1
|
Mahmood NMS, Mahmud AMR, Maulood IM. Vascular actions of Ang 1-7 and Ang 1-8 through EDRFs and EDHFs in non-diabetes and diabetes mellitus. Nitric Oxide 2025; 156:9-26. [PMID: 40032212 DOI: 10.1016/j.niox.2025.02.003] [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: 10/14/2024] [Revised: 02/23/2025] [Accepted: 02/27/2025] [Indexed: 03/05/2025]
Abstract
The renin-angiotensin system (RAS) plays a pivotal role in regulating vascular homeostasis, while angiotensin 1-8 (Ang 1-8) traditionally dominates as a vasoconstrictor factor. However, the discovery of angiotensin 1-7 (Ang 1-7) and Ang 1-8 has revealed counter-regulatory mechanisms mediated through endothelial-derived relaxing factors (EDRFs) and endothelial-derived hyperpolarizing factors (EDHFs). This review delves into the vascular actions of Ang 1-7 and Ang 1-8 in both non-diabetes mellitus (non-DM) and diabetes mellitus (DM) conditions, highlighting their effects on vascular endothelial cell (VECs) function as well. In a non-DM vasculature context, Ang 1-8 demonstrate dual effect including vasoconstriction and vasodilation, respectively. Additionally, Ang 1-7 induces vasodilation upon nitric oxide (NO) production as a prominent EDRFs in distinct mechanisms. Further research elucidating the precise mechanisms underlying the vascular actions of Ang 1-7 and Ang 1-8 in DM will facilitate the development of tailored therapeutic interventions aimed at preserving vascular health and preventing cardiovascular complications.
Collapse
Affiliation(s)
- Nazar M Shareef Mahmood
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq.
| | - Almas M R Mahmud
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Ismail M Maulood
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| |
Collapse
|
2
|
De Bartolo A, Angelone T, Rocca C. Elucidating emerging signaling pathways driving endothelial dysfunction in cardiovascular aging. Vascul Pharmacol 2025; 158:107462. [PMID: 39805379 DOI: 10.1016/j.vph.2025.107462] [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: 10/28/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
The risk for developing cardiovascular diseases dramatically increases in older individuals, and aging vasculature plays a crucial role in determining their morbidity and mortality. Aging disrupts endothelial balance between vasodilators and vasoconstrictors, impairing function and promoting pathological vascular remodeling. In this Review, we discuss the impact of key and emerging molecular pathways that transduce aberrant inflammatory signals (i.e., chronic low-grade inflammation-inflammaging), oxidative stress, and mitochondrial dysfunction in aging vascular compartment. We focus on the interplay between these events, which contribute to generating a vicious cycle driving the progressive alterations in vascular structure and function during cardiovascular aging. We also discuss the primary role of senescent endothelial cells and vascular smooth muscle cells, and the potential link between vascular and myeloid cells, in impairing plaque stability and promoting the progression of atherosclerosis. The aim of this summary is to provide potential novel insights into targeting these processes for therapeutic benefit.
Collapse
Affiliation(s)
- Anna De Bartolo
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Tommaso Angelone
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, Cosenza, Italy; National Institute of Cardiovascular Research (INRC), Bologna, Italy.
| | - Carmine Rocca
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, Cosenza, Italy; National Institute of Cardiovascular Research (INRC), Bologna, Italy.
| |
Collapse
|
3
|
Sun Y, Xiao L, Chen L, Wang X. Doxorubicin-Induced Cardiac Remodeling: Mechanisms and Mitigation Strategies. Cardiovasc Drugs Ther 2025:10.1007/s10557-025-07673-6. [PMID: 40009315 DOI: 10.1007/s10557-025-07673-6] [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] [Accepted: 01/16/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND The therapeutic prowess of doxorubicin in oncology is marred by its cardiotoxic consequences, manifesting as cardiac remodeling. Pathophysiological alterations triggered by doxorubicin include inflammatory cascades, fibrotic tissue deposition, vascular and valvular changes, and finally cardiomyopathy. These multifarious consequences collectively orchestrate the deterioration of cardiac architecture and function. METHOD By charting the molecular underpinnings and remedial prospects, this review aspires to contribute a novel perspective using latest publications to the ongoing quest for cardioprotection in cancer therapy. RESULTS AND DISCUSSION Experimental analyses demonstrate the pivotal roles of oxidative stress and subsequent necrosis and apoptosis of cardiomyocytes, muscle cells, endothelial cells, and small muscle cells in different parts of the heart. In addition, severe and unusual infiltration of macrophages, mast cells, and neutrophils can amplify oxidative damage and subsequent impacts such as chronic inflammatory responses, vascular and valvular remodeling, and fibrosis. These modifications can render cardiomyopathy, ischemia, heart attack, and other disorders. In an endeavor to counteract these ramifications, a spectrum of emerging adjuvants and strategies are poised to fortify the heart against doxorubicin's deleterious effects. CONCLUSION The compendium of mitigation tactics such as innovative pharmacological agents hold the potential to attenuate the cardiotoxic burden.
Collapse
Affiliation(s)
- Yanna Sun
- Department of Cardiology, The First Affiliated of Zhengzhou University, Zhengzhou City Henan Province, 450052, China
| | - Lili Xiao
- Department of Cardiology, The First Affiliated of Zhengzhou University, Zhengzhou City Henan Province, 450052, China
| | - Linlin Chen
- Department of Cardiology, The First Affiliated of Zhengzhou University, Zhengzhou City Henan Province, 450052, China
| | - Xiaofang Wang
- Department of Cardiology, The First Affiliated of Zhengzhou University, Zhengzhou City Henan Province, 450052, China.
| |
Collapse
|
4
|
Ferreira ARO, Ribeiro MVG, Peres MNC, Lopes GKG, Saavedra LPJ, Piovan S, Barbosa LF, Raposo SR, Almeida DL, Malta A, Teixeira JJV, Mathias PCDF, Palma-Rigo K. Hypertension induced by peri-pubertal protein restriction depends on renin-angiotensin system dysfunction in adult male rats. Nutr Metab Cardiovasc Dis 2025; 35:103733. [PMID: 39448316 DOI: 10.1016/j.numecd.2024.09.003] [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: 10/15/2023] [Revised: 08/18/2024] [Accepted: 09/04/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND AND AIMS Hypertension depends on renin-angiotensin system dysfunction; however, little is known about its implications in the outcomes of neurogenic hypertension induced by peri-pubertal insults. This study aimed to evaluate whether hypertension induced by a peri-pubertal low-protein diet is related to renin-angiotensin system dysfunction in adult male Wistar rats. METHODS AND RESULTS Thirty-day-old male Wistar rats were fed a low-protein diet (4 % casein) for 30 days and subsequently fed a 20.5 % normal protein diet for a 60-day dietary recovery (LP group). Control animals (NP group) were fed a 20.5 % protein diet throughout their lives. Cardiovascular and renin-angiotensin system functions were evaluated on postnatal day 120 (6-24 animals per group). Statistical analyses were performed using the Student's t-test. Animals with LP show increased arterial blood pressure. The angiotensin 2 dose-response curve of LP animals showed an increase in the pressor response at a lower dose (50 ng/kg) and a reduction in the pressor response at a higher dose (400 ng/kg) compared with NP animals. Angiotensin 2 type 1 receptor mRNA levels were increased in the hearts of LP animals; however, angiotensin 2 type 2 receptor and MAS receptor mRNA levels were reduced. In the aorta, AT1 and AT2 mRNA levels were increased in LP animals, whereas MAS receptor mRNA levels were decreased in comparison to NP animals. CONCLUSION The renin-angiotensin system is disrupted in hypertension induced by protein restriction exposure during peri-pubertal life.
Collapse
MESH Headings
- Animals
- Male
- Renin-Angiotensin System/drug effects
- Diet, Protein-Restricted/adverse effects
- Rats, Wistar
- Hypertension/physiopathology
- Hypertension/etiology
- Hypertension/metabolism
- Receptor, Angiotensin, Type 2/metabolism
- Receptor, Angiotensin, Type 2/genetics
- Disease Models, Animal
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 1/genetics
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Angiotensin II
- Proto-Oncogene Mas
- Arterial Pressure/drug effects
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Rats
- RNA, Messenger/metabolism
- Age Factors
- Sexual Maturation
Collapse
Affiliation(s)
- Anna Rebeka Oliveira Ferreira
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | - Maiara Vanusa Guedes Ribeiro
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | - Maria Natalia Chimirri Peres
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | - Gabriel Kian Guimarães Lopes
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | - Lucas Paulo Jacinto Saavedra
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | - Silvano Piovan
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | - Leticia Ferreira Barbosa
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | - Scarllet Rodrigues Raposo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | - Douglas Lopes Almeida
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | - Ananda Malta
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | | | - Paulo Cezar de Freitas Mathias
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | - Kesia Palma-Rigo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil; Adventist College of Parana, Ivatuba, Parana, Brazil.
| |
Collapse
|
5
|
Yuan YE, Haas AV, Rosner B, Williams GH, McDonnell ME, Adler GK. The renin-angiotensin-aldosterone system and salt sensitivity of blood pressure offer new insights in obesity phenotypes. Obesity (Silver Spring) 2025; 33:321-330. [PMID: 39828424 PMCID: PMC11774662 DOI: 10.1002/oby.24218] [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: 09/16/2024] [Revised: 10/16/2024] [Accepted: 11/01/2024] [Indexed: 01/22/2025]
Abstract
OBJECTIVE Individuals who have metabolically healthy overweight/obesity (MHOO) do not have cardiometabolic complications despite an elevated BMI. Renin-angiotensin-aldosterone system (RAAS) activation and salt sensitivity of blood pressure (SSBP) are cardiovascular disease (CVD) risks, which are increased in individuals with higher BMI values. Little is known about the differences in RAAS activation and SSBP between MHOO and metabolically unhealthy overweight/obesity (MUOO) phenotypes. METHODS We studied 1430 adults on controlled dietary sodium. Individuals in the MHOO group had BMI ≥ 25 kg/m2 without comorbidities (e.g., diabetes, dyslipidemia, hypertension, CVD), whereas individuals in the MUOO group had BMI ≥ 25 kg/m2 and at least one comorbidity. The control group included healthy individuals (BMI 18.5-24.9 kg/m2). RESULTS BMI was similar between the MHOO (28.9 kg/m2) and MUOO groups (29.3 kg/m2; p = 0.317). On liberal sodium, the MUOO group had activated RAAS compared with the MHOO group, including higher plasma aldosterone concentration (mean [SD], 1.11 [0.48] ng/dL; p = 0.020), plasma angiotensin II levels (4.11 [2.0] pg/mL; p = 0.040), and percentage of individuals with plasma renin activity ≥ 1.0 ng/mL/h (+3.6%; p = 0.017). The MUOO group had higher SSBP than the MHOO group (6.0 [1.9] mm Hg; p = 0.002). Applying a zero-to-six-point metabolic health score found that a worse score was associated with higher measurements of RAAS activity and SSBP (p < 0.001). CONCLUSIONS Compared to the MHOO group, the MUOO group was characterized by an increase in the following two CVD risk factors: higher RAAS activity and SSBP on controlled sodium diets. Therapeutic interventions targeting the effects of angiotensin II and/or aldosterone may offer cardiometabolic protection for individuals with the MUOO phenotype.
Collapse
Affiliation(s)
- Yan Emily Yuan
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrea V. Haas
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Bernard Rosner
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Gordon H. Williams
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Marie E. McDonnell
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gail K. Adler
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Hashemi A, Kwak MJ, Goyal P. Pharmacologic Management of Heart Failure with Preserved Ejection Fraction (HFpEF) in Older Adults. Drugs Aging 2025; 42:95-110. [PMID: 39826050 DOI: 10.1007/s40266-024-01165-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2024] [Indexed: 01/20/2025]
Abstract
There are several pharmacologic agents that have been touted as guideline-directed medical therapy for heart failure with preserved ejection fraction (HFpEF). However, it is important to recognize that older adults with HFpEF also contend with an increased risk for adverse effects from medications due to age-related changes in pharmacokinetics and pharmacodynamics of medications, as well as the concurrence of geriatric conditions such as polypharmacy and frailty. With this review, we discuss the underlying evidence for the benefits of various treatments in HFpEF and incorporate key considerations for older adults, a subpopulation that may be at higher risk for adverse drug events. Key considerations for older adults include: the use of loop diuretics, mineralocorticoid receptor antagonists (MRAs), and sodium glucose co-transporter-2 (SGLT2) inhibitors for most; angiotensin receptor blockers/ angiotensin receptor-neprilysin inhibitors (ARB/ARNIs) and glucagon-like peptide-1 receptor agonists (GLP-1RAs) as add-on therapies for some, though risk of geriatric conditions such as falls, malnutrition, and/or sarcopenia must be considered; and beta blockers for a smaller subset of patients (with consideration of deprescribing for some, though data are lacking on this approach). Naturally, when making clinical decisions for older adults with cardiovascular disease, it is critical to consider the complexity of their conditions, including cognitive and physical function and social and environmental factors, and ensure alignment of care plans with the patient's health goals and priorities.
Collapse
Affiliation(s)
- Ashkan Hashemi
- Program for the Care and Study of the Aging Heart, Department of Medicine, Weill Cornell Medicine, 420 East 70th St, New York, NY, LH-36510063, USA
| | - Min Ji Kwak
- Division of Geriatric and Palliative Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, USA
| | - Parag Goyal
- Program for the Care and Study of the Aging Heart, Department of Medicine, Weill Cornell Medicine, 420 East 70th St, New York, NY, LH-36510063, USA.
| |
Collapse
|
7
|
Fuerlinger A, Stockner A, Sedej S, Abdellatif M. Caloric restriction and its mimetics in heart failure with preserved ejection fraction: mechanisms and therapeutic potential. Cardiovasc Diabetol 2025; 24:21. [PMID: 39827109 PMCID: PMC11742808 DOI: 10.1186/s12933-024-02566-8] [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: 11/25/2024] [Accepted: 12/26/2024] [Indexed: 01/22/2025] Open
Abstract
The global increase in human life expectancy, coupled with an unprecedented rise in the prevalence of obesity, has led to a growing clinical and socioeconomic burden of heart failure with preserved ejection fraction (HFpEF). Mechanistically, the molecular and cellular hallmarks of aging are omnipresent in HFpEF and are further exacerbated by obesity and associated metabolic diseases. Conversely, weight loss strategies, particularly caloric restriction, have shown promise in improving health status in patients with HFpEF and are considered the gold standard for promoting longevity and healthspan (disease-free lifetime) in model organisms. In this review, we implicate fundamental mechanisms of aging in driving HFpEF and elucidate how caloric restriction mitigates the disease progression. Furthermore, we discuss the potential for pharmacologically mimicking the beneficial effects of caloric restriction in HFpEF using clinically approved and emerging caloric restriction mimetics. We surmise that these compounds could offer novel therapeutic avenues for HFpEF and alleviate the challenges associated with the implementation of caloric restriction and other lifestyle modifications to reduce the burden of HFpEF at a population level.
Collapse
Affiliation(s)
- Alexander Fuerlinger
- Department of Cardiology, Medical University of Graz, 8036, Graz, Austria
- BioTechMed-Graz, 8010, Graz, Austria
| | - Alina Stockner
- Department of Cardiology, Medical University of Graz, 8036, Graz, Austria
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, 8036, Graz, Austria
- BioTechMed-Graz, 8010, Graz, Austria
- Faculty of Medicine, University of Maribor, 2000, Maribor, Slovenia
| | - Mahmoud Abdellatif
- Department of Cardiology, Medical University of Graz, 8036, Graz, Austria.
