these issues.
Collapse
Affiliation(s)
- Yu-Jing Huang
- Department of Cardiology, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, PR China
| | - Markus W Ferrari
- Clinic of Internal Medicine 1, HSK, Clinic of the City of Wiesbaden and the HELIOS Group, Wiesbaden, Germany.
| | - Shu Lin
- Centre of Neurological and Metabolic Research, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, PR China; Group of Neuroendocrinology, Garvan Institute of Medical Research, Sydney, Australia.
| | - Zhen-Hua Wang
- Department of Cardiology, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, PR China.
| |
Collapse
|
52
|
Tjandra PM, Ripplinger CM, Christiansen BA. The heart-bone connection: relationships between myocardial infarction and osteoporotic fracture. Am J Physiol Heart Circ Physiol 2024; 326:H845-H856. [PMID: 38305753 PMCID: PMC11062618 DOI: 10.1152/ajpheart.00576.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/03/2024]
Abstract
Myocardial infarction (MI) and osteoporotic fracture (Fx) are two of the leading causes of mortality and morbidity worldwide. Although these traumatic injuries are treated as if they are independent, there is epidemiological evidence linking the incidence of Fx and MI, thus raising the question of whether each of these events can actively influence the risk of the other. Atherosclerotic cardiovascular disease and osteoporosis, the chronic conditions leading to MI and Fx, are known to have shared pathoetiology. Furthermore, sustained systemic inflammation after traumas such as MI and Fx has been shown to exacerbate both underlying chronic conditions. However, the effects of MI and Fx outside their own system have not been well studied. The sympathetic nervous system (SNS) and the complement system initiate a systemic response after MI that could lead to subsequent changes in bone remodeling through osteoclasts. Similarly, SNS and complement system activation following fracture could lead to heart tissue damage and exacerbate atherosclerosis. To determine whether damaging bone-heart cross talk may be important comorbidity following Fx or MI, this review details the current understanding of bone loss after MI, cardiovascular damage after Fx, and possible shared underlying mechanisms of these processes.
Collapse
Affiliation(s)
- Priscilla M Tjandra
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California, United States
- Biomedical Engineering Graduate Group, University of California Davis, Davis, California, United States
| | - Crystal M Ripplinger
- Biomedical Engineering Graduate Group, University of California Davis, Davis, California, United States
- Department of Pharmacology, University of California Davis Health, Davis, California, United States
| | - Blaine A Christiansen
- Biomedical Engineering Graduate Group, University of California Davis, Davis, California, United States
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, California, United States
| |
Collapse
|
53
|
Trauzeddel RF, Rothe LM, Nordine M, Dehé L, Scholtz K, Spies C, Hadzidiakos D, Winterer G, Borchers F, Kruppa J, Treskatsch S. Influence of a chronic beta-blocker therapy on perioperative opioid consumption - a post hoc secondary analysis. BMC Anesthesiol 2024; 24:80. [PMID: 38413849 PMCID: PMC10898005 DOI: 10.1186/s12871-024-02456-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 02/12/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Beta-blocker (BB) therapy plays a central role in the treatment of cardiovascular diseases. An increasing number of patients with cardiovascular diseases undergoe noncardiac surgery, where opioids are an integral part of the anesthesiological management. There is evidence to suggest that short-term intravenous BB therapy may influence perioperative opioid requirements due to an assumed cross-talk between G-protein coupled beta-adrenergic and opioid receptors. Whether chronic BB therapy could also have an influence on perioperative opioid requirements is unclear. METHODS A post hoc analysis of prospectively collected data from a multicenter observational (BioCog) study was performed. Inclusion criteria consisted of elderly patients (≥ 65 years) undergoing elective noncardiac surgery as well as total intravenous general anesthesia without the use of regional anesthesia and duration of anesthesia ≥ 60 min. Two groups were defined: patients with and without BB in their regular preopreative medication. The administered opioids were converted to their respective morphine equivalent doses. Multiple regression analysis was performed using the morphine-index to identify independent predictors. RESULTS A total of 747 patients were included in the BioCog study in the study center Berlin. 106 patients fulfilled the inclusion criteria. Of these, 37 were on chronic BB. The latter were preoperatively significantly more likely to have arterial hypertension (94.6%), chronic renal failure (27%) and hyperlipoproteinemia (51.4%) compared to patients without BB. Both groups did not differ in terms of cumulative perioperative morphine equivalent dose (230.9 (BB group) vs. 214.8 mg (Non-BB group)). Predictive factors for increased morphine-index were older age, male sex, longer duration of anesthesia and surgery of the trunk. In a model with logarithmised morphine index, only gender (female) and duration of anesthesia remained predictive factors. CONCLUSIONS Chronic BB therapy was not associated with a reduced perioperative opioid consumption. TRIAL REGISTRATION This study was registered at ClinicalTrials.gov ( NCT02265263 ) on the 15.10.2014 with the principal investigator being Univ.-Prof. Dr. med. Claudia Spies.
Collapse
Affiliation(s)
- Ralf F Trauzeddel
- Department of Anesthesiology and Intensive Care Medicine, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität and Humboldt Universität zu Berlin, Hindenburgdamm 30, Berlin, 12203, Germany
| | - Luisa M Rothe
- IS Global Campus Cliníc Rosselló, Barcelona Institute for Global Health, 132, 7è, Barcelona, 08036, Spain
| | - Michael Nordine
- Department of Anesthesiology, Intensive Care Medicine, and Pain Therapy, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Lukas Dehé
- Department of Anesthesiology and Intensive Care Medicine, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität and Humboldt Universität zu Berlin, Hindenburgdamm 30, Berlin, 12203, Germany
| | - Kathrin Scholtz
- Department of Anesthesiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität and Humboldt Universität zu Berlin, Augustenburger Platz 1, Berlin, 13353, Germany
| | - Claudia Spies
- Department of Anesthesiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität and Humboldt Universität zu Berlin, Augustenburger Platz 1, Berlin, 13353, Germany
| | - Daniel Hadzidiakos
- Department of Anesthesiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität and Humboldt Universität zu Berlin, Augustenburger Platz 1, Berlin, 13353, Germany
| | - Georg Winterer
- Department of Anesthesiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität and Humboldt Universität zu Berlin, Augustenburger Platz 1, Berlin, 13353, Germany
| | - Friedrich Borchers
- Department of Anesthesiology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität and Humboldt Universität zu Berlin, Augustenburger Platz 1, Berlin, 13353, Germany
| | - Jochen Kruppa
- Hochschule Osnabrück, University of Applied Sciences, Osnabrück, Germany
| | - Sascha Treskatsch
- Department of Anesthesiology and Intensive Care Medicine, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität and Humboldt Universität zu Berlin, Hindenburgdamm 30, Berlin, 12203, Germany.
| |
Collapse
|
54
|
Doiron JE, Li Z, Yu X, LaPenna KB, Quiriarte H, Allerton TD, Koul K, Malek A, Shah SJ, Sharp TE, Goodchild TT, Kapusta DR, Lefer DJ. Early Renal Denervation Attenuates Cardiac Dysfunction in Heart Failure With Preserved Ejection Fraction. J Am Heart Assoc 2024; 13:e032646. [PMID: 38353216 PMCID: PMC11010115 DOI: 10.1161/jaha.123.032646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 12/08/2023] [Indexed: 02/21/2024]
Abstract
BACKGROUND The renal sympathetic nervous system modulates systemic blood pressure, cardiac performance, and renal function. Pathological increases in renal sympathetic nerve activity contribute to the pathogenesis of heart failure with preserved ejection fraction (HFpEF). We investigated the effects of renal sympathetic denervation performed at early or late stages of HFpEF progression. METHODS AND RESULTS Male ZSF1 obese rats were subjected to radiofrequency renal denervation (RF-RDN) or sham procedure at either 8 weeks or 20 weeks of age and assessed for cardiovascular function, exercise capacity, and cardiorenal fibrosis. Renal norepinephrine and renal nerve tyrosine hydroxylase staining were performed to quantify denervation following RF-RDN. In addition, renal injury, oxidative stress, inflammation, and profibrotic biomarkers were evaluated to determine pathways associated with RDN. RF-RDN significantly reduced renal norepinephrine and tyrosine hydroxylase content in both study cohorts. RF-RDN therapy performed at 8 weeks of age attenuated cardiac dysfunction, reduced cardiorenal fibrosis, and improved endothelial-dependent vascular reactivity. These improvements were associated with reductions in renal injury markers, expression of renal NLR family pyrin domain containing 3/interleukin 1β, and expression of profibrotic mediators. RF-RDN failed to exert beneficial effects when administered in the 20-week-old HFpEF cohort. CONCLUSIONS Our data demonstrate that early RF-RDN therapy protects against HFpEF disease progression in part due to the attenuation of renal fibrosis and inflammation. In contrast, the renoprotective and left ventricular functional improvements were lost when RF-RDN was performed in later HFpEF progression. These results suggest that RDN may be a viable treatment option for HFpEF during the early stages of this systemic inflammatory disease.
Collapse
Affiliation(s)
- Jake E. Doiron
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLAUSA
| | - Zhen Li
- Department of Cardiac SurgerySmidt Heart Institute, Cedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Xiaoman Yu
- Department of Cardiac SurgerySmidt Heart Institute, Cedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Kyle B. LaPenna
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLAUSA
| | - Heather Quiriarte
- Department of Vascular MetabolismPennington Biomedical Research CenterBaton RougeLAUSA
| | - Timothy D. Allerton
- Department of Vascular MetabolismPennington Biomedical Research CenterBaton RougeLAUSA
| | - Kashyap Koul
- School of MedicineLouisiana State University Health Sciences Center New OrleansNew OrleansLAUSA
| | - Andrew Malek
- School of MedicineLouisiana State University Health Sciences Center New OrleansNew OrleansLAUSA
| | - Sanjiv J. Shah
- Division of Cardiology, Department of Medicine and Bluhm Cardiovascular InstituteNorthwestern University Feinberg School of MedicineChicagoILUSA
| | - Thomas E. Sharp
- Department of Molecular Pharmacology and Physiology, Morsani College of MedicineUniversity of South FloridaTampaFLUSA
- USF Health Heart InstituteTampaFLUSA
| | - Traci T. Goodchild
- Department of Cardiac SurgerySmidt Heart Institute, Cedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Daniel R. Kapusta
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLAUSA
| | - David J. Lefer
- Department of Cardiac SurgerySmidt Heart Institute, Cedars‐Sinai Medical CenterLos AngelesCAUSA
| |
Collapse
|
55
|
Matsushita H, Saku K, Nishikawa T, Yokota S, Sato K, Morita H, Yoshida Y, Fukumitsu M, Uemura K, Kawada T, Yamaura K. The impact of ECPELLA on haemodynamics and global oxygen delivery: a comprehensive simulation of biventricular failure. Intensive Care Med Exp 2024; 12:13. [PMID: 38361021 PMCID: PMC10869331 DOI: 10.1186/s40635-024-00599-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/18/2024] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND ECPELLA, a combination of veno-arterial (VA) extracorporeal membrane oxygenation (ECMO) and Impella, a percutaneous left ventricular (LV) assist device, has emerged as a novel therapeutic option in patients with severe cardiogenic shock (CS). Since multiple cardiovascular and pump factors influence the haemodynamic effects of ECPELLA, optimising ECPELLA management remains challenging. In this study, we conducted a comprehensive simulation study of ECPELLA haemodynamics. We also simulated global oxygen delivery (DO2) under ECPELLA in severe CS and acute respiratory failure as a first step to incorporate global DO2 into our developed cardiovascular simulation. METHODS AND RESULTS Both the systemic and pulmonary circulations were modelled using a 5-element resistance‒capacitance network. The four ventricles were represented by time-varying elastances with unidirectional valves. In the scenarios of severe LV dysfunction, biventricular dysfunction with normal pulmonary vascular resistance (PVR, 0.8 Wood units), and biventricular dysfunction with high PVR (6.0 Wood units), we compared the changes in haemodynamics, pressure-volume relationship (PV loop), and global DO2 under different VA-ECMO flows and Impella support levels. RESULTS In the simulation, ECPELLA improved total systemic flow with a minimising biventricular pressure-volume loop, indicating biventricular unloading in normal PVR conditions. Meanwhile, increased Impella support level in high PVR conditions rendered the LV-PV loop smaller and induced LV suction in ECPELLA support conditions. The general trend of global DO2 was followed by the changes in total systemic flow. The addition of veno-venous ECMO (VV-ECMO) augmented the global DO2 increment under ECPELLA total support conditions. CONCLUSIONS The optimal ECPELLA support increased total systemic flow and achieved both biventricular unloading. The VV-ECMO effectively improves global DO2 in total ECPELLA support conditions.
Collapse
Affiliation(s)
- Hiroki Matsushita
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Keita Saku
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan.
| | - Takuya Nishikawa
- Department of Research Promotion and Management, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Shohei Yokota
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Kei Sato
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Hidetaka Morita
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Yuki Yoshida
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Masafumi Fukumitsu
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Kazunori Uemura
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
- NTTR-NCVC Bio Digital Twin Centre, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Toru Kawada
- Department of Cardiovascular Dynamics, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Ken Yamaura
- Department of Anesthesiology and Critical Care Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
56
|
Díaz-Jara E, Pereyra K, Vicencio S, Olesen MA, Schwarz KG, Toledo C, Díaz HS, Quintanilla RA, Del Rio R. Superoxide dismutase 2 deficiency is associated with enhanced central chemoreception in mice: Implications for breathing regulation. Redox Biol 2024; 69:102992. [PMID: 38142585 PMCID: PMC10788617 DOI: 10.1016/j.redox.2023.102992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/07/2023] [Indexed: 12/26/2023] Open
Abstract
AIMS In mammals, central chemoreception plays a crucial role in the regulation of breathing function in both health and disease conditions. Recently, a correlation between high levels of superoxide anion (O2.-) in the Retrotrapezoid nucleus (RTN), a main brain chemoreceptor area, and enhanced central chemoreception has been found in rodents. Interestingly, deficiency in superoxide dismutase 2 (SOD2) expression, a pivotal antioxidant enzyme, has been linked to the development/progression of several diseases. Despite, the contribution of SOD2 on O2.- regulation on central chemoreceptor function is unknown. Accordingly, we sought to determine the impact of partial deletion of SOD2 expression on i) O2.-accumulation in the RTN, ii) central ventilatory chemoreflex function, and iii) disordered-breathing. Finally, we study cellular localization of SOD2 in the RTN of healthy mice. METHODS Central chemoreflex drive and breathing function were assessed in freely moving heterozygous SOD2 knockout mice (SOD2+/- mice) and age-matched control wild type (WT) mice by whole-body plethysmography. O2.- levels were determined in RTN brainstem sections and brain isolated mitochondria, while SOD2 protein expression and tissue localization were determined by immunoblot, RNAseq and immunofluorescent staining, respectively. RESULTS Our results showed that SOD2+/- mice displayed reductions in SOD2 levels and high O2.- formation and mitochondrial dysfunction within the RTN compared to WT. Additionally, SOD2+/- mice displayed a heightened ventilatory response to hypercapnia and exhibited overt signs of altered breathing patterns. Both, RNAseq analysis and immunofluorescence co-localization studies showed that SOD2 expression was confined to RTN astrocytes but not to RTN chemoreceptor neurons. Finally, we found that SOD2+/- mice displayed alterations in RTN astrocyte morphology compared to RTN astrocytes from WT mice. INNOVATION & CONCLUSION These findings provide first evidence of the role of SOD2 in the regulation of O2.- levels in the RTN and its potential contribution on the regulation of central chemoreflex function. Our results suggest that reductions in the expression of SOD2 in the brain may contribute to increase O2.- levels in the RTN being the outcome a chronic surge in central chemoreflex drive and the development/maintenance of altered breathing patterns. Overall, dysregulation of SOD2 and the resulting increase in O2.- levels in brainstem respiratory areas can disrupt normal respiratory control mechanisms and contribute to breathing dysfunction seen in certain disease conditions characterized by high oxidative stress.
Collapse
Affiliation(s)
- Esteban Díaz-Jara
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Katherine Pereyra
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Sinay Vicencio
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Margrethe A Olesen
- Laboratory of Neurodegenerative Diseases, Universidad Autónoma de Chile, Santiago, Chile.
| | - Karla G Schwarz
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Camilo Toledo
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile; Institute of Physiology, Universidad Austral de Chile, Valdivia, Chile.
| | - Hugo S Díaz
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Rodrigo A Quintanilla
- Laboratory of Neurodegenerative Diseases, Universidad Autónoma de Chile, Santiago, Chile.