- BioTechMed-Graz, 8010, Graz, Austria.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, 94805, Villejuif, France.
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, 75006, France.
| |
Collapse
|
8
|
Eliopoulos AG, Gkouskou KK, Tsioufis K, Sanoudou D. A perspective on intermittent fasting and cardiovascular risk in the era of obesity pharmacotherapy. Front Nutr 2025; 12:1524125. [PMID: 39895836 PMCID: PMC11782017 DOI: 10.3389/fnut.2025.1524125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/02/2025] [Indexed: 02/04/2025] Open
Abstract
Intermittent fasting has been linked to metabolic health by improving lipid profiles, reducing body weight, and increasing insulin sensitivity. However, several randomized clinical trials have shown that intermittent fasting is not more effective than standard daily caloric restriction for short-term weight loss or cardiometabolic improvements in patients with obesity. Observational studies also suggest cardiovascular benefits from extended rather than reduced eating windows, and indicate that long-term intermittent fasting regimens may increase the risk of cardiovascular disease mortality. In this perspective, we discuss evidence that may support potential adverse effects of intermittent fasting on cardiovascular health through the loss of lean mass, circadian misalignment and poor dietary choices associated with reward-based eating. Given the ongoing revolution in obesity pharmacotherapy, we argue that future research should integrate anti-obesity medications with dietary strategies that confer robust benefits to cardiometabolic health, combine exercise regimens, and consider genetic factors to personalize obesity treatment. Comprehensive approaches combining diet, pharmacotherapy, and lifestyle modifications will become crucial for managing obesity and minimizing long-term cardiovascular risk.
Collapse
Affiliation(s)
- Aristides G. Eliopoulos
- Department of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Genosophy S.A., National and Kapodistrian University of Athens Spin-off Company, Athens, Greece
| | - Kalliopi K. Gkouskou
- Department of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Genosophy S.A., National and Kapodistrian University of Athens Spin-off Company, Athens, Greece
| | - Konstantinos Tsioufis
- 1st Department of Cardiology, Hippokration Hospital of Athens, National and Kapodistrian University of Athens, Athens, Greece
| | - Despina Sanoudou
- Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Clinical Genomics and Pharmacogenomics Unit, 4th Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
9
|
Lehman ML, Domenig O, Ames MK, Morgan JM. Effect of furosemide on comprehensive renin-angiotensin-aldosterone system activity of Thoroughbred horses. J Vet Intern Med 2024; 38:3272-3280. [PMID: 39434560 PMCID: PMC11586568 DOI: 10.1111/jvim.17208] [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: 02/24/2024] [Accepted: 09/19/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Furosemide, a commonly used diuretic, activates the renin-angiotensin-aldosterone system (RAAS) in other species. Little is known about RAAS peptide activation in horses. HYPOTHESIS/OBJECTIVES To evaluate equilibrium analysis as a practical method for RAAS quantification in horses and describe the RAAS response to a single dose of furosemide. We hypothesize that furosemide would cause transient increase in RAAS peptides in horses. ANIMALS 14 healthy adult thoroughbreds from a university teaching herd. METHODS Horses received either furosemide (1 mg/kg IV) or saline IV in a crossover study design. Protease-inhibited samples were compared with equilibrium analysis samples with Deming regression analysis. Renin-angiotensin-aldosterone system hormones were evaluated at 0, 0.25, 0.5, 4, and 24 hours postadministration, via equilibrium analysis. Values were compared with a mixed effects model. RESULTS Correlation between protease inhibition and equilibrium analysis was high for angiotensin I peptide (AngI) and angiotensin II peptide (AngII) (r = .92 and .95, respectively). Baseline RAAS peptide concentrations were below the limit of detection except AngII (median, 7.5 [range, 3.5-14.0] pmol/L). Furosemide administration resulted in an increase in AngI (8.0 [0.5-15.5] pmol/L, P = .03), AngII (33.7 [9.6-57.9] pmol/L, P = .0008), angiotensin III peptide (AngIII) (2.9 [0.9-4.9] pmol/L, P = .0005), angiotensin IV peptide (AngIV) (2.0 [0.6-3.4] pmol/L, P = .0005), and angiotensin 1-5 peptide (Ang1-5) (5.6 [1.2-5.9] pmol/L, P = .003) at 4 hours. Differences are reported as difference in the mean (95% confidence interval [CI]). CONCLUSIONS AND CLINICAL IMPORTANCE Furosemide produced an increase in hormones associated with both the classical and alternative RAAS pathways. Serum equilibrium analysis is practical for RAAS analysis in horses.
Collapse
Affiliation(s)
- Mallory L. Lehman
- Veterinary Medical Teaching Hospital, School of Veterinary MedicineUniversity of California‐DavisDavisCaliforniaUSA
| | | | - Marisa K. Ames
- Department of Medicine and Epidemiology, School of Veterinary MedicineUniversity of California‐DavisDavisCaliforniaUSA
| | - Jessica M. Morgan
- Department of Medicine and Epidemiology, School of Veterinary MedicineUniversity of California‐DavisDavisCaliforniaUSA
| |
Collapse
|
10
|
Shulman RS, Yang W, Cohen DL, Reese PP, Cohen JB. Cardiac Effects of Renin-Angiotensin System Inhibitors in Nonproteinuric CKD. Hypertension 2024; 81:2082-2090. [PMID: 39087321 PMCID: PMC11410532 DOI: 10.1161/hypertensionaha.124.23184] [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: 04/16/2024] [Accepted: 07/09/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND In contrast to proteinuric chronic kidney disease (CKD), the relative cardioprotective benefits of antihypertensive medications in nonproteinuric CKD are unknown. We examined long-term cardiovascular outcomes and mortality in patients with nonproteinuric CKD treated with renin-angiotensin system inhibitors (RASIs) versus other antihypertensive medications. METHODS Among participants of the CRIC study (Chronic Renal Insufficiency Cohort) without proteinuria, we used intention-to-treat analyses with inverse probability of treatment weighting and Cox proportional hazards modeling to determine the association of RASIs versus other antihypertensive medications with a composite cardiovascular outcome (myocardial infarction, stroke, heart failure hospitalization, and death) and mortality. Secondary analyses included per-protocol analyses accounting for continuous adherence and time-updated analyses accounting for the proportion of time using RASIs during follow-up. RESULTS A total of 2806 participants met the inclusion criteria. In the intention-to-treat analyses, RASIs versus other antihypertensive medications were not associated with an appreciable difference in cardiovascular events (adjusted hazard ratio [aHR], 0.94 [95% CI, 0.80-1.11]) or mortality (aHR, 1.06 [95% CI, 0.88-1.28]). In the per-protocol analyses, RASIs were associated with a lower risk of adverse cardiovascular events (aHR, 0.78 [95% CI, 0.63-0.97]) and mortality (aHR, 0.64 [95% CI, 0.48-0.85]). Similarly, in the time-updated analyses, a higher proportion of RASI use over time was associated with a lower mortality risk (aHR, 0.33 [95% CI, 0.14-0.86]). CONCLUSIONS Among individuals with nonproteinuric CKD, after accounting for time-updated use, RASIs are associated with fewer cardiovascular events and a lower mortality risk compared with other antihypertensive medications. Patients with nonproteinuric CKD may benefit from prioritizing RASIs for hypertension management.
Collapse
Affiliation(s)
- Rachel S Shulman
- Renal-Electrolyte and Hypertension Division (R.S.S., D.L.C., P.P.R., J.B.C.), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Wei Yang
- Department of Biostatistics, Epidemiology, and Informatics (W.Y., P.P.R., J.B.C.), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Debbie L Cohen
- Renal-Electrolyte and Hypertension Division (R.S.S., D.L.C., P.P.R., J.B.C.), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Peter P Reese
- Renal-Electrolyte and Hypertension Division (R.S.S., D.L.C., P.P.R., J.B.C.), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Biostatistics, Epidemiology, and Informatics (W.Y., P.P.R., J.B.C.), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jordana B Cohen
- Renal-Electrolyte and Hypertension Division (R.S.S., D.L.C., P.P.R., J.B.C.), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Biostatistics, Epidemiology, and Informatics (W.Y., P.P.R., J.B.C.), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
11
|
Chakraborty P, Nattel S, Nanthakumar K, Connelly KA, Husain M, Po SS, Ha ACT. Sudden cardiac death due to ventricular arrhythmia in diabetes mellitus: A bench to bedside review. Heart Rhythm 2024; 21:1827-1837. [PMID: 38848857 DOI: 10.1016/j.hrthm.2024.05.063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 05/27/2024] [Accepted: 05/31/2024] [Indexed: 06/09/2024]
Abstract
Diabetes mellitus (DM) confers an increased risk of sudden cardiac death (SCD) independent of its associated cardiovascular comorbidities. DM induces adverse structural, electrophysiologic, and autonomic cardiac remodeling that can increase one's risk of ventricular arrhythmias and SCD. Although glycemic control and prevention of microvascular and macrovascular complications are cornerstones in the management of DM, they are not adequate for the prevention of SCD. In this narrative review, we describe the contribution of DM to the pathophysiologic mechanism of SCD beyond its role in atherosclerotic cardiovascular disease and heart failure. On the basis of this pathophysiologic framework, we outline potential preventive and therapeutic strategies to mitigate the risk of SCD in this population of high-risk patients.
Collapse
Affiliation(s)
- Praloy Chakraborty
- Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Kumaraswamy Nanthakumar
- Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Kim A Connelly
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Keenan Research Centre for Biomedical Science, Unity Health Toronto, St Michael's Hospital, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Mansoor Husain
- Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Ontario, Canada
| | - Sunny S Po
- Heart Rhythm Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Andrew C T Ha
- Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
12
|
Mascarenhas LA, Ji Y, Wang W, Inciardi RM, Parikh RR, Eaton AA, Cheng S, Alonso A, Matsushita K, Shah AM, Solomon SD, Meyer ML, Chen LY, Zhang MJ. Association of central arterial stiffness with atrial myopathy: the Atherosclerosis Risk in Communities (ARIC) study. Hypertens Res 2024; 47:2902-2913. [PMID: 39117948 PMCID: PMC11806432 DOI: 10.1038/s41440-024-01831-3] [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: 05/26/2024] [Revised: 07/06/2024] [Accepted: 07/10/2024] [Indexed: 08/10/2024]
Abstract
Atrial myopathy-defined as abnormal left atrial (LA) size and function-is associated with an increased risk of atrial fibrillation, heart failure, and dementia. Central arterial stiffness is associated with increased atrial afterload and fibrosis and may be a risk factor for atrial myopathy. We examined the association of carotid-femoral pulse wave velocity (cfPWV) with LA function and assessed potential causal relationships. We included 2825 Atherosclerosis Risk in Communities (ARIC) study participants from Visit 5 (2011-2013). cfPWV was related to echocardiographic LA function continuously per 1-SD and categorically in quartiles. Mendelian randomization (MR) analysis was performed using U.K. Biobank-derived genetic variants associated with arterial stiffness index and cardiac magnetic resonance measures of LA function. When analyzed per SD increment (297.6 cm/s), higher cfPWV was significantly associated with lower LA reservoir and conduit strain (β = -0.53%, 95% CI [-0.81, -0.25] and β = -0.46%, 95% CI [-0.68, -0.25], respectively) after adjusting for demographics, clinical characteristics, systolic blood pressure, and left ventricular (LV) morphology and function. In MR analyses there was a non-significant inverse association of arterial stiffness index with LA total, passive, and active emptying fractions. Higher cfPWV is associated with lower LA reservoir and conduit strain, independent of systolic blood pressure and LV morphology and function. No evidence for a causal relationship between arterial stiffness index and alterations in LA function was found. Future studies should examine the prospective association of central arterial stiffness with LA function alterations.
Collapse
Affiliation(s)
- Lorraine A Mascarenhas
- Cardiovascular Division, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Yuekai Ji
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Wendy Wang
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Riccardo M Inciardi
- Institute of Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Romil R Parikh
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Anne A Eaton
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Susan Cheng
- Department of Cardiology, Cedars-Sinai Smidt Heart Institute, Los Angeles, CA, USA
| | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Kunihiro Matsushita
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Amil M Shah
- Division of Cardiology, Department of Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Scott D Solomon
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Michelle L Meyer
- Department of Emergency Medicine, University of North Carolina at Chapel Hill, North Carolina, USA
| | - Lin Yee Chen
- Cardiovascular Division, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Michael J Zhang
- Cardiovascular Division, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA.
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN, USA.
| |
Collapse
|
13
|
Apte M, Zambre S, Pisar P, Roy B, Tupe R. Decoding the role of aldosterone in glycation-induced diabetic complications. Biochem Biophys Res Commun 2024; 721:150107. [PMID: 38781658 DOI: 10.1016/j.bbrc.2024.150107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 05/01/2024] [Accepted: 05/12/2024] [Indexed: 05/25/2024]
Abstract
Diabetes-mediated development of micro and macro-vascular complications is a global concern. One of the factors is hyperglycemia induced the non-enzymatic formation of advanced glycation end products (AGEs). Accumulated AGEs bind with receptor of AGEs (RAGE) causing inflammation, oxidative stress and extracellular matrix proteins (ECM) modifications responsible for fibrosis, cell damage and tissue remodeling. Moreover, during hyperglycemia, aldosterone (Aldo) secretion increases, and its interaction with mineralocorticoid receptor (MR) through genomic and non-genomic pathways leads to inflammation and fibrosis. Extensive research on individual involvement of AGEs-RAGE and Aldo-MR pathways in the development of diabetic nephropathy (DN), cardiovascular diseases (CVDs), and impaired immune system has led to the discovery of therapeutic drugs. Despite mutual repercussions, the cross-talk between AGEs-RAGE and Aldo-MR pathways remains unresolved. Hence, this review focuses on the possible interaction of Aldo and glycation in DN and CVDs, considering the clinical significance of mutual molecular targets.
Collapse
Affiliation(s)
- Mayura Apte
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra State, India
| | - Saee Zambre
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra State, India
| | - Pratiksha Pisar
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra State, India
| | - Bishnudeo Roy
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra State, India
| | - Rashmi Tupe
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra State, India.
| |
Collapse
|
14
|
Wattanachayakul P, Srikulmontri T, Prasitsumrit V, Suenghataiphorn T, Danpanichkul P, Polpichai N, Saowapa S, Idowu A, Amanullah A. Vitamin D as a predictor of clinical response among patients with cardiac resynchronization therapy (CRT). J Arrhythm 2024; 40:975-981. [PMID: 39139866 PMCID: PMC11317716 DOI: 10.1002/joa3.13116] [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: 05/29/2024] [Revised: 06/20/2024] [Accepted: 07/05/2024] [Indexed: 08/15/2024] Open
Abstract
Introduction Cardiovascular and noncardiovascular comorbidities have been recognized as predictors of clinical response in patients receiving cardiac resynchronization therapy (CRT). However, data on vitamin D as a predictor of CRT response are conflicting. Method We identified studies from MEDLINE and Embase databases, searching from inception to May 2024, to investigate the association between 25-OH vitamin D levels before CRT implantation and outcomes. Studies had to report 25-OH vitamin D levels or the proportion of patients with vitamin D insufficiency and categorize outcomes as CRT responders or nonresponders. We extracted mean 25-OH vitamin D and standard deviations for both groups from each study and calculated the pooled mean difference (MD). We also retrieved risk ratios, and 95% confidence intervals (CIs) for the association between vitamin D insufficiency and lack of CRT response, combining them using the generic inverse variance method. Results Our meta-analysis included four studies. CRT responders had higher levels of 25-OH vitamin D than nonresponders, with a pooled MD of 8.04 ng/mL (95% CI: 3.16-12.93; I 2 = 48%, p < .001). Patients with vitamin D insufficiency before implantation had higher odds of lacking response to CRT, with a pooled RR of 3.28 (95% CI: 1.43-7.50; I 2 = 0%, p = .005) compared to those with normal vitamin D. Conclusions CRT responders had higher 25-OH vitamin D levels compared to nonresponders. Vitamin D insufficiency was associated with a higher risk of nonresponse to CRT. These findings highlight the importance of monitoring and managing vitamin D levels in these patients.