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile; Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile; Department of Cell Biology and Physiology, School of Medicine, University of Kansas Medical Center, Kansas City, KS, United States.
| |
Collapse
|
57
|
Doumani G, Spanos G, Theofilis P, Vordoni A, Kalaitzidis RG. Cardiorenal syndrome and iron supplementation-more benefits than risks: a narrative review. Int Urol Nephrol 2024; 56:597-606. [PMID: 37606747 DOI: 10.1007/s11255-023-03741-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/09/2023] [Indexed: 08/23/2023]
Abstract
Intravenous iron administration has emerged as a crucial intervention for managing patients with cardiorenal syndrome (CRS) and iron deficiency, with or without the presence of anemia. Multiple studies have demonstrated the benefits of intravenous iron supplementation in improving anemia, symptoms, and functional capacity in patients with HF and iron deficiency. Furthermore, iron supplementation has been associated with a reduction in hospitalizations for HF exacerbation and the improvement of patients' quality of life and clinical outcomes. In addition to its effects on HF management, emerging evidence suggests a potential positive impact on kidney function in patients with CRS. Studies have shown an increase in estimated glomerular filtration rate and improvements in renal function markers in patients receiving intravenous iron therapy, highlighting the potential of this intervention in patients with CRS. This paper reviews the existing literature on the impact of intravenous iron therapy in these patient populations and explores its effects on various clinical outcomes. Future research endeavors are eagerly awaited to further improve our understanding of its clinical implications and optimize patient outcomes.
Collapse
Affiliation(s)
- Georgia Doumani
- Center for Nephrology "G. Papadakis", General Hospital of Nikaia, Piraeus "Agios Panteleimon", 18454, Piraeus, Nikaia, Greece
| | - Georgios Spanos
- Department of Nephrology, General Hospital of Thessaloniki "G. Papanikolaou", Thessaloniki, Greece
| | - Panagiotis Theofilis
- Center for Nephrology "G. Papadakis", General Hospital of Nikaia, Piraeus "Agios Panteleimon", 18454, Piraeus, Nikaia, Greece
| | - Aikaterini Vordoni
- Center for Nephrology "G. Papadakis", General Hospital of Nikaia, Piraeus "Agios Panteleimon", 18454, Piraeus, Nikaia, Greece
| | - Rigas G Kalaitzidis
- Center for Nephrology "G. Papadakis", General Hospital of Nikaia, Piraeus "Agios Panteleimon", 18454, Piraeus, Nikaia, Greece.
| |
Collapse
|
58
|
Sandroni PB, Schroder MA, Hawkins HT, Bailon JD, Huang W, Hagen JT, Montgomery M, Hong SJ, Chin AL, Zhang J, Rodrigo MC, Kim B, Simpson PC, Schisler JC, Ellis JM, Fisher-Wellman KH, Jensen BC. The alpha-1A adrenergic receptor regulates mitochondrial oxidative metabolism in the mouse heart. J Mol Cell Cardiol 2024; 187:101-117. [PMID: 38331556 PMCID: PMC10861168 DOI: 10.1016/j.yjmcc.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 02/10/2024]
Abstract
AIMS The sympathetic nervous system regulates numerous critical aspects of mitochondrial function in the heart through activation of adrenergic receptors (ARs) on cardiomyocytes. Mounting evidence suggests that α1-ARs, particularly the α1A subtype, are cardioprotective and may mitigate the deleterious effects of chronic β-AR activation by shared ligands. The mechanisms underlying these adaptive effects remain unclear. Here, we tested the hypothesis that α1A-ARs adaptively regulate cardiomyocyte oxidative metabolism in both the uninjured and infarcted heart. METHODS We used high resolution respirometry, fatty acid oxidation (FAO) enzyme assays, substrate-specific electron transport chain (ETC) enzyme assays, transmission electron microscopy (TEM) and proteomics to characterize mitochondrial function comprehensively in the uninjured hearts of wild type and α1A-AR knockout mice and defined the effects of chronic β-AR activation and myocardial infarction on selected mitochondrial functions. RESULTS We found that isolated cardiac mitochondria from α1A-KO mice had deficits in fatty acid-dependent respiration, FAO, and ETC enzyme activity. TEM revealed abnormalities of mitochondrial morphology characteristic of these functional deficits. The selective α1A-AR agonist A61603 enhanced fatty-acid dependent respiration, fatty acid oxidation, and ETC enzyme activity in isolated cardiac mitochondria. The β-AR agonist isoproterenol enhanced oxidative stress in vitro and this adverse effect was mitigated by A61603. A61603 enhanced ETC Complex I activity and protected contractile function following myocardial infarction. CONCLUSIONS Collectively, these novel findings position α1A-ARs as critical regulators of cardiomyocyte metabolism in the basal state and suggest that metabolic mechanisms may underlie the protective effects of α1A-AR activation in the failing heart.
Collapse
Affiliation(s)
- Peyton B Sandroni
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America; McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Melissa A Schroder
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Hunter T Hawkins
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Julian D Bailon
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Wei Huang
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - James T Hagen
- Department of Physiology, East Carolina University, Brody School of Medicine, Greenville, NC, United States of America; East Carolina University Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC, United States of America
| | - McLane Montgomery
- Department of Physiology, East Carolina University, Brody School of Medicine, Greenville, NC, United States of America; East Carolina University Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC, United States of America
| | - Seok J Hong
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Andrew L Chin
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Jiandong Zhang
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America; Department of Medicine, Division of Cardiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Manoj C Rodrigo
- Cytokinetics, Inc., South San Francisco, CA, United States of America
| | - Boa Kim
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America; Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Paul C Simpson
- Department of Medicine and Research Service, San Francisco Veterans Affairs Medical Center, San Francisco, CA, United States of America; Cardiovascular Research Institute, University of California, San Francisco, CA, United States of America
| | - Jonathan C Schisler
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America; McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Jessica M Ellis
- Department of Physiology, East Carolina University, Brody School of Medicine, Greenville, NC, United States of America; East Carolina University Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC, United States of America
| | - Kelsey H Fisher-Wellman
- Department of Physiology, East Carolina University, Brody School of Medicine, Greenville, NC, United States of America; East Carolina University Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC, United States of America
| | - Brian C Jensen
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America; McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America; Department of Medicine, Division of Cardiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America.
| |
Collapse
|
59
|
Yang Y, Xu J, Shu S, Wang P, Liang Y, Liu B, Yang B, Zhang H, Zhao Q, Zhao Z, Luo Q, Liu Z, Zeng Q, Xiong C. Circulating acetylcholine serves as a potential biomarker role in pulmonary hypertension. BMC Pulm Med 2024; 24:35. [PMID: 38229103 DOI: 10.1186/s12890-024-02856-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 01/09/2024] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND An increased acetylcholine (ACh) level in the right ventricle tissue of pulmonary hypertension (PH) was revealed, which indicated the important role of ACh in disease pathogenesis. However, the relationship between plasma ACh levels and disease conditions and patients' prognosis has not been investigated. We aimed to explore the association between plasma ACh levels and the prognosis of patients with PH. We also discussed the feasibility of plasma ACh as a biomarker, which may contribute to the management of PH patients in the future. METHODS Patients with confirmed PH in Fuwai Hospital from April 2019 to August 2020 were enrolled. The primary clinical outcome in this study was defined as a composite outcome, including death/lung transplantation, heart failure, and worsening of symptoms. Fasting plasma was collected to detect the ACh levels. The association between ACh levels and patients' prognosis was explored. RESULTS Finally, four hundred and eight patients with PH were enrolled and followed for a mean period of 2.5 years. Patients in the high ACh group had worse World Health Organization Functional Class (WHO-FC), lower 6-minute walk distance (6 MWD), and higher N-terminal pro-brain natriuretic peptide (NT-proBNP). Notably, echocardiographic and hemodynamic parameters in the high metabolite group also suggested a worse disease condition compared with the low ACh group. After adjusting for confounders, compared with low ACh patients, those with high metabolite levels still have worse prognoses characterized as elevated risk of mortality, heart failure, and symptoms worsening. CONCLUSION High circulating ACh levels were associated with severe PH conditions and poor prognosis, which might serve as a potential biomarker in PH.
Collapse
Affiliation(s)
- Yicheng Yang
- Respiratory and Pulmonary Vascular Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, North Lishi Road, Xicheng District, No. 167, Beijing, 100037, China
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Jing Xu
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Songren Shu
- Department of Cardiac Surgery, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100037, China
| | - Peizhi Wang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Yanru Liang
- Respiratory and Pulmonary Vascular Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, North Lishi Road, Xicheng District, No. 167, Beijing, 100037, China
| | - Bingyang Liu
- Respiratory and Pulmonary Vascular Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, North Lishi Road, Xicheng District, No. 167, Beijing, 100037, China
| | - Beilan Yang
- Respiratory and Pulmonary Vascular Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, North Lishi Road, Xicheng District, No. 167, Beijing, 100037, China
| | - Hanwen Zhang
- Respiratory and Pulmonary Vascular Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, North Lishi Road, Xicheng District, No. 167, Beijing, 100037, China
| | - Qing Zhao
- Respiratory and Pulmonary Vascular Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, North Lishi Road, Xicheng District, No. 167, Beijing, 100037, China
| | - Zhihui Zhao
- Respiratory and Pulmonary Vascular Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, North Lishi Road, Xicheng District, No. 167, Beijing, 100037, China
| | - Qin Luo
- Respiratory and Pulmonary Vascular Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, North Lishi Road, Xicheng District, No. 167, Beijing, 100037, China
| | - Zhihong Liu
- Respiratory and Pulmonary Vascular Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, North Lishi Road, Xicheng District, No. 167, Beijing, 100037, China
| | - Qixian Zeng
- Respiratory and Pulmonary Vascular Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, North Lishi Road, Xicheng District, No. 167, Beijing, 100037, China.
| | - Changming Xiong
- Respiratory and Pulmonary Vascular Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, North Lishi Road, Xicheng District, No. 167, Beijing, 100037, China.
| |
Collapse
|
60
|
Nanda D, Pant P, Machha P, Sowpati DT, Kumarswamy R. Transcriptional changes during isoproterenol-induced cardiac fibrosis in mice. Front Mol Biosci 2023; 10:1263913. [PMID: 38178867 PMCID: PMC10765171 DOI: 10.3389/fmolb.2023.1263913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 10/23/2023] [Indexed: 01/06/2024] Open
Abstract
Introduction: β-adrenergic stimulation using β-agonists such as isoproterenol has been routinely used to induce cardiac fibrosis in experimental animal models. Although transcriptome changes in surgical models of cardiac fibrosis such as transverse aortic constriction (TAC) and coronary artery ligation (CAL) are well-studied, transcriptional changes during isoproterenol-induced cardiac fibrosis are not well-explored. Methods: Cardiac fibrosis was induced in male C57BL6 mice by administration of isoproterenol for 4, 8, or 11 days at 50 mg/kg/day dose. Temporal changes in gene expression were studied by RNA sequencing. Results and discussion: We observed a significant alteration in the transcriptome profile across the different experimental groups compared to the saline group. Isoproterenol treatment caused upregulation of genes associated with ECM organization, cell-cell contact, three-dimensional structure, and cell growth, while genes associated with fatty acid oxidation, sarcoplasmic reticulum calcium ion transport, and cardiac muscle contraction are downregulated. A number of known long non-coding RNAs (lncRNAs) and putative novel lncRNAs exhibited differential regulation. In conclusion, our study shows that isoproterenol administration leads to the dysregulation of genes relevant to ECM deposition and cardiac contraction, and serves as an excellent alternate model to the surgical models of heart failure.
Collapse
Affiliation(s)
- Disha Nanda
- Council of Scientific and Industrial Research (CSIR)–Centre for Cellular and Molecular Biology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Priyanka Pant
- Council of Scientific and Industrial Research (CSIR)–Centre for Cellular and Molecular Biology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Pratheusa Machha
- Council of Scientific and Industrial Research (CSIR)–Centre for Cellular and Molecular Biology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Divya Tej Sowpati
- Council of Scientific and Industrial Research (CSIR)–Centre for Cellular and Molecular Biology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Regalla Kumarswamy
- Council of Scientific and Industrial Research (CSIR)–Centre for Cellular and Molecular Biology, Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
61
|
Abele N, Münz F, Zink F, Gröger M, Hoffmann A, Wolfschmitt EM, Hogg M, Calzia E, Waller C, Radermacher P, Merz T. Relation of Plasma Catecholamine Concentrations and Myocardial Mitochondrial Respiratory Activity in Anesthetized and Mechanically Ventilated, Cardiovascular Healthy Swine. Int J Mol Sci 2023; 24:17293. [PMID: 38139121 PMCID: PMC10743631 DOI: 10.3390/ijms242417293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Chronic heart failure is associated with reduced myocardial β-adrenergic receptor expression and mitochondrial function. Since these data coincide with increased plasma catecholamine levels, we investigated the relation between myocardial β-receptor expression and mitochondrial respiratory activity under conditions of physiological catecholamine concentrations. This post hoc analysis used material of a prospective randomized, controlled study on 12 sexually mature (age 20-24 weeks) Early Life Stress or control pigs (weaning at day 21 and 28-35 after birth, respectively) of either sex. Measurements in anesthetized, mechanically ventilated, and instrumented animals comprised serum catecholamine (liquid-chromatography/tandem-mass-spectrometry) and 8-isoprostane levels, whole blood superoxide anion concentrations (electron spin resonance), oxidative DNA strand breaks (tail moment in the "comet assay"), post mortem cardiac tissue mitochondrial respiration, and immunohistochemistry (β2-adrenoreceptor, mitochondrial respiration complex, and nitrotyrosine expression). Catecholamine concentrations were inversely related to myocardial mitochondrial respiratory activity and β2-adrenoceptor expression, whereas there was no relation to mitochondrial respiratory complex expression. Except for a significant, direct, non-linear relation between DNA damage and noradrenaline levels, catecholamine concentrations were unrelated to markers of oxidative stress. The present study suggests that physiological variations of the plasma catecholamine concentrations, e.g., due to physical and/or psychological stress, may affect cardiac β2-adrenoceptor expression and mitochondrial respiration.
Collapse
Affiliation(s)
- Nadja Abele
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89069 Ulm, Germany; (N.A.); (F.Z.); (M.G.); (A.H.); (E.-M.W.); (M.H.); (E.C.)
| | - Franziska Münz
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89069 Ulm, Germany; (N.A.); (F.Z.); (M.G.); (A.H.); (E.-M.W.); (M.H.); (E.C.)
- Clinic for Anesthesiology and Intensive Care, Ulm University Medical Center, 89069 Ulm, Germany
| | - Fabian Zink
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89069 Ulm, Germany; (N.A.); (F.Z.); (M.G.); (A.H.); (E.-M.W.); (M.H.); (E.C.)
| | - Michael Gröger
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89069 Ulm, Germany; (N.A.); (F.Z.); (M.G.); (A.H.); (E.-M.W.); (M.H.); (E.C.)
| | - Andrea Hoffmann
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89069 Ulm, Germany; (N.A.); (F.Z.); (M.G.); (A.H.); (E.-M.W.); (M.H.); (E.C.)
| | - Eva-Maria Wolfschmitt
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89069 Ulm, Germany; (N.A.); (F.Z.); (M.G.); (A.H.); (E.-M.W.); (M.H.); (E.C.)
| | - Melanie Hogg
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89069 Ulm, Germany; (N.A.); (F.Z.); (M.G.); (A.H.); (E.-M.W.); (M.H.); (E.C.)
| | - Enrico Calzia
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89069 Ulm, Germany; (N.A.); (F.Z.); (M.G.); (A.H.); (E.-M.W.); (M.H.); (E.C.)
| | - Christiane Waller
- Clinic for Psychosomatic Medicine and Psychotherapy, Paracelsus Medical Private University, 90402 Nuremberg, Germany;
| | - Peter Radermacher
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89069 Ulm, Germany; (N.A.); (F.Z.); (M.G.); (A.H.); (E.-M.W.); (M.H.); (E.C.)
| | - Tamara Merz
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89069 Ulm, Germany; (N.A.); (F.Z.); (M.G.); (A.H.); (E.-M.W.); (M.H.); (E.C.)
- Clinic for Anesthesiology and Intensive Care, Ulm University Medical Center, 89069 Ulm, Germany
| |
Collapse
|
62
|
Triska J, Tamargo J, Bozkurt B, Elkayam U, Taylor A, Birnbaum Y. An Updated Review on the Role of Non-dihydropyridine Calcium Channel Blockers and Beta-blockers in Atrial Fibrillation and Acute Decompensated Heart Failure: Evidence and Gaps. Cardiovasc Drugs Ther 2023; 37:1205-1223. [PMID: 35357604 DOI: 10.1007/s10557-022-07334-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/24/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE The 2021 European Society of Cardiology guidelines on acute and chronic heart failure (HF) recommend that non-dihydropyridine calcium channel blockers (NDCC) should be avoided in patients with HF with reduced ejection fraction. It also emphasizes that beta-blockers only be initiated in clinically stable, euvolemic patients. Despite these recommendations, NDCC and beta-blockers are often still employed in patients with AF with rapid ventricular response and acute decompensated HF. The relative safety and efficacy of these therapies in this setting is unclear. METHODS To address the question of the safety and efficacy of NDCC and beta-blockers for acute rate control in decompensated HF, we provide a perspective on the literature of NDCC and beta-blockers in chronic HF with reduced and preserved ejection fraction and AF, including trials on the management of AF with rapid ventricular response with and without HF. RESULTS Robust data demonstrates mortality benefits when beta-blockers are used in patients with chronic HF with reduced ejection fraction. The data that inform the contraindication of NDCC in HF with reduced ejection fraction are outdated and were not primarily designed to address the efficacy and safety of rate control of AF in patients with HF. Several studies indicate that for acute rate control, NDCC and beta-blockers are both efficacious therapies, especially in the setting of tachycardia-induced cardiomyopathy. CONCLUSION Future studies are needed to assess the safety and efficacy of beta-blockers and NDCC in both acute and chronic AF with HF with reduced and preserved ejection fraction.
Collapse
Affiliation(s)
- Jeffrey Triska
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| | - Juan Tamargo
- Department of Pharmacology and Toxicology School of Medicine, Institute Gregorio Marañón, Universidad Complutense, Madrid, Spain
| | - Biykem Bozkurt
- Department of Medicine, Section of Cardiology, Baylor College of Medicine, Houston, TX, USA
| | - Uri Elkayam
- Department of Medicine, Division of Cardiovascular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Addison Taylor
- Department of Medicine, Division of Hypertension and Clinical Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Yochai Birnbaum
- Department of Medicine, Section of Cardiology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
63
|
Felippe ISA, Río RD, Schultz H, Machado BH, Paton JFR. Commonalities and differences in carotid body dysfunction in hypertension and heart failure. J Physiol 2023; 601:5527-5551. [PMID: 37747109 PMCID: PMC10873039 DOI: 10.1113/jp284114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 08/29/2023] [Indexed: 09/26/2023] Open
Abstract
Carotid body pathophysiology is associated with many cardiovascular-respiratory-metabolic diseases. This pathophysiology reflects both hyper-sensitivity and hyper-tonicity. From both animal models and human patients, evidence indicates that amelioration of this pathophysiological signalling improves disease states such as a lowering of blood pressure in hypertension, a reduction of breathing disturbances with improved cardiac function in heart failure (HF) and a re-balancing of autonomic activity with lowered sympathetic discharge. Given this, we have reviewed the mechanisms of carotid body hyper-sensitivity and hyper-tonicity across disease models asking whether there is uniqueness related to specific disease states. Our analysis indicates some commonalities and some potential differences, although not all mechanisms have been fully explored across all disease models. One potential commonality is that of hypoperfusion of the carotid body across hypertension and HF, where the excessive sympathetic drive may reduce blood flow in both models and, in addition, lowered cardiac output in HF may potentiate the hypoperfusion state of the carotid body. Other mechanisms are explored that focus on neurotransmitter and signalling pathways intrinsic to the carotid body (e.g. ATP, carbon monoxide) as well as extrinsic molecules carried in the blood (e.g. leptin); there are also transcription factors found in the carotid body endothelium that modulate its activity (Krüppel-like factor 2). The evidence to date fully supports that a better understanding of the mechanisms of carotid body pathophysiology is a fruitful strategy for informing potential new treatment strategies for many cardiovascular, respiratory and metabolic diseases, and this is highly relevant clinically.
Collapse
Affiliation(s)
- Igor S. A. Felippe
- Manaaki Manawa – The Centre for Heart Research, Department of Physiology, Faculty of Health & Medical Sciences, University of Auckland, Grafton, Auckland, 1023, New Zealand
| | - Rodrigo Del Río
- Department of Physiology, Laboratory of Cardiorespiratory Control, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
- Mechanisms of Myelin Formation and Repair Laboratory, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
- Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Harold Schultz
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Benedito H. Machado
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Julian F. R. Paton
- Manaaki Manawa – The Centre for Heart Research, Department of Physiology, Faculty of Health & Medical Sciences, University of Auckland, Grafton, Auckland, 1023, New Zealand
| |
Collapse
|
64
|
Liu Y, Zhang X, Yang S, Zhou Z, Tian L, Li W, Wei J, Abliz Z, Wang Z. Integrated mass spectrometry imaging reveals spatial-metabolic alteration in diabetic cardiomyopathy and the intervention effects of ferulic acid. J Pharm Anal 2023; 13:1496-1509. [PMID: 38223449 PMCID: PMC10785252 DOI: 10.1016/j.jpha.2023.08.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/28/2023] [Accepted: 08/10/2023] [Indexed: 01/16/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is a metabolic disease and a leading cause of heart failure among people with diabetes. Mass spectrometry imaging (MSI) is a versatile technique capable of combining the molecular specificity of mass spectrometry (MS) with the spatial information of imaging. In this study, we used MSI to visualize metabolites in the rat heart with high spatial resolution and sensitivity. We optimized the air flow-assisted desorption electrospray ionization (AFADESI)-MSI platform to detect a wide range of metabolites, and then used matrix-assisted laser desorption ionization (MALDI)-MSI for increasing metabolic coverage and improving localization resolution. AFADESI-MSI detected 214 and 149 metabolites in positive and negative analyses of rat heart sections, respectively, while MALDI-MSI detected 61 metabolites in negative analysis. Our study revealed the heterogenous metabolic profile of the heart in a DCM model, with over 105 region-specific changes in the levels of a wide range of metabolite classes, including carbohydrates, amino acids, nucleotides, and their derivatives, fatty acids, glycerol phospholipids, carnitines, and metal ions. The repeated oral administration of ferulic acid during 20 weeks significantly improved most of the metabolic disorders in the DCM model. Our findings provide novel insights into the molecular mechanisms underlying DCM and the potential of ferulic acid as a therapeutic agent for treating this condition.
Collapse
Affiliation(s)
- Yanhua Liu
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing, 100081, China
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China
| | - Xin Zhang
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing, 100081, China
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China
| | - Shu Yang
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing, 100081, China
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China
| | - Zhi Zhou
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing, 100081, China
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China
| | - Lu Tian
- New Drug Safety Evaluation Center, Institute of Materia Medica, Peking Union Medical College, Beijing, 100050, China
| | - Wanfang Li
- New Drug Safety Evaluation Center, Institute of Materia Medica, Peking Union Medical College, Beijing, 100050, China
| | - Jinfeng Wei
- New Drug Safety Evaluation Center, Institute of Materia Medica, Peking Union Medical College, Beijing, 100050, China
| | - Zeper Abliz
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing, 100081, China
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, 100081, China
| | - Zhonghua Wang
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing, 100081, China
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, 100081, China
| |
Collapse
|
65
|
Yamada Y, Kiyota N, Yoshida M, Omodaka K, Nakazawa T. The Relationship Between Kiritsu-Meijin-Derived Autonomic Function Parameters and Visual-Field Defects in Eyes with Open-Angle Glaucoma. Curr Eye Res 2023; 48:1006-1013. [PMID: 37405397 DOI: 10.1080/02713683.2023.2234105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/04/2023] [Indexed: 07/06/2023]
Abstract
PURPOSE This retrospective cross-sectional study aimed to investigate the association between autonomic parameters measured using the Kiritsu-Meijin device and visual-field defects in patients with open-angle glaucoma. METHODS A total of 79 eyes of 42 patients with open-angle glaucoma were enrolled in this study. Kiritsu-Meijin testing comprised three phases: sitting, standing, and sitting again (2 min, 2 min, and 1 min, respectively). Continuous electrocardiograms were recorded for five minutes. Autonomic parameters were extracted from the resulting data and analyzed, including activity, balance, reaction, switchover, and recovery; these are five representative parameters derived from Kiritsu-Meijin testing. Correlations between these parameters and mean deviation from Humphrey visual field testing were determined. Additionally, we used a linear mixed-effects model to observe sectoral differences in the relationship between total deviation and the Kiritsu-Meijin parameters. In this study, we focused on superior, central, and inferior total deviations. RESULTS Significant positive correlations were observed between activity, balance, and recovery and mean deviation values (β = 0.29-0.38, p < .05). The β value between activity and inferior total deviation was higher than that between activity and superior total deviation (β = 0.22, p < .05). Balance did not show any sectoral differences (p > .05). Recovery was more strongly associated with central to inferior total deviation than superior total deviation (β = 0.17-0.25, p < .05). CONCLUSION Our findings suggest that in patients with open-angle glaucoma, lower activity and recovery are associated with more severe central and/or inferior visual field defects in the superior quadrant. These results imply that measurements of autonomic function made with the Kiritsu-Meijin device may have clinical utility in the management of glaucoma.
Collapse
Affiliation(s)
- Yurina Yamada
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Naoki Kiyota
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mitsuhide Yoshida
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuko Omodaka
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Toru Nakazawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Ophthalmic Imaging and Information Analytics, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
66
|
Boden K, Pongratanakul P, Vogel J, Willemsen N, Jülke EM, Balitzki J, Tinel H, Truebel H, Dinh W, Mondritzki T. Telemetric long-term assessment of autonomic function in experimental heart failure. J Pharmacol Toxicol Methods 2023; 124:107480. [PMID: 37979811 DOI: 10.1016/j.vascn.2023.107480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 11/20/2023]
Abstract
Despite medical advances in the treatment of heart failure (HF), mortality remains high. It has been shown that alterations of the autonomic-nervous-system (ANS) are associated with HF progression and increased mortality. Preclinical models are required to evaluate the effectiveness of novel treatments modulating the autonomic imbalance. However, there are neither standard models nor diagnostic methods established to measure sympathetic and parasympathetic outflow continuously. Digital technologies might be a reliable tool for continuous assessment of autonomic function within experimental HF models. Telemetry devices and pacemakers were implanted in beagle dogs (n = 6). HF was induced by ventricular pacing. Cardiac hemodynamics, plasma catecholamines and parameter describing the ANS ((heart rate variability (HRV), deceleration capacity (DC), and baroreflex sensitivity (BRS)) were continuously measured at baseline, during HF conditions and during recovery phase. The pacing regime led to the expected depression in cardiac hemodynamics. Telemetric assessment of the ANS function showed a significant decrease in Total power, DC, and Heart rate recovery, whereas BRS was not significantly affected. In contrast, plasma catecholamines, revealing sympathetic activity, showed only a significant increase in the recovery phase. A precise diagnostic of the ANS in the context of HF is becoming increasingly important in experimental models. Up to now, these models have shown many limitations. Here we present the continuous assessment of the autonomic function in the progression of HF. We could demonstrate the advantage of highly resolved ANS measurement by HR and BP derived parameters due to early detection of an autonomic imbalance in the progression of HF.
Collapse
Affiliation(s)
- Katharina Boden
- Bayer AG, Wuppertal, Germany; University of Witten/Herdecke, Witten, Germany
| | | | - Julia Vogel
- University of Witten/Herdecke, Witten, Germany; Clinic for Cardiology and Angiology, West-German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Germany
| | - Nicola Willemsen
- Bayer AG, Wuppertal, Germany; University of Duisburg-, Essen, Germany
| | | | - Jakob Balitzki
- Bayer AG, Wuppertal, Germany; Hannover Medical School, Hannover, Germany
| | | | | | - Wilfried Dinh
- Bayer AG, Wuppertal, Germany; University of Witten/Herdecke, Witten, Germany; Department of Cardiology, HELIOS Clinic Wuppertal, University Hospital Witten/Herdecke, Wuppertal, Germany
| | - Thomas Mondritzki
- Bayer AG, Wuppertal, Germany; University of Witten/Herdecke, Witten, Germany.
| |
Collapse
|
67
|
Triposkiadis F, Briasoulis A, Sarafidis P, Magouliotis D, Athanasiou T, Paraskevaidis I, Skoularigis J, Xanthopoulos A. The Sympathetic Nervous System in Hypertensive Heart Failure with Preserved LVEF. J Clin Med 2023; 12:6486. [PMID: 37892623 PMCID: PMC10607346 DOI: 10.3390/jcm12206486] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/09/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
The neurohormonal model of heart failure (HF) pathogenesis states that a reduction in cardiac output caused by cardiac injury results in sympathetic nervous system (SNS) activation, that is adaptive in the short-term and maladaptive in the long-term. This model has proved extremely valid and has been applied in HF with a reduced left ventricular (LV) ejection fraction (LVEF). In contrast, it has been undermined in HF with preserved LVEF (HFpEF), which is due to hypertension (HTN) in the vast majority of the cases. Erroneously, HTN, which is the leading cause of cardiovascular disease and premature death worldwide and is present in more than 90% of HF patients, is tightly linked with SNS overactivity. In this paper we provide a contemporary overview of the contribution of SNS overactivity to the development and progression of hypertensive HF (HHF) as well as the clinical implications resulting from therapeutic interventions modifying SNS activity. Throughout the manuscript the terms HHF with preserved LVEF and HfpEF will be used interchangeably, considering that the findings in most HFpEF studies are driven by HTN.
Collapse
Affiliation(s)
| | - Alexandros Briasoulis
- Department of Therapeutics, Heart Failure and Cardio-Oncology Clinic, National and Kapodistrian University of Athens, 115 27 Athens, Greece;
| | - Pantelis Sarafidis
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece;
| | - Dimitrios Magouliotis
- Unit of Quality Improvement, Department of Cardiothoracic Surgery, University of Thessaly, 411 10 Biopolis, Greece;
| | - Thanos Athanasiou
- Department of Surgery and Cancer, Imperial College London, St Mary’s Hospital, London W2 1NY, UK;
| | | | - John Skoularigis
- Department of Cardiology, University Hospital of Larissa, 411 10 Larissa, Greece;
| | - Andrew Xanthopoulos
- Department of Cardiology, University Hospital of Larissa, 411 10 Larissa, Greece;
| |
Collapse
|
68
|
Scharp D, Harkins SE, Topaz M. Comorbidities of community-dwelling older adults with urinary incontinence: A scoping review. Geriatr Nurs 2023; 53:280-294. [PMID: 37598432 PMCID: PMC10529939 DOI: 10.1016/j.gerinurse.2023.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/22/2023]
Abstract
BACKGROUND Identifying comorbidities is a critical first step to building clinical phenotypes to improve assessment, management, and outcomes. OBJECTIVES 1) Identify relevant comorbidities of community-dwelling older adults with urinary incontinence, 2) provide insights about relationships between conditions. METHODS PubMed, Cumulative Index of Nursing and Allied Health Literature, and Embase were searched. Eligible studies had quantitative designs that analyzed urinary incontinence as the exposure or outcome variable. Critical appraisal was performed using the Joanna Briggs Institute Critical Appraisal Checklists. RESULTS Ten studies were included. Most studies had methodological weaknesses in the measurement of conditions. Comorbidities affecting the neurologic, cardiovascular, psychologic, respiratory, endocrine, genitourinary, and musculoskeletal systems were found to be associated with urinary incontinence. CONCLUSION Existing literature suggests that comorbidities and urinary incontinence are interrelated. Further research is needed to examine symptoms, shared mechanisms, and directionality of relationships to generate clinical phenotypes, evidence-based holistic care guidelines, and improve outcomes.