Collapse
Affiliation(s)
- Phuuwadith Wattanachayakul
- Department of MedicineJefferson Einstein HospitalPhiladelphiaPennsylvaniaUSA
- Sidney Kimmel Medical College, Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Thitiphan Srikulmontri
- Department of MedicineJefferson Einstein HospitalPhiladelphiaPennsylvaniaUSA
- Sidney Kimmel Medical College, Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Vitchapong Prasitsumrit
- Department of MedicineFaculty of Medicine Siriraj Hospital, Mahidol UniversityBangkokThailand
| | | | | | | | | | - Abiodun Idowu
- Department of MedicineJefferson Einstein HospitalPhiladelphiaPennsylvaniaUSA
- Sidney Kimmel Medical College, Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Aman Amanullah
- Sidney Kimmel Medical College, Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
- Division of Cardiovascular DiseaseJefferson Einstein HospitalPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
15
|
Wang RX, Serper M, Taddei TH, Kaplan DE, Mahmud N. The Association Between Angiotensin-Converting Enzyme Inhibitor or Angiotensin Receptor Blocker Exposure and Key Cirrhosis-Related Outcomes. Am J Gastroenterol 2024:00000434-990000000-01269. [PMID: 39051649 PMCID: PMC12036742 DOI: 10.14309/ajg.0000000000002976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024]
Abstract
INTRODUCTION Angiotensin-converting enzyme inhibitors (ACE-I) and angiotensin receptor blockers (ARB) may have hepatic benefits in patients with primarily chronic liver disease. ACE-I/ARB have not been evaluated in broad cohorts inclusive of those with decompensated cirrhosis. We analyzed the real-world association between ACE-I/ARB exposure and cirrhosis-related outcomes in a national cohort. METHODS We performed a retrospective, active comparator new user study of patients with cirrhosis in the Veterans Health Administration. We identified new initiators of ACE-I/ARB or calcium channel blockers (comparator). Inverse probability treatment weighting balanced key confounders and Cox regression evaluated the association between ACE-I/ARB and outcomes of mortality, cirrhosis decompensation, and hepatocellular carcinoma (HCC). In exploratory analysis, cause-specific competing risk models evaluated liver-related vs cardiovascular (CV)-related vs nonliver/non-CV-related mortality. RESULTS There were 904 ACE-I/ARB and 352 calcium channel blocker new initiators. In inverse probability treatment weighting Cox regression, ACE-I/ARB exposure was associated with reduced mortality (hazard ratio [HR] 0.70, 95% confidence interval [CI] 0.61-0.81, P < 0.001). In patients with compensated cirrhosis, ACE-I/ARB were not associated with hepatic decompensation or HCC. Cause-specific hazard models showed ACE-I/ARB exposure was associated with reduction in nonliver/non-CV-related mortality (cause-specific HR 0.49, 95% CI 0.38-0.62, P < 0.001) but not liver-related or CV-related mortality. In Child-Turcotte-Pugh A patients, ACE-I/ARB were associated with decreased CV-related mortality (cause-specific HR 0.41, 95% CI 0.26-0.65, P < 0.001). DISCUSSION ACE-I/ARB exposure was associated with reduced mortality, potentially through CV and other (renal, malignancy-related) mechanisms. In patients with compensated disease, ACE-I/ARB were not associated with hepatic decompensation or HCC. Future research should identify subsets of patients who benefit from ACE-I/ARB exposure.
Collapse
Affiliation(s)
- Roy X. Wang
- Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Marina Serper
- Department of Medicine, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | | | - David E. Kaplan
- Department of Medicine, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Nadim Mahmud
- Department of Medicine, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
16
|
Shah SK, Chaple DR, Masand VH, Jawarkar RD, Chaudhari S, Abiramasundari A, Zaki MEA, Al-Hussain SA. Multi-Target In-Silico modeling strategies to discover novel angiotensin converting enzyme and neprilysin dual inhibitors. Sci Rep 2024; 14:15991. [PMID: 38987327 PMCID: PMC11237057 DOI: 10.1038/s41598-024-66230-7] [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: 04/04/2024] [Accepted: 06/28/2024] [Indexed: 07/12/2024] Open
Abstract
Cardiovascular diseases, including heart failure, stroke, and hypertension, affect 608 million people worldwide and cause 32% of deaths. Combination therapy is required in 60% of patients, involving concurrent Renin-Angiotensin-Aldosterone-System (RAAS) and Neprilysin inhibition. This study introduces a novel multi-target in-silico modeling technique (mt-QSAR) to evaluate the inhibitory potential against Neprilysin and Angiotensin-converting enzymes. Using both linear (GA-LDA) and non-linear (RF) algorithms, mt-QSAR classification models were developed using 983 chemicals to predict inhibitory effects on Neprilysin and Angiotensin-converting enzymes. The Box-Jenkins method, feature selection method, and machine learning algorithms were employed to obtain the most predictive model with ~ 90% overall accuracy. Additionally, the study employed virtual screening of designed scaffolds (Chalcone and its analogues, 1,3-Thiazole, 1,3,4-Thiadiazole) applying developed mt-QSAR models and molecular docking. The identified virtual hits underwent successive filtration steps, incorporating assessments of drug-likeness, ADMET profiles, and synthetic accessibility tools. Finally, Molecular dynamic simulations were then used to identify and rank the most favourable compounds. The data acquired from this study may provide crucial direction for the identification of new multi-targeted cardiovascular inhibitors.
Collapse
Affiliation(s)
- Sapan K Shah
- Department of Pharmaceutical Chemistry, Priyadarshini J. L. College of Pharmacy, Hingna Road, Nagpur, 440016, Maharashtra, India.
| | - Dinesh R Chaple
- Department of Pharmaceutical Chemistry, Priyadarshini J. L. College of Pharmacy, Hingna Road, Nagpur, 440016, Maharashtra, India
| | - Vijay H Masand
- Department of Chemistry, Vidya Bharati Mahavidyalaya, Amravati, 444602, Maharashtra, India
| | - Rahul D Jawarkar
- Department of Medicinal Chemistry and Drug Discovery, Dr. Rajendra Gode Institute of Pharmacy, University Mardi Road, Amravati, 444603, India
| | - Somdatta Chaudhari
- Department of Pharmaceutical Chemistry, Modern College of Pharmacy, Nigdi, Pune, India
| | | | - Magdi E A Zaki
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, 11623, Saudi Arabia.
| | - Sami A Al-Hussain
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, 11623, Saudi Arabia
| |
Collapse
|
17
|
Kalupahana NS, Moustaid-Moussa N. Beyond blood pressure, fluid and electrolyte homeostasis - Role of the renin angiotensin aldosterone system in the interplay between metabolic diseases and breast cancer. Acta Physiol (Oxf) 2024; 240:e14164. [PMID: 38770946 DOI: 10.1111/apha.14164] [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: 01/16/2024] [Revised: 04/16/2024] [Accepted: 04/29/2024] [Indexed: 05/22/2024]
Abstract
The classical renin angiotensin aldosterone system (RAAS), as well as the recently described counter-regulatory or non-canonical RAAS have been well characterized for their role in cardiovascular homeostasis. Moreover, extensive research has been conducted over the past decades on both paracrine and the endocrine roles of local RAAS in various metabolic regulations and in chronic diseases. Clinical evidence from patients on RAAS blockers as well as pre-clinical studies using rodent models of genetic manipulations of RAAS genes documented that this system may play important roles in the interplay between metabolic diseases and cancer, namely breast cancer. Some of these studies suggest potential therapeutic applications and repurposing of RAAS inhibitors for these diseases. In this review, we discuss the mechanisms by which RAAS is involved in the pathogenesis of metabolic diseases such as obesity and type-2 diabetes as well as the role of this system in the initiation, expansion and/or progression of breast cancer, especially in the context of metabolic diseases.
Collapse
Affiliation(s)
- Nishan Sudheera Kalupahana
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences and Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA
| |
Collapse
|
18
|
Swami Vetha BS, Byrum R, Mebane D, Katwa LC, Aileru A. Impact of RAAS Receptors and Membrane-Bound Transporter System in the Left Ventricle during the Long-Term Control of Hypertension. Int J Mol Sci 2024; 25:6997. [PMID: 39000106 PMCID: PMC11241669 DOI: 10.3390/ijms25136997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
The Renin-Angiotensin-Aldosterone System (RAAS) has been implicated in systemic and neurogenic hypertension. The infusion of RAAS inhibitors blunted arterial pressure and efficacy of use-dependent synaptic transmission in sympathetic ganglia. The current investigation aims to elucidate the impact of RAAS-mediated receptors on left ventricular cardiomyocytes and the role of the sarcolemma-bound carrier system in the heart of the hypertensive transgene model. A significant increase in mRNA and the protein expression for angiotensin II (AngII) receptor subtype-1 (AT1R) was observed in (mREN2)27 transgenic compared to the normotensive rodents. Concurrently, there was an upregulation in AT1R and a downregulation in the MAS1 proto-oncogene protein receptor as well as the AngII subtype-2 receptor in hypertensive rodents. There were modifications in the expressions of sarcolemma Na+-K+-ATPase, Na+-Ca2+ exchanger, and Sarcoendoplasmic Reticulum Calcium ATPase in the transgenic hypertensive model. These observations suggest chronic RAAS activation led to a shift in receptor balance favoring augmented cardiac contractility and disruption in calcium handling through modifications of membrane-bound carrier proteins and blood pressure. The study provides insight into mechanisms underlying RAAS-mediated cardiac dysfunction and highlights the potential value of targeting the protective arm of AngII in hypertension.
Collapse
MESH Headings
- Animals
- Renin-Angiotensin System
- Hypertension/metabolism
- Heart Ventricles/metabolism
- Myocytes, Cardiac/metabolism
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 1/genetics
- Rats
- Proto-Oncogene Mas
- Blood Pressure
- Male
- Mice
- Receptor, Angiotensin, Type 2/metabolism
- Receptor, Angiotensin, Type 2/genetics
- Sarcolemma/metabolism
- Sodium-Potassium-Exchanging ATPase/metabolism
- Sodium-Potassium-Exchanging ATPase/genetics
- Sodium-Calcium Exchanger/metabolism
- Sodium-Calcium Exchanger/genetics
- Mice, Transgenic
Collapse
Affiliation(s)
- Berwin Singh Swami Vetha
- Department of Foundational Sciences, East Carolina School of Dental Medicine, Greenville, NC 27834, USA; (R.B.); (D.M.)
| | - Rachel Byrum
- Department of Foundational Sciences, East Carolina School of Dental Medicine, Greenville, NC 27834, USA; (R.B.); (D.M.)
| | - DaQuan Mebane
- Department of Foundational Sciences, East Carolina School of Dental Medicine, Greenville, NC 27834, USA; (R.B.); (D.M.)
| | - Laxmansa C. Katwa
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA;
| | - Azeez Aileru
- Department of Foundational Sciences, East Carolina School of Dental Medicine, Greenville, NC 27834, USA; (R.B.); (D.M.)
| |
Collapse
|
19
|
Kalambokis G, Christaki M, Tsiakas I, Despotis G, Lakkas L, Tsiouris S, Xourgia X, Markopoulos GS, Dova L, Milionis H. Association of left ventricular diastolic dysfunction with inflammatory activity, renal dysfunction, and liver-related mortality in patients with cirrhosis and ascites. Eur J Gastroenterol Hepatol 2024; 36:775-783. [PMID: 38526935 DOI: 10.1097/meg.0000000000002762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Left ventricular diastolic dysfunction (LVDD) is the predominant cardiac abnormality in cirrhosis. We investigated the association of LVDD with systemic inflammation and its impact on renal function, occurrence of hepatorenal syndrome (HRS) and survival in patients with cirrhosis and ascites. We prospectively enrolled 215 patients with cirrhosis and ascites. We evaluated the diagnosis and grading of LVDD by Doppler echocardiography, inflammatory markers, systemic hemodynamics, vasoactive factors, radioisotope-assessed renal function and blood flow, HRS development and liver-related mortality. LVDD was diagnosed in 142 (66%) patients [grade 2/3: n = 61 (43%)]. Serum lipopolysaccharide-binding protein (LBP), plasma renin activity (PRA) and glomerular filtration rate (GFR) were independently associated with the presence of grade 2/3 LVDD and the severity of diastolic dysfunction. Serum tumor necrosis factor-α, cardiac output and plasma noradrenaline were also independently associated with the presence of grade 2/3 LVDD. The diastolic function marker E / e ' was strongly correlated with serum LBP ( r = 0.731; P < 0.001), PRA ( r = 0.714; P < 0.001) and GFR ( r = -0.609; P < 0.001) among patients with LVDD. The 5-year risk of HRS development and death was significantly higher in patients with grade 2/3 LVDD compared to those with grade 1 (35.5 vs. 14.4%; P = 0.01 and 53.3 vs. 28.2%; P = 0.03, respectively). The occurrence and severity of LVDD in patients with cirrhosis and ascites is closely related to inflammatory activity. Advanced LVDD is associated with baseline circulatory and renal dysfunction, favoring HRS development, and increased mortality.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Georgios S Markopoulos
- Hematology Laboratory, Unit of Molecular Biology and Translational Flow Cytometry, Medical School, University of Ioannina, Ioannina, Greece
| | - Lefkothea Dova
- Hematology Laboratory, Unit of Molecular Biology and Translational Flow Cytometry, Medical School, University of Ioannina, Ioannina, Greece
| | | |
Collapse
|
20
|
Boudabbous M, Hammemi R, Gdoura H, Chtourou L, Moalla M, Mnif L, Amouri A, Abid L, Tahri N. Cirrhotic cardiomyopathy: a subject that's always topical. Future Sci OA 2024; 10:FSO954. [PMID: 38817353 PMCID: PMC11137786 DOI: 10.2144/fsoa-2023-0110] [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: 06/13/2023] [Accepted: 12/13/2023] [Indexed: 06/01/2024] Open
Abstract
Cirrhosis is the final stage in the development of hepatic fibrosis in chronic liver disease. It is associated with major hemodynamic disturbances defining the hyperdynamic circulation and may be complicated by specific cardiac involvement or cirrhotic cardiomyopathy which is a silent clinical condition that typically remains asymptomatic until the late stages of liver disease. Recently, new criteria defining CC, based on modern concepts and knowledge of heart failure, have been proposed. Despite knowledge of the main mechanisms behind this entity, there is no specific treatment available for cirrhotic cardiomyopathy. The management approach for symptomatic cardiomyopathy in cirrhotic patients is similar to that for left ventricular failure in non-cirrhotic individuals.
Collapse
Affiliation(s)
- Mona Boudabbous
- Gastroenterology Department, Hédi Chaker Hospital, Sfax, Tunisia
- Medecin sfax university, Sfax university, Tunisia
| | - Rania Hammemi
- Cardiology, Department, Hédi Chaker Hospital, Sfax, Tunisia
- Medecin sfax university, Sfax university, Tunisia
| | - Hela Gdoura
- Gastroenterology Department, Hédi Chaker Hospital, Sfax, Tunisia
- Medecin sfax university, Sfax university, Tunisia
| | - Lassad Chtourou
- Gastroenterology Department, Hédi Chaker Hospital, Sfax, Tunisia
- Medecin sfax university, Sfax university, Tunisia
| | - Manel Moalla
- Gastroenterology Department, Hédi Chaker Hospital, Sfax, Tunisia
- Medecin sfax university, Sfax university, Tunisia
| | - Leila Mnif
- Gastroenterology Department, Hédi Chaker Hospital, Sfax, Tunisia
- Medecin sfax university, Sfax university, Tunisia
| | - Ali Amouri
- Gastroenterology Department, Hédi Chaker Hospital, Sfax, Tunisia
- Medecin sfax university, Sfax university, Tunisia
| | - Leila Abid
- Cardiology, Department, Hédi Chaker Hospital, Sfax, Tunisia
- Medecin sfax university, Sfax university, Tunisia
| | - Nabil Tahri
- Gastroenterology Department, Hédi Chaker Hospital, Sfax, Tunisia
- Medecin sfax university, Sfax university, Tunisia
| |
Collapse
|
21
|
Preston KJ, Kawai T, Torimoto K, Kuroda R, Nakayama Y, Akiyama T, Kimura Y, Scalia R, Autieri MV, Rizzo V, Hashimoto T, Osei-Owusu P, Eguchi S. Mitochondrial fission inhibition protects against hypertension induced by angiotensin II. Hypertens Res 2024; 47:1338-1349. [PMID: 38383894 DOI: 10.1038/s41440-024-01610-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 12/14/2023] [Accepted: 01/27/2024] [Indexed: 02/23/2024]
Abstract
Mitochondrial dysfunction has been implicated in various types of cardiovascular disease including hypertension. Mitochondrial fission fusion balance is critical to mitochondrial quality control, whereas enhanced fission has been reported in several models of cardiovascular disease. However, limited information is available regarding the contribution of mitochondrial fission in hypertension. Here, we have tested the hypothesis that inhibition of mitochondrial fission attenuates the development of hypertension and associated vascular remodeling. In C57BL6 mice infused with angiotensin II for 2 weeks, co-treatment of mitochondrial fission inhibitor, mdivi1, significantly inhibited angiotensin II-induced development of hypertension assessed by radiotelemetry. Histological assessment of hearts and aortas showed that mdivi1 inhibited vessel fibrosis and hypertrophy induced by angiotensin II. This was associated with attenuation of angiotensin II-induced decline in mitochondrial aspect ratio seen in both the endothelial and medial layers of aortas. Mdivi1 also mitigated angiotensin II-induced cardiac hypertrophy assessed by heart weight-to-body weight ratio as well as by echocardiography. In ex vivo experiments, mdivi1 inhibited vasoconstriction and abolished the enhanced vascular reactivity by angiotensin II in small mesenteric arteries. Proteomic analysis on endothelial cell culture media with angiotensin II and/or mdivi1 treatment revealed that mdivi1 inhibited endothelial cell hypersecretory phenotype induced by angiotensin II. In addition, mdivi1 attenuated angiotensin II-induced protein induction of periostin, a myofibroblast marker in cultured vascular fibroblasts. In conclusion, these data suggest that mdivi1 prevented angiotensin II-induced hypertension and cardiovascular remodeling via multicellular mechanisms in the vasculature.
Collapse
Affiliation(s)
- Kyle J Preston
- Department of Cardiovascular Science and Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Tatsuo Kawai
- Lemole Center for Integrated Lymphatics Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Keiichi Torimoto
- Department of Cardiovascular Science and Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Ryohei Kuroda
- Department of Cardiovascular Science and Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Yuki Nakayama
- Department of Cardiovascular Science and Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Tomoko Akiyama
- Advanced Medical Research Center, Yokohama City University, Yokohama, 236-0004, Japan
| | - Yayoi Kimura
- Advanced Medical Research Center, Yokohama City University, Yokohama, 236-0004, Japan
| | - Rosario Scalia
- Lemole Center for Integrated Lymphatics Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Michael V Autieri
- Lemole Center for Integrated Lymphatics Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Victor Rizzo
- Department of Cardiovascular Science and Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Tomoki Hashimoto
- Barrow Aneurysm and AVM Research Center, Departments of Neurosurgery and Neurobiology Barrow Neurological Institute Phoenix AZ, Phoenix, AZ, USA
| | - Patrick Osei-Owusu
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Satoru Eguchi
- Department of Cardiovascular Science and Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA.
| |
Collapse
|
22
|
Takajo D, Przybycien TS, Balakrishnan PL, Natarajan G, Singh GK, Aggarwal S. Left ventricle hypertrophy and re-modeling in children with essential hypertension: does the race matter? Cardiol Young 2024; 34:906-913. [PMID: 37968238 DOI: 10.1017/s1047951123003840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2023]
Abstract
BACKGROUND This is the first study to report on the impact of race on differences in the prevalence of echocardiographic left ventricular hypertrophy and left ventricular adaptation at the time of diagnosis of essential hypertension in children. METHODS This cross-sectional, single-centre study included patients aged 3-18 years who had newly diagnosed essential hypertension. Echocardiography was used to assess left ventricular mass index and left ventricular relative wall thickness. An left ventricular mass index > the 95th percentile for age and gender, and an left ventricular relative wall thickness > 0.42, were used to diagnose left ventricular hypertrophy and concentric adaptation. Various echocardiographic parameters were compared between African Americans and Caucasians. RESULTS The study included 422 patients (289 African Americans and 133 Caucasians) diagnosed with essential hypertension at a median age of 14.6 (interquartile range; 12.1-16.3) years. Eighty-eight patients (20.9%) had left ventricular hypertrophy. There was no statistically significant difference in the prevalence of left ventricular hypertrophy between African Americans and Caucasians (22.5% versus 17.3%, p=0.22). The median left ventricular relative wall thickness was 0.35 (0.29-0.43), and 114 patients (27.0%) had an left ventricular relative wall thickness > 0.42. The presence of an left ventricular relative wall thickness > 0.42 was significantly higher among African Americans compared to Caucasians (30.1% versus 20.3%, p = 0.04). The African American race was a strong predictor for an left ventricular relative wall thickness > 0.42 (odds ratio 1.7, p = 0.04), but not for left ventricular mass index > the 95th percentile (p = 0.22). Overweight/obesity was a strong predictor for an left ventricular mass index > the 95th percentile. CONCLUSIONS There was no difference in the prevalence of left ventricular hypertrophy in children with essential hypertension of different races. Obesity, rather than being African American, is associated with left ventricular hypertrophy.
Collapse
Affiliation(s)
- Daiji Takajo
- Department of Pediatrics, Children's Hospital of Michigan, Detroit, MI, USA
| | - Thomas S Przybycien
- Division of Pediatric Cardiology, Department of Pediatrics, Children's Hospital of Michigan, Detroit, MI, USA
| | - Preetha L Balakrishnan
- Division of Pediatric Cardiology, Department of Pediatrics, Children's Hospital of Michigan, Detroit, MI, USA
| | - Girija Natarajan
- Division of Neonatal & Perinatal Medicine, Department of Pediatrics, Children's Hospital of Michigan, Detroit, MI, USA
| | - Gautam K Singh
- Division of Pediatric Cardiology, Department of Pediatrics, Children's Hospital of Michigan, Detroit, MI, USA
| | - Sanjeev Aggarwal
- Division of Pediatric Cardiology, Department of Pediatrics, Children's Hospital of Michigan, Detroit, MI, USA
| |
Collapse
|
23
|
Huang Z, Duan A, Zhao Z, Zhao Q, Zhang Y, Li X, Zhang S, Gao L, An C, Luo Q, Liu Z. Sleep-disordered breathing patterns and prognosis in pulmonary arterial hypertension: A cluster analysis of nocturnal cardiorespiratory signals. Sleep Med 2024; 113:61-69. [PMID: 37984019 DOI: 10.1016/j.sleep.2023.11.016] [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: 07/17/2023] [Revised: 10/15/2023] [Accepted: 11/08/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND Sleep-disordered breathing (SDB) is common among pulmonary arterial hypertension (PAH) patients and has been associated with unfavorable outcomes. This study aims to cluster overnight cardiorespiratory signals to investigate PAH phenotypes and examining their prognostic implications. METHODS In this retrospective cohort study, we recruited consecutive PAH patients who underwent right heart catheterization and nocturnal cardiorespiratory polygraphy to evaluate SDB. Cluster analysis was employed to classify patients based on their SDB patterns. Cox regression analysis and Kaplan-Meier curves were utilized to assess the association between cluster membership and clinical outcomes. Logistic regression was used to identify risk factors associated with the cluster at higher risk of adverse outcomes. RESULTS The study comprised 386 PAH patients, with a mean age of 44.7 ± 17.0 years, of which 46.6 % were male. Three distinct clusters of PAH patients were identified: Cluster 1 (N = 182) presented with minimal SDB, Cluster 2 (N = 125) displayed obstructive sleep apnea (OSA) without significant hypoxemia, and Cluster 3 (N = 79) exhibited predominantly severe hypoxemic burden along with comorbid OSA. Notably, patients in Cluster 3 had an independent association with an increased risk of clinical worsening (hazard ratio 1.96, 95 % confidence interval [CI] 1.08-3.56, P = 0.027) compared to those in Clusters 1, even after adjusting for common confounders. The rate of clinical worsening for PAH-related events and mortality was higher in Cluster 3 than in Clusters 1 and 2 (26.6 % vs. 12.6 % and 19.2 %, respectively, log-rank P = 0.024). Moreover, the left ventricular mass index was identified as an independent risk factor for Cluster 3 (odds ratios 1.01, 95 % CI 1.00-1.02, P = 0.004). CONCLUSIONS Patients with PAH who have nocturnal hypoxemia and OSA had worse clinical outcomes compared to those with only minimal SDB. Tailored management strategies that address both PAH and nocturnal hypoxemia may be effective in improving clinical outcomes.
Collapse
Affiliation(s)
- Zhihua Huang
- Center for Respiratory and Pulmonary Vascular Diseases, Department of Cardiology, Fuwai Hospital, National Clinical Research Center for Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Anqi Duan
- Center for Respiratory and Pulmonary Vascular Diseases, Department of Cardiology, Fuwai Hospital, National Clinical Research Center for Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhihui Zhao
- Center for Respiratory and Pulmonary Vascular Diseases, Department of Cardiology, Fuwai Hospital, National Clinical Research Center for Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qing Zhao
- Center for Respiratory and Pulmonary Vascular Diseases, Department of Cardiology, Fuwai Hospital, National Clinical Research Center for Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Zhang
- Center for Respiratory and Pulmonary Vascular Diseases, Department of Cardiology, Fuwai Hospital, National Clinical Research Center for Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Li
- Center for Respiratory and Pulmonary Vascular Diseases, Department of Cardiology, Fuwai Hospital, National Clinical Research Center for Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sicheng Zhang
- Center for Respiratory and Pulmonary Vascular Diseases, Department of Cardiology, Fuwai Hospital, National Clinical Research Center for Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Luyang Gao
- Center for Respiratory and Pulmonary Vascular Diseases, Department of Cardiology, Fuwai Hospital, National Clinical Research Center for Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chenhong An
- Center for Respiratory and Pulmonary Vascular Diseases, Department of Cardiology, Fuwai Hospital, National Clinical Research Center for Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qin Luo
- Center for Respiratory and Pulmonary Vascular Diseases, Department of Cardiology, Fuwai Hospital, National Clinical Research Center for Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Zhihong Liu
- Center for Respiratory and Pulmonary Vascular Diseases, Department of Cardiology, Fuwai Hospital, National Clinical Research Center for Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
24
|
Hua R, Gao H, He C, Xin S, Wang B, Zhang S, Gao L, Tao Q, Wu W, Sun F, Xu J. An emerging view on vascular fibrosis molecular mediators and relevant disorders: from bench to bed. Front Cardiovasc Med 2023; 10:1273502. [PMID: 38179503 PMCID: PMC10764515 DOI: 10.3389/fcvm.2023.1273502] [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: 08/06/2023] [Accepted: 11/27/2023] [Indexed: 01/06/2024] Open
Abstract
Vascular fibrosis is a widespread pathologic condition that arises during vascular remodeling in cardiovascular dysfunctions. According to previous studies, vascular fibrosis is characterized by endothelial matrix deposition and vascular wall thickening. The RAAS and TGF-β/Smad signaling pathways have been frequently highlighted. It is, however, far from explicit in terms of understanding the cause and progression of vascular fibrosis. In this review, we collected and categorized a large number of molecules which influence the fibrosing process, in order to acquire a better understanding of vascular fibrosis, particularly of pathologic dysfunction. Furthermore, several mediators that prevent vascular fibrosis are discussed in depth in this review, with the aim that this will contribute to the future prevention and treatment of related conditions.
Collapse
Affiliation(s)
- Rongxuan Hua
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Han Gao
- Department of Clinical Laboratory, Aerospace Center Hospital, Peking University, Beijing, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chengwei He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shuzi Xin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Boya Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, China
| | - Sitian Zhang
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Lei Gao
- Department of Biomedical Informatics, School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Qiang Tao
- Department of Biomedical Informatics, School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Wenqi Wu
- Experimental Center for Morphological Research Platform, Capital Medical University, Beijing, China
| | - Fangling Sun
- Department of Experimental Animal Laboratory, Xuan-Wu Hospital of Capital Medical University, Beijing, China
| | - Jingdong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
25
|
Pesova P, Jiravska Godula B, Jiravsky O, Jelinek L, Sovova M, Moravcova K, Ozana J, Gajdusek L, Miklik R, Sknouril L, Neuwirth R, Sovova E. Exercise-Induced Blood Pressure Dynamics: Insights from the General Population and the Athletic Cohort. J Cardiovasc Dev Dis 2023; 10:480. [PMID: 38132648 PMCID: PMC10743421 DOI: 10.3390/jcdd10120480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/19/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
Blood pressure (BP) dynamics during graded exercise testing provide important insights into cardiovascular health, particularly in athletes. These measurements, taken during intense physical exertion, complement and often enhance our understanding beyond traditional resting BP measurements. Historically, the challenge has been to distinguish 'normal' from 'exaggerated' BP responses in the athletic environment. While basic guidelines have served their purpose, they may not fully account for the complex nature of BP responses in today's athletes, as illuminated by contemporary research. This review critically evaluates existing guidelines in the context of athletic performance and cardiovascular health. Through a rigorous analysis of the current literature, we highlight the multifaceted nature of exercise-induced BP fluctuations in athletes, emphasising the myriad determinants that influence these responses, from specific training regimens to inherent physiological nuances. Our aim is to advocate a tailored, athlete-centred approach to BP assessment during exercise. Such a paradigm shift is intended to set the stage for evidence-based guidelines to improve athletic training, performance and overall cardiovascular well-being.
Collapse
Affiliation(s)
- Petra Pesova
- Faculty of Medicine, Palacky University, Krizkovskeho 511/8, 779 00 Olomouc, Czech Republic; (P.P.); (B.J.G.)