Collapse
Affiliation(s)
- Danielle Scharp
- Columbia University School of Nursing, 560 West 168(th) Street, New York, NY 10032, United States.
| | - Sarah E Harkins
- Columbia University School of Nursing, 560 West 168(th) Street, New York, NY 10032, United States.
| | - Maxim Topaz
- Columbia University School of Nursing, 560 West 168(th) Street, New York, NY 10032, United States.
| |
Collapse
|
69
|
Wafi AM. Nrf2 and autonomic dysregulation in chronic heart failure and hypertension. Front Physiol 2023; 14:1206527. [PMID: 37719456 PMCID: PMC10500196 DOI: 10.3389/fphys.2023.1206527] [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: 04/15/2023] [Accepted: 08/14/2023] [Indexed: 09/19/2023] Open
Abstract
Redox imbalance plays essential role in the pathogenesis of cardiovascular diseases. Chronic heart failure (CHF) and hypertension are associated with central oxidative stress, which is partly mediated by the downregulation of antioxidant enzymes in the central autonomic neurons that regulate sympathetic outflow, resulting in sympathoexcitation. Antioxidant proteins are partially regulated by the transcriptional factor nuclear factor erythroid 2-related factor 2 (Nrf2). Downregulation of Nrf2 is key to disrupting central redox homeostasis and mediating sympathetic nerve activity in the setting of Chronic heart failure and hypertension. Nrf2, in turn, is regulated by various mechanisms, such as extracellular vesicle-enriched microRNAs derived from several cell types, including heart and skeletal muscle. In this review, we discuss the role of Nrf2 in regulating oxidative stress in the brain and its impact on sympathoexcitation in Chronic heart failure and hypertension. Importantly, we also discuss interorgan communication via extracellular vesicle pathways that mediate central redox imbalance through Nrf2 signaling.
Collapse
Affiliation(s)
- Ahmed M. Wafi
- Physiology Department, Faculty of Medicine, Jazan University, Jizan, Saudi Arabia
| |
Collapse
|
70
|
Cincotta AH. Brain Dopamine-Clock Interactions Regulate Cardiometabolic Physiology: Mechanisms of the Observed Cardioprotective Effects of Circadian-Timed Bromocriptine-QR Therapy in Type 2 Diabetes Subjects. Int J Mol Sci 2023; 24:13255. [PMID: 37686060 PMCID: PMC10487918 DOI: 10.3390/ijms241713255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/19/2023] [Accepted: 07/27/2023] [Indexed: 09/10/2023] Open
Abstract
Despite enormous global efforts within clinical research and medical practice to reduce cardiovascular disease(s) (CVD), it still remains the leading cause of death worldwide. While genetic factors clearly contribute to CVD etiology, the preponderance of epidemiological data indicate that a major common denominator among diverse ethnic populations from around the world contributing to CVD is the composite of Western lifestyle cofactors, particularly Western diets (high saturated fat/simple sugar [particularly high fructose and sucrose and to a lesser extent glucose] diets), psychosocial stress, depression, and altered sleep/wake architecture. Such Western lifestyle cofactors are potent drivers for the increased risk of metabolic syndrome and its attendant downstream CVD. The central nervous system (CNS) evolved to respond to and anticipate changes in the external (and internal) environment to adapt survival mechanisms to perceived stresses (challenges to normal biological function), including the aforementioned Western lifestyle cofactors. Within the CNS of vertebrates in the wild, the biological clock circuitry surveils the environment and has evolved mechanisms for the induction of the obese, insulin-resistant state as a survival mechanism against an anticipated ensuing season of low/no food availability. The peripheral tissues utilize fat as an energy source under muscle insulin resistance, while increased hepatic insulin resistance more readily supplies glucose to the brain. This neural clock function also orchestrates the reversal of the obese, insulin-resistant condition when the low food availability season ends. The circadian neural network that produces these seasonal shifts in metabolism is also responsive to Western lifestyle stressors that drive the CNS clock into survival mode. A major component of this natural or Western lifestyle stressor-induced CNS clock neurophysiological shift potentiating the obese, insulin-resistant state is a diminution of the circadian peak of dopaminergic input activity to the pacemaker clock center, suprachiasmatic nucleus. Pharmacologically preventing this loss of circadian peak dopaminergic activity both prevents and reverses existing metabolic syndrome in a wide variety of animal models of the disorder, including high fat-fed animals. Clinically, across a variety of different study designs, circadian-timed bromocriptine-QR (quick release) (a unique formulation of micronized bromocriptine-a dopamine D2 receptor agonist) therapy of type 2 diabetes subjects improved hyperglycemia, hyperlipidemia, hypertension, immune sterile inflammation, and/or adverse cardiovascular event rate. The present review details the seminal circadian science investigations delineating important roles for CNS circadian peak dopaminergic activity in the regulation of peripheral fuel metabolism and cardiovascular biology and also summarizes the clinical study findings of bromocriptine-QR therapy on cardiometabolic outcomes in type 2 diabetes subjects.
Collapse
|
71
|
Hassan MZO, Tawakol A, Wang Y, Alvi RM, Awadalla M, Jones-O’Connor M, B. Bakar R, Banerji D, Rokicki A, Zhang L, Mulligan CP, Osborne MT, Zarif A, Hammad B, Chan AW, Wirth LJ, Warner ET, Pitman RK, Armstrong KA, Addison D, Neilan TG. Amygdalar activity measured using FDG-PET/CT at head and neck cancer staging independently predicts survival. PLoS One 2023; 18:e0279235. [PMID: 37540647 PMCID: PMC10403142 DOI: 10.1371/journal.pone.0279235] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 12/03/2022] [Indexed: 08/06/2023] Open
Abstract
IMPORTANCE The mechanisms underlying the association between chronic stress and higher mortality among individuals with cancer remain incompletely understood. OBJECTIVE To test the hypotheses that among individuals with active head and neck cancer, that higher stress-associated neural activity (ie. metabolic amygdalar activity [AmygA]) at cancer staging associates with survival. DESIGN Retrospective cohort study. SETTING Academic Medical Center (Massachusetts General Hospital, Boston). PARTICIPANTS 240 patients with head and neck cancer (HNCA) who underwent 18F-FDG-PET/CT imaging as part of initial cancer staging. MEASUREMENTS 18F-FDG uptake in the amygdala was determined by placing circular regions of interest in the right and left amygdalae and measuring the mean tracer accumulation (i.e., standardized uptake value [SUV]) in each region of interest. Amygdalar uptake was corrected for background cerebral activity (mean temporal lobe SUV). RESULTS Among individuals with HNCA (age 59±13 years; 30% female), 67 died over a median follow-up period of 3 years (IQR: 1.7-5.1). AmygA associated with heightened bone marrow activity, leukocytosis, and C-reactive protein (P<0.05 each). In adjusted and unadjusted analyses, AmygA associated with subsequent mortality (HR [95% CI]: 1.35, [1.07-1.70], P = 0.009); the association persisted in stratified subset analyses restricted to patients with advanced cancer stage (P<0.001). Individuals within the highest tertile of AmygA experienced a 2-fold higher mortality rate compared to others (P = 0.01). The median progression-free survival was 25 months in patients with higher AmygA (upper tertile) as compared with 36.5 months in other individuals (HR for progression or death [95%CI], 1.83 [1.24-2.68], P = 0.001). CONCLUSIONS AND RELEVANCE AmygA, quantified on routine 18F-FDG-PET/CT images obtained at cancer staging, independently and robustly predicts mortality and cancer progression among patients with HNCA. Future studies should test whether strategies that attenuate AmygA (or its downstream biological consequences) may improve cancer survival.
Collapse
Affiliation(s)
- Malek Z. O. Hassan
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Royal Papworth Hospital, Trumpington, Cambridge, United Kingdom
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ahmed Tawakol
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Nuclear Cardiology, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ying Wang
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Raza M. Alvi
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Magid Awadalla
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Maeve Jones-O’Connor
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Rula B. Bakar
- Department of Medical Sciences, Oxford University, Oxford, United Kingdom
| | - Dahlia Banerji
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Adam Rokicki
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Lili Zhang
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Connor P. Mulligan
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Michael T. Osborne
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Nuclear Cardiology, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Azmaeen Zarif
- Royal Papworth Hospital, Trumpington, Cambridge, United Kingdom
| | - Basma Hammad
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Annie W. Chan
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Lori J. Wirth
- Division of Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Erica T. Warner
- Clinical Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Roger K. Pitman
- Department of Psychiatry, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Katrina A. Armstrong
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Daniel Addison
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Cardiology, Department of Medicine, Ohio State University, Columbus, Ohio, United States of America
| | - Tomas G. Neilan
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
72
|
Oriot P, Hermans MP, Beauloye C, Rogghe PA, Noel S, Paternotte E. Unsuspected diabetic ketoacidosis after myocardial infarction in a patient treated with SGLT2 inhibitor increased length of stay in the hospital: how can it be prevented? A case report. Eur Heart J Case Rep 2023; 7:ytad336. [PMID: 37681058 PMCID: PMC10481769 DOI: 10.1093/ehjcr/ytad336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 07/05/2023] [Accepted: 07/19/2023] [Indexed: 09/09/2023]
Abstract
Background As the use of sodium-glucose co-transporter 2 inhibitors (SGLT2is) has expanded beyond glucose-lowering therapy in type 2 diabetes mellitus (T2DM), including chronic kidney disease and heart failure, there has also been an increase in reported cases of diabetic ketoacidosis (DKA) associated with SGLT2i. Case summary A 77-year-old woman with T2DM presented to the emergency department with ST-segment elevation myocardial infarction (MI) complicated by atrial fibrillation. Her medications included empagliflozin, an SGLT2i, initiated for T2DM. Diabetic ketoacidosis was suspected on the basis of a large anion gap, despite a plasma glucose level below 200 mg/dL (11.1 mmol/L) and the absence of symptoms, including nausea and vomiting. Laboratory tests confirmed metabolic acidosis and high ketones. However, the diagnosis of euglycaemic DKA (eu-DKA) was delayed due to lack of symptoms and moderate hyperglycaemia. The patient was successfully treated according to DKA management guidelines. She was discharged on insulin, and SGLT2i was discontinued. Discussion This is a case of asymptomatic eu-DKA after acute MI (AMI). We discuss the use of SGLT2is in AMI and arrhythmias from a review of the literature and the prophylaxis of eu-DKA. Regular monitoring of blood glucose and ketones should be performed in hospitalized T2DM patients treated with SGLT2i. The SGLT2i should be stopped as soon as possible in the event of critical illness or suspected DKA in the setting of an acute illness such as AMI. To help clinicians prevent this potentially fatal disease, we propose a flowchart for the prophylactic management of eu-DKA among inpatients.
Collapse
Affiliation(s)
- Philippe Oriot
- Department of Diabetology, Mouscron Hospital Centre, Avenue de Fécamp 49, 7700 Mouscron, Belgium
| | - Michel P Hermans
- Division of Endocrinology & Nutrition, Université catholique de louvain, Avenue Hippocrate 10, 1200 Woluwe-Saint-Lambert, Brussels, Belgium
- IREC – Institut de Recherche Expérimentale et Clinique Avenue Hippocrate, 55 bte B1.55.02 - 1200 Woluwé-Saint-Lambert – Belgium
| | - Christophe Beauloye
- Division of Cardiology, Université catholique de louvain, Avenue Hippocrate 10, 1200 Woluwe-Saint-Lambert, Brussels, Belgium
- IREC – Institut de Recherche Expérimentale et Clinique Avenue Hippocrate, 55 bte B1.55.02 - 1200 Woluwé-Saint-Lambert – Belgium
| | - Pierre-Arnaud Rogghe
- Department of Critical Care Unit, Mouscron Hospital Centre, Avenue de Fécamp 49, 7700 Mouscron, Belgium
| | - Sophie Noel
- Université Catholique de Louvain (UCLouvain), Av Hippocrate 10/2806, B-1200 Brussels, Belgium
| | - Emmanuelle Paternotte
- Department of Cardiology, Mouscron Hospital Centre, Avenue de Fécamp 49, 7700 Mouscron, Belgium
| |
Collapse
|
73
|
Martinez A, Lakkimsetti M, Maharjan S, Aslam MA, Basnyat A, Kafley S, Reddy SS, Ahmed SS, Razzaq W, Adusumilli S, Khawaja UA. Beta-Blockers and Their Current Role in Maternal and Neonatal Health: A Narrative Review of the Literature. Cureus 2023; 15:e44043. [PMID: 37746367 PMCID: PMC10517705 DOI: 10.7759/cureus.44043] [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: 07/11/2023] [Accepted: 08/24/2023] [Indexed: 09/26/2023] Open
Abstract
Beta-blockers are a class of medications that act on beta-adrenergic receptors and are categorized as cardio-selective and non-selective. They are principally used to treat cardiovascular conditions such as hypertension and arrhythmias. Beta-blockers have also been used to treat non-cardiogenic indications in non-pregnant individuals and the pediatric population. In pregnancy, labetalol is the mainstay treatment for hypertension and other cardiovascular indications. However, contraindications to certain sub-types of beta-blockers include bradycardia, heart failure, obstructive lung diseases, and hemodynamic instability. There is conflicting evidence of the adverse effects on fetal and neonatal health due to a scarce safety and efficacy profile, and further studies are necessary to understand the pharmacokinetics of the different classes of beta-blockers in pregnancy and fetal health. Understanding the hemodynamic changes during the stages of pregnancy is important to target a more beneficial therapy for both mother and fetus as well as better neonatal outcomes. Beta-blocker use in the pediatric population is less documented in studies but does have the potential to treat various cardiogenic and non-cardiogenic conditions. Future comprehensive studies would further benefit the direction of beta-blocker treatment during pregnancy in neonates and pediatrics.
Collapse
Affiliation(s)
- Andrea Martinez
- Medical School, Universidad Autonoma de Guadalajara, Zapopan, MEX
| | | | - Sameep Maharjan
- General Practice, Patan Academy of Health Sciences, Kathmandu, NPL
| | - Muhammad Ammar Aslam
- Medical School, Sargodha Medical College, University of Health Sciences, Sargodha, PAK
| | - Anouksha Basnyat
- General Practice, Hospital for Advanced Medicine & Surgery (HAMS), Kathmandu, NPL
| | - Shashwat Kafley
- Medical School, Enam Medical College and Hospital, Dhaka, BGD
| | | | - Saima S Ahmed
- Vascular Surgery, Dow International Medical College, Karachi, PAK
| | - Waleed Razzaq
- Internal Medicine, Services Hospital Lahore, Lahore, PAK
| | | | | |
Collapse
|
74
|
Wu Z, Liao J, Liu Q, Zhou S, Chen M. Chronic vagus nerve stimulation in patients with heart failure: challenge or failed translation? Front Cardiovasc Med 2023; 10:1052471. [PMID: 37534273 PMCID: PMC10390725 DOI: 10.3389/fcvm.2023.1052471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 05/31/2023] [Indexed: 08/04/2023] Open
Abstract
Autonomic imbalance between the sympathetic and parasympathetic nervous systems contributes to the progression of chronic heart failure (HF). Preclinical studies have demonstrated that various neuromodulation strategies may exert beneficial cardioprotective effects in preclinical models of HF. Based on these encouraging experimental data, vagus nerve stimulation (VNS) has been assessed in patients with HF with a reduced ejection fraction. Nevertheless, the main trials conducted thus far have yielded conflicting findings, questioning the clinical efficacy of VNS in this context. This review will therefore focus on the role of the autonomic nervous system in HF pathophysiology and VNS therapy, highlighting the potential reasons behind the discrepancy between preclinical and clinical studies.
Collapse
Affiliation(s)
- Zhihong Wu
- Department of Cardiovascular, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jiaying Liao
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qiming Liu
- Department of Cardiovascular, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shenghua Zhou
- Department of Cardiovascular, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Mingxian Chen
- Department of Cardiovascular, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
75
|
Li Y, Xiao W, Huang N, Zhuang Z, Zhang L, Wang W, Song Z, Zhao Y, Dong X, Xu M, Huang T. The effects of blood pressure and antihypertensive drugs on heart failure: A Mendelian randomization study. Nutr Metab Cardiovasc Dis 2023; 33:1420-1428. [PMID: 37156668 DOI: 10.1016/j.numecd.2023.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 05/10/2023]
Abstract
BACKGROUND AND AIMS Heart failure (HF) is often triggered by hypertension and can benefit from antihypertensive treatment. We aimed to investigate whether pulse pressure (PP) could independently raise the risk of HF beyond systolic blood pressure (SBP) and diastolic blood pressure (DBP), as well as explore the potential mechanisms of antihypertensives in HF prevention. METHODS AND RESULTS We generated genetic proxies for SBP, DBP, PP, and five drug classes based on a massive genome-wide association study. We applied two-sample Mendelian randomization (MR) using summary statistics derived from European individuals and conducted summary data-based MR (SMR) with gene expression data. In univariate analysis, PP showed an obvious association with HF risk (OR, 1.24 per 10 mm Hg increment; 95% CI, 1.16 to 1.32), which was largely attenuated in multivariable analysis when adjusted for SBP (0.89; 0.77 to 1.04). A significant decrease in HF risk was obtained with genetically proxied β-blockers (equivalent to a 10 mm Hg reduction in SBP, 0.71; 0.62 to 0.82) and calcium channel blockers (0.71; 0.65 to 0.78), but not with genetically proxied angiotensin-converting enzyme inhibitors (0.69; 0.40 to 1.19) and thiazide diuretics (0.80; 0.47 to 1.37). Additionally, the enrichment of expression for the KCNH2 gene, a target gene of β-blockers, in blood vessels and nerves was significantly associated with HF risk. CONCLUSION Our findings suggest that PP may not be an independent risk factor for HF. β-blockers and calcium channel blockers have a protective effect against HF, which at least partly depends on their blood pressure-lowering effect.
Collapse
Affiliation(s)
- Yueying Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100871, China
| | - Wendi Xiao
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100871, China
| | - Ninghao Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100871, China
| | - Zhenhuang Zhuang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100871, China
| | - Linjing Zhang
- Department of Neurology, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Wenxiu Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100871, China
| | - Zimin Song
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100871, China
| | - Yimin Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100871, China
| | - Xue Dong
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100871, China
| | - Ming Xu
- Department of Cardiology, Institute of Vascular Medicine, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Third Hospital, Beijing 100191, China; State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China.
| | - Tao Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, 100871, China; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Ministry of Education, Beijing, China; Center for Intelligent Public Health, Academy for Artificial Intelligence, Peking University, Beijing, China.
| |
Collapse
|
76
|
Champaneria MK, Patel RS, Oroszi TL. When blood pressure refuses to budge: exploring the complexity of resistant hypertension. Front Cardiovasc Med 2023; 10:1211199. [PMID: 37416924 PMCID: PMC10322223 DOI: 10.3389/fcvm.2023.1211199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 05/25/2023] [Indexed: 07/08/2023] Open
Abstract
Resistant hypertension, defined as blood pressure that remains above goal despite using three or more antihypertensive medications, including a diuretic, affects a significant proportion of the hypertensive population and is associated with increased cardiovascular morbidity and mortality. Despite the availability of a wide range of pharmacological therapies, achieving optimal blood pressure control in patients with resistant hypertension remains a significant challenge. However, recent advances in the field have identified several promising treatment options, including spironolactone, mineralocorticoid receptor antagonists, and renal denervation. In addition, personalized management approaches based on genetic and other biomarkers may offer new opportunities to tailor therapy and improve outcomes. This review aims to provide an overview of the current state of knowledge regarding managing resistant hypertension, including the epidemiology, pathophysiology, and clinical implications of the condition, as well as the latest developments in therapeutic strategies and future prospects.
Collapse
|
77
|
Thanigainathan T, Sharma A, Patel C, Seth S, Roy A, Pandey AK, Gupta P, Kumar R, Kumar P, Bal CS. Imaging of cardiac sympathetic dysfunction with 18F-FDOPA PET/CT in patients with heart failure: a pilot study. J Nucl Cardiol 2023; 30:1147-1157. [PMID: 36474068 DOI: 10.1007/s12350-022-03150-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 10/11/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Routine use of cardiac sympathetic imaging in HF has been limited by the lower availability/sensitivity of radiotracers. This study was aimed to assess the feasibility of 18F-FDOPA (commonly available PET-radiotracer) in assessment of cardiac autonomic dysfunction. METHODS Twenty-four controls (46.5 ± 11.1 years, 16men) and 24 patients (43.5 ± 11.0 years, 18men) with diagnosed HF (Framingham-Criteria) underwent cardiac-PET/CT. Region(s) Of Interest were drawn over entire left ventricular myocardium (LV), individual walls, and mediastinum (M). Coefficient of Variation (CV) was calculated from individual wall counts. RESULTS HF patients had significantly lower myocardial 18F-FDOPA uptake (P < .001, independent t test) than controls [32.4% ± 9.5% global reduction; highest in apex (39.9% ± 7.0%)]. A cut-off of LV/M ≤ 1.68 could differentiate patients from controls with sensitivity and specificity of 100% and 95.8%, respectively. LV/M correlated positively with EF (Pearson coefficient = 0.460, P .031). During follow-up, 3 patients were lost to follow-up, 4 died (survival-20.5 ± 4 months), 2 worsened, and 15 remained stable/showed mild improvement. Patients who worsened/died during follow-up had higher CV than those with stable/improving symptoms [0.16 ± 0.05 vs 0.11 ± 0.05, P value .069 (independent t test); Cox regression P = .084]. CONCLUSION Myocardial 18F-FDOPA uptake in patients with HF is significantly reduced. Higher reduction is seen in those with lower EF. CV, a maker of regional heterogeneity, is a potential prognostic marker.
Collapse
Affiliation(s)
| | - Anshul Sharma
- Department of Nuclear Medicine, HBCHRC (Tata Memorial Centre), Mullanpur, Punjab, India
| | - Chetan Patel
- Department of Nuclear Medicine, AIIMS, New Delhi, India.
| | - Sandeep Seth
- Department of Cardiology, AIIMS, New Delhi, India
| | - Ambuj Roy
- Department of Cardiology, AIIMS, New Delhi, India
| | | | | | - Rajeev Kumar
- Department of Nuclear Medicine, AIIMS, New Delhi, India
| | - Praveen Kumar
- Department of Nuclear Medicine, AIIMS, New Delhi, India
| | | |
Collapse
|
78
|
Cui H, Han S, Dai Y, Xie W, Zheng R, Sun Y, Xia X, Deng X, Cao Y, Zhang M, Shang H. Gut microbiota and integrative traditional Chinese and western medicine in prevention and treatment of heart failure. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 117:154885. [PMID: 37302262 DOI: 10.1016/j.phymed.2023.154885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/27/2023] [Accepted: 05/15/2023] [Indexed: 06/13/2023]
Abstract
BACKGROUND Heart failure (HF) is the terminal stage of multiple cardiovascular diseases, with high mortality and morbidity. More and more studies have proved that gut microbiota may play a role in the process of HF, which is expected to become a new therapeutic target. The combination of traditional Chinese and Western medicine has vast therapeutic potential of complementation against HF. PURPOSE This manuscript expounds on the research progress of mechanisms of gut microbiota participating in the occurrence and prognosis of HF and the role of integrative traditional Chinese and Western medicine from 1987 to 2022. The combination of traditional Chinese and Western medicine in the prevention and treatment of HF from the perspective of gut microbiota has been discussed. METHODS Studies focusing on the effects and their mechanisms of gut microbiota in HF and the role of integrative traditional Chinese and Western medicine were identified and summarized, including contributions from February 1987 until August 2022. The investigation was carried out in accordance with the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) guidelines. We searched PubMed, Embase, Cochrane Library, CNKI, Wanfang, and VIP databases up to April 2023 by using the relevant keywords and operators. RESULTS A total of 34 articles were finally included in this review.16 RCTs and 13 basic researches, and 3 clinical research studies involving 7 relevant outcome indicators(cardiac function evaluation index, changes in gut microbiota, inflammatory factors, metabolites of gut microbiota, serum nutritional index protein, quality of life score, intestinal permeability and all-cause mortality). Compared with healthy controls, serum TNF-α and TMAO levels were significantly higher in patients with heart failure [MD = 5.77, 95%CI(4.97, 6.56), p < 0.0001; SMD = 1.92, 95%CI(1.70, 2.14), p < 0.0001]. Escherichia coli and Thick-walled bacteria increased significantly [SMD = -0.99, 95%CI(-1.38, -0.61), p < 0.0001, SMD = 2.58, 95%CI(2.23, 2.93), p < 0.0001];The number of bacteroides and lactobacillus decreased [SMD = -2.29, 95%CI(-2.54, -2.04), p < 0.0001; SMD = -1.55, 95%CI(-1.8, -1.3), p < 0.0001]. There was no difference in bifidobacterium [SMD = 0.16, 95%CI(-0.22, 0.54), p = 0.42]. In the published literature, it is not difficult to see that most of the results are studied and proved based on animal experiments or clinical trials, involving the cellular level, while the mechanism and mode of action of the molecular biology of traditional Chinese medicine are less elaborated, which is related to the characteristics of multi-components and multi-targets of traditional Chinese medicine. The above are the shortcomings of published literature, which can also be the direction of future research. CONCLUSION Heart failure patients have decreased beneficial bacteria such as Bacillus mimics and Lactobacillus in the intestinal flora and increased harmful flora like thick-walled flora. And increase the inflammatory response of the body and the expression of trimethylamine oxide (TMAO) in the serum. And The prevention and treatment of integrative traditional Chinese and Western medicine against heart failure based on gut microbiota and its metabolites is a promising research direction.
Collapse
Affiliation(s)
- Herong Cui
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China; School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Songjie Han
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yanan Dai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Wei Xie
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Rui Zheng
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yang Sun
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Xiaofeng Xia
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiaopeng Deng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yaru Cao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Mei Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China.
| |
Collapse
|
79
|
Soltani D, Azizi B, Sima S, Tavakoli K, Hosseini Mohammadi NS, Vahabie AH, Akbarzadeh-Sherbaf K, Vasheghani-Farahani A. A systematic review of the effects of transcutaneous auricular vagus nerve stimulation on baroreflex sensitivity and heart rate variability in healthy subjects. Clin Auton Res 2023; 33:165-189. [PMID: 37119426 DOI: 10.1007/s10286-023-00938-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 03/27/2023] [Indexed: 05/01/2023]
Abstract
PURPOSE This systematic review aimed to evaluate the effect of transcutaneous auricular vagus nerve stimulation on heart rate variability and baroreflex sensitivity in healthy populations. METHOD PubMed, Scopus, the Cochrane Library, Embase, and Web of Science were systematically searched for controlled trials that examined the effects of transcutaneous auricular vagus nerve stimulation on heart rate variability parameters and baroreflex sensitivity in apparently healthy individuals. Two independent researchers screened the search results, extracted the data, and evaluated the quality of the included studies. RESULTS From 2458 screened studies, 21 were included. Compared with baseline measures or the comparison group, significant changes in the standard deviation of NN intervals, the root mean square of successive RR intervals, the proportion of consecutive RR intervals that differ by more than 50 ms, high-frequency power, low-frequency to high-frequency ratio, and low-frequency power were found in 86%, 75%, 69%, 47%, 36%, and 25% of the studies evaluating the effects of transcutaneous auricular vagus nerve stimulation on these indices, respectively. Baroreflex sensitivity was evaluated in six studies, of which a significant change was detected in only one. Some studies have shown that the worse the basic autonomic function, the better the response to transcutaneous auricular vagus nerve stimulation. CONCLUSION The results were mixed, which may be mainly attributable to the heterogeneity of the study designs and stimulation delivery dosages. Thus, future studies with comparable designs are required to determine the optimal stimulation parameters and clarify the significance of autonomic indices as a reliable marker of neuromodulation responsiveness.
Collapse
Affiliation(s)
- Danesh Soltani
- Cardiac Primary Prevention Research Center (CPPRC), Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bayan Azizi
- Cardiac Primary Prevention Research Center (CPPRC), Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepehr Sima
- Department of Psychology, University of Tehran, Tehran, Iran
| | - Kiarash Tavakoli
- Cardiac Primary Prevention Research Center (CPPRC), Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Negin Sadat Hosseini Mohammadi
- Cardiac Primary Prevention Research Center (CPPRC), Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Abdol-Hossein Vahabie
- Control and Intelligent Processing Center of Excellence (CIPCE), Cognitive Systems Laboratory, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, Tehran, Iran
- Department of Psychology, Faculty of Psychology and Education, University of Tehran, Tehran, Iran
| | - Kaveh Akbarzadeh-Sherbaf
- Department of Computer Engineering and Information Technology, Imam Reza International University, Mashhad, Iran
| | - Ali Vasheghani-Farahani
- Cardiac Primary Prevention Research Center (CPPRC), Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
80
|
Bereda G. Hypertensive emergency occurred due to forgetting to take antihypertensive medication: A case study. SAGE Open Med Case Rep 2023; 11:2050313X231168287. [PMID: 37113389 PMCID: PMC10126595 DOI: 10.1177/2050313x231168287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
A life-threatening condition known as a "hypertensive emergency" is marked by a severe increase in blood pressure together with acute or significant target-organ damage. On 1 June 2022, a 67-year-old black male farmer was admitted to the emergency department with a major chief complaint of breathing difficulty. The patient was traveling to the village for work and forgetting his medication at home, and he was losing consciousness and motor activity at his workplace. He presented with symptoms of shortness of breath, confusion, dizziness, nausea, vomiting, blurred vision, and faintness. An abnormal cardiac region was visible on chest X-rays, and there were no changes to the pulmonary parenchyma or fluid overload. Upon admission, hydralazine (5 mg) intravenously was administered immediately, and he was reassessed after 20 min and kept at the emergency department. The next day, sustained-release nifedipine (20 mg) was initiated orally twice a day for the patient, and he was transferred to the medical ward. In the medical ward, the patient was assessed for 4 days, and in those 4 days, he showed marked improvement. Hypertensive emergency treatment intends to reverse target-organ damage, readily lowering blood pressure, decreasing adverse clinical complications, and enhancing the quality of life.
Collapse
Affiliation(s)
- Gudisa Bereda
- Pharmacy Department, All Africa Leprosy,
Tuberculosis and Rehabilitation Training Centre, Addis Ababa, Ethiopia
| |
Collapse
|
81
|
Sigurdardottir FD, Bertisch SM, Reid ML, deFilippi CR, Lima JAC, Redline S, Omland T. Association between insomnia phenotypes and subclinical myocardial injury: the Multi-Ethnic Study of Atherosclerosis. Sleep 2023; 46:zsac318. [PMID: 36579654 PMCID: PMC10091090 DOI: 10.1093/sleep/zsac318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/04/2022] [Indexed: 12/30/2022] Open
Abstract
STUDY OBJECTIVES To assess whether the association between insomnia and subclinical myocardial injury, as measured by cardiac troponin T (cTnT), differs across insomnia phenotypes. METHODS We measured cTnT in 2188 participants in the Multi-Ethnic Study of Atherosclerosis study who had completed sleep questionnaires and undergone unattended polysomnography (PSG) and 7-day actigraphy. Insomnia symptoms were defined as reporting at least one of the following ≥5 nights/week over the past 4 weeks: trouble falling asleep, waking up several times a night, having trouble getting back to sleep after waking up too early, or taking sleeping pills to help falling asleep. Obstructive sleep apnea (OSA) was defined as an apnea-hypopnea index (AHI >15 events/h). Participants were classified into insomnia phenotypes, including comorbid insomnia and OSA (COMISA) and insomnia associated with actigraphy-estimated short sleep (<6 h) or sleep fragmentation. RESULTS The mean age was 68.8 (SD 9.2) years, 53.6% were male. In total, 47.8% met threshold levels for insomnia symptoms, and 43.1% had an AHI >15. In adjusted linear regression models COMISA (β 0.08 [standard error (SE) 0.03], p < .01) and insomnia with short sleep duration (β 0.07 [SE 0.03], p < .05) were each associated with higher cTnT compared to a reference group with no insomnia. Insomnia with fragmented sleep (β 0.03 [SE 0.02]) was not associated with higher cTnT (p > .05) in adjusted analyses. OSA was associated with higher cTnT (β 0.09 [SE 0.03], p < .01) in adjusted models. CONCLUSIONS COMISA and insomnia with short sleep duration, but not insomnia symptoms alone or fragmented sleep, were associated with increased circulating cTnT in older adults.