- Sports Cardiology Center, Nemocnice Agel Trinec-Podlesi, Konska 453, 739 61 Trinec, Czech Republic (R.N.)
| | - Bogna Jiravska Godula
- Faculty of Medicine, Palacky University, Krizkovskeho 511/8, 779 00 Olomouc, Czech Republic; (P.P.); (B.J.G.)
- Sports Cardiology Center, Nemocnice Agel Trinec-Podlesi, Konska 453, 739 61 Trinec, Czech Republic (R.N.)
| | - Otakar Jiravsky
- Sports Cardiology Center, Nemocnice Agel Trinec-Podlesi, Konska 453, 739 61 Trinec, Czech Republic (R.N.)
- Faculty of Medicine, Masaryk University, Kamenice 735/5, 625 00 Brno, Czech Republic
| | - Libor Jelinek
- Faculty of Medicine, Palacky University, Krizkovskeho 511/8, 779 00 Olomouc, Czech Republic; (P.P.); (B.J.G.)
| | - Marketa Sovova
- Faculty of Medicine, Palacky University, Krizkovskeho 511/8, 779 00 Olomouc, Czech Republic; (P.P.); (B.J.G.)
| | - Katarina Moravcova
- Faculty of Medicine, Palacky University, Krizkovskeho 511/8, 779 00 Olomouc, Czech Republic; (P.P.); (B.J.G.)
| | - Jaromir Ozana
- Faculty of Medicine, Palacky University, Krizkovskeho 511/8, 779 00 Olomouc, Czech Republic; (P.P.); (B.J.G.)
| | - Libor Gajdusek
- Sports Cardiology Center, Nemocnice Agel Trinec-Podlesi, Konska 453, 739 61 Trinec, Czech Republic (R.N.)
- Faculty of Medicine, University of Ostrava, Syllabova 19, 703 00 Ostrava, Czech Republic
| | - Roman Miklik
- Sports Cardiology Center, Nemocnice Agel Trinec-Podlesi, Konska 453, 739 61 Trinec, Czech Republic (R.N.)
- Faculty of Medicine, Masaryk University, Kamenice 735/5, 625 00 Brno, Czech Republic
| | - Libor Sknouril
- Sports Cardiology Center, Nemocnice Agel Trinec-Podlesi, Konska 453, 739 61 Trinec, Czech Republic (R.N.)
| | - Radek Neuwirth
- Sports Cardiology Center, Nemocnice Agel Trinec-Podlesi, Konska 453, 739 61 Trinec, Czech Republic (R.N.)
- Faculty of Medicine, Masaryk University, Kamenice 735/5, 625 00 Brno, Czech Republic
| | - Eliska Sovova
- Faculty of Medicine, Palacky University, Krizkovskeho 511/8, 779 00 Olomouc, Czech Republic; (P.P.); (B.J.G.)
| |
Collapse
|
26
|
Pu Y, Yang G, Pan X, Zhou Y, Zhong A, Ding N, Su Y, Peng W, Zeng M, Guo T, Chai X. Higher plasma aldosterone concentrations in patients with aortic diseases and hypertension: a retrospective observational study. Eur J Med Res 2023; 28:541. [PMID: 38008731 PMCID: PMC10676595 DOI: 10.1186/s40001-023-01528-2] [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: 03/07/2023] [Accepted: 11/14/2023] [Indexed: 11/28/2023] Open
Abstract
BACKGROUND Aortic diseases remain a highly perilous macrovascular condition. The relationship between circulating aldosterone and aortic diseases is rarely explored, thus we investigated the difference in plasma aldosterone concentration (PAC) between patients with and without aortic disease in hypertensive people. METHODS We analyzed 926 patients with hypertension, ranging in age from 18 to 89 years, who had their PAC measured from the hospital's electronic database. The case group and control group were defined based on inclusion and exclusion criteria. The analysis included general information, clinical data, biochemical data, and medical imaging examination results as covariates. To further evaluate the difference in PAC between primary hypertension patients with aortic disease and those without, we used multivariate logistic regression analysis and also employed propensity score matching to minimize the influence of confounding factors. RESULTS In total, 394 participants were included in the analysis, with 66 individuals diagnosed with aortic diseases and 328 in the control group. The participants were predominantly male (64.5%) and over the age of 50 (68.5%), with an average PAC of 19.95 ng/dL. After controlling for confounding factors, the results showed hypertension patients with aortic disease were more likely to have high PAC levels than those without aortic disease (OR = 1.138, 95% CI [1.062 to 1.238]). Subgroup analysis revealed consistent relationship between PAC and primary hypertensive patients with aortic disease across the different stratification variables. Additionally, hypertensive patients with aortic disease still have a risk of higher PAC levels than those without aortic disease, even after propensity score matching. CONCLUSIONS The results of this study suggest that primary hypertensive patients with aortic diseases have elevated levels of PAC, but the causal relationship between PAC and aortic disease requires further study.
Collapse
Affiliation(s)
- Yuting Pu
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China
- Emergency Medicine and Difficult Disease Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guifang Yang
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China
- Emergency Medicine and Difficult Disease Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaogao Pan
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China
- Emergency Medicine and Difficult Disease Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yang Zhou
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China
- Emergency Medicine and Difficult Disease Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Aifang Zhong
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China
- Emergency Medicine and Difficult Disease Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ning Ding
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China
- Emergency Medicine and Difficult Disease Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yingjie Su
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China
- Emergency Medicine and Difficult Disease Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wen Peng
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China
- Emergency Medicine and Difficult Disease Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mengping Zeng
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China
- Emergency Medicine and Difficult Disease Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tuo Guo
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China
- Emergency Medicine and Difficult Disease Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiangping Chai
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China.
- Emergency Medicine and Difficult Disease Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
27
|
Muendlein A, Heinzle C, Leiherer A, Brandtner EM, Geiger K, Gaenger S, Fraunberger P, Mader A, Saely CH, Drexel H. Circulating glypican-4 is a new predictor of all-cause mortality in patients with heart failure. Clin Biochem 2023; 121-122:110675. [PMID: 37844682 DOI: 10.1016/j.clinbiochem.2023.110675] [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: 06/28/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023]
Abstract
BACKGROUND Heart failure confers a high burden of morbidity and mortality. However, risk prediction in heart failure patients still is limited. Blood-based biomarkers hold promise to improve clinical risk assessment. Recently we have identified circulating glypican-4 (GPC4) as a significant predictor of mortality in coronary angiography patients and patients with peripheral artery disease. The impact of serum GPC4 on mortality in patients with heart failure is unknown and is addressed in this prospective cohort study. METHODS We prospectively recorded all-cause mortality in 288 patients with heart failure. GPC4 levels were measured using an enzyme-linked immunosorbent assay at baseline. RESULTS During the 24-month follow-up period, 28.1% (n = 81) of the patients died. Serum GPC4 significantly predicted all-cause mortality (hazard ratio (HR) per doublingof GPC4 = 3.57 [2.31-5.53]; P < 0.001). Subgroup analysis showed that GPC4 was significantly associated with all-cause mortality in patients with reduced ejection fraction (HR per doubling = 3.25 [1.75-6.04]; P < 0.001) as well as in those with preserved ejection fraction (HR per doubling = 3.07 [1.22-7.70]; P = 0.017). The association between serum GPC4 and all-cause mortality remained significant in multivariable Cox regression analysis correcting for traditional risk factors (P = 0.035). Results from C-statistics indicated an additional prognostic value of GPC4 relative to NT-proBNP for the prediction of two-year all-cause mortality (P = 0.030). CONCLUSION Circulating GPC4 independently predicts all-cause mortality in patients with heart failure.
Collapse
Affiliation(s)
- Axel Muendlein
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria.
| | - Christine Heinzle
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria; Medical Central Laboratories, Feldkirch, Austria
| | - Andreas Leiherer
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria; Medical Central Laboratories, Feldkirch, Austria; Private University in the Principality of Liechtenstein, Triesen, Liechtenstein
| | - Eva Maria Brandtner
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
| | - Kathrin Geiger
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria; Medical Central Laboratories, Feldkirch, Austria
| | - Stella Gaenger
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
| | | | - Arthur Mader
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria; Department of Internal Medicine I, Academic Teaching Hospital Feldkirch, Feldkirch, Austria
| | - Christoph H Saely
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria; Private University in the Principality of Liechtenstein, Triesen, Liechtenstein; Department of Internal Medicine I, Academic Teaching Hospital Feldkirch, Feldkirch, Austria
| | - Heinz Drexel
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria; Private University in the Principality of Liechtenstein, Triesen, Liechtenstein; Vorarlberger Landeskrankenhausbetriebsgesellschaft, Feldkirch, Austria; Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
28
|
Abdellatif M, Rainer PP, Sedej S, Kroemer G. Hallmarks of cardiovascular ageing. Nat Rev Cardiol 2023; 20:754-777. [PMID: 37193857 DOI: 10.1038/s41569-023-00881-3] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/21/2023] [Indexed: 05/18/2023]
Abstract
Normal circulatory function is a key determinant of disease-free life expectancy (healthspan). Indeed, pathologies affecting the cardiovascular system, which are growing in prevalence, are the leading cause of global morbidity, disability and mortality, whereas the maintenance of cardiovascular health is necessary to promote both organismal healthspan and lifespan. Therefore, cardiovascular ageing might precede or even underlie body-wide, age-related health deterioration. In this Review, we posit that eight molecular hallmarks are common denominators in cardiovascular ageing, namely disabled macroautophagy, loss of proteostasis, genomic instability (in particular, clonal haematopoiesis of indeterminate potential), epigenetic alterations, mitochondrial dysfunction, cell senescence, dysregulated neurohormonal signalling and inflammation. We also propose a hierarchical order that distinguishes primary (upstream) from antagonistic and integrative (downstream) hallmarks of cardiovascular ageing. Finally, we discuss how targeting each of the eight hallmarks might be therapeutically exploited to attenuate residual cardiovascular risk in older individuals.
Collapse
Affiliation(s)
- Mahmoud Abdellatif
- Department of Cardiology, Medical University of Graz, Graz, Austria.
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- BioTechMed Graz, Graz, Austria.
| | - Peter P Rainer
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| |
Collapse
|
29
|
Ding K, Li Z, Lu Y, Sun L. Efficacy and safety assessment of mineralocorticoid receptor antagonists in patients with chronic kidney disease. Eur J Intern Med 2023; 115:114-127. [PMID: 37328398 DOI: 10.1016/j.ejim.2023.05.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/09/2023] [Accepted: 05/29/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND The objective of our study is to evaluate the efficacy and safety of mineralocorticoid receptor antagonists (MRAs) and determine the optimal MRA treatment regimen in patients with chronic kidney disease (CKD). METHODS We searched PubMed, Embase, Web of Science, and the Cochrane Library from their inception to June 20, 2022. The composite kidney outcome, cardiovascular events, urinary albumin to creatinine ratio (UACR), estimated glomerular filtration rate (EGFR), serum potassium, systolic blood pressure (SBP), diastolic blood pressure (DBP), creatine and creatine clearance were included for analysis. We conducted pairwise meta-analyses and Bayesian network meta-analyses (NMA) and calculated the surface under the cumulative ranking curve (SUCRA). RESULTS We included 26 studies with 15,531 participants. By pairwise meta-analyses, we found that MRA treatment could significantly reduce UACR in CKD patients with or without diabetes. Notably, compared to placebo, Finerenone was associated with a lower risk of composite kidney outcome and cardiovascular events. Data from NMA demonstrated an overt UACR reduction without increasing serum potassium by Apararenone, Esaxerenone, and Finerenone in CKD patients. Spironolactone decreased SBP and DBP but elevated CKD patients' serum potassium. CONCLUSIONS Compared to placebo, Apararenone, Esaxerenone, and Finerenone might ameliorate albuminuria in CKD patients without causing elevated serum potassium levels. Remarkably, Finerenone conferred a cardiovascular benefit, and Spironolactone lowered blood pressure in CKD patients.
Collapse
Affiliation(s)
- Kaiyue Ding
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Zhuoyu Li
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Yingying Lu
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Lin Sun
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China.
| |
Collapse
|
30
|
Tian C, Ziegler JN, Zucker IH. Extracellular Vesicle MicroRNAs in Heart Failure: Pathophysiological Mediators and Therapeutic Targets. Cells 2023; 12:2145. [PMID: 37681877 PMCID: PMC10486980 DOI: 10.3390/cells12172145] [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: 07/18/2023] [Revised: 08/20/2023] [Accepted: 08/22/2023] [Indexed: 09/09/2023] Open
Abstract
Extracellular vesicles (EVs) are emerging mediators of intracellular and inter-organ communications in cardiovascular diseases (CVDs), especially in the pathogenesis of heart failure through the transference of EV-containing bioactive substances. microRNAs (miRNAs) are contained in EV cargo and are involved in the progression of heart failure. Over the past several years, a growing body of evidence has suggested that the biogenesis of miRNAs and EVs is tightly regulated, and the sorting of miRNAs into EVs is highly selective and tightly controlled. Extracellular miRNAs, particularly circulating EV-miRNAs, have shown promising potential as prognostic and diagnostic biomarkers for heart failure and as therapeutic targets. In this review, we summarize the latest progress concerning the role of EV-miRNAs in HF and their application in a therapeutic strategy development for heart failure.
Collapse
Affiliation(s)
- Changhai Tian
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA;
| | - Jessica N. Ziegler
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40536, USA;
| | - Irving H. Zucker
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| |
Collapse
|
31
|
Dattani A, Brady EM, Alfuhied A, Gulsin GS, Steadman CD, Yeo JL, Aslam S, Banovic M, Jerosch-Herold M, Xue H, Kellman P, Costet P, Cvijic ME, Zhao L, Ebert C, Liu L, Gunawardhana K, Gordon D, Chang CP, Arnold JR, Yates T, Kelly D, Hogrefe K, Dawson D, Greenwood J, Ng LL, Singh A, McCann GP. Impact of diabetes on remodelling, microvascular function and exercise capacity in aortic stenosis. Open Heart 2023; 10:e002441. [PMID: 37586847 PMCID: PMC10432628 DOI: 10.1136/openhrt-2023-002441] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 08/04/2023] [Indexed: 08/18/2023] Open
Abstract
OBJECTIVE To characterise cardiac remodelling, exercise capacity and fibroinflammatory biomarkers in patients with aortic stenosis (AS) with and without diabetes, and assess the impact of diabetes on outcomes. METHODS Patients with moderate or severe AS with and without diabetes underwent echocardiography, stress cardiovascular magnetic resonance (CMR), cardiopulmonary exercise testing and plasma biomarker analysis. Primary endpoint for survival analysis was a composite of cardiovascular mortality, myocardial infarction, hospitalisation with heart failure, syncope or arrhythmia. Secondary endpoint was all-cause death. RESULTS Diabetes (n=56) and non-diabetes groups (n=198) were well matched for age, sex, ethnicity, blood pressure and severity of AS. The diabetes group had higher body mass index, lower estimated glomerular filtration rate and higher rates of hypertension, hyperlipidaemia and symptoms of AS. Biventricular volumes and systolic function were similar, but the diabetes group had higher extracellular volume fraction (25.9%±3.1% vs 24.8%±2.4%, p=0.020), lower myocardial perfusion reserve (2.02±0.75 vs 2.34±0.68, p=0.046) and lower percentage predicted peak oxygen consumption (68%±21% vs 77%±17%, p=0.002) compared with the non-diabetes group. Higher levels of renin (log10renin: 3.27±0.59 vs 2.82±0.69 pg/mL, p<0.001) were found in diabetes. Multivariable Cox regression analysis showed diabetes was not associated with cardiovascular outcomes, but was independently associated with all-cause mortality (HR 2.04, 95% CI 1.05 to 4.00; p=0.037). CONCLUSIONS In patients with moderate-to-severe AS, diabetes is associated with reduced exercise capacity, increased diffuse myocardial fibrosis and microvascular dysfunction, but not cardiovascular events despite a small increase in mortality.