Collapse
Affiliation(s)
- Fjola D Sigurdardottir
- Department of Cardiology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Suzanne M Bertisch
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Michelle L Reid
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, USA
| | | | - Joao A C Lima
- Division of Cardiology, Department of Medicine, Johns Hopkins Hospital and School of Medicine, Baltimore, MD, USA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Torbjørn Omland
- Department of Cardiology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
82
|
Ma Z, Liu Z, Li X, Zhang H, Han D, Xiong W, Zhou H, Yang X, Zeng Q, Ren H, Xu D. Metformin Collaborates with PINK1/Mfn2 Overexpression to Prevent Cardiac Injury by Improving Mitochondrial Function. BIOLOGY 2023; 12:biology12040582. [PMID: 37106782 PMCID: PMC10135998 DOI: 10.3390/biology12040582] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/05/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023]
Abstract
Both mitochondrial quality control and energy metabolism are critical in maintaining the physiological function of cardiomyocytes. When damaged mitochondria fail to be repaired, cardiomyocytes initiate a process referred to as mitophagy to clear defective mitochondria, and studies have shown that PTEN-induced putative kinase 1 (PINK1) plays an important role in this process. In addition, previous studies indicated that peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) is a transcriptional coactivator that promotes mitochondrial energy metabolism, and mitofusin 2 (Mfn2) promotes mitochondrial fusion, which is beneficial for cardiomyocytes. Thus, an integration strategy involving mitochondrial biogenesis and mitophagy might contribute to improved cardiomyocyte function. We studied the function of PINK1 in mitophagy in isoproterenol (Iso)-induced cardiomyocyte injury and transverse aortic constriction (TAC)-induced myocardial hypertrophy. Adenovirus vectors were used to induce PINK1/Mfn2 protein overexpression. Cardiomyocytes treated with isoproterenol (Iso) expressed high levels of PINK1 and low levels of Mfn2, and the changes were time dependent. PINK1 overexpression promoted mitophagy, attenuated the Iso-induced reduction in MMP, and reduced ROS production and the apoptotic rate. Cardiac-specific overexpression of PINK1 improved cardiac function, attenuated pressure overload-induced cardiac hypertrophy and fibrosis, and facilitated myocardial mitophagy in TAC mice. Moreover, metformin treatment and PINK1/Mfn2 overexpression reduced mitochondrial dysfunction by inhibiting ROS generation leading to an increase in both ATP production and mitochondrial membrane potential in Iso-induced cardiomyocyte injury. Our findings indicate that a combination strategy may help ameliorate myocardial injury by improving mitochondrial quality.
Collapse
Affiliation(s)
- Zhuang Ma
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510080, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
| | - Zuheng Liu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510080, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
- Xiamen Key Laboratory of Cardiac Electrophysiology, Department of Cardiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361013, China
| | - Xudong Li
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510080, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
| | - Hao Zhang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510080, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
| | - Dunzheng Han
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510080, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Wenjun Xiong
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510080, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
| | - Haobin Zhou
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510080, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
| | - Xi Yang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510080, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
| | - Qingchun Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510080, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
| | - Hao Ren
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
- Department of Rheumatology, Nanfang Hospital, Southern Medical University, Guangzhou 516006, China
| | - Dingli Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510080, China
- Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou 510515, China
| |
Collapse
|
83
|
Regitz-Zagrosek V, Gebhard C. Gender medicine: effects of sex and gender on cardiovascular disease manifestation and outcomes. Nat Rev Cardiol 2023; 20:236-247. [PMID: 36316574 PMCID: PMC9628527 DOI: 10.1038/s41569-022-00797-4] [Citation(s) in RCA: 100] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/26/2022] [Indexed: 11/06/2022]
Abstract
Despite a growing body of evidence, the distinct contributions of biological sex and the sociocultural dimension of gender to the manifestations and outcomes of ischaemic heart disease and heart failure remain unknown. The intertwining of sex-based differences in genetic and hormonal mechanisms with the complex dimension of gender and its different components and determinants that result in different disease phenotypes in women and men needs to be elucidated. The relative contribution of purely biological factors, such as genes and hormones, to cardiovascular phenotypes and outcomes is not yet fully understood. Increasing awareness of the effects of gender has led to efforts to measure gender in retrospective and prospective clinical studies and the development of gender scores. However, the synergistic or opposing effects of sex and gender on cardiovascular traits and on ischaemic heart disease and heart failure mechanisms have not yet been systematically described. Furthermore, specific considerations of sex-related and gender-related factors in gender dysphoria or in heart-brain interactions and their association with cardiovascular disease are still lacking. In this Review, we summarize contemporary evidence on the distinct effects of sex and gender as well as of their interactions on cardiovascular disease and how they favourably or unfavourably influence the pathogenesis, clinical manifestations and treatment responses in patients with ischaemic heart disease or heart failure.
Collapse
Affiliation(s)
- Vera Regitz-Zagrosek
- Institute for Gender in Medicine, Charité University Medicine Berlin, Berlin, Germany.
- Faculty of Medicine, University of Zurich, Zurich, Switzerland.
| | - Catherine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
- Department of Cardiology, Inselspital Bern University Hospital, Bern, Switzerland
| |
Collapse
|
84
|
Schulz R, Schlüter KD. Importance of Mitochondria in Cardiac Pathologies: Focus on Uncoupling Proteins and Monoamine Oxidases. Int J Mol Sci 2023; 24:ijms24076459. [PMID: 37047436 PMCID: PMC10095304 DOI: 10.3390/ijms24076459] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
On the one hand, reactive oxygen species (ROS) are involved in the onset and progression of a wide array of diseases. On the other hand, these are a part of signaling pathways related to cell metabolism, growth and survival. While ROS are produced at various cellular sites, in cardiomyocytes the largest amount of ROS is generated by mitochondria. Apart from the electron transport chain and various other proteins, uncoupling protein (UCP) and monoamine oxidases (MAO) have been proposed to modify mitochondrial ROS formation. Here, we review the recent information on UCP and MAO in cardiac injuries induced by ischemia-reperfusion (I/R) as well as protection from I/R and heart failure secondary to I/R injury or pressure overload. The current data in the literature suggest that I/R will preferentially upregulate UCP2 in cardiac tissue but not UCP3. Studies addressing the consequences of such induction are currently inconclusive because the precise function of UCP2 in cardiac tissue is not well understood, and tissue- and species-specific aspects complicate the situation. In general, UCP2 may reduce oxidative stress by mild uncoupling and both UCP2 and UCP3 affect substrate utilization in cardiac tissue, thereby modifying post-ischemic remodeling. MAOs are important for the physiological regulation of substrate concentrations. Upon increased expression and or activity of MAOs, however, the increased production of ROS and reactive aldehydes contribute to cardiac alterations such as hypertrophy, inflammation, irreversible cardiomyocyte injury, and failure.
Collapse
|
85
|
Escobar Cervantes C, Esteban Fernández A, Recio Mayoral A, Mirabet S, González Costello J, Rubio Gracia J, Núñez Villota J, González Franco Á, Bonilla Palomas JL. Identifying the patient with heart failure to be treated with vericiguat. Curr Med Res Opin 2023; 39:661-669. [PMID: 36897009 DOI: 10.1080/03007995.2023.2189857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
The pathophysiology of heart failure with reduced ejection fraction (HFrEF) is a complex process in which a number of neurohormonal systems are involved. Targeting only some of these systems, but not all, translates into a partial benefit of HF treatment. The nitric oxide-soluble guanylate cyclase (sGC)-cGMP pathway is impaired in HF, leading to cardiac, vascular and renal disturbances. Vericiguat is a once-daily oral stimulator of sGC that restores this system. No other disease-modifying HF drugs act on this system. Despite guidelines recommendations, a substantial proportion of patients are not taking all recommended drugs or when taking them, they do so at low doses, limiting their potential benefits. In this context, treatment should be optimized considering different parameters, such as blood pressure, heart rate, renal function, or potassium, as they may interfere with their implementation at the recommended doses. The VICTORIA trial showed that adding vericiguat to standard therapy in patients with HFrEF significantly reduced the risk of cardiovascular death or HF hospitalization by 10% (NNT 24). Furthermore, vericiguat does not interfere with heart rate, renal function or potassium, making it particularly useful for improving the prognosis of patients with HFrEF in specific settings and clinical profiles.
Collapse
Affiliation(s)
| | | | | | - Sonia Mirabet
- Cardiology Department, Hospital de Sant Pau, Barcelona, Spain
| | - José González Costello
- Department of Cardiology, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
- BIOHEART-Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Department of Clinical Sciences, School of Medicine, Universitat de Barcelona, Spain
- Ciber Cardiovascular (CIBERCV), Instituto Salud Carlos III, Madrid, Spain
| | - Jorge Rubio Gracia
- Internal Medicine Department, Hospital Clínico Univeristario Lozano Blesa, University of Zaragoza, Spain
| | - Julio Núñez Villota
- Cardiology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
- Universidad de Valencia, INCLIVA, CIBER Cardiovascular, Valencia, Spain
| | | | | |
Collapse
|
86
|
de Almeida LB, Laterza MC, Rondon MUPB, Toschi-Dias E, de Matos LDNJ, Oliveira CC, Trevizan PF, Martinez DG. High-dose Chemotherapy Impairs Cardiac Autonomic Control of Hospitalized Cancer Patients Undergoing Autologous Hematopoietic Stem Cell Transplantation. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:e131-e138. [PMID: 36604245 DOI: 10.1016/j.clml.2022.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/10/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Autologous hematopoietic stem cell transplantation (HSCT) patients have intermediary and late cardiac autonomic dysfunction, which is an independent mortality predictor. However, it is unknown when this HSCT-related autonomic dysfunction begins during hospitalization for HSCT and whether cardiac autonomic control (CAC) is related to cardiotoxicity in these patients. PATIENTS AND METHODS CAC was assessed in 36 autologous-HSCT inpatients (HSCT group) and 23 cancer-free outpatients (CON group) using heart rate variability analysis. The HSCT group was assessed at five time-points from admission to hospital discharge during hospitalization period. The CON group was assessed once. The severity of cardiotoxicity (CTCAE 5.0) and cardiac troponin I were recorded. RESULTS The CAC was significantly reduced after high-dose chemotherapy (HDC) (reduction of MNN, SDNN, RMSSD, LFms2 and HFnu, and increase of LFnu and LF/HF; P<0.05). At the onset of neutropenia, pNN50 and HFms2 were also reduced (P<0.05) compared to the admission ones. Although both groups were similar regarding CAC at hospital admission, the HSCT patients showed impaired CAC at hospital discharge (P<0.05). The LF/HF was positively associated with cardiac troponin I and RMSSD was inversely associated with the severity of cardiotoxicity (P≤0.05). CONCLUSION CAC worsened during hospitalization for autologous-HSCT, mainly after HDC. In addition, it seems associated to early signs of cardiotoxicity in these patients.
Collapse
Affiliation(s)
- Leonardo Barbosa de Almeida
- Cardiovascular Research Unit and Exercise Physiology, Federal University of Juiz de Fora, Juiz de Fora, Brazil.
| | - Mateus Camaroti Laterza
- Cardiovascular Research Unit and Exercise Physiology, Federal University of Juiz de Fora, Juiz de Fora, Brazil; Master´s Program on Rehabilitation Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | | | | | | | | | | | - Daniel Godoy Martinez
- Cardiovascular Research Unit and Exercise Physiology, Federal University of Juiz de Fora, Juiz de Fora, Brazil; Master´s Program on Rehabilitation Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| |
Collapse
|
87
|
Lapointe MO, Caru M, Curnier D, Raboisson MJ, Andelfinger G, Krajinovic M, Laverdière C, Sinnett D, Périé D. Dexrazoxane Treatments Limits Subclinical Cardiac Dysfunction in Childhood Acute Lymphoblastic Leukemia Survivors Exposed to Doxorubicin Treatments. J Pediatr Hematol Oncol 2023; 45:70-77. [PMID: 36161876 DOI: 10.1097/mph.0000000000002538] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 07/11/2022] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Doxorubicin leads to dose dependent cardiotoxicity in childhood acute lymphoblastic leukemia (ALL) survivors. We investigated survivors' heart health using echocardiography and evaluated doxorubicin and dexrazoxane treatments on cardiac function. METHODS A total of 196 childhood ALL survivors were stratified (standard risk [SR], high risk with and without dexrazoxane (HR+DEX and HR). We performed a complete transthoracic echocardiographic assessment with M-mode echocardiography, Doppler, and Tissue Doppler. We used 2-dimensional and 3-dimensional echocardiography to measure the left ventricular ejection fraction, whereas myocardial strain imaging was used to obtain global strain indices. RESULTS Although most cardiac and arterial dimension parameters were not different between groups, a difference was observed in posterior intima of the right carotid ( P =0.017). Diastolic functions analyses reported that LV shortening fraction and left and right ventricular lateral S' wave amplitudes were lower in HR than in SR and HR+DEX groups ( P =0.028, P =0.048, and P =0.005, respectively). The LV lateral E' in diastolic function was lower in the HR than in SR and HR+DEX groups ( P =0.036). The LV end-systolic wall stress was higher in HR than in SR and HR+DEX groups ( P =0.009). A decrease contractility was observed, while the effect was not group specific. Strain rate was not different between groups, as opposed to tissue Doppler measurements. CONCLUSIONS This study showed that dexrazoxane treatments could limit subclinical cardiac dysfunction in childhood ALL survivors, whereas survivors in HR group who did not receive dexrazoxane had potential subclinical cardiac damage observable in heart failure patients. Echocardiographic screening for survivors must be part of the follow-up routine in cardio-oncology.
Collapse
Affiliation(s)
- Marc-Olivier Lapointe
- Department of Mechanical Engineering, Polytechnique
- Sainte-Justine University Health Center, Research Center
| | - Maxime Caru
- Department of Mechanical Engineering, Polytechnique
- Sainte-Justine University Health Center, Research Center
| | - Daniel Curnier
- Sainte-Justine University Health Center, Research Center
- School of Kinesiology and Physical Activity Sciences, Faculty of Medicine
| | | | - Gregor Andelfinger
- Sainte-Justine University Health Center, Research Center
- Department of Pediatrics, University of Montreal, Montreal, QC, Canada
| | - Maja Krajinovic
- Sainte-Justine University Health Center, Research Center
- Department of Pediatrics, University of Montreal, Montreal, QC, Canada
| | - Caroline Laverdière
- Sainte-Justine University Health Center, Research Center
- Department of Pediatrics, University of Montreal, Montreal, QC, Canada
| | - Daniel Sinnett
- Sainte-Justine University Health Center, Research Center
- Department of Pediatrics, University of Montreal, Montreal, QC, Canada
| | - Delphine Périé
- Department of Mechanical Engineering, Polytechnique
- Sainte-Justine University Health Center, Research Center
| |
Collapse
|
88
|
Bencivenga L, Komici K, Paolillo S, Nappi C, Gargiulo P, Assante R, Gambino G, Santillo F, Femminella GD, Corbi GM, Ferrara N, Cuocolo A, Perrone-Filardi P, Rengo G. Cardiac sympathetic innervation and mortality risk scores in patients with heart failure. Eur J Clin Invest 2023; 53:e13948. [PMID: 36576359 DOI: 10.1111/eci.13948] [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: 07/27/2022] [Revised: 11/15/2022] [Accepted: 11/19/2022] [Indexed: 12/29/2022]
Abstract
INTRODUCTION In the risk stratification and selection of patients with heart failure (HF) eligible for implantable cardioverter-defibrillator (ICD) therapy, 123 I-meta-IodineBenzylGuanidine (123 I-mIBG) scintigraphy has emerged as an effective non-invasive method to assess cardiac adrenergic innervation. Similarly, clinical risk scores have been proposed to identify patients with HF at risk of all-cause mortality, for whom the net clinical benefit of device implantation would presumably be lower. Nevertheless, the association between the two classes of tools, one suggestive of arrhythmic risk, the other of all-cause mortality, needs further investigation. OBJECTIVE To test the relationship between the risk scores for predicting mortality and cardiac sympathetic innervation, assessed through myocardial 123 I-mIBG imaging, in a population of patients with HF. METHODS In HF patients undergoing 123 I-mIBG scintigraphy, eight risk stratification models were assessed: AAACC, FADES, MADIT, MADIT-ICD non-arrhythmic mortality score, PACE, Parkash, SHOCKED and Sjoblom. Cardiac adrenergic impairment was assessed by late heart-to-mediastinum ratio (H/M) <1.6. RESULTS Among 269 patients suffering from HF, late H/M showed significant negative correlation with all the predicting models, although generally weak, ranging from -0.15 (p = .013) for PACE to -0.32 (p < .001) for FADES. The scores showed poor discrimination for cardiac innervation, with areas under the curve (AUC) ranging from 0.546 for Parkash to 0.621 for FADES. CONCLUSION A weak association emerged among mortality risk scores and cardiac innervation, suggesting to integrate in clinical practice tools indicative of both arrhythmic and general mortality risks, when evaluating patients affected by HF eligible for device implantation.
Collapse
Affiliation(s)
- Leonardo Bencivenga
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy.,Gérontopôle de Toulouse, Institut du Vieillissement, CHU de Toulouse, Toulouse, France
| | - Klara Komici
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | - Stefania Paolillo
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Carmela Nappi
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Paola Gargiulo
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Roberta Assante
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Giuseppina Gambino
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Fabio Santillo
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | | | - Grazia Maria Corbi
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Nicola Ferrara
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Alberto Cuocolo
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Pasquale Perrone-Filardi
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy.,Mediterranea Cardiocentro, Naples, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy.,Istituti Clinici Scientifici Maugeri SpA Società Benefit, Telese Terme, Italy
| |
Collapse
|
89
|
Wernhart S, Papathanasiou M, Rassaf T, Luedike P. The controversial role of beta-blockers in heart failure with preserved ejection fraction. Pharmacol Ther 2023; 243:108356. [PMID: 36750166 DOI: 10.1016/j.pharmthera.2023.108356] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/22/2023] [Accepted: 02/01/2023] [Indexed: 02/07/2023]
Abstract
Beta-blocker (BB) therapy is a main pillar in treating patients with heart failure and reduced ejection fraction and has shown a prognostic benefit. However, evidence for application of BB in heart failure with preserved ejection fraction (HFpEF), especially in the absence of coronary artery disease, atrial fibrillation or arterial hypertension, is scarce. HFpEF is characterized by elevations in left atrial pressure and reduced compliance of the left ventricle leading to a hampered increase of cardiac output (CO) during exercise, which results in exertional dyspnea. This may be due to either a limited increase in stroke volume or reduced chronotropy during physical activity. We critically discuss the pathophysiological background of HFpEF, current data on BB in heart failure therapy, as well as the potential benefits and harms of BB therapy in HFpEF. Furthermore, we argue that non-cardio selective BB with peripheral activity to reduce afterload may be more suitable in this population than cardio-selective BB. Although preliminary data on BB in HFpEF are available, multicenter prospective trials to assess a reduction of cardiovascular morbidity are warranted. Future trials need to focus on phenotyping HFpEF patients and assess who may benefit most from tailored BB therapy.
Collapse
Affiliation(s)
- Simon Wernhart
- University Hospital Essen, University Duisburg-Essen, West German Heart- and Vascular Center, Department of Cardiology and Vascular Medicine, Hufelandstrasse 55, 45147 Essen, Germany
| | - Maria Papathanasiou
- University Hospital Essen, University Duisburg-Essen, West German Heart- and Vascular Center, Department of Cardiology and Vascular Medicine, Hufelandstrasse 55, 45147 Essen, Germany
| | - Tienush Rassaf
- University Hospital Essen, University Duisburg-Essen, West German Heart- and Vascular Center, Department of Cardiology and Vascular Medicine, Hufelandstrasse 55, 45147 Essen, Germany
| | - Peter Luedike
- University Hospital Essen, University Duisburg-Essen, West German Heart- and Vascular Center, Department of Cardiology and Vascular Medicine, Hufelandstrasse 55, 45147 Essen, Germany.
| |
Collapse
|
90
|
Eslami A, Nassif NT, Lal S. Neuropsychological Performance and Cardiac Autonomic Function in Blue- and White-Collar Workers: A Psychometric and Heart Rate Variability Evaluation. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:4203. [PMID: 36901213 PMCID: PMC10002343 DOI: 10.3390/ijerph20054203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/20/2023] [Accepted: 02/20/2023] [Indexed: 06/18/2023]
Abstract
The 21st century has brought a growing and significant focus on performance and health within the workforce, with the aim of improving the health and performance of the blue- and white-collar workforce. The present research investigated heart rate variability (HRV) and psychological performance between blue and white-collar workers to determine if differences were evident. A total of 101 workers (n = 48 white-collar, n = 53 blue-collar, aged 19-61 years) underwent a three lead electrocardiogram to obtain HRV data during baseline (10 min) and active (working memory and attention) phases. The Cambridge Neuropsychological Test Automated Battery, specifically the spatial working memory, attention switching task, rapid visual processing and the spatial span, were used. Differences in neurocognitive performance measures indicated that white-collar workers were better able to detect sequences and make less errors than blue-collar workers. The heart rate variability differences showed that white-collar workers exhibit lower levels of cardiac vagal control during these neuropsychological tasks. These initial findings provide some novel insights into the relationship between occupation and psychophysiological processes and further highlight the interactions between cardiac autonomic variables and neurocognitive performance in blue and white-collar workers.
Collapse
Affiliation(s)
- Ardalan Eslami
- Neuroscience Research Unit, School of Life Sciences, University of Technology Sydney, Sydney 2007, Australia
- Neuroscience Research Unit, Transdisciplinary School, School of Public Health, University of Technology Sydney, Sydney 2007, Australia
| | - Najah T. Nassif
- School of Life Sciences, University of Technology Sydney, Sydney 2007, Australia
| | - Sara Lal
- Neuroscience Research Unit, Transdisciplinary School, School of Public Health, University of Technology Sydney, Sydney 2007, Australia
- Honorary, School of Psychology, Faculty of Science, University of New South Wales, Sydney 2052, Australia
| |
Collapse
|
91
|
Guckel D, Eitz T, El Hamriti M, Braun M, Khalaph M, Imnadze G, Fink T, Sciacca V, Sohns C, Sommer P, Nölker G. Baroreflex activation therapy in advanced heart failure therapy: insights from a real-world scenario. ESC Heart Fail 2023; 10:284-294. [PMID: 36208130 PMCID: PMC9871720 DOI: 10.1002/ehf2.14190] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 09/01/2022] [Accepted: 09/21/2022] [Indexed: 01/29/2023] Open
Abstract
AIMS Baroreflex activation therapy (BAT) is an innovative treatment option for advanced heart failure (HFrEF). We analysed patients' BAT acceptance and the outcome of BAT patients compared with HFrEF patients solely treated with a guideline-directed medical therapy (GDMT) and studied effects of sacubitril/valsartan (ARNI). METHODS In this prospective study, 40 HFrEF patients (71 ± 3 years, 20% female) answered a questionnaire on the acceptance of BAT. Follow-up visits were performed after 3, 6, and 12 months. Primary efficacy endpoints included an improvement in QoL, NYHA class, LVEF, HF hospitalization, NT-proBNP levels, and 6MHWD. RESULTS Twenty-nine patients (73%) showed interest in BAT. Ten patients (25%) opted for implantation. BAT and BAT + ARNI patients developed an increase in LVEF (BAT +10%, P-value (P) = 0.005*; BAT + ARNI +9%, P = 0.049*), an improved NYHA class (BAT -88%, P = 0.014*, BAT + ARNI -90%, P = 0.037*), QoL (BAT +21%, P = 0.020*, BAT + ARNI +22%, P = 0.012*), and reduced NT-proBNP levels (BAT -24%, P = 0.297, BAT + ARNI -37%, P = 0.297). BAT HF hospitalization rates were lower (50%) compared with control group patients (83%) (P = 0.020*). CONCLUSIONS Although BAT has generated considerable interest, acceptance appears to be ambivalent. BAT improves outcome with regard to LVEF, NYHA class, QoL, NT-proBNP levels, and HF hospitalization rates. BAT + ARNI resulted in more pronounced effects than ARNI alone.
Collapse
Affiliation(s)
- Denise Guckel
- Clinic for Electrophysiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Thomas Eitz
- Clinic for Thoracic and Cardiovascular Surgery, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Mustapha El Hamriti
- Clinic for Electrophysiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Martin Braun
- Clinic for Electrophysiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Moneeb Khalaph
- Clinic for Electrophysiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Guram Imnadze
- Clinic for Electrophysiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Thomas Fink
- Clinic for Electrophysiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Vanessa Sciacca
- Clinic for Electrophysiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Christian Sohns
- Clinic for Electrophysiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Philipp Sommer
- Clinic for Electrophysiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Georg Nölker
- Clinic for Electrophysiology, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
- Clinic for Internal Medicine II/Cardiology, Christliches Klinikum Unna Mitte, Unna, Germany
| |
Collapse
|
92
|
Tamisier R, Pépin JL, Woehrle H, Salvat M, Barone-Rochette G, Rocca C, Vettorazzi E, Teschler H, Cowie M, Lévy P. Effect of adaptive servo-ventilation for central sleep apnoea in systolic heart failure on muscle sympathetic nerve activity: a SERVE-HF randomised ancillary study. Eur Respir J 2023; 61:13993003.00384-2022. [PMID: 36229051 PMCID: PMC9909211 DOI: 10.1183/13993003.00384-2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 09/18/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND Adaptive servo-ventilation (ASV) effectively suppresses central sleep apnoea (CSA) but has been associated with increased all-cause and cardiovascular mortality in chronic heart failure patients with reduced ventricular ejection fraction (HFrEF). All-cause and, especially, cardiovascular mortality in chronic heart failure is highly correlated with sympathetic tone. This analysis of SERVE-HF data investigated the effect of ASV on sympathetic tone in patients with HFrEF and CSA. METHODS HFrEF patients in the SERVE-HF trial (left ventricular ejection fraction (LVEF) ≤45%, apnoea-hypopnoea index (AHI) ≥15 events·h-1 with predominant CSA) were randomly assigned to receive guideline-based heart failure treatment alone (controls) or plus ASV. For this analysis, the primary outcome was change in muscle sympathetic nerve activity (MSNA) at 3-month follow-up. The effects of baseline MSNA and change in MSNA over time on mortality in the main study were also assessed. RESULTS 40 patients with HFrEF were included in this analysis (age 71.3±11.7 years, LVEF 34.2±7.7%, 57.5% in New York Heart Association (NYHA) Functional Class II, 42.5% in NYHA Functional Class III, AHI 35.2±11 events·h-1). Sympathetic tone evolution during follow-up did not differ between groups (controls: 47.6±8.3 bursts·min-1 at baseline to 44.6±11.2 bursts·min-1; ASV group: 43.0±9.0 bursts·min-1 at baseline to 42.74±9.45 bursts·min-1). The reduction in sympathetic tone was associated with significantly increased cardiovascular mortality in the ASV group, whereas in the control group reduced sympathetic tone appeared to be protective. CONCLUSIONS Suppression of CSA with ASV did not seem to have a significant effect on chronic heart failure-related sympathetic activation. Simultaneous suppression of CSA and reduction in MSNA was associated with increased cardiovascular mortality.
Collapse
Affiliation(s)
- Renaud Tamisier
- University Grenoble Alpes, HP2, Inserm 1300, Grenoble Alpes University Hospital, Grenoble, France
- Co-first authors
| | - Jean-Louis Pépin
- University Grenoble Alpes, HP2, Inserm 1300, Grenoble Alpes University Hospital, Grenoble, France
- Co-first authors
| | - Holger Woehrle
- ResMed Science Center, ResMed Germany, Martinsried, Germany
- Sleep and Ventilation Center Blaubeuren, Respiratory Center Ulm, Ulm, Germany
| | - Muriel Salvat
- Cardiology Clinic, Pole Thorax et Vaisseaux, CHU Grenoble Alpes University Hospital, Grenoble, France
| | - Gilles Barone-Rochette
- INSERM, U1039, Radiopharmaceutiques Biocliniques, University Grenoble Alpes, Department of Cardiology, Grenoble Alpes University Hospital and French Alliance Clinical Trial, French Clinical Research Infrastructure Network, Grenoble, France
| | - Cécile Rocca
- Cardiac Re-education Unit, Pole Thorax et Vaisseaux, Grenoble Alpes University Hospital, Grenoble, France
| | - Eik Vettorazzi
- Department of Medical Biometry and Epidemiology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Helmut Teschler
- Department of Pneumology, Ruhrlandklinik, West German Lung Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Martin Cowie
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, UK
- Co-senior authors
| | - Patrick Lévy
- University Grenoble Alpes, HP2, Inserm 1300, Grenoble Alpes University Hospital, Grenoble, France
- Co-senior authors
| |
Collapse
|
93
|
Mishkina AI, Saushkin VV, Atabekov TA, Sazonova SI, Shipulin VV, Massalha S, Batalov RE, Popov SV, Zavadovsky KV. The value of cardiac sympathetic activity and mechanical dyssynchrony as cardiac resynchronization therapy response predictors: comparison between patients with ischemic and non-ischemic heart failure. J Nucl Cardiol 2023; 30:371-382. [PMID: 35834158 DOI: 10.1007/s12350-022-03046-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 06/12/2022] [Indexed: 10/17/2022]
Abstract
BACKGROUND Impaired cardiac sympathetic activity and mechanical dyssynchrony (MD) are associated with poor prognosis in patients with heart failure (HF) after cardiac resynchronization therapy (CRT). The study aims to assess the significance of scintigraphic evaluation of cardiac sympathetic innervation and contractility in predicting response to CRT in patients with ischemic and non-ischemic chronic HF. METHODS AND RESULTS The study includes 58 HF patients, who were referred for CRT. Prior to CRT all patients underwent 123I-metaiodobenzylguanidine (123I-MIBG) imaging and gated myocardial perfusion imaging (MPI) using a cadmium-zinc-telluride (CZT) SPECT/CT device. At a one-year follow-up post-CRT, the delayed heart-to-mediastinum 123I-MIBG uptake ratio was an independent predictor of CRT response in non-ischemic HF patients (OR 1.469; 95% CI 1.076-2.007, p = .003). In ischemic HF patients the MD index histogram bandwidth (HBW) obtained by CZT-gated MPI had a predictive value (OR 1.06, 95% CI 1.001-1.112, p = .005) to CRT response. CONCLUSION CRT response can be predicted by cardiac 123I-MIBG scintigraphy, specifically by the heart-to-mediastinum ratio in non-ischemic HF and by the MD index HBW in ischemic HF. These results suggest the value of a potentially useful algorithm to improve outcomes in HF patients who are candidates for CRT.
Collapse
Affiliation(s)
- Anna I Mishkina
- Department of Nuclear Medicine, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Kievskaya Str 111A, Tomsk, Russia, 634012
| | - Victor V Saushkin
- Department of Nuclear Medicine, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Kievskaya Str 111A, Tomsk, Russia, 634012
| | - Tariel A Atabekov
- Department of Interventional Arrhythmology, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia
| | - Svetlana I Sazonova
- Department of Nuclear Medicine, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Kievskaya Str 111A, Tomsk, Russia, 634012
| | - Vladimir V Shipulin
- Department of Nuclear Medicine, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Kievskaya Str 111A, Tomsk, Russia, 634012
| | | | - Roman E Batalov
- Department of Interventional Arrhythmology, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia
| | - Sergey V Popov
- Department of Interventional Arrhythmology, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia
| | - Konstantin V Zavadovsky
- Department of Nuclear Medicine, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Kievskaya Str 111A, Tomsk, Russia, 634012.
| |
Collapse
|
94
|
Elia A, Fossati S. Autonomic nervous system and cardiac neuro-signaling pathway modulation in cardiovascular disorders and Alzheimer's disease. Front Physiol 2023; 14:1060666. [PMID: 36798942 PMCID: PMC9926972 DOI: 10.3389/fphys.2023.1060666] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 01/19/2023] [Indexed: 01/31/2023] Open
Abstract
The heart is a functional syncytium controlled by a delicate and sophisticated balance ensured by the tight coordination of its several cell subpopulations. Accordingly, cardiomyocytes together with the surrounding microenvironment participate in the heart tissue homeostasis. In the right atrium, the sinoatrial nodal cells regulate the cardiac impulse propagation through cardiomyocytes, thus ensuring the maintenance of the electric network in the heart tissue. Notably, the central nervous system (CNS) modulates the cardiac rhythm through the two limbs of the autonomic nervous system (ANS): the parasympathetic and sympathetic compartments. The autonomic nervous system exerts non-voluntary effects on different peripheral organs. The main neuromodulator of the Sympathetic Nervous System (SNS) is norepinephrine, while the principal neurotransmitter of the Parasympathetic Nervous System (PNS) is acetylcholine. Through these two main neurohormones, the ANS can gradually regulate cardiac, vascular, visceral, and glandular functions by turning on one of its two branches (adrenergic and/or cholinergic), which exert opposite effects on targeted organs. Besides these neuromodulators, the cardiac nervous system is ruled by specific neuropeptides (neurotrophic factors) that help to preserve innervation homeostasis through the myocardial layers (from epicardium to endocardium). Interestingly, the dysregulation of this neuro-signaling pathway may expose the cardiac tissue to severe disorders of different etiology and nature. Specifically, a maladaptive remodeling of the cardiac nervous system may culminate in a progressive loss of neurotrophins, thus leading to severe myocardial denervation, as observed in different cardiometabolic and neurodegenerative diseases (myocardial infarction, heart failure, Alzheimer's disease). This review analyzes the current knowledge on the pathophysiological processes involved in cardiac nervous system impairment from the perspectives of both cardiac disorders and a widely diffused and devastating neurodegenerative disorder, Alzheimer's disease, proposing a relationship between neurodegeneration, loss of neurotrophic factors, and cardiac nervous system impairment. This overview is conducive to a more comprehensive understanding of the process of cardiac neuro-signaling dysfunction, while bringing to light potential therapeutic scenarios to correct or delay the adverse cardiovascular remodeling, thus improving the cardiac prognosis and quality of life in patients with heart or neurodegenerative disorders.