Collapse
Affiliation(s)
- Abhishek Dattani
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Emer M Brady
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Aseel Alfuhied
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Gaurav S Gulsin
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Christopher D Steadman
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
- Department of Cardiology, Poole Hospital NHS Foundation Trust, Poole, UK
| | - Jian L Yeo
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Saadia Aslam
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Marko Banovic
- Cardiology Department, Clinical Centre of Serbia, Belgrade, Serbia
| | | | - Hui Xue
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Peter Kellman
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | - Lei Zhao
- Bristol Myers Squibb Co, Princeton, New Jersey, USA
| | | | - Laura Liu
- Bristol Myers Squibb Co, Princeton, New Jersey, USA
| | | | - David Gordon
- Bristol Myers Squibb Co, Princeton, New Jersey, USA
| | | | - J Ranjit Arnold
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Thomas Yates
- Diabetes Research Centre, University of Leicester, Leicester, UK
| | - Damian Kelly
- Cardiology Department, Royal Derby Hospital, Derby, UK
| | - Kai Hogrefe
- Cardiology Department, Kettering General Hospital NHS Foundation Trust, Kettering, UK
| | - Dana Dawson
- Cardiovascular Medicine Research Unit, University of Aberdeen, Aberdeen, UK
| | - John Greenwood
- Department of Cardiology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
- Department of Biomedical Imaging Sciences, University of Leeds, Leeds, UK
| | - Leong L Ng
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Anvesha Singh
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Gerry P McCann
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| |
Collapse
|
32
|
Mkhize BC, Mosili P, Ngubane PS, Sibiya NH, Khathi A. The Relationship between Renin-Angiotensin-Aldosterone System (RAAS) Activity, Osteoporosis and Estrogen Deficiency in Type 2 Diabetes. Int J Mol Sci 2023; 24:11963. [PMID: 37569338 PMCID: PMC10419188 DOI: 10.3390/ijms241511963] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/15/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Type 2 diabetes (T2D) is associated with a plethora of comorbidities, including osteoporosis, which occurs due to an imbalance between bone resorption and formation. Numerous mechanisms have been explored to understand this association, including the renin-angiotensin-aldosterone system (RAAS). An upregulated RAAS has been positively correlated with T2D and estrogen deficiency in comorbidities such as osteoporosis in humans and experimental studies. Therefore, research has focused on these associations in order to find ways to improve glucose handling, osteoporosis and the downstream effects of estrogen deficiency. Upregulation of RAAS may alter the bone microenvironment by altering the bone marrow inflammatory status by shifting the osteoprotegerin (OPG)/nuclear factor kappa-Β ligand (RANKL) ratio. The angiotensin-converting-enzyme/angiotensin II/Angiotensin II type 1 receptor (ACE/Ang II/AT1R) has been evidenced to promote osteoclastogenesis and decrease osteoblast formation and differentiation. ACE/Ang II/AT1R inhibits the wingless-related integration site (Wnt)/β-catenin pathway, which is integral in bone formation. While a lot of literature exists on the effects of RAAS and osteoporosis on T2D, the work is yet to be consolidated. Therefore, this review looks at RAAS activity in relation to osteoporosis and T2D. This review also highlights the relationship between RAAS activity, osteoporosis and estrogen deficiency in T2D.
Collapse
Affiliation(s)
- Bongeka Cassandra Mkhize
- Human Physiology, Health Science, Westville Campus, University of KwaZulu-Natal, Westville 4041, South Africa; (B.C.M.); (P.M.); (P.S.N.)
| | - Palesa Mosili
- Human Physiology, Health Science, Westville Campus, University of KwaZulu-Natal, Westville 4041, South Africa; (B.C.M.); (P.M.); (P.S.N.)
| | - Phikelelani Sethu Ngubane
- Human Physiology, Health Science, Westville Campus, University of KwaZulu-Natal, Westville 4041, South Africa; (B.C.M.); (P.M.); (P.S.N.)
| | | | - Andile Khathi
- Human Physiology, Health Science, Westville Campus, University of KwaZulu-Natal, Westville 4041, South Africa; (B.C.M.); (P.M.); (P.S.N.)
| |
Collapse
|
33
|
Gnudi L, Fountoulakis N, Panagiotou A, Corcillo A, Maltese G, Rife MF, Ntalas I, Franks R, Chiribiri A, Ayis S, Karalliedde J. Effect of active vitamin-D on left ventricular mass index: Results of a randomized controlled trial in type 2 diabetes and chronic kidney disease. Am Heart J 2023; 261:1-9. [PMID: 36934979 DOI: 10.1016/j.ahj.2023.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 03/03/2023] [Accepted: 03/12/2023] [Indexed: 05/26/2023]
Abstract
BACKGROUND Active vitamin-D deficiency is a potential modifiable risk factor for increased ventricular mass. We explored the effects of active vitamin-D (calcitriol) treatment on left ventricular mass in patients with type-2 diabetes (T2D) and chronic kidney disease (CKD). METHODS We performed a 48-week duration single center randomized double-blind parallel group trial examining the impact of calcitriol, 0.5 mcg once daily, as compared to placebo on a primary endpoint of change from baseline in left ventricular mass index (LVMI) measured by magnetic resonance imaging . Patients with T2D, CKD stage-3 and raised left ventricular mass on stable renin angiotensin aldosterone system blockade, who all had elevated intact parathyroid hormone were eligible. Secondary endpoints included interstitial myocardial fibrosis, assessed with cardiac magnetic resonance imaging. In total, 45 (male 73%) patients with T2D and stage-3 CKD were studied (calcitriol n = 19, placebo n = 26). RESULTS Following 48-weeks calcitriol treatment, the median difference and the (95% CI) of LVMI between the 2 treatment arms was 1.84 (-1.28, 4.96), similar between the 2 groups studied. Intact parathyroid hormone fell only in the calcitriol group from 142 pg/mL (80-293) to 76 pg/mL (41-204)(median, interquartile range, P= .04). No significant differences were observed in interstitial myocardial fibrosis or other secondary endpoints. CONCLUSIONS The study did not provide evidence that treatment with calcitriol as compared to placebo might improve LVMI in patients with T2D, mild left ventricular hypertrophy and stable CKD. Our data does not support the routine use of active vitamin-D for LVMI regression and cardiovascular protection in patients with T2D and stage-3 CKD.
Collapse
Affiliation(s)
- Luigi Gnudi
- School of Cardiovascular and Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom.
| | - Nikolaos Fountoulakis
- School of Cardiovascular and Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Angeliki Panagiotou
- School of Cardiovascular and Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Antonella Corcillo
- School of Cardiovascular and Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Giuseppe Maltese
- School of Cardiovascular and Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Maria Flaquer Rife
- School of Cardiovascular and Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Ioannis Ntalas
- School of Biomedical Engineering & Imaging Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Russell Franks
- School of Biomedical Engineering & Imaging Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Amedeo Chiribiri
- School of Biomedical Engineering & Imaging Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Salma Ayis
- School of Population Health & Environmental Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Janaka Karalliedde
- School of Cardiovascular and Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| |
Collapse
|
34
|
Bueno Junior CR, Bano A, Tang Y, Sun X, Abate A, Hall E, Mitri J, Morieri ML, Shah H, Doria A. Rapid kidney function decline and increased risk of heart failure in patients with type 2 diabetes: findings from the ACCORD cohort : Rapid kidney function decline and heart failure in T2D. Cardiovasc Diabetol 2023; 22:131. [PMID: 37365586 PMCID: PMC10291814 DOI: 10.1186/s12933-023-01869-6] [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: 04/01/2023] [Accepted: 05/28/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND Impaired kidney function and albuminuria are associated with increased risk of heart failure (HF) in patients with type 2 diabetes (T2D). We investigated whether rapid kidney function decline over time is an additional determinant of increased HF risk in patients with T2D, independent of baseline kidney function, albuminuria, and other HF predictors. METHODS Included in the study were 7,539 participants in the Action to Control Cardiovascular Risk in Diabetes (ACCORD) study with baseline urinary albumin-to-creatinine ratio (UACR) data, who had completed 4 years of follow-up and had ≥ 3 eGFR measurements during that period (median eGFR/year = 1.9, IQR 1.7-3.2). The association between rapid kidney function decline (eGFR loss ≥ 5 ml/min/1.73 m2/year) and odds of HF hospitalization or HF death during the first 4 years of follow-up was estimated by logistic regression. The improvement in risk discrimination provided by adding rapid kidney function decline to other HF risk factors was evaluated as the increment in the area under the Receiving Operating Characteristics curve (ROC AUC) and integrated discrimination improvement (IDI). RESULTS Over 4 years of follow-up, 1,573 participants (20.9%) experienced rapid kidney function decline and 255 (3.4%) experienced a HF event. Rapid kidney function decline was associated with a ~ 3.2-fold increase in HF odds (3.23, 95% CI, 2.51-4.16, p < 0.0001), independent of baseline CVD history. This estimate was not attenuated by adjustment for potential confounders, including eGFR and UACR at baseline as well as at censoring (3.74; 95% CI 2.63-5.31). Adding rapid kidney function decline during follow-up to other clinical predictors (WATCH-DM score, eGFR, and UACR at study entry and end of follow-up) improved HF risk classification (ROC AUC = + 0.02, p = 0.027; relative IDI = + 38%, p < 0.0001). CONCLUSIONS In patients with T2D, rapid kidney function decline is associated with a marked increase in HF risk, independent of starting kidney function and/or albuminuria. These findings highlight the importance of serial eGFR measurements over time to improve HF risk estimation in T2D.
Collapse
Affiliation(s)
- Carlos Roberto Bueno Junior
- Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- School of Physical Education and Sport, Medical School, College of Nursing of Ribeirao Preto, University of Sao Paulo (USP), Ribeirao Preto, Sao Paulo, Brazil
| | - Arjola Bano
- Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, Switzerland
- Department of Cardiology, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Yaling Tang
- Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Xiuqin Sun
- Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Endocrine and Metabolism, Peking University Shuang Hospital, Beijing, China
| | - Alex Abate
- Research Division, Joslin Diabetes Center, Boston, MA, USA
| | - Elizabeth Hall
- Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Joanna Mitri
- Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Mario Luca Morieri
- Department of Medicine, Metabolic Disease Unit, University of Padova, University Hospital of Padova, Padova, Italy
| | - Hetal Shah
- Research Division, Joslin Diabetes Center, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Alessandro Doria
- Research Division, Joslin Diabetes Center, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Research Division, Joslin Diabetes Center, One Joslin Place, Boston, MA, 02215, USA.
| |
Collapse
|
35
|
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
|
36
|
Salama ABM, Abouleisa RRE, Ou Q, Tang XL, Alhariry N, Hassan S, Gebreil A, Dastagir M, Abdulwali F, Bolli R, Mohamed TMA. Transient gene therapy using cell cycle factors reverses renin-angiotensin-aldosterone system activation in heart failure rat model. Mol Cell Biochem 2023; 478:1245-1250. [PMID: 36282351 PMCID: PMC10126184 DOI: 10.1007/s11010-022-04590-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/13/2022] [Indexed: 10/31/2022]
Abstract
The loss of cardiomyocytes after myocardial infarction (MI) leads to heart failure. Recently, we demonstrated that transient overexpression of 4 cell cycle factors (4F), using a polycistronic non-integrating lentivirus (TNNT2-4F-NIL) resulted in significant improvement in cardiac function in a rat model of MI. Yet, it is crucial to demonstrate the reversal of the heart failure-related pathophysiological manifestations, such as renin-angiotensin-aldosterone system activation (RAAS). To assess that, Fisher 344 rats were randomized to receive TNNT2-4F-NIL or control virus seven days after coronary occlusion for 2 h followed by reperfusion. 4 months after treatment, N-terminal pro-brain natriuretic peptide, plasma renin activity, and aldosterone levels returned to the normal levels in rats treated with TNNT2-4F-NIL but not in vehicle-treated rats. Furthermore, the TNNT2-4F-NIL-treated group showed significantly less liver and kidney congestion than vehicle-treated rats. Thus, we conclude that in rat models of MI, TNNT2-4F-NIL reverses RAAS activation and subsequent systemic congestion.
Collapse
Affiliation(s)
- Abou Bakr M Salama
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, Louisville, KY, USA
- Department of Cardiology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
- Department of Cardiac Surgery, University of Verona, Verona, Italy
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
- Diabetes and Obesity Center, Department of Medicine, Envirome Institute, University of Louisville, Louisville, KY, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Riham R E Abouleisa
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, Louisville, KY, USA
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
- Diabetes and Obesity Center, Department of Medicine, Envirome Institute, University of Louisville, Louisville, KY, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Qinghui Ou
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, Louisville, KY, USA
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
- Diabetes and Obesity Center, Department of Medicine, Envirome Institute, University of Louisville, Louisville, KY, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Xian-Liang Tang
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, Louisville, KY, USA
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
- Diabetes and Obesity Center, Department of Medicine, Envirome Institute, University of Louisville, Louisville, KY, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Nashwah Alhariry
- Department of Pathology, Faculty of Medicine, Suez University, Ismailia, Egypt
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
- Diabetes and Obesity Center, Department of Medicine, Envirome Institute, University of Louisville, Louisville, KY, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Sarah Hassan
- Department of Electron Microscopy, Theodor Bilharz Research Institute, Imbaba Giza, Egypt
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
- Diabetes and Obesity Center, Department of Medicine, Envirome Institute, University of Louisville, Louisville, KY, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Ahmad Gebreil
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, Louisville, KY, USA
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
- Diabetes and Obesity Center, Department of Medicine, Envirome Institute, University of Louisville, Louisville, KY, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Muzammil Dastagir
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, Louisville, KY, USA
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
- Diabetes and Obesity Center, Department of Medicine, Envirome Institute, University of Louisville, Louisville, KY, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Fareeha Abdulwali
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, Louisville, KY, USA
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
- Diabetes and Obesity Center, Department of Medicine, Envirome Institute, University of Louisville, Louisville, KY, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Roberto Bolli
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, Louisville, KY, USA
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
- Diabetes and Obesity Center, Department of Medicine, Envirome Institute, University of Louisville, Louisville, KY, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Tamer M A Mohamed
- Department of Medicine, Institute of Molecular Cardiology, University of Louisville, Louisville, KY, USA.
- Department of Electron Microscopy, Theodor Bilharz Research Institute, Imbaba Giza, Egypt.
- Department of Bioengineering, University of Louisville, Louisville, KY, USA.
- Diabetes and Obesity Center, Department of Medicine, Envirome Institute, University of Louisville, Louisville, KY, USA.
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA.
- Institute of Cardiovascular Sciences, University of Manchester, Manchester, UK.