Collapse
|
95
|
Comparison of Hemodynamic Response between Patients with Systolic Heart Failure Differing in Serum Aldosterone Concentrations during and after a 6-Minute Walk Test. J Clin Med 2023; 12:jcm12031007. [PMID: 36769655 PMCID: PMC9917580 DOI: 10.3390/jcm12031007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/16/2023] [Accepted: 01/24/2023] [Indexed: 02/03/2023] Open
Abstract
Aldosterone regulates hemodynamics, including blood pressure (BP), and is involved in the development and progression of cardiovascular diseases, including systolic heart failure (HF). While exercise intolerance is typical for HF, neither BP nor heart rate (HR) have specific characteristics in HF patients. This study compares BP and HR profiles during and after standardized exercise between patients with systolic HF with either lower or higher aldosterone concentrations. We measured BP and HR in 306 ambulatory adults with systolic HF (left ventricular ejection fraction (LVEF) <50%) during and after a 6 min walk test (6MWT). All patients underwent a resting transthoracic echocardiography, and venous blood samples were collected for biochemical analyses. The patients were also divided into tertiles of serum aldosterone concentration: T1 (<106 pg/mL), T2 (106 and 263 pg/mL) and T3 (>263 pg/mL), respectively. Individuals from T1 and T2 were combined into T1-T2 as the reference group for comparisons with patients from T3. The individuals from T3 had significantly lower systolic, mean and diastolic BPs at rest, at the end and at 1 and 3 min post-6MWT recovery, as well as a more dilated left atrium and right ventricle alongside a higher concentration of N-terminal pro-B-type natriuretic peptide (NT-proBNP). Higher serum aldosterone concentration in HF patients with an LVEF < 50% is associated with a lower 6MWT BP but not an HR profile.
Collapse
|
96
|
Higashikuni Y, Liu W, Numata G, Tanaka K, Fukuda D, Tanaka Y, Hirata Y, Imamura T, Takimoto E, Komuro I, Sata M. NLRP3 Inflammasome Activation Through Heart-Brain Interaction Initiates Cardiac Inflammation and Hypertrophy During Pressure Overload. Circulation 2023; 147:338-355. [PMID: 36440584 DOI: 10.1161/circulationaha.122.060860] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Mechanical stress on the heart, such as high blood pressure, initiates inflammation and causes hypertrophic heart disease. However, the regulatory mechanism of inflammation and its role in the stressed heart remain unclear. IL-1β (interleukin-1β) is a proinflammatory cytokine that causes cardiac hypertrophy and heart failure. Here, we show that neural signals activate the NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing 3) inflammasome for IL-1β production to induce adaptive hypertrophy in the stressed heart. METHODS C57BL/6 mice, knockout mouse strains for NLRP3 and P2RX7 (P2X purinoceptor 7), and adrenergic neuron-specific knockout mice for SLC17A9, a secretory vesicle protein responsible for the storage and release of ATP, were used for analysis. Pressure overload was induced by transverse aortic constriction. Various animal models were used, including pharmacological treatment with apyrase, lipopolysaccharide, 2'(3')-O-(4-benzoylbenzoyl)-ATP, MCC950, anti-IL-1β antibodies, clonidine, pseudoephedrine, isoproterenol, and bisoprolol, left stellate ganglionectomy, and ablation of cardiac afferent nerves with capsaicin. Cardiac function and morphology, gene expression, myocardial IL-1β and caspase-1 activity, and extracellular ATP level were assessed. In vitro experiments were performed using primary cardiomyocytes and fibroblasts from rat neonates and human microvascular endothelial cell line. Cell surface area and proliferation were assessed. RESULTS Genetic disruption of NLRP3 resulted in significant loss of IL-1β production, cardiac hypertrophy, and contractile function during pressure overload. A bone marrow transplantation experiment revealed an essential role of NLRP3 in cardiac nonimmune cells in myocardial IL-1β production and cardiac phenotype. Pharmacological depletion of extracellular ATP or genetic disruption of the P2X7 receptor suppressed myocardial NLRP3 inflammasome activity during pressure overload, indicating an important role of ATP/P2X7 axis in cardiac inflammation and hypertrophy. Extracellular ATP induced hypertrophic changes of cardiac cells in an NLRP3- and IL-1β-dependent manner in vitro. Manipulation of the sympathetic nervous system suggested sympathetic efferent nerves as the main source of extracellular ATP. Depletion of ATP release from sympathetic efferent nerves, ablation of cardiac afferent nerves, or a lipophilic β-blocker reduced cardiac extracellular ATP level, and inhibited NLRP3 inflammasome activation, IL-1β production, and adaptive cardiac hypertrophy during pressure overload. CONCLUSIONS Cardiac inflammation and hypertrophy are regulated by heart-brain interaction. Controlling neural signals might be important for the treatment of hypertensive heart disease.
Collapse
Affiliation(s)
- Yasutomi Higashikuni
- Department of Cardiovascular Medicine (Y.H., W.L., G.N., K. Tanaka, T.I., E.T., I.K.), The University of Tokyo, Japan
| | - Wenhao Liu
- Department of Cardiovascular Medicine (Y.H., W.L., G.N., K. Tanaka, T.I., E.T., I.K.), The University of Tokyo, Japan
| | - Genri Numata
- Department of Cardiovascular Medicine (Y.H., W.L., G.N., K. Tanaka, T.I., E.T., I.K.), The University of Tokyo, Japan
| | - Kimie Tanaka
- Department of Cardiovascular Medicine (Y.H., W.L., G.N., K. Tanaka, T.I., E.T., I.K.), The University of Tokyo, Japan.,Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan (K. Tanaka)
| | - Daiju Fukuda
- Department of Cardiovascular Medicine, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan (D.F.)
| | - Yu Tanaka
- Department of Pediatrics (Y. Tanaka, Y.H.), The University of Tokyo, Japan
| | - Yoichiro Hirata
- Department of Pediatrics (Y. Tanaka, Y.H.), The University of Tokyo, Japan
| | - Teruhiko Imamura
- Department of Cardiovascular Medicine (Y.H., W.L., G.N., K. Tanaka, T.I., E.T., I.K.), The University of Tokyo, Japan.,Second Department of Medicine, University of Toyama, Japan (T.I.)
| | - Eiki Takimoto
- Department of Cardiovascular Medicine (Y.H., W.L., G.N., K. Tanaka, T.I., E.T., I.K.), The University of Tokyo, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine (Y.H., W.L., G.N., K. Tanaka, T.I., E.T., I.K.), The University of Tokyo, Japan
| | - Masataka Sata
- Department of Cardiovascular Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Japan (M.S.)
| |
Collapse
|
97
|
Continuous exposure to isoprenaline reduced myotube size by delaying myoblast differentiation and fusion through the NFAT-MEF2C signaling pathway. Sci Rep 2023; 13:436. [PMID: 36624121 PMCID: PMC9829891 DOI: 10.1038/s41598-022-22330-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 10/13/2022] [Indexed: 01/11/2023] Open
Abstract
We aimed to explore whether superfluous sympathetic activity affects myoblast differentiation, fusion, and myofiber types using a continuous single-dose isoprenaline exposure model in vitro and to further confirm the role of distinct NFATs in ISO-mediated effects. Compared with delivery of single and interval single, continuous single-dose ISO most obviously diminished myotube size while postponing myoblast differentiation/fusion in a time- and dose-dependent pattern, accompanied by an apparent decrease in nuclear NFATc1/c2 levels and a slight increase in nuclear NFATc3/c4 levels. Overexpression of NFATc1 or NFATc2, particularly NFATc1, markedly abolished the inhibitory effects of ISO on myoblast differentiation/fusion, myotube size and Myh7 expression, which was attributed to a remarkable increase in the nuclear NFATc1/c2 levels and a reduction in the nuclear NFATc4 levels and the associated increase in the numbers of MyoG and MEF2C positive nuclei within more than 3 nuclei myotubes, especially in MEF2C. Moreover, knockdown of NFATc3 by shRNA did not alter the inhibitory effect of ISO on myoblast differentiation/fusion or myotube size but partially recovered the expression of Myh7, which was related to the slightly increased nuclear levels of NFATc1/c2, MyoG and MEF2C. Knockdown of NFATc4 by shRNA prominently increased the number of MyHC +, MyoG or MEF2C + myoblast cells with 1 ~ 2 nuclei, causing fewer numbers and smaller myotube sizes. However, NFATc4 knockdown further deteriorated the effects of ISO on myoblast fusion and myotube size, with more than 5 nuclei and Myh1/2/4 expression, which was associated with a decrease in nuclear NFATc2/c3 levels. Therefore, ISO inhibited myoblast differentiation/fusion and myotube size through the NFAT-MyoG-MEF2C signaling pathway.
Collapse
|
98
|
Relationships among norepinephrine levels, exercise capacity, and chronotropic responses in heart failure patients. Heart Fail Rev 2023; 28:35-45. [PMID: 35325323 DOI: 10.1007/s10741-022-10232-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/15/2022] [Indexed: 02/07/2023]
Abstract
In heart failure (HF) patients, the pathophysiological mechanisms of severe exercise intolerance and impaired exercise capacity are related to both central and peripheral abnormalities. The central abnormalities in HF patients include impaired cardiac function and chronotropic incompetence (CI). Indeed, CI, the inability to adequately increase heart rate (HR) from rest to exercise often exhibited by HF patients, is related to activation of the sympathetic nervous system (SNS) yielding a rise in circulating norepinephrine (NE). CI may result from downregulation of β-adrenergic receptors, β-blocker usage, high baseline HR, or due to a combination of factors. This paper discusses the role of elevated NE in altering chronotropic responses in HF patients and consequently resulting in impaired exercise capacity. We suggest that future research should focus on the potential treatment of CI with rate-adaptive pacing, using a sensor to measure physical activity, without inducing deleterious hormonal activation of the sympathetic system.
Collapse
|
99
|
Saavedra-Alvarez A, Pereyra KV, Toledo C, Iturriaga R, Del Rio R. Vascular dysfunction in HFpEF: Potential role in the development, maintenance, and progression of the disease. Front Cardiovasc Med 2022; 9:1070935. [PMID: 36620616 PMCID: PMC9810809 DOI: 10.3389/fcvm.2022.1070935] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 11/17/2022] [Indexed: 12/24/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a complex, heterogeneous disease characterized by autonomic imbalance, cardiac remodeling, and diastolic dysfunction. One feature that has recently been linked to the pathology is the presence of macrovascular and microvascular dysfunction. Indeed, vascular dysfunction directly affects the functionality of cardiomyocytes, leading to decreased dilatation capacity and increased cell rigidity, which are the outcomes of the progressive decline in myocardial function. The presence of an inflammatory condition in HFpEF produced by an increase in proinflammatory molecules and activation of immune cells (i.e., chronic low-grade inflammation) has been proposed to play a pivotal role in vascular remodeling and endothelial cell death, which may ultimately lead to increased arterial elastance, decreased myocardium perfusion, and decreased oxygen supply to the tissue. Despite this, the precise mechanism linking low-grade inflammation to vascular alterations in the setting of HFpEF is not completely known. However, the enhanced sympathetic vasomotor tone in HFpEF, which may result from inflammatory activation of the sympathetic nervous system, could contribute to orchestrate vascular dysfunction in the setting of HFpEF due to the exquisite sympathetic innervation of both the macro and microvasculature. Accordingly, the present brief review aims to discuss the main mechanisms that may be involved in the macro- and microvascular function impairment in HFpEF and the potential role of the sympathetic nervous system in vascular dysfunction.
Collapse
Affiliation(s)
- Andrea Saavedra-Alvarez
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Katherine V. Pereyra
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Camilo Toledo
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo Iturriaga
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile,Facultad de la Salud, Centro de Investigación en Fisiología y Medicina de Altura (MedAlt), Universidad de Antofagasta, Antofagasta, Chile
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile,Facultad de la Salud, Centro de Investigación en Fisiología y Medicina de Altura (MedAlt), Universidad de Antofagasta, Antofagasta, Chile,*Correspondence: Rodrigo Del Rio
| |
Collapse
|
100
|
Oh T, Ogawa K, Nagoshi T, Minai K, Ogawa T, Kawai M, Yoshimura M. Relationship between haemodynamic indicators and haemogram in patients with heart failure. ESC Heart Fail 2022; 10:955-964. [PMID: 36478404 PMCID: PMC10053360 DOI: 10.1002/ehf2.14258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 11/08/2022] [Accepted: 11/27/2022] [Indexed: 12/12/2022] Open
Abstract
AIMS Pulmonary congestion, reduced cardiac output, neurohumoral factor activation, and decreased renal function associated with decreased cardiac function may have various effects on haemograms. The relationship between these factors and haemograms in patients with heart failure has not been sufficiently investigated. Recently, it was suggested that the lungs are an important site for platelet (Plt) biosynthesis and that it is necessary to study the relationship between pulmonary congestion and Plt count in heart failure in detail. In this study, we examined the relationship between various haemodynamic indicators and haemograms in detail using statistical analyses. METHODS AND RESULTS A total of 345 patients who underwent cardiac catheterization for the evaluation of cardiac function between 1 January 2015 and 31 December 2020 were included in the study. Haemodynamic indices, including left ventricular end-diastolic pressure (LVEDP) and cardiac index (CI), were measured. Plasma noradrenaline (Nor) concentration, estimated glomerular filtration rate (eGFR), white blood cell (WBC) count, haemoglobin (Hb) level, and Plt count were measured using blood samples collected at the same time. Structural equation modelling (SEM) was used to examine the relationship between LVEDP, CI, plasma Nor concentration, eGFR, WBC count, Hb level, and Plt count. Bayesian inference using SEM was performed for Plt count. A total of 345 patients (mean age: 66.0 ± 13.2 years) were included in this study, and 251 (73%) patients were men. After simple and multiple regression analyses, path diagrams were drawn and analysed using SEM. LVEDP showed a significant negative relationship with Plt count (standardized estimate: -0.129, P = 0.015), and CI showed a significant negative relationship with Hb level (standardized estimate: -0.263, P < 0.001). Plasma Nor concentration showed a significant positive relationship with WBC count (standardized estimate: 0.165, P = 0.003) and Plt count (standardized estimate: 0.198, P < 0.001). The eGFR had a significant positive relationship with Hb level (standardized estimate: 0.274, P < 0.001). Bayesian inference using SEM revealed no relationship between LVEDP and Hb level or WBC count but a significant negative relationship between LVEDP and Plt count. CONCLUSIONS LVEDP, CI, plasma Nor concentration, and eGFR were related to WBC count, Hb level, and Plt count in patients with heart failure. There was a strong relationship between elevated LVEDP and decreased Plt count, suggesting that pressure overload on the lungs may interfere with the function of the lung as a site of Plt biosynthesis.
Collapse
Affiliation(s)
- Takuya Oh
- Division of Cardiology, Department of Internal Medicine The Jikei University School of Medicine 3‐25‐8 Nishi‐shinbashi, Minato‐ku Tokyo 105‐8461 Japan
| | - Kazuo Ogawa
- Division of Cardiology, Department of Internal Medicine The Jikei University School of Medicine 3‐25‐8 Nishi‐shinbashi, Minato‐ku Tokyo 105‐8461 Japan
| | - Tomohisa Nagoshi
- Division of Cardiology, Department of Internal Medicine The Jikei University School of Medicine 3‐25‐8 Nishi‐shinbashi, Minato‐ku Tokyo 105‐8461 Japan
| | - Kosuke Minai
- Division of Cardiology, Department of Internal Medicine The Jikei University School of Medicine 3‐25‐8 Nishi‐shinbashi, Minato‐ku Tokyo 105‐8461 Japan
| | - Takayuki Ogawa
- Division of Cardiology, Department of Internal Medicine The Jikei University School of Medicine 3‐25‐8 Nishi‐shinbashi, Minato‐ku Tokyo 105‐8461 Japan
| | - Makoto Kawai
- Division of Cardiology, Department of Internal Medicine The Jikei University School of Medicine 3‐25‐8 Nishi‐shinbashi, Minato‐ku Tokyo 105‐8461 Japan
| | - Michihiro Yoshimura
- Division of Cardiology, Department of Internal Medicine The Jikei University School of Medicine 3‐25‐8 Nishi‐shinbashi, Minato‐ku Tokyo 105‐8461 Japan
| |
Collapse
|