- Institute of Molecular Cardiology, University of Louisville, 580 South Preston Street, Louisville, KY, 40202, USA.
| |
Collapse
|
37
|
Maines E, Moretti M, Vitturi N, Gugelmo G, Fasan I, Lenzini L, Piccoli G, Gragnaniello V, Maiorana A, Soffiati M, Burlina A, Franceschi R. Understanding the Pathogenesis of Cardiac Complications in Patients with Propionic Acidemia and Exploring Therapeutic Alternatives for Those Who Are Not Eligible or Are Waiting for Liver Transplantation. Metabolites 2023; 13:563. [PMID: 37110221 PMCID: PMC10143878 DOI: 10.3390/metabo13040563] [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: 02/13/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
The guidelines for the management of patients affected by propionic acidemia (PA) recommend standard cardiac therapy in the presence of cardiac complications. A recent revision questioned the impact of high doses of coenzyme Q10 on cardiac function in patients with cardiomyopathy (CM). Liver transplantation is a therapeutic option for several patients since it may stabilize or reverse CM. Both the patients waiting for liver transplantation and, even more, the ones not eligible for transplant programs urgently need therapies to improve cardiac function. To this aim, the identification of the pathogenetic mechanisms represents a key point. Aims: This review summarizes: (1) the current knowledge of the pathogenetic mechanisms underlying cardiac complications in PA and (2) the available and potential pharmacological options for the prevention or the treatment of cardiac complications in PA. To select articles, we searched the electronic database PubMed using the Mesh terms "propionic acidemia" OR "propionate" AND "cardiomyopathy" OR "Long QT syndrome". We selected 77 studies, enlightening 12 potential disease-specific or non-disease-specific pathogenetic mechanisms, namely: impaired substrate delivery to TCA cycle and TCA dysfunction, secondary mitochondrial electron transport chain dysfunction and oxidative stress, coenzyme Q10 deficiency, metabolic reprogramming, carnitine deficiency, cardiac excitation-contraction coupling alteration, genetics, epigenetics, microRNAs, micronutrients deficiencies, renin-angiotensin-aldosterone system activation, and increased sympathetic activation. We provide a critical discussion of the related therapeutic options. Current literature supports the involvement of multiple cellular pathways in cardiac complications of PA, indicating the growing complexity of their pathophysiology. Elucidating the mechanisms responsible for such abnormalities is essential to identify therapeutic strategies going beyond the correction of the enzymatic defect rather than engaging the dysregulated mechanisms. Although these approaches are not expected to be resolutive, they may improve the quality of life and slow the disease progression. Available pharmacological options are limited and tested in small cohorts. Indeed, a multicenter approach is mandatory to strengthen the efficacy of therapeutic options.
Collapse
Affiliation(s)
- Evelina Maines
- Division of Pediatrics, Santa Chiara General Hospital, APSS, 38122 Trento, Italy
| | - Michele Moretti
- Division of Cardiology, Santa Chiara General Hospital, APSS, 38122 Trento, Italy
| | - Nicola Vitturi
- Division of Metabolic Diseases, Department of Medicine-DIMED, University Hospital, 35128 Padova, Italy
| | - Giorgia Gugelmo
- Division of Clinical Nutrition, Department of Medicine-DIMED, University Hospital, 35128 Padova, Italy
| | - Ilaria Fasan
- Division of Clinical Nutrition, Department of Medicine-DIMED, University Hospital, 35128 Padova, Italy
| | - Livia Lenzini
- Emergency Medicine Unit, Department of Medicine-DIMED, University Hospital, 35128 Padova, Italy
| | - Giovanni Piccoli
- CIBIO, Department of Cellular, Computational and Integrative Biology, Italy & Dulbecco Telethon Institute, Università degli Studi di Trento, 38123 Trento, Italy
| | - Vincenza Gragnaniello
- Division of Inherited Metabolic Diseases, Reference Centre Expanded Newborn Screening, Department of Women’s and Children’s Health, University Hospital, 35128 Padova, Italy
| | - Arianna Maiorana
- Division of Metabolism and Research Unit of Metabolic Biochemistry, Bambino Gesù Children’s Hospital-IRCCS, 00165 Rome, Italy
| | - Massimo Soffiati
- Division of Pediatrics, Santa Chiara General Hospital, APSS, 38122 Trento, Italy
| | - Alberto Burlina
- Division of Inherited Metabolic Diseases, Reference Centre Expanded Newborn Screening, Department of Women’s and Children’s Health, University Hospital, 35128 Padova, Italy
| | - Roberto Franceschi
- Division of Pediatrics, Santa Chiara General Hospital, APSS, 38122 Trento, Italy
| |
Collapse
|
38
|
Ghosh N, Chacko L, Bhattacharya H, Vallamkondu J, Nag S, Dey A, Karmakar T, Reddy PH, Kandimalla R, Dewanjee S. Exploring the Complex Relationship between Diabetes and Cardiovascular Complications: Understanding Diabetic Cardiomyopathy and Promising Therapies. Biomedicines 2023; 11:biomedicines11041126. [PMID: 37189744 DOI: 10.3390/biomedicines11041126] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 05/17/2023] Open
Abstract
Diabetes mellitus (DM) and cardiovascular complications are two unmet medical emergencies that can occur together. The rising incidence of heart failure in diabetic populations, in addition to apparent coronary heart disease, ischemia, and hypertension-related complications, has created a more challenging situation. Diabetes, as a predominant cardio-renal metabolic syndrome, is related to severe vascular risk factors, and it underlies various complex pathophysiological pathways at the metabolic and molecular level that progress and converge toward the development of diabetic cardiomyopathy (DCM). DCM involves several downstream cascades that cause structural and functional alterations of the diabetic heart, such as diastolic dysfunction progressing into systolic dysfunction, cardiomyocyte hypertrophy, myocardial fibrosis, and subsequent heart failure over time. The effects of glucagon-like peptide-1 (GLP-1) analogues and sodium-glucose cotransporter-2 (SGLT-2) inhibitors on cardiovascular (CV) outcomes in diabetes have shown promising results, including improved contractile bioenergetics and significant cardiovascular benefits. The purpose of this article is to highlight the various pathophysiological, metabolic, and molecular pathways that contribute to the development of DCM and its significant effects on cardiac morphology and functioning. Additionally, this article will discuss the potential therapies that may be available in the future.
Collapse
Affiliation(s)
- Nilanjan Ghosh
- Molecular Pharmacology Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Leena Chacko
- BioAnalytical Lab, Meso Scale Discovery, Rockville, MD 20850-3173, USA
| | - Hiranmoy Bhattacharya
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | | | - Sagnik Nag
- Department of Biotechnology, Vellore Institute of Technology (VIT), School of Biosciences & Technology, Tiruvalam Road, Vellore 632014, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata 700073, India
| | - Tanushree Karmakar
- Dr. B C Roy College of Pharmacy and Allied Health Sciences, Durgapur 713206, India
| | | | - Ramesh Kandimalla
- Department of Biochemistry, Kakatiya Medical College, Warangal 506007, India
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| |
Collapse
|
39
|
Theobald D, Nair AR, Sriramula S, Francis J. Cardiomyocyte-specific deletion of TLR4 attenuates angiotensin II-induced hypertension and cardiac remodeling. Front Cardiovasc Med 2023; 10:1074700. [PMID: 37034342 PMCID: PMC10079917 DOI: 10.3389/fcvm.2023.1074700] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 02/28/2023] [Indexed: 04/11/2023] Open
Abstract
Toll-like receptor 4 (TLR4) is an integral factor in the initiation of the innate immune response and plays an important role in cardiovascular diseases such as hypertension and myocardial infarction. Previous studies from our lab demonstrated that central TLR4 blockade reduced cardiac TLR4 expression, attenuated hypertension, and improved cardiac function. However, the contribution of cardiac specific TLR4 to the development of hypertension and cardiac remodeling is unknown. Therefore, we hypothesized that cardiomyocyte specific knockdown of TLR4 would have beneficial effects on hypertension, cardiac hypertrophy, and remodeling. To test this hypothesis, cardiomyocyte-specific TLR4 knockdown (cTLR4KO) mice were generated by crossing floxed TLR4 mice with Myh6-Cre mice, and subjected to angiotensin II (Ang II, 1 µg/kg/min or vehicle for 14 days) hypertension model. Blood pressure measurements using radio telemetry revealed no differences in baseline mean arterial pressure between control littermates and cTLR4KO mice (103 ± 2 vs. 105 ± 3 mmHg, p > 0.05). Ang II-induced hypertension (132 ± 2 vs. 151 ± 3 mmHg, p < 0.01) was attenuated and cardiac hypertrophy (heart/body weight; 4.7 vs. 5.8 mg/g, p < 0.01) was prevented in cTLR4KO mice when compared with control mice. In addition, the level of myocardial fibrosis was significantly reduced, and the cardiac function was improved in cTLR4KO mice infused with Ang II. Furthermore, cardiac inflammation, as evidenced by elevated gene expression of TNF, IL-6, and MCP-1 in the left ventricle, was attenuated in cTLR4KO mice infused with Ang II. Together, this data revealed a protective role for cardiomyocyte-specific deletion of TLR4 against Ang II-induced hypertension and cardiac dysfunction through inhibition of proinflammatory cytokines.
Collapse
Affiliation(s)
- Drew Theobald
- Department of Pharmacology and Toxicology, Brody School of Medicine at East Carolina University, Greenville, NC, United States
| | - Anand R. Nair
- Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Srinivas Sriramula
- Department of Pharmacology and Toxicology, Brody School of Medicine at East Carolina University, Greenville, NC, United States
| | - Joseph Francis
- Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
40
|
Recent Developments in the Evaluation and Management of Cardiorenal Syndrome: A Comprehensive Review. Curr Probl Cardiol 2023; 48:101509. [PMID: 36402213 DOI: 10.1016/j.cpcardiol.2022.101509] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 11/11/2022] [Indexed: 11/18/2022]
Abstract
Cardiorenal syndrome (CRS) is an increasingly recognized diagnostic entity associated with high morbidity and mortality among acutely ill heart failure (HF) patients with acute and/ or chronic kidney diseases (CKD). While traditionally viewed as a state of decline in glomerular filtration rate (GFR) due to decreased renal perfusion, mainly due to therapeutic interventions to relieve congestive in HF, recent insights into the underlying pathophysiologic mechanisms of CRS led to a broader definition and further classification of CRS into 5 distinct types. In this comprehensive review, we discuss the classification of CRS, highlighting the underlying common pathogenetic pathways of heart failure and kidney injury, including increased congestion, neurohormonal dysregulation, oxidative stress as well as inflammation, and cytokine storm that are particularly evident in COVID-19 patients with multiorgan failure and also in those with other disorders including sepsis, systemic lupus erythematosus and amyloidosis. In this review we also present the recent advances in the diagnostic strategies of CRS including cardiac and renal biomarkers as well as advanced cardiac and renal imaging techniques that are available to aid in the diagnosis as well as in the prognostication of this disorder. Finally, we discuss the various therapeutic options available to-date, including fluid optimization, hemofiltration, renal replacement therapy as well as the role of SGLT2 inhibitors in light of recent data from RCTs. It is important to note that, CRS population are either excluded or underrepresented, at best, in major RCTs and therefore, therapeutic recommendations are largely extrapolated from HF and CKD clinical trials.
Collapse
|
41
|
Litwin SE, East CA. Assessing clinical and biomarker characteristics to optimize the benefits of sacubitril/valsartan in heart failure. Front Cardiovasc Med 2022; 9:1058998. [PMID: 36620638 PMCID: PMC9815716 DOI: 10.3389/fcvm.2022.1058998] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Of the various medical therapies for heart failure (HF), sacubitril/valsartan is a first-in-class angiotensin receptor-neprilysin inhibitor that combines sacubitril, a pro-drug that is further metabolized to the neprilysin inhibitor sacubitrilat, and the angiotensin II type 1 receptor blocker valsartan. Inhibition of neprilysin and blockade of the angiotensin II type 1 receptor with sacubitril/valsartan increases vasoactive peptide levels, increasing vasodilation, natriuresis, and diuresis. Left ventricular ejection fraction (LVEF) is widely used to classify HF, to assist with clinical decision-making, for patient selection in HF clinical trials, and to optimize the benefits of sacubitril/valsartan in HF. However, as HF is a complex syndrome that occurs on a continuum of overlapping and changing phenotypes, patient classification based solely on LVEF becomes problematic. LVEF measurement can be imprecise, have low reproducibility, and often changes over time. LVEF may not accurately reflect inherent disease heterogeneity and complexity, and the addition of alternate criteria to LVEF may improve phenotyping of HF and help guide treatment choices. Sacubitril/valsartan may work, in part, by mechanisms that are not directly related to the LVEF. For example, this drug may exert antifibrotic and neurohumoral modulatory effects through inhibition or activation of several signaling pathways. In this review, we discuss markers of cardiac remodeling, fibrosis, systemic inflammation; activation of neurohormonal pathways, including the natriuretic system and the sympathetic nervous system; the presence of comorbidities; patient characteristics; hemodynamics; and HF signs and symptoms that may all be used to (1) better understand the mechanisms of action of sacubitril/valsartan and (2) help to identify subsets of patients who might benefit from treatment, regardless of LVEF.
Collapse
Affiliation(s)
- Sheldon E. Litwin
- Division of Cardiology, Medical University of South Carolina, Charleston, SC, United States,Ralph H. Johnson Veterans Affairs Health Network, Charleston, SC, United States,*Correspondence: Sheldon E. Litwin,
| | - Cara A. East
- Baylor Soltero Cardiovascular Research Center, Baylor Scott and White Research Institute, Dallas, TX, United States
| |
Collapse
|
42
|
Matthia EL, Setteducato ML, Elzeneini M, Vernace N, Salerno M, Kramer CM, Keeley EC. Circulating Biomarkers in Hypertrophic Cardiomyopathy. J Am Heart Assoc 2022; 11:e027618. [PMID: 36382968 PMCID: PMC9851432 DOI: 10.1161/jaha.122.027618] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Hypertrophic cardiomyopathy is the most common genetic heart disease. Biomarkers, molecules measurable in the blood, could inform the clinician by aiding in diagnosis, directing treatment, and predicting outcomes. We present an updated review of circulating biomarkers in hypertrophic cardiomyopathy representing key pathologic processes including wall stretch, myocardial necrosis, fibrosis, inflammation, hypertrophy, and endothelial dysfunction, in addition to their clinical significance.
Collapse
Affiliation(s)
| | | | | | | | - Michael Salerno
- Department of Medicine, Cardiovascular DivisionUniversity of VirginiaCharlottesvilleVA
| | - Christopher M. Kramer
- Department of Medicine, Cardiovascular DivisionUniversity of VirginiaCharlottesvilleVA
| | - Ellen C. Keeley
- Department of MedicineUniversity of FloridaGainesvilleFL,Division of Cardiovascular MedicineUniversity of FloridaGainesvilleFL
| |
Collapse
|
43
|
Parfianowicz D, Shah S, Nguyen C, Maitz TN, Hajra A, Goel A, Sreenivasan J, Aronow WS, Vyas A, Gupta R. Finerenone: A New Era for Mineralocorticoid Receptor Antagonism and Cardiorenal Protection. Curr Probl Cardiol 2022; 47:101386. [PMID: 36057315 DOI: 10.1016/j.cpcardiol.2022.101386] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/03/2022]
Abstract
The renin-angiotensin-aldosterone system is a neurohormonal system responsible for maintaining homeostasis of fluid regulation, sodium balance, and blood pressure. The complexity of this pathway enables it to be a common target for blood pressure and volume-regulating medications. The mineralocorticoid receptor is one of these targets, and is found not only in the kidney, but also tissues making up the heart, blood vessels, and adipose. Mineralocorticoid receptor antagonists have been shown to slow progression of chronic kidney disease, treat refractory hypertension and primary aldosteronism, and improve morbidity and mortality in management of heart failure with reduced ejection fraction. The more well-studied medications were derived from steroid-based compounds, and thus come with a distinct side-effect profile. To avoid these adverse effects, developing a mineralocorticoid receptor antagonist (MRA) from a non-steroidal base compound has gained much interest. This review will focus on the novel non-steroidal MRA, Finerenone, to describe its unique mechanism of action while summarizing the available clinical trials supporting its use in patients with various etiologies of cardiorenal disease.
Collapse
Affiliation(s)
| | - Swara Shah
- Department of Medicine, Lehigh Valley Health Network, Allentown, PA
| | - Catherine Nguyen
- Department of Medicine, Lehigh Valley Health Network, Allentown, PA
| | - Theresa N Maitz
- Department of Medicine, Lehigh Valley Health Network, Allentown, PA
| | - Adrija Hajra
- Department of Medicine, Jacobi Medical Center, Bronx, NY
| | - Akshay Goel
- Department of Cardiology, Westchester Medical Center and New York Medical College, Valhalla, NY
| | - Jayakumar Sreenivasan
- Department of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT
| | - Wilbert S Aronow
- Department of Cardiology, Westchester Medical Center and New York Medical College, Valhalla, NY
| | - Apurva Vyas
- Department of Cardiology, Lehigh Valley Heart Institute, Lehigh Valley Health Network, Allentown, PA
| | - Rahul Gupta
- Department of Cardiology, Lehigh Valley Heart Institute, Lehigh Valley Health Network, Allentown, PA.
| |
Collapse
|
44
|
Ranjbar T, Oza PP, Kashfi K. The Renin-Angiotensin-Aldosterone System, Nitric Oxide, and Hydrogen Sulfide at the Crossroads of Hypertension and COVID-19: Racial Disparities and Outcomes. Int J Mol Sci 2022; 23:ijms232213895. [PMID: 36430371 PMCID: PMC9699619 DOI: 10.3390/ijms232213895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Coronavirus disease 2019 is caused by SARS-CoV-2 and is more severe in the elderly, racial minorities, and those with comorbidities such as hypertension and diabetes. These pathologies are often controlled with medications involving the renin-angiotensin-aldosterone system (RAAS). RAAS is an endocrine system involved in maintaining blood pressure and blood volume through components of the system. SARS-CoV-2 enters the cells through ACE2, a membrane-bound protein related to RAAS. Therefore, the use of RAAS inhibitors could worsen the severity of COVID-19's symptoms, especially amongst those with pre-existing comorbidities. Although a vaccine is currently available to prevent and reduce the symptom severity of COVID-19, other options, such as nitric oxide and hydrogen sulfide, may also have utility to prevent and treat this virus.
Collapse
Affiliation(s)
- Tara Ranjbar
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA
| | - Palak P. Oza
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA
- Graduate Program in Biology, City University of New York Graduate Center, New York, NY 10016, USA
- Correspondence:
| |
Collapse
|
45
|
Kim SM, Kang M, Kang E, Kim JH, Kim Y, Ryu H, Han SS, Lee H, Kim YC, Oh KH. Associations among body composition parameters and quality of life in peritoneal dialysis patients. Sci Rep 2022; 12:19192. [PMID: 36357419 PMCID: PMC9649675 DOI: 10.1038/s41598-022-19715-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 09/02/2022] [Indexed: 11/11/2022] Open
Abstract
Health-related quality of life (HRQOL) is an important issue among patients undergoing dialysis treatment. Peritoneal dialysis (PD) is associated with a number of adverse body composition changes. However, whether body composition is associated with HRQOL is uncertain. The purpose of this study was to analyze the effects of body composition on HRQOL in PD patients. We performed a cross-sectional observational study on the association between body composition and HRQOL in PD patients at a single center. Body composition was determined by multifrequency bioimpedance spectroscopy. HRQOL is summarized to three composite scores: kidney disease component summary (KDCS), physical component summary (PCS), and mental component summary (MCS). The relationships between HRQOL and the hydration index, lean tissue index (LTI), and fat tissue index (FTI) were analyzed by regression analysis. One hundred and ninety-seven PD patients were included in the present study. Patients with severe fluid overload showed a lower PCS. The hydration index and FTI showed statistically significant negative associations with PCS. In subgroup analysis, the associations between the hydration index and PCS remained robust after stratifying according to sex, age, and residual urine. Our results indicated that both the hydration index and FTI were negatively associated with HRQOL, especially PCS.
Collapse
Affiliation(s)
- Seon-Mi Kim
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daehakro, Jongno-Gu, Seoul, 03080 Republic of Korea
| | - Minjung Kang
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daehakro, Jongno-Gu, Seoul, 03080 Republic of Korea
| | - Eunjeong Kang
- grid.255649.90000 0001 2171 7754Department of Internal Medicine, Ewha Womans University Seoul Hospital, Seoul, Republic of Korea
| | - Ji Hye Kim
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daehakro, Jongno-Gu, Seoul, 03080 Republic of Korea
| | - Yunmi Kim
- grid.411625.50000 0004 0647 1102Department of Internal Medicine, Inje University Busan Paik Hospital, Busan, Republic of Korea
| | - Hyunjin Ryu
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daehakro, Jongno-Gu, Seoul, 03080 Republic of Korea
| | - Seung Seok Han
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daehakro, Jongno-Gu, Seoul, 03080 Republic of Korea
| | - Hajeong Lee
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daehakro, Jongno-Gu, Seoul, 03080 Republic of Korea
| | - Yong Chul Kim
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daehakro, Jongno-Gu, Seoul, 03080 Republic of Korea
| | - Kook-Hwan Oh
- grid.412484.f0000 0001 0302 820XDepartment of Internal Medicine, Seoul National University Hospital, 101 Daehakro, Jongno-Gu, Seoul, 03080 Republic of Korea
| |
Collapse
|
46
|
Salim H, Jones AM. Angiotensin II receptor blockers (ARBs) and manufacturing contamination: A retrospective National Register Study into suspected associated adverse drug reactions. Br J Clin Pharmacol 2022; 88:4812-4827. [PMID: 35585835 PMCID: PMC9796460 DOI: 10.1111/bcp.15411] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 01/01/2023] Open
Abstract
AIMS The aim of this study was to determine if any suspected adverse drug reactions (ADRs) observed with the use of angiotensin II receptor blockers (ARBs) could be linked to either (a) their unique respective physicochemical and pharmacological profiles and (b) the recently disclosed suspected carcinogenic manufacturing contaminants found in certain sartan drug class batches. METHODS The pharmacology profiles of ARBs were data-mined from the Chemical Database of bioactive molecules with drug-like properties, European Molecular Biology Laboratory (ChEMBL). Suspected ADR data (from 01/2016-10/2022, inclusive) and prescribing rates of ARBs over a 5-year prescribing window (from 09/2016 to 08/2021, inclusive) were obtained via analysis of the United Kingdom Medicines and Healthcare products Regulatory Authority (MHRA) Yellow Card drug analysis profile and Open prescribing databases, respectively. RESULTS The overall suspected ADRs and fatalities per 100 000 prescriptions identified across the ARBs studied were found to be different between the sartan drug class members (chi-squared test, P < .05). There is a greater relative rate of reports for valsartan across all investigated organ classes of ADRs, than other ARBs, despite valsartan's more limited pharmacological profile and similar physicochemical properties to other sartans. The disparity in ADR reporting rates with valsartan vs other ARBs could be due to the dissimilarity in formulation excipients, patient factors and publicity surrounding batch contaminations, amongst others. Cancer-related ADRs and fatalities per 100 000 prescriptions identified across the ARBs studied are not statistically significant (chi-squared test, P > .05) based on the datasets used over the 5-year period. CONCLUSION No connection between ARB pharmacology and their suspected ADRs could be found. No conclusion between sartan batch contaminations and increased suspected cancer-related ADRs was found.
Collapse
Affiliation(s)
- Hamisha Salim
- Medicines Safety Research Group (MSRG), School of PharmacyUniversity of BirminghamBirminghamUnited Kingdom
| | - Alan M. Jones
- Medicines Safety Research Group (MSRG), School of PharmacyUniversity of BirminghamBirminghamUnited Kingdom
| |
Collapse
|
47
|
Tadic M, Cuspidi C. Right Ventricle in Arterial Hypertension: Did We Forget Something? J Clin Med 2022; 11:6257. [PMID: 36362485 PMCID: PMC9655282 DOI: 10.3390/jcm11216257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 10/18/2022] [Accepted: 10/21/2022] [Indexed: 08/30/2023] Open
Abstract
Right ventricular remodeling has been neglected in patients with arterial hypertension as all studies have concentrated on the left ventricle and left atrial-ventricular and ventricular-arterial coupling. The development of novel imaging techniques has revealed significant impairment in the RV structure, systolic and diastolic function, and, afterwards, RV longitudinal mechanics. However, these changes are subclinical and can be detected only after comprehensive imaging analysis. The latest findings confirm the importance of RV hypertrophy, systolic, and diastolic dysfunction in the prediction of cardiovascular adverse events in the hypertensive population, representing an important clinical implication of these parameters. In clinical practice, 2D echocardiography is widely used for the evaluation of RV remodeling. However, existing techniques are largely underused and limited to a few basic parameters (RV thickness and TAPSE), which are not nearly enough for a detailed assessment of RV remodeling. In addition, 3D echocardiography provides the possibility of accurate evaluation of RV volumes and ejection fraction, which are comparable with results obtained by cardiac magnetic resonance (CMR)-a gold standard for the evaluation of the RV. The use of 3D echocardiography is limited due to its low availability, the lack of adequate software necessary for the calculation of results, and the necessity for a higher level of expertise. CMR provides all information required for a detailed assessment of RV structural, functional, and mechanical remodeling, and it is considered the reference method for this type of evaluation. Furthermore, it is the only technique that may provide tissue characterization and evaluation of the interstitial space, which is essential for hypertensive heart disease. The aim of this review is to provide the current level of evidence regarding RV remodeling in patients with arterial hypertension evaluated with different imaging techniques and various parameters from each method.
Collapse
Affiliation(s)
- Marijana Tadic
- Klinik für Innere Medizin II, Universitätsklinikum Ulm, Albert-Einstein Allee 23, 89081 Ulm, Germany
| | - Cesare Cuspidi
- Department of Medicine and Surgery, University of Milano-Bicocca, 20126 Milano, Italy
| |
Collapse
|
48
|
Ma J, Chen X. Advances in pathogenesis and treatment of essential hypertension. Front Cardiovasc Med 2022; 9:1003852. [PMID: 36312252 PMCID: PMC9616110 DOI: 10.3389/fcvm.2022.1003852] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/26/2022] [Indexed: 11/23/2022] Open
Abstract
Hypertension is a significant risk factor for cardiovascular and cerebrovascular diseases and the leading cause of premature death worldwide. However, the pathogenesis of the hypertension, especially essential hypertension, is complex and requires in-depth studies. Recently, new findings about essential hypertension have emerged, and these may provide important theoretical bases and therapeutic tools to break through the existing bottleneck of essential hypertension. In this review, we demonstrated important advances in the different pathogenesis areas of essential hypertension, and highlighted new treatments proposed in these areas, hoping to provide insight for the prevention and treatment of the essential hypertension.
Collapse
|
49
|
Iovanovici DC, Bungau SG, Vesa CM, Moisi M, Babes EE, Tit DM, Horvath T, Behl T, Rus M. Reviewing the Modern Therapeutical Options and the Outcomes of Sacubitril/Valsartan in Heart Failure. Int J Mol Sci 2022; 23:11336. [PMID: 36232632 PMCID: PMC9570001 DOI: 10.3390/ijms231911336] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/22/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Sacubitril/valsartan (S/V) is a pharmaceutical strategy that increases natriuretic peptide levels by inhibiting neprilysin and regulating the renin-angiotensin-aldosterone pathway, blocking AT1 receptors. The data for this innovative medication are mainly based on the PARADIGM-HF study, which included heart failure with reduced ejection fraction (HFrEF)-diagnosed patients and indicated a major improvement in morbidity and mortality when S/V is administrated compared to enalapril. A large part of the observed favorable results is related to significant reverse cardiac remodeling confirmed in two prospective trials, PROVE-HF and EVALUATE-HF. Furthermore, according to a subgroup analysis from the PARAGON-HF research, S/V shows benefits in HFrEF and in many subjects having preserved ejection fraction (HFpEF), which indicated a decrease in HF hospitalizations among those with a left ventricular ejection fraction (LVEF) < 57%. This review examines the proven benefits of S/V and highlights continuing research in treating individuals with varied HF characteristics. The article analyses published data regarding both the safeness and efficacy of S/V in patients with HF, including decreases in mortality and hospitalization, increased quality of life, and reversible heart remodeling. These benefits led to the HF guidelines recommendations updating and inclusion of S/V combinations a key component of HFrEF treatment.
Collapse
Affiliation(s)
- Diana-Carina Iovanovici
- Doctoral School of Biomedical Sciences, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Simona Gabriela Bungau
- Doctoral School of Biomedical Sciences, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
| | - Cosmin Mihai Vesa
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Madalina Moisi
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Elena Emilia Babes
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Delia Mirela Tit
- Doctoral School of Biomedical Sciences, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
| | - Tunde Horvath
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
| | - Tapan Behl
- School of Health Sciences &Technology, University of Petroleum and Energy Studies, Bidholi, Dehradun 248007, India
| | - Marius Rus
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| |
Collapse
|
50
|
Teer E, Dominick L, Mukonowenzou NC, Essop MF. HIV-Related Myocardial Fibrosis: Inflammatory Hypothesis and Crucial Role of Immune Cells Dysregulation. Cells 2022; 11:cells11182825. [PMID: 36139400 PMCID: PMC9496784 DOI: 10.3390/cells11182825] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Although the underlying mechanisms driving human immunodeficiency virus (HIV)-mediated cardiovascular diseases (CVD) onset and progression remain unclear, the role of chronic immune activation as a significant mediator is increasingly being highlighted. Chronic inflammation is a characteristic feature of CVD and considered a contributor to diastolic dysfunction, heart failure, and sudden cardiac death. This can trigger downstream effects that result in the increased release of pro-coagulant, pro-fibrotic, and pro-inflammatory cytokines. Subsequently, this can lead to an enhanced thrombotic state (by platelet activation), endothelial dysfunction, and myocardial fibrosis. Of note, recent studies have revealed that myocardial fibrosis is emerging as a mediator of HIV-related CVD. Together, such factors can eventually result in systolic and diastolic dysfunction, and an increased risk for CVD. In light of this, the current review article will focus on (a) the contributions of a chronic inflammatory state and persistent immune activation, and (b) the role of immune cells (mainly platelets) and cardiac fibrosis in terms of HIV-related CVD onset/progression. It is our opinion that such a focus may lead to the development of promising therapeutic targets for the treatment and management of CVD in HIV-positive patients.
Collapse
Affiliation(s)
- Eman Teer
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Leanne Dominick
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Nyasha C. Mukonowenzou
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch 7600, South Africa
| | - M. Faadiel Essop
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 8000, South Africa
- Correspondence: ; Tel.: +27-21-938-9388
| |
Collapse
|